Résumé

La congélation du sperme et des embryons est une biotechnologie importante pour la conservation et propagation de la génétique. L’objectif de cet article est discuter les techniques de congélation établies ainsi que les acquisitions récentes dans ce domaine chez les ovins, caprins et petits ruminant sauvages. L’utilisation du sperme congelé en production est plus développée chez les caprins que chez les ovins. Ceci est principalement dû au besoin de l’utilisation de l’insémination par laparoscopie chez les brebis. La plus grande différence entre les caprins et les ovins est le comportement du plasma séminal vis as vis des protéines du jaune d’œuf. Récemment, les efforts de recherches se sont penchés principalement sur le développement de nouvelles approches pour l’amélioration des dilueurs de congélation. Les axes les plus importants étant le remplacement des protéines animales (jaune d’œuf et lait) par les lécithines végétales, l’utilisation de cryoprotcteurs autres que le glycérol, et la réduction des effets de l’oxydation des lipides. L’introduction de méthodes d’analyses de la morphologie et fonction des spermatozoïdes, telles que les techniques de fluorescence, les tests hypopsmotiques et l’évaluation numérique de la mobilité, permet une évaluation plus rigoureuse des effets de ces facteurs sur la congélabilité du sperme La conservation des embryons des petits ruminants par vitrification se développe de plus en plus et pourrait remplacer les techniques lentes, plus laborieuses et coûteuses. Chez les ovins, les morulas et blastocystes semblent être plus résistants à la congélation. Par contre chez les caprins, les blastocystes en expansion et éclos sont plus résistants à la congélation que les morulas et jeunes blastocyste. Les taux de gestation obtenus après transfert d’embryons congelés sont très intéressants surtout quand la gestion des receveuses est adéquate et les techniques de transfert sont maîtrisées.


Mots-clés: Congélation, sperme, embryons, insémination, fertilité, dilueurs.

INTRODUCTION

Cryopreservation of sperm and embryos of small ruminants is an important reproductive biotechnology for the preservation of rare or local breeds and the propagation of genetic material from imported high performing individual animals. Another advantage of these technologies is the introduction in a flock or herd of new genetics without breaching biosecurity measures if donor animals are submitted to rigorous veterinary inspection and disease testing. The objective of this paper is to review the general principles of sperm and embryo cryopreservation and discuss new research aimed at improvement of these technologies in small ruminants.

SEMEN CRYOPRESERVATION

Although sperm preservation is considered common practice in both sheep and goat, new improvements have been introduced mostly to simplify the technique for field use. Improvement in post-thaw quality aims to bypass laparoscopic insemination particularly in sheep. Advances in cryopreservation of small ruminant sperm include development of new extenders to eliminate animal products such as milk or egg yolk. Differences in sperm cryopreservation exist between sheep and goat. Goat sperm is known for the toxic reaction to high levels of egg yolk in the extender.

Cryopreservation of ejaculated sperm

One of the primary conditions for successful cryopreservation of sperm is the initial quality of the ejaculate. Rams and bucks that are candidates for semen cryopreservation and use in artificial insemination programs should undergo a thorough breeding soundness examination (BSE) and disease testing prior to enrollment. Cryopreservation causes biological and functional changes that can affect sperm viability and fertility. These changes occur primarily at the membrane levels and are due to ice crystal formation and changes in membrane permeability. A significant proportion (20 to 30%) of cryopreserved spermatozoa lose their fertilizing ability after thawing. Therefore, only ejaculates of excellent quality (>90% motility and >70% normal morphology) should be used for cryopreservation (Dorado et al., 2010). Principles for BSE in rams and bucks are described in detail elsewhere in the present publication (Tibary et al., 2018). It is important to take into account seasonal effects on initial quality of the ejaculate and seminal plasma proteins and their interaction with membrane stability and ability to withstand freezing and thawing (Goularte et al., 2014; Sobrinho et al., 2014; Wang et al., 2015). Melatonin treatment of bucks improved fresh semen quality but not cryopreservation (Gallego-Calvo et al., 2015).

Cryopreservation of small ruminant sperm has been practiced for decades. Several extenders have been developed and are either egg yolk (EY) or skim milk based (Table 1) (Cseh et al., 2012; Kucuk et al., 2014; Emamverdi et al., 2015). The main buffers used are citric acid and Tris. Zwitterionic buffer have also been used successfully for small ruminant semen preservation (Tuli and Holtz, 1992). Egg yolk low-density lipoproteins (lecithin and phosohatidylcholines) protect the sperm membrane during the freezing and thawing processes. A major difference between ram and buck semen is their behavior toward egg yolk, which tends to coagulate in presence of seminal plasma from bucks. This effect has been attributed to the presence of a specific lipase belonging to the pancreatic lipase-related protein 2 family that is secreted by the bulbourethral glands (egg yolk coagulating enzyme or BUSgp60) (Cseh et al., 2012). Goat ejaculates are washed by centrifugation (700 x g for 15 minutes) in PBS or extender without egg yolk prior to dilution with egg yolk containing extenders (Batista et al., 2014). Alternately, semen can be diluted with extenders containing very low level of egg yolk (2 to 3%) (Shamsuddin et al., 2000; Bispo et al., 2011; Anand and Yadav, 2016). Other researchers have used up to 12% egg yolk without washing (Cabrera et al., 2005).

Following dilution, semen is slowly cooled to 5°C for a minimum of 1.5 to 2 hours, loaded in straws and frozen in liquid nitrogen vapors (Cseh et al., 2012). The final dilution of semen prior to cryopreservation is generally between 100 and 200 million sperm per mL. Increasing the concentration above 400 million per mL results in a decrease in post-thaw quality (Alvarez et al., 2012a).

There are few studies on the optimal freezing rate for ram and buck sperm. The optimal freezing rate should avoid intracellular ice formation, cell shrinkage and long exposure to high osmotic and ionic strength. Freezing rates commonly used for small ruminant sperm cryopreservation range from -10 to -100°C/min. In a recent study on ram semen, the best freezing rate was determined to be -40°C/minute when semen was diluted in Tris-glucose EY and cooled at 4°C for 2 hours in 0.25 mL straws (Fang et al., 2016). Although protocols for cryopreservation of ram and goat spermatozoa have been successfully used for several decades, introduction of technologies such as sex-sorted semen requires improvement of cryopreservation (Bathgate et al., 2013; Quan et al., 2015). Factors that have been studied recently include the type and method of addition of cryoprotectant (Silva et al., 2012b; Pelufo et al., 2015), cooling rate and equilibration time (Ahmad et al., 2015) and individual male variability. Cryoprotectants are generally added in a single step or in two steps (after cooling). Cooling rates and equilibration times from 2 to 8 hours have been tested (Ahmad et al., 2015). Equilibration time of 2 hours seems to be sufficient for most protocols (da Silva et al., 2014). Equilibrated semen is placed in 0.25 or 0.5 mL straws and cooled at a rate of 3°C/min from 5°C to -8°C and at 25°C /minute from -8 to -120°C.

Individual male variation exists in the ability of sperm to withstand freezing. This variation may be linked to differences in protein content of seminal plasma and membrane stability (Moura et al., 2010; Romon et al., 2013). These individual variations are illustrated by difference in changes in sperm head volume during the freezing process (Romon et al., 2013). Genetic variation may also affect cryopreservation of semen. Recently, the heat shock protein 70 (HSP70) gene has been suspected to have an effect on semen quality and possibly freezing ability in Boer goats under tropical conditions (Nikbin et al., 2014).

Table 1: Extenders used for cryopreservation of small ruminant sperm

New developments in sperm cryobiology

Replacement of egg yolk

As stated above, addition of egg yolk to extenders provides cryoprotection to the sperm through its low-density lipoprotein. Egg yolk from different species has been tested resulting in some differences (Kulaksiz et al., 2010). Egg yolk, as a membrane protective agent in semen extenders, has become progressively undesirable because of its antigenic proprieties (Ustuner et al., 2014) and the reluctance of some countries to import semen frozen with egg yolk containing extenders for biosecurity reasons. This led to its replacement by lecithins from vegetal origin such as soybean (Del Valle et al., 2013; Emamverdi et al., 2013; Vidal et al., 2013; Salmani et al., 2014; Bohlool et al., 2015; Chelucci et al., 2015; Sharafi et al., 2015; Masoudi et al., 2016a; Toker et al., 2016). The combination of soy milk and glycerol has been used successfully for cryopreservation of ram sperm (Jerez et al., 2016). The use of soy extender (Bioxcell®) for goat semen has been shown to remove the need for washing of ejaculates prior to dilution and freezing (Sariozkan et al., 2010; Roof et al., 2012).

Addition of antioxidants

Sperm membrane has a high concentration of polyunsaturated fatty acids and is extremely sensitive to oxidative stress during cryopreservation (Del Olmo et al., 2015; Amidi et al., 2016; Toker et al., 2016). Research has focused, in the last decade, on improvement in cryopreservation of ram and buck semen through addition of various antioxidants to extenders with variable results. Enzymatic antioxidants such as glutathione (GSH), superoxide dismutase (SOD) and catalase (Silva et al., 2011; Santiani et al., 2014) have been used in various concentrations to improve post-thaw sperm quality. In rams, addition of SOD, GSH and MNTBAP (a superoxide-dismutase mimetic) seems to reduce acrosomal damage (see review by Budai et al., 2014). Addition of Trolox and catalase to tris-egg yolk extender significantly reduced lipid-peroxidation in ram semen (Maia et al., 2010; Sicherle et al., 2011; Silva et al., 2013; Camara et al., 2016). Trolox was also beneficial for preservation of sperm of plasma membrane and mitochondrial sheath integrity of goat sperm frozen in a skim milk based extender (Soares et al., 2015).

Other antioxidants (zinc, tocopherol, albumin, glutathione, taurine, hypotaurine, carnitine, carotenoids, cysteine, urate and prostasomes) (Mata-Campuzano et al., 2011; Mata-Campuzano et al., 2012; Budai et al., 2014; Mata-Campuzano et al., 2015) have also been tested in small ruminant sperm cryopreservation with variable results (Budai et al., 2014). Oxidized gluthatione (Camara et al., 2011; Razliqi et al., 2015; Hazarika et al., 2016), cysteamine (Najafi et al., 2014a; Alcay et al., 2016), methionine (Tuncer et al., 2010; Alcay et al., 2016), catalase, cysteamine (Bucak et al., 2007), Butylated hydroxytoluene (BHT) (Naijian et al., 2013; Alcay et al., 2016), ergothioneine (Coyan et al., 2011; Najafi et al., 2014a), hypotaurine (Bucak et al., 2013), taurine (Bucak et al., 2007) and trehalose (Bucak et al., 2007; Tuncer et al., 2010; Najafi et al., 2013; Tuncer et al., 2013; Bohlool et al., 2015) have all been used as antioxidants and reactive oxygen species scavengers. Resveratrol and quercetin have also been shown to improve post-thaw viability and mitochondrial membrane potential (Silva et al., 2012a). Dithiothreitol, a protamine disulfide bond reducing agent, has been shown to have a positive effect on post-thaw motility of ram semen (Baspinar et al., 2011). All these approaches proved to be beneficial in increasing viability, post-thaw motility and acrosome integrity. However, the true effect of some these antioxidants on sperm viability and post-thaw fertility remains controversial. In one study, addition of cysteamine to electroejaculated ram semen was detrimental to cryosurvival while addition of iodixanol was beneficial (Cirit et al., 2013). Glutathione addition had no beneficial effect on buck semen extended in soybean lecithin containing extender (Salmani et al., 2013). In one study on ram semen, no advantage was found with addition of gluthatione, superoxide dismutase or catalase (Camara et al., 2011). Supplementation of extender with vitamin C has been shown to improve post-thaw buck sperm quality (Gangwar et al., 2015). Other studies have shown little benefit or negative effect of some antioxidants on post thaw sperm quality (Yildiz et al., 2015; Zhandi and Sharafi, 2015). The effects of antioxidants on post-thaw sperm quality merits however further studies to elucidate the interaction between specific extender and concentration of antioxidants (Mata-Campuzano et al., 2015; Zhandi and Sharafi, 2015).

Another approach to reduce reactive oxygen species due lipid peroxidation in semen is through nutrition. Addition of fish oil to ram diet has been shown to improve sperm quality and in vitro fertilization results (Behzad et al., 2014; Esmaeili et al., 2014; Masoudi et al., 2016b). Semen quality was also shown to be improved in rams supplemented orally with vitamins E and C (Memon et al., 2013; Cofre-Narbona et al., 2016). Dietary supplementation with n-3 polyunsaturated fatty acid of rams had limited effects on the quality of liquid stored semen (Towhidi et al., 2013; Fair et al., 2014).

Alternative cryoprotectants

Glycerol is the most commonly used cryoprotectant for sperm. However, exposure to high levels of glycerol is toxic and can be detrimental to sperm function and viability in utero. Therefore, research efforts have focused on alternative cryoprotectants. In goats, dimethylsulfoxide (DMSO) and ethylene glycol (EG) did not present any added advantage in sperm cryopreservation (Buyukleblebici et al., 2014). In rams, EG at concentrations of 3 or 5% was beneficial but not as good as glycerol (Silva et al., 2012b). DMSO was ineffective in ram semen preservation (Bezerra et al., 2011; Moustacas et al., 2011). In rams, propanediol, sucrose and trehalose did not perform as well as glycerol (Nur et al., 2010). However, trehalose supplementation to extenders improved cryopreservation of Boer semen in Tris-citric acid extender (Naing et al., 2010). Amino acids (L-glutamine and L-proline) have been used successfully to improve cryoprotection and reduce lipid peroxidation (Farshad and Hosseini, 2013; Mehr and Noori, 2013; Sangeeta et al., 2015). Addition of cholesterol-loaded cyclodextrin may allow reduction of the concentration of glycerol to 3% (Moce et al., 2010; Awad, 2011; Konyali et al., 2013; Moce et al., 2014; Motamedi-Mojdehi et al., 2014).

Counteracting capacitation-like changes

One of the major effects of cryopreservation on sperm is induction of capacitation-like changes (i.e. cryocapacitation). Several studies have shown that semen collected by electroejaculation (i.e. higher seminal plasma content) has better cryotolerance than semen collected by artificial vagina (Jimenez-Rabadan et al., 2013; Ledesma et al., 2015). However other studies did not show any difference between the two methods of collection while others have shown that AV collected semen from bucks has better post-thaw motility (Jimenez-Rabadan et al., 2012). Addition of seminal plasma to cryopreserved sperm was found to improve fertility in ewes inseminated transcervically (Maxwell et al., 1999). Some proteins that are provided by seminal plasma such as spermadhesins and SPINK3 (Serine Protease Inhibitor Kazal Type 3 or caltrin) interfere with the capacitation pathways by either stabilizing the membrane or inhibiting calcium transport (Maxwell et al., 2007; Zalazar et al., 2016). SPINK3 was found in high concentration in seminal plasma from rams with good freezing ability spermatozoa (Rickard et al., 2016). This protein is known to modulate calcium increase and to downregulate the signal transduction pathway leading to capacitation. Addition of heterologous recombinant SPINK3 to the freezing extender or after thawing was shown to improve post-thaw progressive motility and reduce acrosomal loss (Zalazar et al., 2016). Post-thaw addition of seminal plasma was also shown to reverse cryopreservation damage (Bernardini et al., 2011). There is a seasonal variation in the protective effect of seminal plasma on frozen-thawed ram spermatozoa (Leahy et al., 2010). Despite these studies, the effect of addition of seminal plasma to cryopreserved sperm remains controversial (Prado et al., 2013; Rovegno et al., 2013).

Other additives

Fruits juices from pineapple and orange (Daramola et al., 2016b), coconut water (Daramola et al., 2016a), honey (Jerez-Ebensperger et al., 2015) as well as herbal antioxidants (Mascaro et al., 2013; Zanganeh et al., 2013; Baghshahi et al., 2014) have been used with various degrees of success.

Cryopreservation of semen from other small ruminants

Commercial extenders developed for the cryopreservation of ram and buck semen have been used successfully for the cryopreservation of semen from farmed cervids (i.e. elk, white-tailed deer) and wild small ruminants (i.e. Bighorn sheep, roe deer, mouflon...). Semen is generally collected by electroejaculation with or without ultrasound guided rectal massage under general anesthesia. In farmed white tail deer, soybean (Andromed®) and liposome-based (Optixcell®) extenders were found to be superior to Tris-based extenders (Biladyl® or Triladyl®) (Stewart et al., 2016). Electroejaculated semen from captive roe deer (Capreolus capreolus) showed better post-thaw motility and acrosome integrity when from frozen in Berliner Cryomedium® (TES-Tris-fructose-lactose-EY) compared to Tris-citric acid-glucose or TES-Tris-glucose (Prieto-Pablos et al., 2016). Similar extenders have been used successfully for the cryopreservation of semen from endangered gazelles (Gazella cuvieri, G. dama mhorr, and G. dorcas neglecta) (Garde et al., 2003; Garde et al., 2008), ibex (Capra pyrenaica) (Santiago-Moreno et al., 2009b; Coloma et al., 2010a; Coloma et al., 2010b; Coloma et al., 2011; Pradiee et al., 2015), chamois (Rupicapra pyrenaica), mouflon (Ovis musimon) and aoudad (Ammotragus lervia sahariensis) (Santiago-Moreno et al., 2013; Pradiee et al., 2016). In our laboratory, electroejaculated semen from Bighorn sheep (Ovis canadensis) has been successfully cryopreserved in ram semen extenders and used for artificial insemination (Rodriguez et al., 2009; Subramaniam et al., 2014).

Cryopreservation of epididymal sperm

Post-mortem cryopreservation of cauda epididymis sperm and its use for artificial insemination is an important area of research. Optimization of the technique allows salvage of genetics from terminal animals or from rare domestic and wild small ruminants (Vasquez et al., 2013). Epididymal sperm can be harvested from severely injured valuable males or wildlife and cryopreserved. Testes can be cooled to 5°C and stored for up to 48 hours before processing (Turri et al., 2014). This technique has been used successfully in a variety of species including the ibex (Santiago-Moreno et al., 2006a; Santiago-Moreno et al., 2006b; Santiago-Moreno et al., 2007; Santiago-Moreno et al., 2008; Santiago-Moreno et al., 2009a; Fernández-Santos et al., 2011; Pradiee et al., 2014; López-Saucedo et al., 2015), Iberian red deer, roe deer, catabrian chamois (Martinez-Pastor et al., 2005; Martínez et al., 2008), and Bighorn sheep (Rodriguez et al., 2010; Hansen et al., 2012; Campbell et al., 2014) (Figure 1).

Advances in method of evaluation of cryopreserved sperm and relationships with fertility

Method to evaluate post-thaw semen quality has been a subject of research for several decades. Post-thaw progressive motility is often used as the primary criterion. Velocity and linearity determined by computer assisted sperm analysis (CASA) seems to be a more reliable indicator of quality and correlates with fertility (Furstoss et al., 2010; Del Olmo et al., 2013). In addition to motility, evaluation of membrane integrity, viability and functional tests have become more and more commonly used to objectively evaluate sperm quality. Viability or membrane integrity can be determined with eosin-nigrosin staining (Rekha et al., 2016). However, fluorescence techniques such as SYBR-14 and prodium iodide offer a more repeatable measure. SYBR-14 is used as a membrane-permeable DNA intercalating agent with maximum emission of 516 nm (green) and readily stains all nuclei. Propidium iodide is used as an intercalating agent with maximum emission of 617 nm (red), but it only stains the nucleus if the sperm plasma membrane is damaged which makes it a useful counter stain for dead cells (Yaniz et al., 2013; Stewart et al., 2016) (Figure 2). Membrane integrity can also be evaluated with carboxyflurescien diacetate and propidium iodide (Soares et al., 2015).

Membrane integrity can also be assessed by the hypo-osmotic swelling test (HOST). The HOST solution is prepared using a mixture of fructose (9 g/L) and sodium citrate (4.9 g/L) (Alcay et al., 2016; Quan et al., 2016; Rekha et al., 2016) or a 100 mOsm solution of sucrose. Semen (20 µl) is added to the solution (200 µl) and incubated for 30 to 60 minutes at 37ºC. Spermatozoa (200) are evaluated under the microscope (400×). Sperm with swollen membranes (curled tails) are considered to have an intact membrane (Figure 3).

Figure 2: SBYR-14 and propidium iodide viability staining: Red sperm (stained with PI) have damaged plasma membrane, Green sperm (SYBR-14 stained are viable).

Acrosome status can be assessed with Chlortetracyclin staining (Najafi et al., 2014b) or by a combination of Fluorescein isothiocyanate-conjugate peanut (Pisum sativum) agglutinin (FITC-PNA) and PI. This stain has a maximum emission of 521 nm (green) and targets the inner leaflet of the outer acrosomal membrane identifying damages acrosomes. PI is used as a counterstain to allow for simultaneous sperm viability and condition of acrosome (Soares et al., 2015; Alcay et al., 2016; Stewart et al., 2016) (Figure 4).

Figure 3: Hypoosmotic swelling test: sperm with damaged membranes do not swell (straight), normal sperm shows various coiling of the tail and mid-piece due to swelling.

Figure 4: FITC-PNA damaged acrosome (green fluorescence).

Sperm mitochondrial status can be assessed using mitochondrial stain rhodamine l23 or 5,5′,6,6′tetrachloro-1,1′,3,3′-tetraethylbenzimidazolyl-carbocyanine iodide (JC-1). For staining, 300 μL of semen is mixed with 2.5 μL of 1.53 mM solution of JC-1 in DMSO and 2.5 μL of 0.2 % solution of PI in distilled water. The sample is gently mixed and incubated at 37°C in dark for 20 minutes. JC-1 aggregates inside the functional mitochondria. Highly functional mitochondria are more penetrable by the stain and glow yellow-orange compared to green fluorescing weak functioning mitochondria (Figure 5).

Figure 5: Mitochondrial activity assessed with JC-1. Sperm with midpiece yellow-orange fluorescence have highly functional mitochondria. Green fluorescence denotes weak activity

Another sperm quality test that has gained a lot of interest is sperm chromatin structure assay (SCSA) using acridine orange stained sample analyzed by flow cytometry under 488 nm argon excitation laser. Acridine orange is a cell permeant nucleic acid binding dye that emits green fluorescence when bound to double stranded DNA (normal) and red when bound to single stranded DNA (abnormal) (Stewart et al., 2016).

Sperm death can be determined using apoptosis detection kits. One such kits uses PI and Annexin V. PI can only penetrate membrane of necrotic cells to stain nucleus. PI positive population, dead; Annexin V-FITC positive and PI negative population, early apoptotic; both Annexin V-FITC and PI positive population, late apoptotic; both Annexin V-FITC and PI negative population, normal (Quan et al., 2016). In situ cell death detection can also be determined enzymatically using a Terminal deoxynucleotydyl transferease dUTP nick end labeling (TUNEL) assay to detect DNA fragmentation (Alcay et al., 2016). Apoptotic cells can also be identified by mitochondrial membrane potential using specific stains such as MitoLight® (5,5_,6,6_-tetrachloro-1,1_,3,3_-tetraethylbenzimidazolylcarbocyanine chloride) that differentially stains mitochondria of healthy cells red while it remains as monomers in the cytoplasm and fluoresces green (Quan et al., 2016).

FERTILITY

Pregnancy rates using frozen-thawed semen vary from 50 to 70% in goats. Several factors, including breed, season, male, dose of semen, method of synchronization and flock/herd management (Anel et al., 2005; Arrebola et al., 2014). Artificial insemination is performed at a fixed time following synchronization or 12 hours after onset of estrus. In goats, most breeds can be inseminated by transcervical catheterization except for very small breeds where laparoscopic insemination is required in order to deposit semen in utero (Nordstoga et al., 2010b).

In sheep, the main challenge in artificial insemination with frozen-thawed semen has been the difficulty in placing semen in utero via transcervical catheterization because of the morphology of the cervix (Moura et al., 2011; Richardson et al., 2011; Robinson et al., 2011). Traditionally, ewes are inseminated with frozen-thawed seem via laparoscopy (Masoudi et al., 2017). Laparoscopic insemination does not seem to be affected by side of insemination (ipsilateral vs contralateral to ovulation) (Anakkul et al., 2014). In recent years, transcervical or vaginal inseminations gained more popularity. Several treatments have been tested to improve pregnancy rate following intracervical insemination. These include the use of estrogens, relaxin, oxytocin (IM) (Bartlewski and Candappa, 2015), topical application of PGE2 on the cervix (Horta et al., 2010; Barbas et al., 2013; Bartlewski and Candappa, 2015) and treatment with carazolol (a beta adrenergic blocking agent) (Gündüz et al., 2010). However, these treatments result in only modest improvement of pregnancy rates or sometimes a reduced fertility. Deposition of semen in the vaginal fornix can result in pregnancy although the overall conception rates are generally 10 to 20% lower than with intracervical insemination (Richardson et al., 2012a). A recent small study on vaginal insemination using a dose of 400 million spermatozoa per insemination resulted in a 50 % conception rate (Richardson et al., 2012a). Despite a reduced pregnancy rate with vaginal insemination, the technique is more attractive given the lesser cost and animal welfare issues compared to laparoscopic insemination (Paulenz et al., 2007). However, excellent semen quality is required with vaginal insemination (Paulenz et al., 2007; Richardson et al., 2012b). In sheep, epididymal sperm produced equal pregnancy rates as semen collected with artificial vagina. However, semen collected by electroejaculation produced lower pregnancy rates (Alvarez et al., 2012b).

CRYOPRESERVATION OF EMBRYOS

Embryo cryopreservation has been used in small ruminants since the late 1970’s. There are two methods of embryo cryopreservation; the conventional slow method and vitrification (Massip, 2001). The first step in embryo cryopreservation is selection of excellent quality embryos (Grade I or II) (Dalcin et al., 2013) (Figure 6). The main recent advances are in the development of simple procedures of vitrification making the process cheaper and faster for field use.

Cryopreservation of embryos by the slow cooling methods

The conventional method for embryo cryopreservation is often referred to as a slow-freezing method because it involves several steps which include, dehydration and equilibration with cryoprotectant, slow cooling to the seeding point, seeding and holding then further cooling at a slow rate prior to plunging the embryos in liquid nitrogen (Youngs, 2011).

Cryoprotectant addition and equilibration

The cryoprotectant can be added in a single step or in multiple steps with increasing concentration. Embryos are equilibrated in each solution for 5 to 10 minutes depending on the cryoprotectant used. The most common cryoprotectants used are glycerol of ethylene glycol. After equilibration, the embryos (2 per straw) are loaded in 0.25 mL straws (Youngs, 2011).

Initial cooling and seeding

The slow embryo freezing methods requires the use of a controlled rate biological freezer (Figure 7). These devices are designed to cool the embryos at a rate of 1°C/min from room temperature to the seeding point (-6 to -6.5°C). Seeding is the process of induction of ice crystal formation. This is achieved by touching the meniscus of the medium in the straw with very cold small tweezers or cotton-tipped stick that have been placed in liquid nitrogen (Figure 8). The formation of ice crystal in the medium surrounding the embryo results in further dehydration of the embryo. The straw is held at the seeding temperature for approximately 10 minutes before resuming the cooling process (Youngs, 2011).

Final cooling and storage in liquid nitrogen

After holding at seeding temperature, the straws must be cooled at a rate of 0.5°C/min to approximately -34 or -35°C where they are held for approximately 10 minutes prior to direct plunging in liquid nitrogen (-196°C) then transferred to a liquid nitrogen tank for storage (Youngs, 2011).

Thawing of embryos frozen with the conventional method

Cryopreserved embryos must be thawed following a strictly defined method. Straws are usually removed from the liquid nitrogen tank and held at room temperature for 3 to 5 seconds. This would reduce the risk of cracking the zona pellucida and explosion of the straws. Straws are then placed in a water bath at 35 to 38°C for 25 to 30 seconds. The embryos are removed from the straw and placed in a rehydration (cryoprotectant removal) medium prior to transfer. The process of cryoprotectant removal or rehydration is performed in a stepwise or single step fashion. The stepwise technique requires embryos to be placed successively in media with lower concentrations of cryoprotectant (1.5, 1.0 and 0.5 Molar) for 10 minutes per step. In the single step technique, the embryos are placed in a 1.0 Molar solution of sucrose (a non-penetrating sugar) for 10 minutes (Youngs, 2011).

In recent years, the trend has been to freeze embryos using the direct transfer method which does not require removing the embryos from the straws. The direct transfer method requires the use of ethylene glycol as the cryoprotectant. Because ethylene glycol is a small molecule, it diffuses quickly out of the embryo upon transfer to the recipient. It is very important to minimize the time from thawing to transfer into the uterus when using this method. In sheep, a one-step addition of 1.5 M ethylene glycol followed by the one step removal in 1 M sucrose after controlled freezing and thawing yielded very high survival rate (>70%) (McGinnis et al., 1993).

In goats, the maximum survival of frozen-thawed embryos is attained with expanded, hatching or hatched blastocysts (Li et al., 1990) (Figure 8).

Embryo vitrification

Vitrification is an ultrafast fast method for freezing (2500°C/min) resulting in “glass” (i.e. vitrous state) instead of ice crystal formation. The technique was first described for mammalian embryos in 1985 (Rall and Fahy, 1985). This avoidance of intracellular and extracellular ice crystal formation reduces cellular damage and improves embryo viability (Depaz et al., 1994). In addition, this technique does not require any sophisticated equipment, which reduces the cost of cryopreservation of embryos. Vitrification uses media containing high concentration of permeating cryoprotective agents. Embryos are placed in a first medium of 2 Molar of the cryoprotectant. Following equilibration, the embryos are placed immediately in a medium with much higher concentration of cryoprotectant (e.g 7 Molar) for a very short period of time (30 to 45 second) then they are plunged in liquid nitrogen (Massip, 2001). Several vitrification protocols have been reported for sheep and goat embryos. There seems to be an important difference between species in terms of behavior of embryos towards the vitrification medium (goats (El-Gayar and Holtz, 2001; Al Ahmad et al., 2012; Araujo-Lemos et al., 2015); sheep (Ali and Shelton, 1993; Bettencourt et al., 2009; de Araujo-Lemos et al., 2014; Santos Neto et al., 2015)).

In sheep, vitrification of embryos was reported to be as efficient as conventional freezing (Depaz et al., 1994; Martinez and Matkovic, 1998; Baril et al., 2001; Isachenko et al., 2003; Bettencourt et al., 2009; Green et al., 2009). Several improvements of the vitrification protocol have been achieved in recent years resulting in improved viability of embryos. There is a general agreement amongst authors that ovine embryos are more resistant to cryodamage than those of other species (Bettencourt et al., 2009).

A recently described protocol resulted in good survival of embryos. Embryos are placed in a basal solution of Hepes-TCM 199 supplemented with 20% Fetal Bovine Serum (FBS) for 5 minutes then transferred into a medium containing 10 % ethylene glycol (EG) and 10 % dimethylformamide (DMF) for 1 minute and moved to a solution of 20 % EG, 20% DMF and 0.5M sucrose and frozen using the open pulled straw technique by plunging in liquid nitrogen (Araujo-Lemos et al., 2015).

Sheep and goat embryos have been vitrified successfully using a standard simple commercial protocol used for bovine embryo vitrification. Embryos are placed in a commercial holding medium (Syngro®) until vitrification. The commercial vitrification protocol (Bovipro®, Minitube, Germany) consisted of a basic medium supplemented with 20 % FBS. The embryos are placed for 5 minutes in BM+10% glycerol then transferred into BM + 20 % glycerol +20 % EG for 5 minutes. Finally, the embryos are placed in the vitrification solution (BM+25 % glycerol+ 25 % EG) for 30 seconds. Embryos are loaded in 1 µl of vitrification medium in the tip of a micropipette and then placed in a 3.6 ml cryotube filled with liquid nitrogen. For thawing, the micropipette tips are warmed between the thumb and the middle finger for 10 seconds and the embryos are immersed in BM at 25°C in three dilution steps for 5 minutes each (12.5 % G+12.5 % EG+ 0.5 M sucrose, 0.5 M sucrose and 0.25 M sucrose). The embryos are then placed in BM solution for 5 minutes prior to transfer (Gibbons et al., 2011).

The stage of the embryo is very important. In sheep, morulas and blastocysts have similar survival rates while day 2 embryos have lower survival rate after vitrification (Santos Neto et al., 2015). In goats, blastocysts seem to tolerate vitrification better than morulae and hatched blastocysts (Al Yacoub et al., 2010).

Factors affecting pregnancy rate of frozen-thawed embryos

Several factors may impact the success of embryo cryopreservation and need to be further evaluated. These include method of production of embryos (in vivo vs in vitro), stage of embryo development, breed, age and diet of the donor. Vitrification protocols have been hindered by the toxicity of cryoprotectants. Research efforts have been centered on testing new, less toxic cryoprotectants. Pregnancy rates from vitrified embryos have improved to reach levels that are very comparable to transfer of fresh embryos.

Pregnancy rates following transfer of cryopreserved embryos vary between 38-73% for slow freezing and between 52-79% for vitrification and are not significantly different from those obtained with fresh embryos (50 to 90%) (Baril et al., 2001; Guignot et al., 2006; Hong et al., 2007; Green et al., 2009; Gibbons et al., 2011). There is however an interaction between method of freezing and cryoprotectant used and some vitrification techniques may produce lower pregnancy rates than conventional slow freezing (Varago et al., 2014). In sheep, direct transfer of vitrified embryos resulted in higher pregnancy rates than for embryos frozen by the slow method (Green et al., 2009).

Several factors may affect results of embryo cryopreservation. In vitro produced embryos have lower survival rates than in vivo produced embryos (Zhu et al., 2001; Martinez et al., 2006; Bhat et al., 2015). Pregnancy and birth rates achieved with in vitro produced cryopreserved embryos is also affected by method of culture during production (Mara et al., 2015). The poor survival of in vitro produced embryos may be due to epigenetic alterations (Nieddu et al., 2015; Romão et al., 2015). Also, in goats more advanced stages of development (expanded and hatched blastocysts) survive cryopreservation better than morulas (Gibbons et al., 2011). The effect of breed on embryo ability to withstand cryopreservation merits further studies (Fair et al., 2006). Some the breed effect may be due to differences in lipid content of oocytes and embryos (Romão et al., 2016).

CONCLUSION

Sperm and embryo cryopreservation are established techniques in small ruminants. Incorporation of these biotechnologies in a strategy for the preservation of genetic diversity of small ruminants and propagation of elite sires and dams is a reality. Improvements in sperm preservation through better extenders and reduction of oxidative stress may render the need for laparoscopic artificial insemination in sheep absolute and expand the use of the technique in the field. The development of new more objective methods for evaluation of cryopreserved semen allowed a better understanding of factors affecting post-thaw semen quality. Embryo preservation by vitrification produces similar results as conventional slow methods and reduces the cost of investment in specialized equipment. Further research is needed to refine vitrification techniques for use in the field for direct transfer.

REFERENCES

Abdi-Benemar H., Jafaroghli M., Khalili B., Zamiri M.J., Ezazi, H., Shadparvar, A.A. (2015). Effects of DHA supplementation of the extender containing egg yolk and alpha- tocopherol on the freezability and post-thawing fertility of ram semen. Small Ruminant Res., 130: 166-170.

Ahmad M., Nasrullah R., Ahmad N. (2015). Effect of cooling rate and equilibration time on pre-freeze and post-thaw survival of buck sperm. Cryobiology 70: 233-238.

Al Ahmad M.Z.A., Chebloune Y., Chatagnon G., Pellerin J.L., Fieni F. (2012). Is caprine arthritis encephalitis virus (CAEV) transmitted vertically to early embryo development stages (morulae or blastocyst) via in vitro infected frozen semen? Theriogenology, 77: 1673-1678.

Al Yacoub A.N., Gauly M., Holtz W. (2010). Open pulled straw vitrification of goat embryos at various stages of development. Theriogenology, 73: 1018-1023.

Alcay S., Gokce E., Toker M.B., Onder N.T., Ustuner B., Uzabaci E., Gui Z., Cavus S. (2016). Freeze-dried egg yolk based extenders containing various antioxidants improve post-thawing quality and incubation resilience of goat spermatozoa. Cryobiology, 72: 269-273.

Ali J., Shelton J.N. (1993). Successful Vitrification of Day-6 Sheep Embryos. J. Reprod. Fertil., 99: 65-70.

Alvarez M., Tamayo-Canul J., Anel E., Boixo J.C., Mata-Campuzano M., Martinez-Pastor F., Anel L., de Paz P. (2012a). Sperm concentration at freezing affects post-thaw quality and fertility of ram semen. Theriogenology, 77: 1111-1118.

Alvarez M., Tamayo-Canul J., Martinez-Rodriguez C., Lopez-Uruena E., Gomes-Alves S., Anel L., Martinez-Pastor F., de Paz P. (2012b). Specificity of the extender used for freezing ram sperm depends of the spermatozoa source (ejaculate, electroejaculate or epididymis). Anim. Reprod. Sci., 132: 145-154.

Amidi F., Pazhohan A., Nashtaei M.S., Khodarahmian M., Nekoonam S. (2016). The role of antioxidants in sperm freezing: a review. Cell Tissue Bank, 17: 745-756.

Anakkul N., Suwimonteerabutr J., Tharasanit T., Khunmanee S., Diloksumpan P., Berg D.K., Techakumphu M. (2014). Sperm distribution and fertilization after unilateral and bilateral laparoscopic artificial insemination with frozen-thawed goat semen. Theriogenology, 82: 1137-1144.

Anand M., Yadav S. (2016). Assessment of motion and kinematic characteristics of frozen-thawed Sirohi goat semen using computer-assisted semen analysis. Vet. World, 9: 203-206.

Anel L., Kaabi M., Abroug B., Alvarez M., Anel E., Boixo J.C., de la Fuente L.F., de Paz P. (2005). Factors influencing the success of vaginal and laparoscopic artificial insemination in churra ewes: a field study. Theriogenology, 63: 1235-1247.

Araujo-Lemos P.F.B., Neto L.M.F., Moura M.T., Melo J.V., Lima P.F., Oliveira M.A.L. (2015). Comparison of vitrification and conventional freezing for cryopreservation of caprine embryos. Zygote, 23: 594-602.

Arrebola F., Gonzalez O., Torres R., Abecia J.A. (2014). Artificial insemination in Payoya goats: factors affecting fertility. Anim. Prod. Sci., 54: 356-362.

Awad M.M. (2011). Effects of sub-optimal glycerol concentration and cholesterol-loaded cyclodextrin in a Tris-based diluent on cryopreserved ram sperm longevity and acrosomal integrity. Small Ruminant Res., 100: 164-168.

Baghshahi H., Riasi A., Mandavi A.H., Shirazi A. (2014). Antioxidant effects of clove bud (Syzygium aromaticum) extract used with different extenders on ram spermatozoa during cryopreservation. Cryobiology, 69: 482-487.

Barbas, J.P., Horta, A.E.M., Marques, C.C., Baptista, M.C., Mascarenhas, R.D., Martins, D.O., Vasques, M.I., Pereira, R.M., Cavaco-Gonçalves, S. (2013). The fertility increase after misoprostol administration is differently expressed when sheep are inseminated with chilled or frozen-thawed semen. Small Ruminant Res., 113: 398-401.

Baril, G., Cognie, Y., Pougnard, J.L., Leboeuf, B., Traldi, A.S., Guignot, F., Beckers, J.F., Mermillod, P. (2001). Improvement of embryo, cryopreservation and embryo transfer methods in small ruminants. Eighth Conference on Ruminant Research, 365-368.

Bartlewski, P.M., Candappa, I.B.R. (2015). Assessing the usefulness of prostaglandin E2 (Cervidil) for transcervical artificial insemination in ewes. Theriogenology, 84: 1594-1602.

Baspinar, N., Coyan, K., Bucak, M.N., Tuncer, P.B. (2011). Effects of dithioerythritol on ram semen after the freeze-thawing process. Cryobiology, 63: 152-156.

Bathgate, R., Mace, N., Heasman, K., Evans, G., Maxwell, W.M.C., de Graaf, S.P. (2013). Birth of Kids After Artificial Insemination with Sex-Sorted, Frozen-Thawed Goat Spermatozoa. Reprod. Domest. Anim., 48: 893-898.

Batista M., Nino T., Santana M., Alamo D., Cabrera F., Gonzalez F., Gracia A. (2014). Post-thaw quality of buck semen samples cooled at 5 degrees C up to 2 days before cryopreservation. Small Ruminant Res., 121: 101-105.

Behzad A.M., Ebrahimi B., Alizadeh A.R., Esmaeili V., Dalman A., Rashki L., Shahverdi A.H. (2014). Improvement in In Vitro Fertilization Rate, Decrease in Reactive Oxygen Species and Spermatozoa Death Incidence in Rams by Dietary Fish Oil. Reprod. Domest. Anim., 49: 599-605.

Bernardini A., Hozbor F., Sanchez E., Fornes M.W., Alberio R.H., Cesari A. (2011). Conserved ram seminal plasma proteins bind to the sperm membrane and repair cryopreservation damage. Theriogenology, 76: 436-447.

Bettencourt E.M., Bettencourt C.M., Silva J.C.E., Ferreira P., Matos C.P., Romao R.J., Rocha A. (2009). Fertility rates following the transfer of ovine embryos cryopreserved using three protocols. Small Ruminant Res., 82: 112-116.

Bezerra F.S.B., Castelo T.S., Alves H.M., Oliveira I.R.S., Lima G.L., Peixoto G.C.X., Bezerra A.C.S.D., Silva A.R. (2011). Objective assessment of the cryoprotective effects of dimethylformamide for freezing goat semen. Cryobiology, 63: 263-266.

Bhat M.H., Sharma V., Khan F.A., Naykoo N.A., Yaqoob S.H., Vajta G., Khan H.M., Fazili M.R., Ganai N.A., Shah R.A. (2015). Open pulled straw vitrification and slow freezing of sheep IVF embryos using different cryoprotectants. Reprod. Fert. Develop., 27: 1175-1180.

Bispo C.A.S., Pugliesi G., Galvao P., Rodrigues M.T., Ker P.G., Filgueiras B., Carvalho G.R. (2011). Effect of low and high egg yolk concentrations in the semen extender for goat semen cryopreservation. Small Ruminant Res., 100: 54-58.

Bohlool Z., Mohammadi M., Mehr M.R.A., Hossein-Zadeh N.G. (2015). Effect of different concentrations of trehalose and glycerol on the freezability of ram semen using soybean lecithin-based diluents. Anim. Prod. Sci., 55: 666-671.

Bucak M.N., Atessahin A., Varish O., Yuce A., Tekin N., Akcay A. (2007). The influence of trehalose, taurine, cysteamine and hyaluronan on ram semen - Microscopic and oxidative stress parameters after freeze-thawing process. Theriogenology, 67: 1060-1067.

Bucak M.N., Keskin N., Taspinar M., Coyan K., Baspinar N., Cenariu M.C., Bilgili A., Ozturk C., Kursunlu A.N. (2013). Raffinose and hypotaurine improve the post-thawed Merino ram sperm parameters. Cryobiology, 67: 34-39.

Budai C., Egerszegi I., Olah J., Javor A., Kovacs A. (2014). The protective effect of antioxidants on liquid and frozen stored ram semen - review. Scientific Papers: Animal Science and Biotechnologies, 47: 46-52.

Buyukleblebici S., Tuncer P.B., Tasdemir U., Ozgurtas T., Durmaz E., Buyukleblebici O. (2014). The Comparison of Three Different Cryoprotectants in Cryopreservation of Angora Goat Semen. Kafkas Univ. Vet. Fak., 20: 613-619.

Cabrera F., González F., Batista M., Calero P., Medrano A., Gracia A. (2005). The effect of removal of seminal plasma, egg yolk level and season on sperm freezability of Canary buck (Capra hircus). Reprod. Domest. Anim., 40: 191-195.

Camara D.R., Pinto L.C., Pinto M.M.C.M., Kastelic J.P., Nunes J.F., Barbosa J.M.P., Guerra M.M.P. (2016). Influence of catalase and pre-freezing equilibration on post-thaw semen quality and conception rate in ewes laparoscopically inseminated. Anim. Reprod., 13: 21-27.

Camara, D.R., Silva, S.V., Almeida, F.C., Nunes, J.F., Guerra, M.M.P. (2011). Effects of antioxidants and duration of pre-freezing equilibration on frozen-thawed ram semen. Theriogenology, 76: 342-350.

Campbell, A., Pearson, L.K., Ciccarelli, M., Tibary, A. (2014). Comparison of two semen extenders for cryopreservation of Bighorn sheep (Ovis canadensis canadensis) epididymal sperm. Clinical Theriogenology, 6.

Chelucci, S., Pasciu, V., Succu, S., Addis, D., Leoni, G.G., Manca, M.E., Naitana, S., Berlinguer, F. (2015). Soybean lecithin-based extender preserves spermatozoa membrane integrity and fertilizing potential during goat semen cryopreservation. Theriogenology, 83: 1064-1074.

Cirit, U., Bagis, H., Demir, K., Agca, C., Pabuccuoglu, S., Varisli, O., Clifford-Rathert, C., Agca, Y. (2013). Comparison of cryoprotective effects of iodixanol, trehalose and cysteamine on ram semen. Anim. Reprod. Sci., 139: 38-44.

Cofre-Narbona, E.J., Peralta-Troncoso, O.A., Urquieta-Mangiola, B.E., Raggi-Saini, L.A., Benavides-Aguila, N., Parraguez-Gamboa, V.H. (2016). Improvement of antioxidant status and semen quality by oral supplementation with vitamins c and e in rams. Rev. Scient-Fac Cien, 26: 156-163.

Coloma, M.A., Gómez-Brunet, A., Velázquez, R., Toledano-Díaz, A., López-Sebastián, A., Santiago-Moreno, J. (2010a). Freezability of Iberian ibex (Capra pyrenaica) spermatozoa according to the glycerolization temperature and plasma testosterone concentration. Cryobiology, 61: 204-210.

Coloma, M.A., Toledano-Díaz, A., Castaño, C., Velázquez, R., Gómez-Brunet, A., López-Sebastián, A., Santiago-Moreno, J. (2011). Seasonal variation in reproductive physiological status in the Iberian ibex (Capra pyrenaica) and its relationship with sperm freezability. Theriogenology, 76: 1695-1705.

Coloma, M.A., Toledano-Díaz, A., López-Sebastián, A., Santiago-Moreno, J. (2010b). The influence of washing Spanish ibex (Capra pyrenaica) sperm on the effects of cryopreservation in dependency of the photoperiod. Theriogenology, 73: 900-908.

Coyan, K., Baspinar, N., Bucak, M.N., Akalin, P.P. (2011). Effects of cysteine and ergothioneine on post-thawed Merino ram sperm and biochemical parameters. Cryobiology, 63: 1-6.

Cseh S., Faigl, V., Amiridis, G.S. (2012). Semen processing and artificial insemination in health management of small ruminants. Anim. Reprod. Sci., 130: 187-192.

da Silva M.C., Moura L.C.D., de Melo M.I.V., Mambrini J.V.D., Neves M.M., Henry M.R.J.M., Snoeck P.P.D. (2014). Prolonged post cooling but not pre-cooling equilibrium length improves the viability of ram sperm cryopreserved in an extender containing low-density lipoproteins. Small Ruminant Res., 119: 88-95.

Dalcin L., Silva R.C., Paulini F., Silva B.D.M., Neves J.P., Lucci C.M. (2013). Cytoskeleton structure, pattern of mitochondrial activity and ultrastructure of frozen or vitrified sheep embryos. Cryobiology, 67: 137-145.

Daramola J.O., Adekunle E.O., Oke O.E., Onagbesan O.M., Oyewusi I.K., Oyewusi J.A. (2016a). Effects of coconut (Cocos nucifera) water with or without egg-yolk on viability of cryopreserved buck spermatozoa. Anim. Reprod., 13: 57-62.

Daramola J.O., Adekunle E.O., Onagbesan O.M., Oke, O.E. Ladokun A.O., Abiona J.A., Abioja M.O., Oyewusi I.K., Oyewusi J.A., Isah O.A., Sogunle O.M., Adeleke M.A. (2016b). Protective effects of fruit-juices on sperm viability of West African Dwarf goat bucks during cryopreservation. Anim. Reprod., 13: 7-13.

de Araujo-Lemos P.F.B., Neto L.M.D., de Melo J.V., Moura M.T., Lima P.F., Oliveira M.A.L. (2014). Comparison of different cryoprotectant regimes for vitrification of ovine embryos produced in vivo. Small Ruminant Res., 119: 100-106.

Del Olmo E., Bisbal A., Garcia-Alvarez O., Maroto-Morales A., Ramon M., Jimenez-Rabadan P., Anel-Lopez L., Soler A.J., Garde J.J., Fernandez-Santos M.R. (2015). Free-radical production after post- thaw incubation of ram spermatozoa is related to decreased in vivo fertility. Reprod. Fert. Develop., 27: 1187-1196.

Del Olmo E., Bisbal A., Maroto-Morales A., Garcia-Alvarez O., Ramon M., Jimenez-Rabadan P., Martinez-Pastor F., Soler A.J., Garde J.J., Fernandez-Santos M.R. (2013). Fertility of cryopreserved ovine semen is determined by sperm velocity. Anim. Reprod. Sci.,138: 102-109.

Del Valle I., Souter A., Maxwell W.M.C., Muino-Blanco T., Cebrian-Perez J.A. (2013). Function of ram spermatozoa frozen in diluents supplemented with casein and vegetable oils. Anim. Reprod. Sci., 138: 213-219.

Depaz P., Sanchez A.J., Fernandez J.G., Carbajo M., Dominguez J.C., Chamorro C.A., Anel L. (1994). Sheep Embryo Cryopreservation by Vitrification and Conventional Freezing. Theriogenology, 42: 327-338.

Dorado J., Munoz-Serrano A., Hidalgo M. (2010). The effect of cryopreservation on goat semen characteristics related to sperm freezability. Anim. Reprod. Sci., 121: 115-123.

El-Gayar M., Holtz W. (2001). Technical note: Vitrification of goat embryos by the open pulled-straw method. J. Anim. Sci., 79: 2436-2438.

Emamverdi M., Zhandi M., Shahneh A.Z., Sharafi M., Akbari-Sharif A. (2013). Optimization of Ram Semen Cryopreservation Using a Chemically Defined Soybean Lecithin-Based Extender. Reprod. Domest. Anim., 48: 899-904.

Emamverdi M., Zhandi M., Shahneh A.Z., Sharafi M., Akhlaghi A., Motlagh M.K., Dadkhah F., Davachi N.D. (2015). Flow cytometric and microscopic evaluation of post-thawed ram semen cryopreserved in chemically defined home-made or commercial extenders. Anim. Prod. Sci., 55: 551-558.

Esmaeili V., Shahverdi A.H., Alizadeh A.R., Alipour H., Chehrazi M. (2014). Saturated, omega-6 and omega-3 dietary fatty acid effects on the characteristics of fresh, frozen-thawed semen and blood parameters in rams. Andrologia, 46: 42-49.

Fair S., Doyle D.N., Diskin M.G., Hennessy A.A., Kenny D.A. (2014). The effect of dietary n-3 polyunsaturated fatty acids supplementation of rams on semen quality and subsequent quality of liquid stored semen. Theriogenology, 81: 210-219.

Fair S., Hanrahan J.P., Ward F., O’Meara C.M., Duffy P., Donovan A., Lonergan P., Evans A.C.O. (2006). The difference in embryo quality between Belclare and Suffolk ewes is not due to differences in oocyte quality. Theriogenology, 66: 191-197.

Fang Y., Blair H., Zhong R.Z., Sun H.X., Zhou D.W. (2016). Optimizing the freezing rate for ovine semen cryopreservation: Phospholipid profiles and functions of the plasma membrane and quality and fertilization of spermatozoa. Small. Rumin. Res., 139: 46-51.

Farshad A., Hosseini Y. (2013). The cryoprotective effects of amino acids supplementation on cooled and post-thaw Markhoz bucks semen quality. Small Ruminant Res., 114: 258-263.

Fernández-Santos M.R., Soler A.J., Ramón M., Ros-Santaella J.L., Maroto-Morales A., García-Álvarez O., Bisbal A., Garde J.J., Coloma M.A., Santiago-Moreno J. (2011). Effect of post-mortem time on post-thaw characteristics of Spanish ibex (Capra pyrenaica) spermatozoa. Anim. Reprod. Sci., 129: 56-66.

Forouzanfar M., Sharafi M., Hosseini S.M., Ostadhosseini S., Hajian M., Hosseini L., Abedi P., Nili N., Rahmani H.R., Nasr-Esfahani M.H. (2010). In vitro comparison of egg yolk-based and soybean lecithin-based extenders for cryopreservation of ram semen. Theriogenology, 73: 480-487.

Furstoss V., Borderes F., Forgerit Y., Guillouet P., Leboeuf B. (2010). The value of the percentage of motile sperm in predicting a significant portion of the fertility variation of frozen-thawed buck semen. Theriogenology, 74: 1197-1206.

Gallego-Calvo L., Gatica M.C., Santiago-Moreno J., Guzman J.L., Zarazaga L.A. (2015). Exogenous melatonin does not improve the freezability of Blanca Andaluza goat semen over exposure to two months of short days. Anim. Reprod. Sci., 157: 24-32.

Gangwar C., Kharche S.D., Ranjan R., Kumar S., Goel A.K., Jindal, S.K., Agarwal, S.K. (2015). Effect of vitamin C supplementation on freezability of Barbari buck semen. Small Ruminant Res., 129: 104-107.

Garde J.J., Olmo A.d., Soler A.J., Espeso G., Gomendio M., Roldan E.R.S. (2008). Effect of egg yolk, cryoprotectant, and various sugars on semen cryopreservation in endangered Cuvier’s gazelle (Gazella cuvieri). Anim. Reprod. Sci., 108: 384-401.

Garde J.J., Soler A.J., Cassinello J., Crespo C., Malo A.F., Espeso G., Gomendio M., Roldan E.R.S. (2003). Sperm cryopreservation in three species of endangered gazelles (Gazella cuvieri, G. dama mhorr, and G. dorcas neglecta). Biol. Reprod., 69: 602-611.

Gibbons A., Cueto M.I., Bonnet F.P. (2011). A simple vitrification technique for sheep and goat embryo cryopreservation. Small Ruminant Res., 95: 61-64.

Goularte K.L., Gastal G.D.A., Schiavon R.S., Goncalves A.O., Schneider J.R., Corcini C.D., Lucia T. (2014). Association between the presence of protein bands in ram seminal plasma and sperm tolerance to freezing. Anim. Reprod. Sci., 146: 165-169.

Green R.E., Santos B.F.S., Sicherle C.C., Landim-Alvarenga F.C., Bicudo S.D. (2009). Viability of OPS Vitrified Sheep Embryos After Direct Transfer. Reprod. Domest. Anim., 44:406-410.

Guignot F., Bouttier A., Baril G., Salvetti P., Pignon P., Beckers J.F., Touze J.L., Cognie J., Traldi A.S., Cognie Y., Mermillod P. (2006). Improved vitrification method allowing direct transfer of goat embryos. Theriogenology, 66: 1004-1011.

Gündüz M.C., Turna Ö., Cİrİt Ü., Uçmak M., Tek Ç., Sabuncu A., Bacınoğlu S. (2010). Lambing rates and litter size following carazolol administration prior to insemination in Kivircik ewes. Anim. Reprod. Sci., 118:32-36.

Hansen W., Pearson L., Rodriguez J., Sandoval S.A.T. (2012). Effects of a second freeze-thaw cycle on bighorn sheep (Ovis canadensis canadensis) semen motility and membrane integrity. Clinical Theriogenology, 4: 425.

Hazarika S.B., Bhuyan D., Deka B.C., Sinha S., Biswas R.K., Dutta D.J., Das A., Borah P., Dewry R.K. (2016). Effect of glutathione on the quality of frozen buck semen. Indian J. Anim. Sci., 86: 535-538.

Hong Q.H., Tian S.J., Zhu S.E., Feng J.Z., Yan C.L., Zhao X.M., Liu, G.S., Zheng, S.M. (2007). Vitrification of boer goat morulae and early blastocysts by straw and open-pulled straw method. Reprod. Domest. Anim., 42: 34-38.

Horta A.E.M., Barbas J.P., Marques C.C., Baptista M.C., Vasques M.I., Pereira R.M., Mascarenhas R.D., Cavaco-Gonçalves S. (2010). Improvement of fertility in artificially inseminated ewes following vaginal treatment with misoprostol plus terbutaline sulphate. Reprod. Domest. Anim., 45: e412-e416.

Isachenko V., Alabart J.L., Dattena M., Nawroth F., Cappai P., Isachenko E., Cocero M.J., Oliveira J., Roche A., Accardo C., Krivokharchenko A., Folch J. (2003). New technology for vitrification and field (microscopefree) warming and transfer of small ruminant embryos. Theriogenology, 59: 1209-1218.

Jerez-Ebensperger R.A., Luno V., Olaciregui M., Gonzalez, N., de Blas I., Gil L. (2015). Effect of pasteurized egg yolk and rosemary honey supplementation on quality of cryopreserved ram semen. Small Ruminant Res., 130: 153-156.

Jerez R., Gonzalez N., Olaciregui M., Luno V., de Blas I., Gil L. (2016). Use of soy milk combined with different cryoprotectants for the ram semen cryopreservation. Small Ruminant Res., 134: 34-38.

Jimenez-Rabadan P., Ramon M., Garcia-Alvarez O., Maroto-Morales A., Alvaro-Garcia P.J., Del Olmo E., Perez-Guzman M.D., Fernandez-Santos M.R., Garde J.J., Soler A.J. (2013). Improved cryopreservation protocol for Blanca-Celtiberica buck semen collected by electroejaculation. Cryobiology, 67: 251-257.

Jimenez-Rabadan P., Ramon M., Garcia-Alvarez O., Maroto-Morales A., del Olmo E., Perez-Guzman M.D., Bisbal A., Fernandez-Santos M.R., Garde J.J., Soler A.J. (2012). Effect of semen collection method (artificial vagina vs. electroejaculation), extender and centrifugation on post-thaw sperm quality of Blanca-Celtiberica buck ejaculates. Anim. Reprod. Sci., 132, 88-95.

Konyali C., Tomas C., Blanch E., Gomez E.A., Graham J.K., Moce E. (2013). Optimizing conditions for treating goat semen with cholesterol-loaded cyclodextrins prior to freezing to improve cryosurvival. Cryobiology, 67: 124-131.

Kucuk N., Aksoy M., Ucan U., Ahmad E., Naseer Z., Ceylan A., Serin I. (2014). Comparison of two different cryopreservation protocols for freezing goat semen. Cryobiology, 68: 327-331.

Kulaksiz R., Cebi C., Akcay E., Daskin A. (2010). The protective effect of egg yolk from different avian species during the cryopreservation of Karayaka ram semen. Small Ruminant Res., 88: 12-15.

Leahy T., Marti J.I., Evans G., Maxwell W.M.C. (2010). Seasonal variation in the protective effect of seminal plasma on frozen-thawed ram spermatozoa. Anim. Reprod. Sci., 119: 147-153.

Ledesma A., Manes J., Rios G., Aller J., Cesari A., Alberio R., Hozbor F. (2015). Effect of Seminal Plasma on Post-Thaw Quality and Functionality of Corriedale Ram Sperm Obtained by Electroejaculation and Artificial Vagina. Reprod. Domest. Anim., 50: 386-392.

Li R., Cameron A., Batt P., Trounson A. (1990). Maximum survival of frozen goat embryos is attained at the expanded, hatching and hatched blastocyst stages of development. Reprod. Fertil. Dev., 2: 345-350.

López-Saucedo, J., Santiago-Moreno, J., Fierro, R., Izquierdo, D., Coloma, M.A., Catalá, M.G., Jiménez, I., Paramio, M.T. (2015). Fertilization capacity of cryopreserved Iberian ibex epididymal sperm in a heterologous in vitro fertilization assay. Zygote, 23: 136-144.

Maia M.D., Bicudo S.D., Sicherle C.C., Rodello L., Gallego I.C.S. (2010). Lipid peroxidation and generation of hydrogen peroxide in frozen-thawed ram semen cryopreserved in extenders with antioxidants. Anim. Reprod. Sci., 122: 118-123.

Mara L., Sanna D., Dattena M., Muñoz I.M.M. (2015). Different in vitro culture systems affect the birth weight of lambs from vitrified ovine embryos. Zygote, 23: 53-57.

Martinez-Pastor F., Guerra C., Kaabi M., Garcia-Macias V., de Paz P., Alvarez M. (2005). Season effect on genitalia and epididymal sperm from Iberian red deer, roe deer and Cantabrian chamois. Theriogenology, 63: 1857-1875.

Martínez A.F., Martínez-Pastor F., Alvarez M., Fernández-Santos M.R., Esteso M.C., de Paz P. (2008). Sperm parameters on Iberian red deer: electroejaculation and post-mortem collection. Theriogenology, 70: 216-226.

Martinez A.G., Matkovic M. (1998). Cryopreservation of ovine embryos: Slow freezing and vitrification. Theriogenology, 49: 1039-1049.

Martinez A.G., Valcarcel A., Furnus C.C., de Matos D.G., Iorio G., de las Heras M.A. (2006). Cryopreservation of in vitro-produced ovine embryos. Small Ruminant Res., 63: 288-296.

Mascaro F., Gil L., Malo C., Gonzalez N., Martinez F., de Blas I. (2013). Effect of Pasteurized Egg and Rosmarinus Officinalis Supplementation on Quality of Cryopreserved Ram Semen. Cryoletters, 34: 422-431.

Masoudi R., Shahneh A.Z., Towhidi A., Kohram H., Akbarisharif A., Sharafi M. (2017). Fertility response of artificial insemination methods in sheep with fresh and frozen-thawed semen. Cryobiology, 4: 77-80.

Masoudi R., Sharafi M., Shahneh A.Z., Towhidi A., Kohram H., Esmaeili V., Shahverdi A., Davachi N.D. (2016a). Fertility and flow cytometry study of frozen-thawed sperm in cryopreservation medium supplemented with soybean lecithin. Cryobiology, 73: 69-72.

Masoudi R., Sharafi M., Shahneh A.Z., Towhidi A., Kohram H., Zhandi M., Esmaeili V., Shahverdi A. (2016b). Effect of dietary fish oil supplementation on ram semen freeze ability and fertility using soybean lecithin- and egg yolk-based extenders. Theriogenology, 86: 1583-1588.

Massip A. (2001). Cryopreservation of embryos of farm animals. Reprod. Domest. Anim., 36: 49-55.

Mata-Campuzano M., Alvarez-Rodriguez M., Alvarez M., Anel L., de Paz P., Garde J.J., Martinez-Pastor F. (2012). Effect of Several Antioxidants on Thawed Ram Spermatozoa Submitted to 37 degrees C up to Four Hours. Reprod. Domest. Anim., 47: 907-914.

Mata-Campuzano M., Alvarez-Rodriguez M., Alvarez M., de Paz P., Anel L., Garde J., Martinez-Pastor F. (2011). Comparison of the effect of different antioxidants on frozen-thawed and incubated ram semen samples. Reprod. Domest. Anim., 46: 128-128.

Mata-Campuzano M., Alvarez-Rodriguez M., Alvarez M., Tamayo-Canul J., Anel L., de Paz P., Martinez-Pastor F. (2015). Post-thawing quality and incubation resilience of cryopreserved ram spermatozoa are affected by antioxidant supplementation and choice of extender. Theriogenology, 83: 520-528.

Maxwell W.M., de Graaf S.P., Ghaoui Rel H., Evans G. (2007). Seminal plasma effects on sperm handling and female fertility. . Soc. Reprod.Fertil., 64 (Suppl.): 13-38.

Maxwell W.M., Evans G., Mortimer S.T., Gillan L., Gellatly E.S., McPhie C.A. (1999). Normal fertility in ewes after cervical insemination withfrozen-thawed spermatozoa supplemented with seminal plasma. . Reprod. Fertil. Dev., 11: 123-126.

McGinnis L., Duplantis S.J., Youngs C. (1993). Cryopreservation of sheep embryos using ethylene glycol. Anim. Reprod. Sci., 30: 273-280.

Mehr M.R.A., Noori H. (2013). Effect of different levels of L-Glutamine and glycerol on freezing of ram spermatozoa. Small Ruminant Res., 115: 103-107.

Memon A.A., Wahid H., Rosnina Y., Goh Y.M., Ebrahimi M., Nadia F.M. (2013). Effect of Ascorbic Acid Concentrations, Methods of Cooling and Freezing on Boer Goat Semen Cryopreservation. Reprod. Domest. Anim., 48: 325-330.

Moce E., Purdy P.H., Graham J.K. (2010). Treating ram sperm with cholesterol-loaded cyclodextrins improves cryosurvival. Anim. Reprod. Sci., 118: 236-247.

Moce E., Tomas C., Blanch E., Graham J.K. (2014). Effect of cholesterol-loaded cyclodextrins on bull and goat sperm processed with fast or slow cryopreservation protocols. Animal, 8: 771-776.

Motamedi-Mojdehi R., Mehr M.R.A., Rajabi-Toustani R. (2014). Effect of Different Levels of Glycerol and Cholesterol-Loaded Cyclodextrin on Cryosurvival of Ram Spermatozoa. Reprod. Domest. Anim., 49: 65-70.

Moura D.S., Lourenco T.T., Moscardini M.M., Scott C., Fonseca P.O., Souza F.F. (2011). Morphological aspects of ovine cervix. Pesquisa Vet. Brasil, 31: 33-38.

Moura P.P., Franco M.M., Silva T.A.D.N., Rocha T.L., Leal D.R., Passos P.I.B., Neves J.P. (2010). Characterization of seminal plasma proteins and its relationship with quality parameters of frozen semen in ram. Cienc. Rural, 40: 1154-1159.

Moustacas V.S., Cruz B.C., Varago F.C., Miranda D.A., Lage P.G., Henry M. (2011). Extenders Containing Dimethylformamide Associated or Not with Glycerol are Ineffective for Ovine Sperm Cryopreservation. Reprod. Domest. Anim., 46: 924-925.

Naijian H.R., Kohram H., Shahneh A.Z., Sharafi M., Bucak M.N. (2013). Effects of different concentrations of BHT on microscopic and oxidative parameters of Mahabadi goat semen following the freeze-thaw process. Cryobiology, 66: 151-155.

Naing S.W., Wahid H., Azam K.M., Rosnina Y., Zuki A.B., Kazhal S., Bukar M.M., Thein M., Kyaw T., San M.M. (2010). Effect of sugars on characteristics of Boer goat semen after cryopreservation. Anim. Reprod. Sci., 122: 23-28.

Najafi A., Kia H.D., Mohammadi H., Najafi M.H., Zanganeh Z., Sharafi M., Martinez-Pastor F., Adeldust H. (2014a). Different concentrations of cysteamine and ergothioneine improve microscopic and oxidative parameters in ram semen frozen with a soybean lecithin extender. Cryobiology, 69: 68-73.

Najafi A., Najafi M.H., Zanganeh Z., Sharafi M., Martinez-Pastor F., Adeldust H. (2014b). Cryopreservation of Ram Semen in Extenders Containing Soybean Lecithin as Cryoprotectant and Hyaluronic Acid as Antioxidant. Reprod. Domest. Anim., 49: 934-940.

Najafi A., Zhandi M., Towhidi A., Sharafi M., Sharif A.A., Motlagh M.K., Martinez-Pastor F. (2013). Trehalose and glycerol have a dose-dependent synergistic effect on the post-thawing quality of ram semen cryopreserved in a soybean lecithin-based extender. Cryobiology, 66: 275-282.

Nieddu S.M., Mossa F., Strina A., Ariu F., Pau S., Ledda M., Sotgia S., Carru C., Ledda S. (2015). Differences in amniotic amino acid concentrations between pregnancies obtained with transfer of vitrified thawed in vitro-produced embryos and with natural mating in sheep. Theriogenology, 83: 687-692.

Nikbin S., Panandam J.M., Yaakub H., Murugaiyah M., Sazili A.Q. (2014). Novel SNPs in heat shock protein 70 gene and their association with sperm quality traits of Boer goats and Boer crosses. Anim. Reprod. Sci., 146: 176-181.

Nordstoga A.B., Soderquist L., Adnoy T., Farstad W., Paulenz H. (2010a). Vaginal deposition of frozen-thawed semen in Norwegian Dairy goats: Comparison of single and double insemination with equal total number of spermatozoa. Theriogenology, 74: 895-900.

Nordstoga A.B., Soderquist L., Adnoy T., Paulenz H. (2011). Fertility Results after Vaginal Deposition of Frozen-Thawed Buck Semen Diluted with Two Different Extenders Using One- or Two-Step Procedures. Reprod. Domest. Anim., 46: 82-86.

Nur Z., Zik B., Ustuner B., Sagirkaya H., Ozguden C.G. (2010). Effects of different cryoprotective agents on ram sperm morphology and DNAintegrity. Theriogenology, 73: 1267-1275.

Paulenz H., Ǻdnøy T., Söderquist L. (2007). Comparison of fertility results after vaginal insemination using different thawing procedures and packages for frozen ram semen. Acta Vet. Scand., 49.

Pelufo V., Armengol M.F.L., Malcotti V., Venturino A., Aisen E.G. (2015). Effects of glycerol and sugar mixing temperature on the morphologic and functional integrity of cryopreserved ram sperm. Theriogenology, 83: 144-151.

Pradiee J., Esteso M.C., Castaño C., Toledano-Díaz A., López-Sebastián, A., Santiago-Moreno, J. (2014). Cryopreservation of epididymal sperm from ibexes (Capra pyrenaica) using short equilibration time with glycerol. Theriogenology, 82: 525-528.

Pradiee J., Esteso M.C., Lopez-Sebastián A., Toledano-Díaz A., Castaño C., Carrizosa J.A., Urrutia B., Santiago-Moreno J. (2015). Successful ultrarapid cryopreservation of wild Iberian ibex (Capra pyrenaica) spermatozoa. Theriogenology, 84: 1513-1522.

Pradiee J., O’Brien E., Esteso M.C., Castaño C., Toledano-Díaz A., Lopez-Sebastián A., Marcos-Beltrán J.L., Vega R.S., Guillamón F.G., Martínez-Nevado E., Guerra R., Santiago-Moreno J. (2016). Effect of shortening the prefreezing equilibration time with glycerol on the quality of chamois (Rupicapra pyrenaica), ibex (Capra pyrenaica), mouflon (Ovis musimon) and aoudad (Ammotragus lervia) ejaculates. Anim. Reprod. Sci., 171: 121-128.

Prado O.R., Bastos G.M., Monteiro A.L.G., Saab B.B., Gilaverte S., Pierobom C.C., Hentz F., Martins L.H.S., Silva C.J.A., Dranca G.S., Stivari T.S.S., Cerqueira G. (2013). Addition of seminal plasma to frozen-thawed semen and pregnancy rate of fixed time inseminated ewes. Arq. Bras. Med. Vet. Zoo., 65: 13-18.

Prieto-Pablos M.T., Sanchez-Calabuig M.J., Hildebrandt T.B., Goritz F., Ortmann S., Eder S., Santiago-Moreno J., Hermes R., Saragusty J. (2016). Cryopreservation of captive roe deer (Capreolus capreolus) semen. Theriogenology, 86: 695-703.

Quan G.B., Ma Y., Li J., Wu G.Q., Li D.J., Ni Y.N., Lv C.R., Zhu L., Hong Q.H. (2015). Effects of Hoechst33342 staining on the viability and flow cytometric sex-sorting of frozen-thawed ram sperm. Cryobiology, 70: 23-31.

Quan G.B., Wu G.Q., Wang Y.J., Li D.J., Ma Y., Hong,Q.H. (2016). Effects of the Tris, Tes, or skim milk based extender on in vitro parameters of ram spermatozoa during liquid storage. Small Ruminant Res., 134: 14-21.

Rall W.F., Fahy G.M. (1985). Ice-free cryopreservation of mouse embryos at -196ºC by vitrification. Nature 313: 573-575.,

Razliqi R.N., Zhandi M., Shakeri M., Towhidi A., Sharafi M., Emamverdi M., Motlagh M.K. (2015). Protective role of glutathione in buck semen cryopreservation. Iran J. Vet. Res., 16: 298-300.

Rekha A., Zohara B.F., Bari F., Alam M.G.S. (2016). Comparison of commercial Triladyl extender with a tris-fructose-egg-yolk extender on the quality of frozen semen and pregnancy rate after transcervical AI in Bangladeshi indigenous sheep (Ovis aries). Small Ruminant Res., 134: 39-43.

Richardson L., Hanrahan J.P., Donovan A., Martí J.I., Fair, S., Evans A.C.O., Lonerg P. (2012a). Effect of site of deposition on the fertility of sheep inseminated with frozen-thawed semen. Anim. Reprod. Sci., 131: 160-164.

Richardson L., Hanrahan J.P., O’Hara L., Donovan A., Fair S., O’Sullivan M., Carrington S.D., Lonergan P., Evans A.C.O. (2011). Ewe breed differences in fertility after cervical AI with frozen-thawed semen and associated differences in sperm penetration and physicochemical properties of cervical mucus. Anim. Reprod. Sci., 129: 37-43.

Rickard P., Schmidt R.E., Maddison J.W., Bathgate R., Lynch G.W., Druart X., de Graaf S.P. (2016). Variation in seminal plasma alters the ability of ram spermatozoa to survive cryopreservation. Reprod. Fert. Develop., 28: 516-523.

Robinson J.J., McKelvey W.A.C., King M.E., Mitchell S.E., Mylne M.J.A., McEvoy T.G., Dingwall W.S., Williams L.M. (2011). Traversing the ovine cervix - a challenge for cryopreserved semen and creative science. Animal, 5: 1791-1804.

Rodriguez J.S., Pearson L.K., Sandoval S., Kasimanickam R.K., Tibary, A. (2010). Cryopreservation and fertility of Bighorn (Ovis canadensis c.) cauda epididymis semen. Clinical Theriogenology, 2: 385.

Rodriguez J.S., Tibary A., Sandoval S., Nielsen S., Subramanian R., Srikumaran S., Foreyt E., Knowles D.P. (2009). Production of hybrid (Bighorn x domestic sheep) lambs by laparoscopic artificial insemination using Bighorn fresh semen collected by electro ejaculation. Clinical Theriogenology, 1.

Romão R., Bettencourt E., Pereira R.M.L.N., Marques C.C., Baptista M.C., Barbas J.P., Oliveira E., Bettencourt C., Sousa M. (2016). Ultrastructural characterization of fresh and vitrified in vitro- and in vivo-produced sheep embryos. Anatomia Histologia Embryologia, 45: 231-239.

Romão R., Marques C.C., Baptista M.C., Barbas J.P., Horta A.E.M., Carolino N., Bettencourt E., Pereira R.M. (2015). Cryopreservation of in vitro-produced sheep embryos: effects of different protocols of lipid reduction. Theriogenology, 84: 118-126.

Romon M., Perez-Guzman M.D., Jimenez-Rabadan P., Esteso M.C., Garcia-Alvarez O., Maroto-Morales A., Anel-Lopez L., Soler A.J., Fernandez-Santos M.R., Garde J.J. (2013). Sperm Cell Population Dynamics in Ram Semen during the Cryopreservation Process. Plos One 8.

Roof D.J., Bowley S., Price L.L., Matsas D.J. (2012). Comparison of two commercial extenders for cryopreservation of goat semen without sperm washing. Theriogenology, 77: 412-420.

Rovegno M., Feitosa W.B., Rocha A.M., Mendes C.M., Visintin, J.A., Assumpcao, M.E.O.D. (2013). Assessment of post-thawed ram sperm viability after incubation with seminal plasma. Cell Tissue Bank, 14: 333-339.

Salmani H., Nabi M.M., Vaseghi-Dodaran H., Rahman M.B., Mohammadi-Sangcheshmeh A., Shakeri M., Towhidi A., Shahneh A.Z., Zhandi M. (2013). Effect of glutathione in soybean lecithin-based semen extender on goat semen quality after freeze-thawing. Small Ruminant Res., 112: 123-127.

Salmani, H., Towhidi, A., Zhandi, M., Bahreini, M., Sharafi, M. (2014). In vitro assessment of soybean lecithin and egg yolk based diluents for cryopreservation of goat semen. Cryobiology, 68: 276-280.

Sangeeta S., Arangasamy A., Kulkarni S., Selvaraju S. (2015). Role of amino acids as additives on sperm motility, plasma membrane integrity and lipid peroxidation levels at pre-freeze and post-thawed ram semen. Anim. Reprod. Sci., 161: 82-88.

Santiago-Moreno J., Astorga R.J., Luque I., Coloma M.A., Toledano-Díaz A., Pulido-Pastor A., Gómez-Guillamon F., Salas-Vega R., López-Sebastián A. (2009a). Influence of recovery method and microbial contamination on the response to freezing-thawing in ibex (Capra pyrenaica) epididymal spermatozoa. Cryobiology, 59: 357-362.

Santiago-Moreno J., Castaño C., Toledano-Díaz A., Esteso M.C., López-Sebastián A., Guerra R., Ruiz, M.J., Mendoza N., Luna C., Cebrián-Pérez J.A., Hildebrandt T.B. (2013). Cryopreservation of aoudad (Ammotragus lervia sahariensis) sperm obtained by transrectal ultrasound-guided massage of the accessory sex glands and electroejaculation. Theriogenology, 79: 383-391.

Santiago-Moreno J., Coloma M.A., Dorado J., Pulido-Pastor A., Gómez-Guillamon F., Salas-Vega R., Gómez-Brunet A., López-Sebastián A. (2009b). Cryopreservation of Spanish ibex (Capra pyrenaica) sperm obtained by electroejaculation outside the rutting season. Theriogenology, 71: 1253-1260.

Santiago-Moreno J., Coloma M.A., Toledano-Díaz A., Gómez-Brunet A., Pulido-Pastor A., Zamora-Soria A., Carrizosa J.A., Urrutia B., López-Sebastián A. (2008). A comparison of the protective action of chicken and quail egg yolk in the cryopreservation of spanish ibex epididymal spermatozoa. Cryobiology, 57: 25-29.

Santiago-Moreno J., Toledano-Díaz A., Dorado J., Pulido-Pastor A., Coloma M.A., López-Sebastián A. (2007). Recovery and cryopreservation of Spanish ibex epididymal spermatozoa. Arch. Andrology, 53: 309-316.

Santiago-Moreno J., Toledano-Díaz A., Pulido-Pastor,A., Dorado J., Gómez-Brunet A., López-Sebastián,A. (2006a). Effect of egg yolk concentration on cryopreserving Spanish ibex (Capra pyrenaica) epididymal spermatozoa. Theriogenology, 66: 1219-1226.

Santiago-Moreno J., Toledano-Díaz A., Pulido-Pastor A., Gómez-Brunet A., López-Sebastián A. (2006b). Birth of live Spanish ibex (Capra pyrenaica hispanica) derived from artificial insemination with epididymal spermatozoa retrieved after death. Theriogenology, 66: 283-291.

Santiani A., Evangelista S., Sepulveda N., Risopatron J., Villegas J., Sanchez R. (2014). Addition of superoxide dismutase mimics during cooling process prevents oxidative stress and improves semen quality parameters in frozen/thawed ram spermatozoa. Theriogenology, 82: 884-889.

Santos Neto P.C.d., Vilariño M., Barrera N., Cuadro F., Crispo M., Menchaca A. (2015). Cryotolerance of Day 2 or Day 6 in vitro produced ovine embryos after vitrification by Cryotop or Spatula methods. Cryobiology, 70: 17-22.

Sariozkan S., Bucak M.N., Tuncer P.B., Tasdemir U., Kinet H., Ulutas P.A. (2010). Effects of different extenders and centrifugation/washing on postthaw microscopic-oxidative stress parameters and fertilizing ability of Angora buck sperm. Theriogenology, 73: 316-323.

Shamsuddin M., Amiri Y., Bhuivan M.M.U. (2000). Characteristics of buck semen with regard to ejaculate numbers, collection intervals, diluents and preservation periods. Reprod. Domest. Anim., 35: 53-57.

Sharafi M., Zhandi M., Sharif A.A. (2015). Supplementation of soybean lecithin-based semen extender by antioxidants: complementary flowcytometric study on post-thawed ram spermatozoa. Cell Tissue Bank, 16: 261-269.

Sicherle C.C., Maia M.S., Bicudo S.D., Rodello L., Azevedo H.C. (2011). Lipid peroxidation and generation of hydrogen peroxide in frozen-thawed ram semen supplemented with catalase or Trolox. Small Ruminant Res., 95: 144-149.

Silva E.C.B., Cajueiro J.F.P., Silva S.V., Soares P.C., Guerra M.M.P. (2012a). Effect of antioxidants resveratrol and quercetin on in vitro evaluation of frozen ram sperm. Theriogenology, 77: 1722-1726.

Silva E.C.B., Cajueiro J.F.P., Silva S.V., Vidal A.H., Soares P.C., Guerra M.M.P. (2012b). In vitro evaluation of ram sperm frozen with glycerol, ethylene glycol or acetamide. Anim. Reprod. Sci., 132: 155-158.

Silva S.V., Soares A.T., Batista A.M., Almeida F.C., Nunes J.F., Peixoto C.A., Guerra M.M.P. (2011). In Vitro and In Vivo Evaluation of Ram Sperm Frozen in Tris Egg-yolk and Supplemented with Superoxide Dismutase and Reduced Glutathione. Reprod. Domest. Anim., 46: 874-881.

Silva S.V., Soares A.T., Batista A.M., Almeida F.C., Nunes J.F., Peixoto C.A., Guerra M.M.P. (2013). Vitamin E (Trolox) addition to Tris-egg yolk extender preserves ram spermatozoon structure and kinematics after cryopreservation. Anim. Reprod. Sci. 137: 37-44.

Soares A.T., Silva S.V., Batista A.M., Almeida F.C., Nunes, J.F., Peixoto, C.A., Guerra, M.M.P. (2015). Ultrastructure evaluation of goat spermatozoa after freezing in a skim milk-based extender with Trolox supplementation. Andrologia. 47: 470-476.

Sobrinho J.M.F., Branco M.A.C., Sousa A., Nascimento I.M.R., Mota L.H.C.M., Carvalho Y.N.T., Ferreira S.B., Costa D.N.M., Moraes F.J., Souza A.T. (2014). Characteristics of the semen of Dorper, Santa Ines and undefined breed sheep, pre-and post-freezing, in the rainy and dry period. Arq. Bras. Med. Vet. Zoo. 66: 969-976.

Stewart J.L., Shipley C.F., Katich A.S., Po, E., Ellerbrock, R.E., Lima, F.S., Canisso, I.F. (2016). Cryopreservation of white-tailed deer (Odocoileus virginianus) semen using soybean-, liposome, and egg yolk-based extenders. Anim. Reprod. Sci. 171: 7-16.

Subramaniam R., Shanthalingam S., Bavananthasivam J., Kugadas A., Raghavan B., Batra S.A., Herndon C.N., Rodriguez J., Tibary A., Nelson D., Potter K.A., Foreyt W.J., Srikumaran S. (2014). Bighorn sheep x domestic sheep hybrids survive Mannheimia haemolytica challenge in the absence of vaccination. Veterinary Microbiology. 170: 278-283.

Tibary A., Boukhliq R., El Allali K. (2018). Ram and Buck breeding soundness examination. Rev. Mar. Sci. Agr. Vét., 6: 241-255

Toker M.B., Alcay S., Gokce E., Ustuner B. (2016). Cryopreservation of ram semen with antioxidant supplemented soybean lecithin-based extenders and impacts on incubation resilience. Cryobiology. 72: 205-209.

Towhidi A., Zeinoaldini S., Ardebili R., Davachi N.D., Nasiri, A.H. (2013). Combined n-3 Fatty Acids and alpha-Tocopherol Supplementation Improved the Ovine Sperm Cryosurvival. Iran J. Biotechnol. 11: 238-243.

Tuli R., Holtz W. (1992). The effect of zwitterions buffers on the freezability of Boer goat semen. Theriogenology. 37: 674-951.

Tuncer P.B., Bucak M.N., Sariozkan S., Sakin F., Yeni D., Cigerci I.H., Atessahin A., Avdatek F., Gundogan M., Buyukleblebici O. (2010). The effect of raffinose and methionine on frozen/thawed Angora buck (Capra hircus ancryrensis) semen quality, lipid peroxidation and antioxidant enzyme activities. Cryobiology. 61: 89-93.

Tuncer P.B., Tasdemir U., Buyukleblebici S., Ozgurtas T., Coskun E., Erol H., Aydin F.N., Gurcan I.S. (2013). Effects of different doses of trehalose supplementation in egg yolk extender in frozen-thawed Angora buck semen. Small Ruminant Res. 113: 383-389.

Turri F., Madeddu M., Gliozzi T.M., Gandini G., Pizzi F. (2014). Effect of testicle postmortem storage on goat frozen-thawed epididymal sperm quality as a tool to improve genebanking in local breeds. Animal. 8: 440-447.

Ustuner B., Alcay S., Nur Z., Sagirkaya H., Soylu M.K. (2014). Effect of Egg Yolk and Soybean Lecithin on Tris-Based Extender in Post-Thaw Ram Semen Quality and in vitro Fertility. Kafkas Univ. Vet. Fak. 20: 393-398.

Varago F.C., Moutacas V.S., Carvalho B.C., Serapiao R.V., Vieira F., Chiarini-Garcia H., Brandao F.Z., Camargo L.S., Henry M., Lagares M.A. (2014). Comparison of Conventional Freezing and Vitrification with Dimethylformamide and Ethylene Glycol for Cryopreservation of Ovine Embryos. Reprod. Domest. Anim. 49: 839-844.

Vasquez J.H., Nunez V.H., Florentini E.A., Gonzales J.M., Camargo L.A., Valdivia M.E. (2013). Effects of five cryoprotective agents on quality of sheep epididymal spermatozoa during pre-freezing. Livest. Sci., 152: 94-99.

Vidal A.H., Batista A.M., da Silva E.C.B., Gomes W.A., Pelinca M.A., Silva S.V., Guerra M.M.P. (2013). Soybean lecithin-based extender as an alternative for goat sperm cryopreservation. Small Ruminant Res. 109: 47-51.

Wang W., Luo J., Sun, S., Xi L., Gao, Q., Haile A.B., Shi H., Zhang W., Shi H. (2015). The Effect of Season on Spermatozoa Motility, Plasma Membrane and Acrosome Integrity in Fresh and Frozen-Thawed Semen from Xinong Saanen Bucks. Reprod. Domest. Anim. 50: 23-28.

Yaniz J.L., Palacin I., Vicente-Fiel S., Gosalvez J., Lopez-Fernandez C., Santolaria P. (2013). Comparison of Membrane-Permeant Fluorescent Probes for Sperm Viability Assessment in the Ram. Reprod. Domest. Anim. 48: 598-603.

Yildiz, S., Ozturkler, Y., Ari, U.C., Lehimcioglu, N.C., Atakisi, E., Kulaksiz, R. (2015). The Effects of L-Ergothioneine, N-acetylcystein and Cystein on Freezing of Ram Semen. Kafkas Univ. Vet. Fak. 21: 81-86.

Youngs C.R. (2011). Cryopreservation of Preimplantation Embryos of Cattle, Sheep, and Goats. Jove J. Vis. Exp.

Zalazar L., Ledesma A., Hozbor F., Cesari A. (2016). Heterologous recombinant protein with decapacitating activity prevents and reverts cryodamage in ram sperm: An emerging biotechnological tool for cryobiology. Anim. Reprod. Sci. 164: 31-39.

Zanganeh Z., Zhandi M., Zare-Shahneh A., Najafi A., Nabi M.M., Mohammadi-Sangcheshmeh A. (2013). Does rosemary aqueous extract improve buck semen cryopreservation? Small Ruminant Res. 114: 120-125.

Zhandi M., Sharafi M. (2015). Negative effect of combined cysteine and glutathione in soy lecithin-based extender on post-thawed ram spermatozoa. Cell Tissue Bank. 16: 443-448.

Zhu S.E., Zeng S.M., Yu W.L., Li S.J., Zhang Z.C., Chen Y.F. (2001). Vitrification of in vivo and in vitro produced ovine blastocysts. Anim. Biotechnol. 12: 193-203.