Next Article in Journal
A Heterozygous Missense Variant in MAP2K2 in a Stillborn Romagnola Calf with Skeletal-Cardio-Enteric Dysplasia
Previous Article in Journal
Antioxidant and Antimicrobial Activity of Cleome droserifolia (Forssk.) Del. and Its Biological Effects on Redox Status, Immunity, and Gut Microflora
Previous Article in Special Issue
V-Set and Immunoglobulin Domain-Containing 1 (VSIG1), Predominantly Expressed in Testicular Germ Cells, Is Dispensable for Spermatogenesis and Male Fertility in Mice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Overview on the Antioxidants, Egg Yolk Alternatives, and Mesenchymal Stem Cells and Derivatives Used in Canine Sperm Cryopreservation

by
Feriel Yasmine Mahiddine
and
Min-Jung Kim
*
Department of Research and Development, Mjbiogen Corp., Gwangnaru-ro 144, Seoul 14788, Korea
*
Author to whom correspondence should be addressed.
Animals 2021, 11(7), 1930; https://doi.org/10.3390/ani11071930
Submission received: 26 May 2021 / Revised: 22 June 2021 / Accepted: 22 June 2021 / Published: 28 June 2021
(This article belongs to the Special Issue Reproduction, Fertility and Embryonic Development of Animals)

Abstract

:

Simple Summary

Canine sperm cryopreservation is a method commonly used in veterinary clinics and laboratories. The present article reviews the antioxidants used in canine sperm cryopreservation, the egg yolk alternatives used for preventing cross-contamination, and the application of mesenchymal stem cells (MSCs) and their derivatives in dog sperm cryopreservation.

Abstract

Sperm cryopreservation is a widely used assisted reproductive technology for canine species. The long-term storage of dog sperm is effective for the breeding of dogs living far apart, scheduling the time of artificial insemination that suits the female, and preventing diseases of the reproductive tract. However, spermatozoa functions are impaired during the freeze–thaw processes, which may decrease reproductive performance. Numerous attempts have been made to restore such impairments, including the use of cryoprotectants to prevent the damage caused by ice crystal formation, and supplementation of antioxidants to reduce reactive oxygen species generation due to osmotic stress during the procedure. Egg yolk derivatives, antioxidants, and, more recently, mesenchymal stem cells (MSCs) and their derivatives have been proposed in this research field. This review article will summarize the current literature available on the topic.

1. Introduction

The use of assisted reproductive technologies (ARTs) in canine species is becoming more frequent due to the possibility of reducing the omnipresent threat of canine reproductive disorders, such as brucellosis [1], venereal transmissible tumors [2]; overcoming the limitations related to the characteristics of the female reproductive cycles; shipping canine semen to other facilities; and enabling the breeding of dogs living far apart. Practitioners using ARTs in their clinics can schedule semen collection or artificial insemination (AI) according to the male’s general condition, the female’s estrus cycle or vaccination schedule, or a suitable parturition date. These options allow the clinicians and breeders to save both time and money, schedule a cesarean delivery if required, and avoid transmissible diseases. Dog sperm cryopreservation is a widely used ART in clinics. However, the post-thaw parameters of cryopreserved sperm are more variable and poorer than those of fresh sperm [3,4,5]. Therefore, a standard and an efficient cryopreservation protocol is warranted for canine species.
The damages caused by reactive oxygen species (ROS) and oxidative stress during the freezing process are the main obstacles to sperm cryopreservation [6,7]. Moreover, thermotropic phase transitions during cooling–thawing processes induce a reorganization of the lipid phase [8], with a subsequent loss of cholesterol and polyunsaturated fatty acids [9]. These modifications are also associated with an influx of intracellular calcium, which triggers protein phosphorylation and later sperm capacitation-like changes, also referred to as cryocapacitation [10,11]. To overcome this limitation, supplementation of various antioxidants in freezing extenders has been attempted for decades [12]. In addition, although egg yolk is the most widely used cryoprotectant for sperm, issues of cross-contamination and the manual separation of the egg yolk have been addressed [13,14]. To overcome these shortcomings, skim milk [15], Equex STM paste [16], low-density lipoproteins (LDLs) [17], and Soybean lecithin [13,18] have been proposed as alternatives to egg yolk in the cryopreservation buffer. Recently, mesenchymal stem cells (MSCs) and their derivatives have been used in sperm cryopreservation research [19]. In 1976, Friedenstein et al. isolated MSCs from bone marrow for the first time [20]. Since then, these cells have been used in clinical trials and proven useful for treating various conditions, such as arthritis, diabetes, cancer, and neurological disorders in humans [21]. Subsequently, the use of MSC derivatives in veterinary medicine has garnered much attention; however, their use in theriogenology remained limited compared to that in other clinical fields.
The present review discusses the current trends in cryoprotectants and the chemicals used in canine sperm cryopreservation, with a particular focus on antioxidant supplementation and egg yolk alternatives, and summarizes the innovative emerging technologies using MSCs and their derivatives.

2. Traditional Cryoprotectants Supplemented with Antioxidants

During cryopreservation, a sudden decrease in temperature induces deleterious changes in sperm function and morphology. This is due to the osmotic stress induced by ice crystal formation, which further promotes ROS production, membrane damage, and intracellular content leakage. To counteract these events, the supplementation of cryoprotectants and antioxidants is essential. In sperm cryopreservation, various chemicals have been tested for their antioxidant potential and proven useful in recovering optimal post-thaw sperm quality parameters in different species (Table 1). In canine sperm cryopreservation, effects of anti-oxidants such as butylated hydroxytoluene [22,23], taurine [24,25], vitamin E [24,26], and ascorbic acid [24,26,27] have been investigated and have similar effects on post-thaw sperm quality-related parameters. In this part, less known antioxidants will be reviewed, and their optimal concentrations and effects in sperm cryopreservation in dogs and other species will be compared (Table 1).

2.1. Astaxanthin

Astaxanthin is a red carotenoid used for its anti-cancer, antioxidant, anti-aging effects, and for its role in delaying or preventing degenerative conditions such as aging, eye diseases, or atherosclerosis [84,85]. In canine sperm cryopreservation, astaxanthin supplementation in the freezing media significantly enhanced the post-thaw quality-related sperm parameters [30]. Carotenoids are known for their antioxidant activity and are commonly taken as dietary supplements. However, the effects of dietary supplementation with carotenoids on sperm quality are variable and inconclusive in different species [86,87,88,89,90]. Astaxanthin exhibits the highest antioxidant activity among carotenoids [84], and its direct addition to the freezing media significantly protected the sperm parameters in dogs and other species as well (Table 1). Through these results, it can be concluded that carotenoids, especially astaxanthin, have a better protective effect on sperm parameters when added directly to the samples, than being included in the diet. Additionally, in boars, astaxanthin increased the sperm oocytes penetration ratio, in vitro fertilization (IVF) efficiency, and cleavage and blastocyst rates (Table 1). These results suggest that astaxanthin could be used to enhance the dog sperm fertilization rate, and more research using this chemical to enhance canine IVF is warranted.

2.2. Resveratrol

Resveratrol is a polyphenol found in plants, with powerful antioxidant effects. Dietary inclusion of resveratrol was associated with a higher oxidative stress resistance [91]. In canine sperm cryopreservation, supplementation of resveratrol in the freezing media improved the effects of cryopreservation [35]. Since in other species, resveratrol supplementation enhanced the antioxidant capacities (Table 1), resveratrol’s effects on canine sperm’s post-thaw parameters can be attributed to its antioxidant activity. Moreover, chromatin integrity was well preserved in the resveratrol-treated samples, which can enhance the male fertility potential [92]. However, its effect on oxidative stress and fertility in canine frozen–thawed sperm has not been assessed yet.

2.3. Quercetin

Quercetin is a flavonoid with anti-bacterial, anti-carcinogenic, anti-inflammatory, and antioxidant activities [93]. It has been used in canine sperm cryopreservation and protected the post-thaw quality-related kinematic parameters and fertility [39]. However, artificial insemination was used as a fertility assessment in this study, and an evaluation of in vitro fertilizing abilities is still needed to confirm quercetin’s effects on dog fertility [39]. Moreover, sperm viability, chromatin integrity, plasma membrane and acrosome integrity, oxidative stress levels, and mitochondrial activity were not assessed in this study. Therefore, more complete studies on its effect on canine sperm are needed.

2.4. Myoinositol

Myoinositol is an active form of inositol, which belongs to the vitamin B complex. It shows antioxidant properties and has been used to treat male infertility due to its enhancing effects on sperm quality [94,95]. Myoinositol is a component of the seminiferous tubule fluid, and it is involved in sperm maturation, capacitation, motility, mitochondrial function, viability, and acrosomal reaction [45,96]. It is also present in the seminal plasma, where it acts as an osmo-regulator [97]. In canine semen cryopreservation, the supplementation of myoinositol in the freezing medium significantly protected the post-thaw quality parameters from the effects of cryopreservation, including motility; linearity; straightness; amplitude of lateral head displacement; live sperm percentage; membrane, chromatin, and acrosome integrity; and mucus-penetrating ability in comparison with a control [42]. Therefore, myoinositol protects the sperm survival rate, hyperactivation, and kinematic parameters, making it a potent supplement for cryopreservation media due its enhancing effects on sperm fertilization ability. Additionally, at the gene level, myoinositol significantly decreased the expression of pro-apoptotic and mitochondrial ROS modulator genes and increased the expression of protamine and anti-apoptotic genes in sperm [42]. However, whether or not myoinositol had a real effect on sperm genes expressions is controversial, as mature sperm cells are presumed to have no translation machinery [98].

2.5. Curcumin

This polyphenol is the active ingredient in turmeric (Curcuma longa) and has been used for its therapeutical potential since ancient times [99]. Even though the protective and antioxidant effects of curcumin have been documented previously, its use in sperm cryopreservation is recent. In dogs, the effects of curcumin supplementation during sperm cryopreservation were evaluated recently [51]. In this study, the addition of curcumin significantly protected sperm the post-thaw kinematic parameters, DNA integrity, and oxidative defenses. Interestingly, the authors investigated NADPH oxidase 5 (NOX5) gene expression, which has been identified in human spermatocytes and spermatids [100] and has been linked to ROS production in human sperm but has not been studied in canine sperm yet. Curcumin supplementation resulted in low expression levels of NOX5 genes. However, as mentioned previously, cautions should be taken when discussing gene expression analysis results in spermatozoa.

2.6. Iodixanol

Traditionally, iodixanol is commonly used as a contrast medium [101]. Nowadays, it is also used in density-gradient centrifugation [102,103], and more recently for sperm cryopreservation [54,56]. In dogs, 1.5% iodixanol conserved post-thaw sperm motility and mucus-penetrating ability, relieved oxidative stress, and enhanced protamine-related gene expression in sperm [56]. Furthermore, it also reduced protamine deficiency levels, and the relative expression of pro-apoptotic and mitochondrial ROS modulator genes in sperm [56]. When incubated in a canine capacitation medium supplemented with iodixanol, the sperm viability and acrosome integrity were significantly higher than the control group [56]. The supplementation of a canine capacitation medium with iodixanol significantly improved the sperm viability and the acrosome integrity in comparison with the control. Overall, iodixanol protects the general post-thaw sperm kinematics and sperm during capacitation and can thus be used in canine sperm capacitation media.

2.7. Spermine

The seminal plasma is an important fraction of the semen, which is involved in sperm regulation, homeostasis, and ROS modulation [104]. Spermine is a polyamine present in the seminal plasma, which exerts protective effects on cells [105]. Despite its antioxidant properties, however, the sperm fraction is usually separated from the seminal plasma during cryopreservation. Consequently, spermine is withdrawn from the sperm [106]. This protein is specific to human and rat seminal plasma [107]. However, the addition of spermine to the canine sperm cryopreservation medium could regulate oxidative stress, conserve sperm motility while maintaining membrane integrity, and preventing apoptosis-like changes and the cryocapacitation of dog sperm [62]. In this study too, the gene expression of NOX5 and other genes associated with oxidative stress and apoptosis was lower when compared with the control group.

2.8. Kinetin

Kinetin, a member of the cytokinin family, exhibits immune and antioxidant potential [108]. However, studies reporting the use of kinetin as a supplement in ARTs are scarce. Kinetin is more commonly added to the embryo culture media for plants than for animals, and its use has been reported only once in porcine embryos [109]. In semen experiments, kinetin has been used once in canine sperm cryopreservation and once in ram sperm storage at a refrigerator temperature [65]. In dogs, kinetin could protect the sperm from oxidative stress and could significantly protect sperm kinematics, mitochondrial activity, and membrane and acrosome integrity [64]. Therefore, the use of kinetin as an antioxidant in sperm cryopreservation should be considered in future research.

2.9. Melatonin

This hormone is synthesized by the pineal gland and is involved in many biological and physiological processes [110]. Melatonin and its metabolites exert both direct and indirect antioxidant effects in cells. Melatonin is a potent antioxidant and free radical scavenger [110,111,112], and it is commonly used in ARTs [113,114,115] including sperm cryopreservation. The use of melatonin as an oral or in vitro supplement in semen experiments is well documented [116,117,118]. Its protective and beneficial effects on the sperm during cryopreservation have been confirmed in many species, including humans, stallions, bulls, rams, mice, pigs, rabbits [68,71,75,77,78,119,120], and, more recently, in dogs [69]. Melatonin alleviates oxidative stress and reduces ROS levels, plasma membrane lipid peroxidation, DNA fragmentation, and apoptosis-like changes [116]. In addition, it significantly protects post-thaw sperm motility, membrane and acrosome integrity, and mitochondrial activity [116]. In canine sperm cryopreservation, melatonin conserved the post-thaw sperm plasma membrane integrity, kinematic parameters, mitochondrial activity, and conception rates when compared to the control group [69]. However, more experiments with canine sperm are warranted to unveil the precise properties and actions of melatonin in this system.

2.10. Metformin

Despite being extracted from a toxic plant [121,122,123], metformin has been used to treat various disorders, including type 2 diabetes mellitus [124] and cancer [125]. In ART and reproductive biology, metformin has been used for its antioxidant and therapeutic properties [126]. However, it has only been used for sperm cryopreservation in mice [81] and canines [80]. In mouse sperm, metformin did not affect the post-thaw quality parameters, although a higher fertilization rate and lower abnormal zygote rate were recorded in the treatment groups [81]. In contrast, in dog sperm, the supplementation of cryopreservation medium with metformin protected the post-thaw quality markers and parameters, including motility and oxidative defense [80]. Subsequently, these beneficial effects of metformin on canine sperm parameters were proposed to be produced through the activation of the 5′ adenosine monophosphate-activated protein kinase (AMPK) pathway [80]. To date, that study remains the only one to demonstrate the beneficial effects of metformin on sperm kinematics.

2.11. Olive-Derived Antioxidants

Among olive derivatives, 3,4-dihydroxyphenyl glycol (DHPG) is a phenol isolated from olive oil waste, with powerful antioxidant properties [127]. Its use in sperm cryopreservation is still new as it has only been used in two species (Table 1). In canine sperm cryopreservation, the addition of DHPG significantly protected the frozen–thawed sperm kinematic parameters, and reduced DNA damage [82]. The activities of enzymatic antioxidants were also enhanced in the treated samples. However, these results are not in accordance with the ones found in ram post-thawed sperm when the same concentrations were used (Table 1), and more studies are needed to confirm these results, and evaluate the effects of DHPG on in vivo and in vitro fertility.

3. Egg Yolk Alternatives

The application of cryopreservation to store and preserve sperm cells requires the use of cryoprotectants. Glycerol, egg yolk, skim milk, and Equex (Sodium Dodecyl Sulfate-SDS) are the major cryoprotectants used in the current standardized protocols [128]. Typically, extenders used for cryopreservation can be prepared in laboratories or purchased from commercial suppliers. The extenders used for canine sperm require the addition of fresh egg yolk, as it is the most common external cryoprotectant used. However, the use of egg yolk is associated with potential cross-contamination, and the manual separation of egg yolk from albumen is difficult and time-consuming [129]. Therefore, an increasing number of recent studies have explored egg yolk alternatives and assessed their efficacy; however, effects of these alternatives on fertility rates have not yet been assessed.

3.1. Low-Density Lipoproteins

Egg yolk contains LDLs, which can prevent cholesterol efflux and lower tyrosine-containing protein phosphorylation, thereby inhibiting sperm capacitation [130]. The importance of LDL in sperm cryopreservation has been demonstrated, as it is the key component of egg yolk that is responsible for sperm protection [131,132]. In previous studies, 4–12% LDL in semen extenders for different species could achieve better results than the percentage of egg yolk typically used in extenders (20%) [17,130,133,134]. Several explanations and hypotheses regarding the mechanisms through which LDLs protect sperm have been proposed.
In canine sperm cryopreservation, 6% LDL in Tris–citric acid–fructose medium did not reduce the fertilization ability of spermatozoa [17]. Moreover, the use of 6% LDL instead of egg yolk successfully preserved the motility parameters and DNA, membrane, and acrosome integrity [17]. In another study, 6% LDL combined with 20 mmol/mL glutamine was more efficient in protecting sperm during cryopreservation than egg yolk, 6% LDL alone, or the Equex extender [16]. These results suggest that LDL addition to extenders is a promising alternative to the conventional use of egg yolk. However, LDL use is limited due to the complexity of its extraction and purification methods [135].

3.2. Egg Yolk Plasma

LDL represents the protective fraction of egg yolk, and a major portion of LDL is present in the plasma (approximately 85%) [129]. Liquefied or lyophilized egg yolk plasma can be used as a substitute for LDL in canine sperm cryopreservation. Unlike whole egg yolk, the plasma fraction can be sterilized using gamma irradiation. Egg yolk plasma has been proven efficient as a substitute for whole egg yolk in stallion sperm cryopreservation. Equine sperm cryopreservation medium kits that include egg yolk plasma are already being commercialized [129]. In canine sperm, egg yolk plasma was more efficient in preserving the post-thaw quality parameters than whole egg yolk [136,137]. Egg yolk plasma well preserved motility, morphology, and membrane and acrosome integrity when the sperm were cooled for up to 10 h before cryopreservation [136]. However, the precise mechanism and extended effects of egg yolk plasma on canine sperm cryopreservation remain unknown and warrant further research.

3.3. Soybean Lecithin

Lecithin contains 65% phospholipids and triglycerides and is a component of egg yolk and soybeans [138]. The lecithin extracted from soybean oil has traditionally been used in food and non-food products as a substitute for animal fat [138,139]. It can act as a thickener, emulsifier, or crystallization control agent [139]. Soybean lecithin has been proposed as an alternative to egg yolk and its derivatives in sperm freezing media, and many new extenders containing lecithin have been developed and used in different species, including rams, sheep, bovids, equines, boars, and humans [140,141,142,143,144,145,146]. Indeed, lecithin could shield sperm from physical damage by forming a protective layer, preventing ice crystal formation, and replacing sperm membrane phospholipids [147].
In dog sperm cryopreservation, high concentrations of lecithin (1% and 2%) showed no positive effects on the post-thaw quality parameters in comparison with the egg yolk extender [148,149]. When supplemented with 0.04% lecithin without Equex, the frozen–thawed canine sperm parameters were similar to the ones obtained with a Tris–egg yolk–Equex extender [150]. In other mammals, effective lecithin concentrations ranged between 0.08 and 6% [145,147,151,152]. At concentrations of 0.01, 0.05, or 0.1%, lecithin showed a similar protective ability to egg yolk for canine sperm [18]. Moreover, egg yolk exacerbated the lipid peroxidation, consequently increasing the stress susceptibility of the frozen samples [18,153]. Therefore, lecithin offers more advantages than the conventional whole egg yolk. In a previous study, however, the higher lecithin concentrations used (0.05–0.1%) were detrimental to the post-thaw quality parameters of sperm [18]. This confirms that low concentrations of lecithin should be used for canine sperm cryopreservation.

3.4. Skim Milk

A skim milk extender is commonly used in mouse [154] and goat [155] sperm cryopreservation. It is free of lipids, and its protective constituent, casein micelles, reduces lipid loss and reduces the binding of seminal plasma proteins to sperm [132,156]. The main advantages of using skim milk-based extenders in comparison with egg yolk and its derivatives, are its simple storage conditions, cheap costs, availability, and easy acquisition. It has been reported as a good alternative to egg yolk in canine sperm cryopreservation and showed no adverse effect on in vivo fertility [15,157]. It was also hypothesized that the fertility of spermatozoa frozen with skim milk might be maintained longer than those frozen with egg yolk [15]. However, in vitro fertility assays should be performed to confirm these results.

3.5. Polyvinyl Alcohol

Polyvinyl alcohol (PVA) is a synthetic polymer with ice recrystallization inhibition properties in pure water, at concentrations as low as 1 mg/mL [158]. In an egg-yolk-free extender supplemented with PVA, the frozen–thawed canine sperm total motility, progressive motility, and acrosome integrity were protected [159]. However, the control group used in this study was an egg-yolk-free extender containing only glycerol as a cryoprotectant. More studies using egg-yolk-based extenders as a control group, and other conventional non-penetrant cryoprotectants, should be conducted. Moreover, sperm motility has been assessed by visual evaluation only, which does not give a full screening of the sperm kinematic parameters. Therefore, further evaluations of PVA effects on canine sperm cryopreservation are needed.

4. Cryoprotectants Supplemented with MSCs or Their Derivatives

MSCs exert their therapeutic effects through diverse mechanisms, which have been widely studied [160,161]. They can release factors involved in oxidative stress defense, apoptosis, cell survival, and cell metabolism [162]. They act through different pathways, such as the PI3K/Akt pathway that enhances MSCs survival ability [163], by activating ERK1/2 that promotes cell proliferation [164], or by suppressing the p38MAPK pathway to enhance oxidative defenses [164].
Since their discovery, MSCs have been extensively studied, characterized, and used in different clinical and experimental studies. The therapeutic potential of MSCs and their derivatives has been demonstrated in multiple studies, but their use in ARTs is still new and warrants further investigation to unveil their full potential (Table 2). Recently, an increasing number of studies have demonstrated the utility of MSCs as well as their conditioned media (CM) and extracellular vesicles in reproductive medicine and biotechnology, such as fertility recovery trials [165,166,167,168,169], sperm cryopreservation [19,170,171,172,173,174], oocytes in vitro maturation [175,176,177,178], and embryo production and maturation [167,177]. In canine sperm cryopreservation, the following two types of MSC have been used: adipose-derived MSCs (Ad-MSCs) [170,171] and amniotic-membrane-derived MSCs (AMSCs) [172,173].

4.1. Ad-MSCs Supplementation

In a recent study, Ad-MSCs were directly added to an egg yolk–Tris extender for the first time [171]; the results indicated that Ad-MSCs could preserve post-thaw viability; motility; and plasma membrane, chromatin, and acrosome integrity. Additionally, as a potential mechanism underlying this effect, the authors hypothesized that Ad-MSCs secrete repair factors in the medium to protect sperm cells from cryodamage [171], as the expression of genes related to membrane and chromatin repair was significantly enhanced when the sperm were treated with an extender supplemented with Ad-MSCs. These results confirm that Ad-MSCs can significantly enhance post-thaw sperm quality. However, the precise mechanisms involved remain unknown.
Of note is the fact that the main limiting factor of that study was the presence of cells other than sperm cells in the straws. Before analysis or use for insemination, the samples must be thoroughly washed; however, the risk of the contamination of sperm with Ad-MSCs remains high, and a new separation method is required. Furthermore, the packing proportion must be taken into account, as the sperm concentration in the straw is an important aspect of cryopreservation. The typical sperm concentrations used a range between 100 × 106 and 200 × 106 cells/mL, and this range is suitable for canine sperm cryopreservation protocols [181,182,183]. However, the addition of live cells to the medium alters the cell density, concentration, and nutrient availability. Indeed, in a previous study, the addition of high concentrations of Ad-MSCs (5 × 106 cells/mL) increased the sperm cell damage and decreased the viability and kinematics [171]. A higher cell density promotes the production of ROS [184] and release of pro-apoptotic factors [185,186], producing negative effects on cell recovery [186,187].

4.2. Ad-MSCs Derivatives Supplementation

A good alternative to the use of stem cells-based therapies, is the use of their derivatives (conditioned medium or extracellular vesicles). Recently, several MSC derivatives have been extracted, characterized, and processed for use as alternatives to stem cells in various therapies [188]. The major advantages of these alternatives are that, compared with stem cells, their derivatives present greater innocuity, longer viability, and easier manipulation and storage, and also lack teratogenic potential [189,190,191,192].
Exosomes are single-membrane extracellular vesicles secreted by cells, with diameters ranging from 30 to 200 nm [193,194]. They transduce cellular messages to the neighboring cells and tissues and contain a broad array of proteins, RNAs, lipids, and DNA [194]. In sperm cryopreservation, the use of exosomes is advantageous because they do not cause an increase in cell density, which is detrimental, unlike the use of whole MSCs [171]. Exosomes derived from Ad-MSCs have been used in canine sperm cryopreservation and shown to successfully maintain high post-thaw quality [170]. The addition of 50 µg/mL exosomes to the freezing extenders positively affected sperm motility, progressive motility, acrosome and membrane integrity, and viability [170]. These protective effects were likely produced by the action of exosomal mRNAs and proteins. With the recent published data about extracellular vesicles’ uptake by spermatozoa [195,196], more research using them in sperm cryopreservation is expected. Nonetheless, further studies are warranted to evaluate the precise mechanism of action of exosomes, and their effects on fertility.

4.3. Supplementation of AMSCs Derivatives

Amniotic-membrane-derived MSC derivatives have also been used in canine sperm cryopreservation [172,173]. A CM derived from AMSCs (AMSC-CM) has been characterized and used at different concentrations in the freezing medium [173]. CM is obtained through stem cell starvation, during which the cells release important paracrine factors involved in cell protection [197,198,199]. Even though it contains extracellular vesicles, the use of CM itself is more advantageous as it is less expensive to obtain, and the current extracellular vesicle extraction methods do not offer a satisfactory yield [200]. Therefore, CMs have been used in many clinical trials [201]. In canine sperm cryopreservation, 10% AMSC-CM was sufficient to recover the post-thaw sperm functions, and sperm motility, membrane integrity, mitochondrial function, and viability were enhanced in the treated groups. Moreover, some of the 86 proteins present in the AMSC-CM were involved in pathways related to various processes, including cell repair, sperm motility and metabolism, and cell defense [202,203,204,205,206,207]. Therefore, MSC-CM addition to the cryopreservation extenders is safe and can produce positive post-thaw outcomes. However, as there are no in vivo and in vitro fertility assessments using MSC-CM, more studies need to be conducted to evaluate the effects of MSC-CM on fertility.
Extracellular vesicles, and more precisely exosomes, have been extracted from AMSC-CM and used in canine sperm cryopreservation [172]. However, only low concentrations, ranging from 0 to 2 µg/mL, have been tested, with no effects observed [172]. Further studies using higher concentrations, similar to those of Ad-MSC exosomes [170], combined with the characterization of AMSC-derived exosomes are warranted.

5. Conclusions

Numerous studies have attempted to address the deleterious effects of sperm cryopreservation, including post-thaw low motility, viability, and fertility, due to the damage caused during the freeze–thawing procedures. As oxidative stress is one of the main results of osmotic changes, various antioxidants have been used in canine sperm cryopreservation. Despite the associated risk of cross-contamination, the conventional methods for canine sperm cryopreservation have used whole egg yolk for years. To avoid the risks associated with the use of whole egg yolk, different substitutes, including egg yolk derivatives and soybean lecithin, have been investigated. However, the extraction methods used to obtain these chemicals are complex and expensive, limiting their clinical application. Recently, the effects of MSCs and their derivatives on canine sperm have been evaluated, and they have proven promising as freezing extender supplements. Given their wide therapeutic effects, MSCs or their derivatives warrant further research for application in sperm cryopreservation, with particular focus on finding ways to overcome the current limitations associated with their use and elucidate the precise underlying mechanisms of their effects. In addition, although the currently available data are promising and interesting, many further experiments, specifically in vivo assays, are imperative to confirm the actual clinical effects of these chemicals on fertility.

Author Contributions

Conceptualization, F.Y.M. and M.-J.K.; methodology, F.Y.M. and M.-J.K.; validation, M.-J.K.; investigation, F.Y.M.; resources, M.-J.K.; writing—original draft preparation, F.Y.M.; writing—review and editing, F.Y.M. and M.-J.K.; visualization, F.Y.M. and M.-J.K.; supervision, M.-J.K.; project administration, M.-J.K.; funding acquisition, M.-J.K. All authors have read and agreed to the published version of the manuscript.

Funding

Cooperative research program of Rural Development Administration (#PJ014786012021).

Institutional Review Board Statement

Not applicable.

Acknowledgments

The authors would like to thank Heekee Park for his assistance.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Santos, R.L.; Souza, T.D.; Mol, J.P.S.; Eckstein, C.; Paixao, T.A. Canine Brucellosis: An Update. Front. Vet. Sci. 2021, 8, 594291. [Google Scholar] [CrossRef] [PubMed]
  2. Abeka, Y.T. Review on Canine Transmissible Venereal Tumor (CTVT). Cancer Ther. Oncol. Int. J. 2019, 14, 86–94. [Google Scholar]
  3. Prinosilova, P.; Rybar, R.; Zajicova, A.; Hlavicova, J. DNA integrity in fresh, chilled and frozen-thawed canine spermatozoa. Vet. Med. Czech. 2012, 57, 133–142. [Google Scholar] [CrossRef] [Green Version]
  4. Sicherle, C.C.; de Souza, F.F.; Freitas-Dell’Aqua, C.P.; Mothe, G.B.; Padovani, C.R.; Papa, F.O.; Lopes, M.D. Effects of the cryopreservation process on dog sperm integrity. Anim. Reprod. 2020, 17, e20190081. [Google Scholar] [CrossRef] [Green Version]
  5. Núñez-Martínez, I.; Moran, J.; Peña, F. A three-step statistical procedure to identify sperm kinematic subpopulations in canine ejaculates: Changes after cryopreservation. Reprod. Domest. Anim. 2006, 41, 408–415. [Google Scholar] [CrossRef] [PubMed]
  6. PERROS, C. Oxidative stress challenges during the sperm cryopreservation in dogs. J. Vet. Androl. 2017, 2, 1–7. [Google Scholar]
  7. Tatone, C.; Di Emidio, G.; Vento, M.; Ciriminna, R.; Artini, P.G. Cryopreservation and oxidative stress in reproductive cells. Gynecol. Endocrinol. 2010, 26, 563–567. [Google Scholar] [CrossRef]
  8. Drobnis, E.Z.; Crowe, L.M.; Berger, T.; Anchordoguy, T.J.; Overstreet, J.W.; Crowe, J.H. Cold Shock Damage Is Due to Lipid Phase-Transitions in Cell-Membranes—A Demonstration Using Sperm as a Model. J. Exp. Zool. 1993, 265, 432–437. [Google Scholar] [CrossRef]
  9. Maldjian, A.; Pizzi, F.; Gliozzi, T.; Cerolini, S.; Penny, P.; Noble, R. Changes in sperm quality and lipid composition during cryopreservation of boar semen. Theriogenology 2005, 63, 411–421. [Google Scholar] [CrossRef]
  10. Singh, V.K.; Atreja, S.K.; Kumar, R.; Chhillar, S.; Singh, A.K. Assessment of intracellular Ca2+, cAMP and 1,2-diacylglycerol in cryopreserved buffalo (Bubalus bubalis) spermatozoa on supplementation of taurine and trehalose in the extender. Reprod. Domest. Anim. 2012, 47, 584–590. [Google Scholar] [CrossRef]
  11. Kumar, R.; Atreja, S.K. Effect of incorporation of additives in tris-based egg yolk extender on buffalo (Bubalus bubalis) sperm tyrosine phosphorylation during cryopreservation. Reprod. Domest. Anim. 2012, 47, 485–490. [Google Scholar] [CrossRef]
  12. Majzoub, A.; Agarwal, A. Antioxidants in sperm cryopreservation. In Male Infertility; Springer: Berlin/Heidelberg, Germany, 2020; pp. 671–678. [Google Scholar]
  13. Sanchez-Calabuig, M.J.; Maillo, V.; Beltran-Brena, P.; Martinez, J.D.; Calera-Carrillo, S.; Perez-Gutierrez, J.F.; Perez-Cerezales, S. Cryopreservation of canine sperm using egg yolk and soy bean based extenders. Reprod. Biol. 2017, 17, 233–238. [Google Scholar] [CrossRef] [PubMed]
  14. Bousseau, S.; Brillard, J.P.; Marquant-Le Guienne, B.; Guerin, B.; Camus, A.; Lechat, M. Comparison of bacteriological qualities of various egg yolk sources and the in vitro and in vivo fertilizing potential of bovine semen frozen in egg yolk or lecithin based diluents. Theriogenology 1998, 50, 699–706. [Google Scholar] [CrossRef]
  15. Abe, Y.; Yokozawa, S.; Umemiya-Shirafuji, R.; Moumouni, P.; Suwa, Y.; Suzuki, H. Fertilizing ability of canine spermatozoa cryopreserved with skim milk-based extender in a retrospective study. Reprod. Domest. Anim. 2018, 53, 237–242. [Google Scholar] [CrossRef]
  16. Bencharif, D.; Amirat-Briand, L.; Garand, A.; Anton, M.; Schmitt, E.; Desherces, S.; Delhomme, G.; Langlois, M.-L.; Barrière, P.; Destrumelle, S. The advantages of using a combination of LDL and glutamine in comparison with TRIS egg yolk and Equex® STAMP extenders in the cryopreservation of canine semen. Res. Vet. Sci. 2012, 93, 440–447. [Google Scholar] [CrossRef]
  17. Bencharif, D.; Amirat, L.; Anton, M.; Schmitt, E.; Desherces, S.; Delhomme, G.; Langlois, M.L.; Barriere, P.; Larrat, M.; Tainturier, D. The advantages of LDL (low density lipoproteins) in the cryopreservation of canine semen. Theriogenology 2008, 70, 1478–1488. [Google Scholar] [CrossRef]
  18. Dalmazzo, A.; Losano, J.D.A.; Rocha, C.C.; Tsunoda, R.H.; Angrimani, D.S.R.; Mendes, C.M.; Assumpcao, M.; Nichi, M.; Barnabe, V.H. Effects of Soy Lecithin Extender on Dog Sperm Cryopreservation. Anim. Biotechnol. 2018, 29, 174–182. [Google Scholar] [CrossRef]
  19. Saadeldin, I.M.; Khalil, W.A.; Alharbi, M.G.; Lee, S.H. The Current Trends in Using Nanoparticles, Liposomes, and Exosomes for Semen Cryopreservation. Animals 2020, 10, 2281. [Google Scholar] [CrossRef]
  20. Friedenstein, A.J.; Gorskaja, J.F.; Kulagina, N.N. Fibroblast precursors in normal and irradiated mouse hematopoietic organs. Exp. Hematol. 1976, 4, 267–274. [Google Scholar]
  21. Hmadcha, A.; Martin-Montalvo, A.; Gauthier, B.R.; Soria, B.; Capilla-Gonzalez, V. Therapeutic Potential of Mesenchymal Stem Cells for Cancer Therapy. Front. Bioeng. Biotechnol. 2020, 8, 43. [Google Scholar] [CrossRef] [PubMed]
  22. Sun, L.; Wu, C.; Xu, J.; Zhang, S.; Dai, J.; Zhang, D. Addition of butylated hydroxytoluene (BHT) in tris-based extender improves post-thaw quality and motion dynamics of dog spermatozoa. Cryobiology 2020, 97, 71–75. [Google Scholar] [CrossRef] [PubMed]
  23. Neagu, V.R.; Garcia, B.M.; Sandoval, C.S.; Rodriguez, A.M.; Ferrusola, C.O.; Fernandez, L.G.; Tapia, J.A.; Pena, F.J. Freezing dog semen in presence of the antioxidant butylated hydroxytoluene improves postthaw sperm membrane integrity. Theriogenology 2010, 73, 645–650. [Google Scholar] [CrossRef] [PubMed]
  24. Michael, A.; Alexopoulos, C.; Pontiki, E.; Hadjipavlou-Litina, D.; Saratsis, P.; Boscos, C. Effect of antioxidant supplementation on semen quality and reactive oxygen species of frozen-thawed canine spermatozoa. Theriogenology 2007, 68, 204–212. [Google Scholar] [CrossRef] [PubMed]
  25. Martins-Bessa, A.; Rocha, A.; Mayenco-Aguirre, A. Effects of taurine and hypotaurine supplementation and ionophore concentrations on post-thaw acrosome reaction of dog spermatozoa. Theriogenology 2009, 71, 248–253. [Google Scholar] [CrossRef] [PubMed]
  26. Lecewicz, M.; Strzezek, R.; Kordan, W.; Majewska, A. Effect of Extender Supplementation with Low-molecular-weight Antioxidants on Selected Quality Parameters of Cryopreserved Canine Spermatozoa. J. Vet. Res. 2018, 62, 221–227. [Google Scholar] [CrossRef] [Green Version]
  27. Monteiro, J.C.; Goncalves, J.S.; Rodrigues, J.A.; Lucio, C.F.; Silva, L.C.; Assumpcao, M.E.; Vannucchi, C.I. Influence of ascorbic acid and glutathione antioxidants on frozen-thawed canine semen. Reprod. Domest. Anim. 2009, 44 (Suppl. 2), 359–362. [Google Scholar] [CrossRef] [PubMed]
  28. Guo, H.T.; Wang, J.R.; Sun, L.Z.; Jin, X.H.; Shi, X.Y.; Lin, J.Y.; Yue, S.L.; Zhou, J.B. Effects of astaxanthin on plasma membrane function and fertility of boar sperm during cryopreservation. Theriogenology 2021, 164, 58–64. [Google Scholar] [CrossRef]
  29. Basioura, A.; Tsakmakidis, I.A.; Martinez, E.A.; Roca, J.; Li, J.; Molina, M.F.; Theodoridis, A.; Boscos, C.M.; Parrilla, I. Effect of astaxanthin in extenders on sperm quality and functional variables of frozen-thawed boar semen. Anim. Reprod. Sci. 2020, 218, 106478. [Google Scholar] [CrossRef] [PubMed]
  30. Qamar, A.Y.; Fang, X.; Bang, S.; Shin, S.T.; Cho, J. The effect of astaxanthin supplementation on the post-thaw quality of dog semen. Reprod. Domest. Anim. 2020, 55, 1163–1171. [Google Scholar] [CrossRef]
  31. Lee, E.; Kim, D. Effects of Astaxanthin on Miniature Pig Sperm Cryopreservation. Biomed. Res. Int. 2018, 2018, 6784591. [Google Scholar] [CrossRef]
  32. Abdi-Benemar, H.; Khalili, B.; Zamiri, M.J.; Ezazi, H.; Ardabili, G.S.; Moghadam, S.H.; Simanoor, N. Effects of astaxanthin supplementation on the freezability, lipid peroxidation, antioxidant enzyme activities and post-thawing fertility of ram semen. Small Rumin. Res. 2020, 192, 106213. [Google Scholar] [CrossRef]
  33. Zhu, Z.; Li, R.; Fan, X.; Lv, Y.; Zheng, Y.; Hoque, S.; Wu, D.; Zeng, W. Resveratrol Improves Boar Sperm Quality via 5AMP-Activated Protein Kinase Activation during Cryopreservation. Oxidative Med. Cell. Longev. 2019, 2019, 5921503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Longobardi, V.; Zullo, G.; Salzano, A.; De Canditiis, C.; Cammarano, A.; De Luise, L.; Puzio, M.V.; Neglia, G.; Gasparrini, B. Resveratrol prevents capacitation-like changes and improves in vitro fertilizing capability of buffalo frozen-thawed sperm. Theriogenology 2017, 88, 1–8. [Google Scholar] [CrossRef] [PubMed]
  35. Bang, S.; Qamar, A.Y.; Tanga, B.M.; Fang, X.; Cho, J. Resveratrol supplementation into extender protects against cryodamage in dog post-thaw sperm. J. Vet. Med. Sci. 2021, 21-0125. [Google Scholar] [CrossRef]
  36. Nouri, H.; Shojaeian, K.; Samadian, F.; Lee, S.; Kohram, H.; Lee, J.I. Using Resveratrol and Epigallocatechin-3-Gallate to Improve Cryopreservation of Stallion Spermatozoa With Low Quality. J. Equine Vet. Sci. 2018, 70, 18–25. [Google Scholar] [CrossRef]
  37. Garcez, M.E.; dos Santos Branco, C.; Lara, L.V.; Pasqualotto, F.F.; Salvador, M. Effects of resveratrol supplementation on cryopreservation medium of human semen. Fertil. Steril. 2010, 94, 2118–2121. [Google Scholar] [CrossRef]
  38. Avdatek, F.; Yeni, D.; Inanc, M.E.; Cil, B.; Tuncer, B.P.; Turkmen, R.; Tasdemir, U. Supplementation of quercetin for advanced DNA integrity in bull semen cryopreservation. Andrologia 2018, 50, e12975. [Google Scholar] [CrossRef]
  39. Kawasaki, Y.; Sakurai, D.; Yoshihara, T.; Tsuchida, M.; Harakawa, S.; Suzuki, H. Effect of quercetin on the motility of cryopreserved canine spermatozoa. Cryobiology 2020, 96, 50–54. [Google Scholar] [CrossRef]
  40. Seifi-Jamadi, A.; Kohram, H.; Shahneh, A.Z.; Ansari, M.; Macias-Garcia, B. Quercetin Ameliorate Motility in Frozen-Thawed Turkmen Stallions Sperm. J. Equine Vet. Sci. 2016, 45, 73–77. [Google Scholar] [CrossRef]
  41. Zribi, N.; Chakroun, N.F.; Ben Abdallah, F.; Elleuch, H.; Sellami, A.; Gargouri, J.; Rebai, T.; Fakhfakh, F.; Keskes, L.A. Effect of freezing-thawing process and quercetin on human sperm survival and DNA integrity. Cryobiology 2012, 65, 326–331. [Google Scholar] [CrossRef]
  42. Qamar, A.Y.; Fang, X.; Kim, M.J.; Cho, J. Myoinositol Supplementation of Freezing Medium Improves the Quality-Related Parameters of Dog Sperm. Animals 2019, 9, 1038. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Affonso, F.J.; Carvalho, H.F.; Lanconi, R.; Lemes, K.M.; Leite, T.G.; Oliveira, L.Z.; Celeghini, E.C.C.; de Arruda, R.P. Addition of Antioxidants Myoinositol, Ferulic Acid, and Melatonin and Their Effects on Sperm Motility, Membrane Integrity, and Reactive Oxygen Species Production in Cooled Equine Semen. J. Equine Vet. Sci. 2017, 59, 57–63. [Google Scholar] [CrossRef]
  44. Condorelli, R.A.; La Vignera, S.; Bellanca, S.; Vicari, E.; Calogero, A.E.J.U. Myoinositol: Does it improve sperm mitochondrial function and sperm motility? Urology 2012, 79, 1290–1295. [Google Scholar] [CrossRef] [PubMed]
  45. Condorelli, R.A.; La Vignera, S.; Di Bari, F.; Unfer, V.; Calogero, A.E. Effects of myoinositol on sperm mitochondrial function in-vitro. Eur. Rev. Med. Pharmacol. Sci. 2011, 15, 129–134. [Google Scholar]
  46. Mohammadi, F.; Varanloo, N.; Heydari Nasrabadi, M.; Vatannejad, A.; Amjadi, F.S.; Javedani Masroor, M.; Bajelan, L.; Mehdizadeh, M.; Aflatoonian, R.; Zandieh, Z. Supplementation of sperm freezing medium with myoinositol improve human sperm parameters and protects it against DNA fragmentation and apoptosis. Cell Tissue Bank 2019, 20, 77–86. [Google Scholar] [CrossRef]
  47. Saleh, R.; Assaf, H.; Abd El Maged, W.M.; Elsuity, M.; Fawzy, M. Increased cryo-survival rate in ejaculated human sperm from infertile men following pre-freeze in vitro myo-inositol supplementation. Clin. Exp. Reprod. Med. 2018, 45, 177–182. [Google Scholar] [CrossRef]
  48. Kulaksız, R.; Bucak, M.N.; Akçay, E.; Sakin, F.; Daşkın, A.; Ateşşahin, A. The Effects of Different Extenders and Myo-Inositol on Post-thaw Quality of Ram Semen. Kafkas Univ. Vet. Fak. Derg. 2011, 17, 217–222. [Google Scholar]
  49. Chanapiwat, P.; Kaeoket, K.J.A.S.J. The effect of C urcuma longa extracted (curcumin) on the quality of cryopreserved boar semen. Anim. Sci. J. 2015, 86, 863–868. [Google Scholar]
  50. Bucak, M.N.; Baspinar, N.; Tuncer, P.B.; Coyan, K.; Sariozkan, S.; Akalin, P.P.; Buyukleblebici, S.; Kucukgunay, S. Effects of curcumin and dithioerythritol on frozen-thawed bovine semen. Andrologia 2012, 44 (Suppl. 1), 102–109. [Google Scholar] [CrossRef] [PubMed]
  51. Aparnak, P.; Saberivand, A. Effects of curcumin on canine semen parameters and expression of NOX5 gene in cryopreserved spermatozoa. In Veterinary Research Forum; Urmia University: Urmia, Iran, 2019; p. 221. [Google Scholar]
  52. Santonastaso, M.; Mottola, F.; Iovine, C.; Colacurci, N.; Rocco, L. Protective Effects of Curcumin on the Outcome of Cryopreservation in Human Sperm. Reprod. Sci. 2021, 1–11. [Google Scholar] [CrossRef]
  53. Chuawongboon, P.; Sirisathien, S.; Pongpeng, J.; Sakhong, D.; Nagai, T.; Vongpralub, T. Effects of supplementation of iodixanol to semen extender on quality and fertilization ability of frozen-thawed Thai native bull sperm. Anim. Sci. J. 2017, 88, 1311–1320. [Google Scholar] [CrossRef] [PubMed]
  54. Saragusty, J.; Gacitua, H.; Rozenboim, I.; Arav, A. Protective effects of iodixanol during bovine sperm cryopreservation. Theriogenology 2009, 71, 1425–1432. [Google Scholar] [CrossRef]
  55. Swami, D.S.; Kumar, P.; Malik, R.K.; Saini, M.; Kumar, D.; Jan, M.H. The cryoprotective effect of iodixanol in buffalo semen cryopreservation. Anim. Reprod. Sci. 2017, 179, 20–26. [Google Scholar] [CrossRef] [PubMed]
  56. Abdillah, D.A.; Setyawan, E.M.N.; Oh, H.J.; Ra, K.; Lee, S.H.; Kim, M.J.; Lee, B.C. Iodixanol supplementation during sperm cryopreservation improves protamine level and reduces reactive oxygen species of canine sperm. J. Vet. Sci. 2019, 20, 79–86. [Google Scholar] [CrossRef] [PubMed]
  57. Beehan, D.P. The Effects of Iodixanol Present during Equine Semen Cryopreservation. Master’s Thesis, Louisiana State University and Agricultural and Mechanical College, Baton Rouge, LA, USA, 2012. [Google Scholar]
  58. Kim, S.; Hooper, S.; Agca, C.; Agca, Y. Post-thaw ATP supplementation enhances cryoprotective effect of iodixanol in rat spermatozoa. Reprod. Biol. Endocrinol. 2016, 14, 5. [Google Scholar] [CrossRef] [Green Version]
  59. Cirit, U.; Bagis, H.; Demir, K.; Agca, C.; Pabuccuoglu, S.; Varisli, O.; Clifford-Rathert, C.; Agca, Y. Comparison of cryoprotective effects of iodixanol, trehalose and cysteamine on ram semen. Anim. Reprod. Sci. 2013, 139, 38–44. [Google Scholar] [CrossRef]
  60. Ozmen, M.F.; Cirit, U.; Arici, R.; Demir, K.; Kurt, D.; Pabuccuoglu, S.; Ak, K. Evaluation of synergic effects of iodixanol and trehalose on cryosurvival of electroejaculated ram semen. Andrologia 2020, 52, e13656. [Google Scholar] [CrossRef] [PubMed]
  61. Siddique, R.A.; Atreja, S.K. Effect of L-Arginine and spermine-NONOate on motility, viability, membrane integrity and lipid peroxidation of Murrah buffalo (Bubalus bubalis) spermatozoa. Livest. Sci. 2013, 153, 147–153. [Google Scholar] [CrossRef]
  62. Setyawan, E.M.N.; Kim, M.J.; Oh, H.J.; Kim, G.A.; Jo, Y.K.; Lee, S.H.; Choi, Y.B.; Lee, B.C. Spermine reduces reactive oxygen species levels and decreases cryocapacitation in canine sperm cryopreservation. Biochem. Biophys. Res. Commun. 2016, 479, 927–932. [Google Scholar] [CrossRef] [PubMed]
  63. Olğaç, K.T.; Akçay, E. Effects of Spermine and Spermidine supplemented extenders on post-thaw Spermatological Parameters in Stallion Semen Cryopreservation. Cryobiology 2021, 100, 72–76. [Google Scholar] [CrossRef]
  64. Qamar, A.Y.; Fang, X.; Bang, S.; Kim, M.J.; Cho, J. Effects of kinetin supplementation on the post-thaw motility, viability, and structural integrity of dog sperm. Cryobiology 2020, 95, 90–96. [Google Scholar] [CrossRef]
  65. Hashem, E.Z.; Eslami, M. Kinetin improves motility, viability and antioxidative parameters of ram semen during storage at refrigerator temperature. Cell Tissue Bank. 2018, 19, 97–111. [Google Scholar] [CrossRef]
  66. Inyawilert, W.; Rungruangsak, J.; Liao, Y.J.; Tang, P.C.; Paungsukpaibool, V. Melatonin supplementation improved cryopreserved Thai swamp buffalo semen. Reprod. Domest. Anim. 2021, 56, 83–88. [Google Scholar] [CrossRef] [PubMed]
  67. Ashrafi, I.; Kohram, H.; Ardabili, F.F. Antioxidative effects of melatonin on kinetics, microscopic and oxidative parameters of cryopreserved bull spermatozoa. Anim. Reprod. Sci. 2013, 139, 25–30. [Google Scholar] [CrossRef] [PubMed]
  68. ChaithraShree, A.R.; Ingole, S.D.; Dighe, V.D.; Nagvekar, A.S.; Bharucha, S.V.; Dagli, N.R.; Kekan, P.M.; Kharde, S.D. Effect of melatonin on bovine sperm characteristics and ultrastructure changes following cryopreservation. Vet. Med. Sci. 2020, 6, 177–186. [Google Scholar] [CrossRef]
  69. Martinez-Rodriguez, J.A.; Carbajal, F.J.; Martinez-De-Anda, R.; Alcantar-Rodriguez, A.; Medrano, A. Melatonin added to freezing diluent improves canine (Bulldog) sperm cryosurvival. J. Reprod. 2020, 1, 11–19. [Google Scholar] [CrossRef]
  70. Mehaisen, G.M.K.; Partyka, A.; Ligocka, Z.; Nizanski, W. Cryoprotective effect of melatonin supplementation on post-thawed rooster sperm quality. Anim. Reprod. Sci. 2020, 212, 106238. [Google Scholar] [CrossRef]
  71. Lanconi, R.; Celeghini, E.C.C.; Alves, M.B.R.; Lemes, K.M.; Gonella-Diaza, A.M.; Oliveira, L.Z.; de Arruda, R.P. Melatonin Added to Cryopreservation Extenders Improves the Mitochondrial Membrane Potential of Postthawed Equine Sperm. J. Equine Vet. Sci. 2018, 69, 78–83. [Google Scholar] [CrossRef]
  72. Balao da Silva, C.M.; Macías-García, B.; Miró-Morán, A.; González-Fernández, L.; Morillo-Rodriguez, A.; Ortega-Ferrusola, C.; Gallardo-Bolaños, J.M.; Stilwell, G.; Tapia, J.A.; Peña, F. Melatonin reduces lipid peroxidation and apoptotic-like changes in stallion spermatozoa. J. Pineal Res. 2011, 51, 172–179. [Google Scholar] [CrossRef]
  73. El-Battawy, K. Preservation of goat semen at 5 C with emphasis on its freezability and the impact of melatonin. Int. J. Vet. Sci. Res. 2019, 5, 035–038. [Google Scholar] [CrossRef] [Green Version]
  74. Najafi, A.; Adutwum, E.; Yari, A.; Salehi, E.; Mikaeili, S.; Dashtestani, F.; Abolhassani, F.; Rashki, L.; Shiasi, S.; Asadi, E. Melatonin affects membrane integrity, intracellular reactive oxygen species, caspase3 activity and AKT phosphorylation in frozen thawed human sperm. Cell Tissue Res. 2018, 372, 149–159. [Google Scholar] [CrossRef] [PubMed]
  75. Karimfar, M.H.; Niazvand, F.; Haghani, K.; Ghafourian, S.; Shirazi, R.; Bakhtiyari, S. The protective effects of melatonin against cryopreservation-induced oxidative stress in human sperm. Int. J. Immunopathol. Pharmacol. 2015, 28, 69–76. [Google Scholar] [CrossRef] [PubMed]
  76. Deng, S.L.; Sun, T.C.; Yu, K.; Wang, Z.P.; Zhang, B.L.; Zhang, Y.; Wang, X.X.; Lian, Z.X.; Liu, Y.X. Melatonin reduces oxidative damage and upregulates heat shock protein 90 expression in cryopreserved human semen. Free Radic. Biol. Med. 2017, 113, 347–354. [Google Scholar] [CrossRef] [PubMed]
  77. Chen, X.J.; Zhang, Y.; Jia, G.X.; Meng, Q.G.; Bunch, T.D.; Liu, G.S.; Zhu, S.E.; Xhou, G.B. Effect of melatonin supplementation on cryopreserved sperm quality in mouse. Cryo Lett. 2016, 37, 115–122. [Google Scholar]
  78. Zhu, Z.; Li, R.; Lv, Y.; Zeng, W. Melatonin protects rabbit spermatozoa from cryo-damage via decreasing oxidative stress. Cryobiology 2019, 88, 1–8. [Google Scholar] [CrossRef] [PubMed]
  79. Succu, S.; Berlinguer, F.; Pasciu, V.; Satta, V.; Leoni, G.G.; Naitana, S. Melatonin protects ram spermatozoa from cryopreservation injuries in a dose-dependent manner. J. Pineal Res. 2011, 50, 310–318. [Google Scholar] [CrossRef]
  80. Grandhaye, J.; Partyka, A.; Ligocka, Z.; Dudek, A.; Nizanski, W.; Jeanpierre, E.; Estienne, A.; Froment, P. Metformin Improves Quality of Post-Thaw Canine Semen. Animals 2020, 10, 287. [Google Scholar] [CrossRef] [Green Version]
  81. Bertoldo, M.J.; Guibert, E.; Tartarin, P.; Guillory, V.; Froment, P. Effect of metformin on the fertilizing ability of mouse spermatozoa. Cryobiology 2014, 68, 262–268. [Google Scholar] [CrossRef] [PubMed]
  82. Shakouri, N.; Soleimanzadeh, A.; Rakhshanpour, A.; Bucak, M.N. Antioxidant effects of supplementation of 3,4-dihydroxyphenyl glycol on sperm parameters and oxidative markers following cryopreservation in canine semen. Reprod. Domest. Anim. 2021. [Google Scholar] [CrossRef] [PubMed]
  83. Arando, A.; Delgado, J.V.; Fernandez-Prior, A.; Leon, J.M.; Bermudez-Oria, A.; Nogales, S.; Perez-Marin, C.C. Effect of different olive oil-derived antioxidants (hydroxytyrosol and 3,4-dihydroxyphenylglycol) on the quality of frozen-thawed ram sperm. Cryobiology 2019, 86, 33–39. [Google Scholar] [CrossRef]
  84. Naguib, Y.M. Antioxidant activities of astaxanthin and related carotenoids. J. Agric. Food Chem. 2000, 48, 1150–1154. [Google Scholar] [CrossRef]
  85. Bennedsen, M.; Wang, X.; Willén, R.; Wadström, T.; Andersen, L.P. Treatment of H. pylori infected mice with antioxidant astaxanthin reduces gastric inflammation, bacterial load and modulates cytokine release by splenocytes. Immunol. Lett. 2000, 70, 185–189. [Google Scholar] [CrossRef]
  86. Foo, Y.Z.; Rhodes, G.; Simmons, L.W. The carotenoid beta-carotene enhances facial color, attractiveness and perceived health, but not actual health, in humans. Behav. Ecol. 2017, 28, 570–578. [Google Scholar] [CrossRef]
  87. Almbro, M.; Dowling, D.K.; Simmons, L.W. Effects of vitamin E and beta-carotene on sperm competitiveness. Ecol. Lett. 2011, 14, 891–895. [Google Scholar] [CrossRef] [PubMed]
  88. Tizkar, B.; Kazemi, R.; Alipour, A.; Seidavi, A.; Naseralavi, G.; Ponce-Palafox, J.T. Effects of dietary supplementation with astaxanthin and β-carotene on the semen quality of goldfish (Carassius auratus). Theriogenology 2015, 84, 1111–1117. [Google Scholar] [CrossRef]
  89. Bahmanzadeh, M.; Vahidinia, A.; Mehdinejadiani, S.; Shokri, S.; Alizadeh, Z. Dietary supplementation with astaxanthin may ameliorate sperm parameters and DNA integrity in streptozotocin-induced diabetic rats. Clin. Exp. Reprod. Med. 2016, 43, 90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Keogh, L.M.; Byrne, P.G.; Silla, A.J. Effect of long-term dietary beta-carotene supplementation on sperm concentration and motility in an endangered amphibian. Anim. Reprod. Sci. 2018, 195, 259–265. [Google Scholar] [CrossRef]
  91. King, R.E.; Bomser, J.A.; Min, D.B. Bioactivity of resveratrol. Compr. Rev. Food Sci. Food Saf. 2006, 5, 65–70. [Google Scholar] [CrossRef]
  92. Erenpreiss, J.; Spano, M.; Erenpreisa, J.; Bungum, M.; Giwercman, A. Sperm chromatin structure and male fertility: Biological and clinical aspects. Asian J. Androl. 2006, 8, 11–29. [Google Scholar] [CrossRef] [PubMed]
  93. Ay, M.; Charli, A.; Jin, H.; Anantharam, V.; Kanthasamy, A.; Kanthasamy, A.G. Quercetin. In Nutraceuticals; Elsevier: Amsterdam, The Netherlands, 2021; pp. 749–755. [Google Scholar]
  94. Korosi, T.; Barta, C.; Rokob, K.; Torok, T. Physiological Intra-Cytoplasmic Sperm Injection (PICSI) outcomes after oral pretreatment and semen incubation with myo-inositol in oligoasthenoteratozoospermic men: Results from a prospective, randomized controlled trial. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 66–72. [Google Scholar]
  95. Palmieri, M.; Papale, P.; Della Ragione, A.; Quaranta, G.; Russo, G.; Russo, S. In Vitro Antioxidant Treatment of Semen Samples in Assisted Reproductive Technology: Effects of Myo-Inositol on Nemaspermic Parameters. Int. J. Endocrinol. 2016, 2016, 2839041. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Bevilacqua, A.; Carlomagno, G.; Gerli, S.; Montanino Oliva, M.; Devroey, P.; Lanzone, A.; Soulange, C.; Facchinetti, F.; Carlo Di Renzo, G.; Bizzarri, M.J.G.E. Results from the International Consensus Conference on myo-inositol and D-chiro-inositol in Obstetrics and Gynecology–assisted reproduction technology. Gynecol. Endocrinol. 2015, 31, 441–446. [Google Scholar] [CrossRef]
  97. Chauvin, T.R.; Griswold, M.D. Characterization of the expression and regulation of genes necessary for myo-inositol biosynthesis and transport in the seminiferous epithelium. Biol. Reprod. 2004, 70, 744–751. [Google Scholar] [CrossRef] [PubMed]
  98. Ren, X.; Chen, X.; Wang, Z.; Wang, D. Is transcription in sperm stationary or dynamic? J. Reprod. Dev. 2017, 2016–2093. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Hatcher, H.; Planalp, R.; Cho, J.; Torti, F.M.; Torti, S.V. Curcumin: From ancient medicine to current clinical trials. Cell. Mol. Life Sci. 2008, 65, 1631–1652. [Google Scholar] [CrossRef] [PubMed]
  100. Banfi, B.; Molnar, G.; Maturana, A.; Steger, K.; Hegedus, B.; Demaurex, N.; Krause, K.H. A Ca2+-activated NADPH oxidase in testis, spleen, and lymph nodes. J. Biol. Chem. 2001, 276, 37594–37601. [Google Scholar] [CrossRef] [Green Version]
  101. Spencer, C.M.; Goa, K.L. Iodixanol. A review of its pharmacodynamic and pharmacokinetic properties and diagnostic use as an x-ray contrast medium. Drugs 1996, 52, 899–927. [Google Scholar] [CrossRef]
  102. Kjeken, R.; Mousavi, S.A.; Brech, A.; Gjoen, T.; Berg, T. Fluid phase endocytosis of [I-125]iodixanol in rat liver parenchymal, endothelial and Kupffer cells. Cell Tissue Res. 2001, 304, 221–230. [Google Scholar] [CrossRef]
  103. Stuhtmann, G.; Oldenhof, H.; Peters, P.; Klewitz, J.; Martinsson, G.; Sieme, H. Iodixanol density gradient centrifugation for selecting stallion sperm for cold storage and cryopreservation. Anim. Reprod. Sci. 2012, 133, 184–190. [Google Scholar] [CrossRef]
  104. Rodríguez-Martínez, H.; Kvist, U.; Ernerudh, J.; Sanz, L.; Calvete, J.J. Seminal plasma proteins: What role do they play? Am. J. Reprod. Immunol. 2011, 66, 11–22. [Google Scholar] [CrossRef] [Green Version]
  105. Pegg, A.E. The function of spermine. IUBMB Life 2014, 66, 8–18. [Google Scholar] [CrossRef] [PubMed]
  106. Stănescu, M.; Bîrţoiu, A.I. Comparative Studies of Canine Semen Freezing Protocols; Bulletin of University: Cook County, IL, USA, 2010. [Google Scholar] [CrossRef]
  107. Mann, T.; Lutwak-Mann, C. Male Reproductive Function and Semen: Themes and Trends in Physiology, Biochemistry and Investigative Andrology; Springer Science & Business Media: London, UK, 2012. [Google Scholar] [CrossRef]
  108. Barciszewski, J.; Massino, F.; Clark, B.F. Kinetin—A multiactive molecule. J. Int. J. Biol. Macromol. 2007, 40, 182–192. [Google Scholar] [CrossRef]
  109. Won, C.; Park, S.K.; Cho, S.G.; Min, B.M.; Roh, S. Kinetin enhances in vitro development of parthenogenetic and nuclear transfer porcine embryos. Mol. Reprod. Dev. 2008, 75, 1701–1709. [Google Scholar] [CrossRef] [PubMed]
  110. Hacışevki, A.; Baba, B. An overview of melatonin as an antioxidant molecule: A biochemical approach. J. Melatonin Mol. Biol. Clin. Pharm. Approaches 2018, 59–85. [Google Scholar] [CrossRef] [Green Version]
  111. Marshall, K.A.; Reiter, R.J.; Poeggeler, B.; Aruoma, O.I.; Halliwell, B. Evaluation of the antioxidant activity of melatonin in vitro. Free Radic Biol. Med. 1996, 21, 307–315. [Google Scholar] [CrossRef]
  112. Wang, J.; Wang, X.; He, Y.; Jia, L.; Yang, C.S.; Reiter, R.J.; Zhang, J. Antioxidant and Pro-Oxidant Activities of Melatonin in the Presence of Copper and Polyphenols In Vitro and In Vivo. Cells 2019, 8, 903. [Google Scholar] [CrossRef] [Green Version]
  113. Zhao, X.M.; Wang, N.; Hao, H.S.; Li, C.Y.; Zhao, Y.H.; Yan, C.L.; Wang, H.Y.; Du, W.H.; Wang, D.; Liu, Y.; et al. Melatonin improves the fertilization capacity and developmental ability of bovine oocytes by regulating cytoplasmic maturation events. J. Pineal Res. 2018, 64, e12445. [Google Scholar] [CrossRef] [Green Version]
  114. Kim, E.H.; Ridlo, M.R.; Lee, B.C.; Kim, G.A. Melatonin-Nrf2 Signaling Activates Peroxisomal Activities in Porcine Cumulus Cell-Oocyte Complexes. Antioxidants 2020, 9, 1080. [Google Scholar] [CrossRef] [PubMed]
  115. Song, Y.; Wu, H.; Wang, X.; Haire, A.; Zhang, X.; Zhang, J.; Wu, Y.; Lian, Z.; Fu, J.; Liu, G.; et al. Melatonin improves the efficiency of super-ovulation and timed artificial insemination in sheep. PeerJ 2019, 7, e6750. [Google Scholar] [CrossRef]
  116. Medrano, A.; Contreras, C.F.B.; Herrera, F.M.; Alcantar-Rodriguez, A.M. Melatonin as an antioxidant preserving sperm from domestic animals. J. Asian Pacific J. Reprod. 2017, 6, 241. [Google Scholar] [CrossRef]
  117. Cebrián-Pérez, J.A.; Casao, A.; González-Arto, M.; dos Santos Hamilton, T.; Pérez-Pé, R.; Muiño-Blanco, T. Melatonin in sperm biology: Breaking paradigms. J. Reprod. Domest. Anim. 2014, 49, 11–21. [Google Scholar] [CrossRef] [PubMed]
  118. Luboshitzky, R.; Shen-Orr, Z.; Nave, R.; Lavi, S.; Lavie, P. Melatonin administration alters semen quality in healthy men. J. Androl. 2002, 23, 572–578. [Google Scholar] [PubMed]
  119. Fang, Y.; Zhao, C.; Xiang, H.; Zhao, X.; Zhong, R. Melatonin Inhibits Formation of Mitochondrial Permeability Transition Pores and Improves Oxidative Phosphorylation of Frozen-Thawed Ram Sperm. Front. Endocrinol. 2019, 10, 896. [Google Scholar] [CrossRef] [Green Version]
  120. Pezo, F.; Zambrano, F.; Uribe, P.; Moya, C.; de Andrade, A.F.C.; Risopatron, J.; Yeste, M.; Burgos, R.A.; Sanchez, R. Oxidative and nitrosative stress in frozen-thawed pig spermatozoa. I: Protective effect of melatonin and butylhydroxytoluene on sperm function. Res. Vet. Sci. 2021, 136, 143–150. [Google Scholar] [CrossRef] [PubMed]
  121. Puyt, J.D.; Faliu, L.; Keck, G.; Gedfrain, J.C.; Pinault, L.; Tainturier, D. Fatal poisoning of sheep by Galega officinalis (French honeysuckle). Vet. Hum. Toxicol. 1981, 23, 410–412. [Google Scholar]
  122. Keeler, R.F.; Baker, D.C.; Evans, J.O. Individual animal susceptibility and its relationship to induced adaptation or tolerance in sheep to Galega officinalis L. Vet. Hum. Toxicol. 1988, 30, 420–423. [Google Scholar]
  123. Rasekh, H.R.; Nazari, P.; Kamli-Nejad, M.; Hosseinzadeh, L. Acute and subchronic oral toxicity of Galega officinalis in rats. J. Ethnopharmacol. 2008, 116, 21–26. [Google Scholar] [CrossRef]
  124. Bailey, C.J.; Day, C. Metformin: Its botanical background. J. Pract. Diabetes Int. 2004, 21, 115–117. [Google Scholar] [CrossRef]
  125. Dowling, R.J.O.; Goodwin, P.J.; Stambolic, V. Understanding the benefit of metformin use in cancer treatment. BMC Med. 2011, 9, 1–6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Faure, M.; Bertoldo, M.J.; Khoueiry, R.; Bongrani, A.; Brion, F.; Giulivi, C.; Dupont, J.; Froment, P. Metformin in Reproductive Biology. Front. Endocrinol. 2018, 9, 675. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Rodriguez, G.; Rodriguez, R.; Fernandez-Bolanos, J.; Guillen, R.; Jimenez, A. Antioxidant activity of effluents during the purification of hydroxytyrosol and 3,4-dihydroxyphenyl glycol from olive oil waste. Eur. Food Res. Technol. 2007, 224, 733–741. [Google Scholar] [CrossRef]
  128. World Health Organization. WHO Laboratory Manual for the Examination and Processing of Human Semen; WHO: Geneva, Switzerland, 2010. [Google Scholar]
  129. Pillet, E.; Duchamp, G.; Batellier, F.; Beaumal, V.; Anton, M.; Desherces, S.; Schmitt, E.; Magistrini, M. Egg yolk plasma can replace egg yolk in stallion freezing extenders. Theriogenology 2011, 75, 105–114. [Google Scholar] [CrossRef]
  130. Dalal, J.; Chandolia, R.K.; Pawaria, S.; Kumar, A.; Kumar, D.; Selokar, N.L.; Andonissamy, J.; Yadav, P.S.; Kumar, P. Low-density lipoproteins protect sperm during cryopreservation in buffalo: Unraveling mechanism of action. Mol. Reprod. Dev. 2020, 87, 1231–1244. [Google Scholar] [CrossRef]
  131. Peruma, P. Low density lipoprotein in cryopreservation of semen. J. Asian Pacific J. Reprod. 2018, 7, 103. [Google Scholar] [CrossRef]
  132. Bergeron, A.; Manjunath, P. New insights towards understanding the mechanisms of sperm protection by egg yolk and milk. Mol. Reprod. Dev. 2006, 73, 1338–1344. [Google Scholar] [CrossRef] [PubMed]
  133. Iaffaldano, N.; Di Iorio, M.; Rosato, M.P.; Manchisi, A. Cryopreservation of rabbit semen using non-permeable cryoprotectants: Effectiveness of different concentrations of low-density lipoproteins (LDL) from egg yolk versus egg yolk or sucrose. Anim. Reprod. Sci. 2014, 151, 220–228. [Google Scholar] [CrossRef] [PubMed]
  134. Shahverdi, A.; Sharafi, M.; Gourabi, H.; Yekta, A.A.; Esmaeili, V.; Sharbatoghli, M.; Janzamin, E.; Hajnasrollahi, M.; Mostafayi, F. Fertility and flow cytometric evaluations of frozen-thawed rooster semen in cryopreservation medium containing low-density lipoprotein. Theriogenology 2015, 83, 78–85. [Google Scholar] [CrossRef] [PubMed]
  135. Moustacas, V.; Zaffalon, F.; Lagares, M.; Loaiza-Eccheverri, A.; Varago, F.; Neves, M.; Heneine, L.; Arruda, R.; Henry, M.J.T. Natural, but not lyophilized, low density lypoproteins were an acceptable alternative to egg yolk for cryopreservation of ram semen. Theriogenology 2011, 75, 300–307. [Google Scholar] [CrossRef]
  136. Corcini, C.D.; Goularte, K.L.; Bongalhardo, D.C.; Lucia, T., Jr.; Jardim, R.D.; Varela Junior, A.S. Effect of egg yolk plasma on dog sperm cryopreservation. Andrologia 2016, 48, 114–115. [Google Scholar] [CrossRef]
  137. Belala, R.; Briand-Amirat, L.; Martinot, A.; Thorin, C.; Michaud, S.; Desherces, S.; Youngs, C.R.; Bencharif, D. A comparison of liquid and lyophilized egg yolk plasma to low density lipoproteins for freezing of canine spermatozoa. Reprod. Domest. Anim. 2019, 54, 1131–1138. [Google Scholar] [CrossRef]
  138. Wu, Y.Z.; Wang, T. Soybean lecithin fractionation and functionality. J. Am. Oil Chem Soc. 2003, 80, 319–326. [Google Scholar] [CrossRef]
  139. List, G. Soybean lecithin: Food, industrial uses, and other applications. J. Polar Lipids 2015, 1–33. [Google Scholar] [CrossRef]
  140. Gil, J.; Rodriguez-Irazoqui, M.; Lundeheim, N.; Söderquist, L.; Rodríguez-Martínez, H. Fertility of ram semen frozen in Bioexcell® and used for cervical artificial insemination. Theriogenology 2003, 59, 1157–1170. [Google Scholar] [CrossRef]
  141. Fukui, Y.; Kohno, H.; Togari, T.; Hiwasa, M.; Okabe, K. Fertility after artificial insemination using a soybean-based semen extender in sheep. J. Reprod. Dev. 2008, 54, 286–289. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Forouzanfar, M.; Sharafi, M.; Hosseini, S.M.; Ostadhosseini, S.; Hajian, M.; Hosseini, L.; Abedi, P.; Nili, N.; Rahmani, H.R.; Nasr-Esfahani, M.H. In vitro comparison of egg yolk-based and soybean lecithin-based extenders for cryopreservation of ram semen. Theriogenology 2010, 73, 480–487. [Google Scholar] [CrossRef]
  143. Aires, V.A.; 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. [Google Scholar] [CrossRef]
  144. Papa, F.O.; Felicio, G.B.; Melo-Ona, C.M.; Alvarenga, M.A.; De Vita, B.; Trinque, C.; Puoli-Filho, J.N.; Dell’Aqua, J.A., Jr. Replacing egg yolk with soybean lecithin in the cryopreservation of stallion semen. Anim. Reprod. Sci. 2011, 129, 73–77. [Google Scholar] [CrossRef]
  145. Zhang, S.S.; Hu, J.H.; Li, Q.W.; Jiang, Z.L.; Zhang, X.Y. The cryoprotective effects of soybean lecithin on boar spermatozoa quality. Afr. J. Biotechnol. 2009, 8, 6476–6480. [Google Scholar]
  146. Jeyendran, R.S.; Acosta, V.C.; Land, S.; Coulam, C.B. Cryopreservation of human sperm in a lecithin-supplemented freezing medium. Fertil. Steril. 2008, 90, 1263–1265. [Google Scholar] [CrossRef]
  147. Emamverdi, M.; Zhandi, M.; Zare Shahneh, A.; Sharafi, M.; Akbari-Sharif, A. Optimization of Ram semen cryopreservation using a chemically defined soybean lecithin-based extender. Reprod. Domest. Anim. 2013, 48, 899–904. [Google Scholar] [CrossRef]
  148. Axner, E.; Lagerson, E. Cryopreservation of Dog Semen in a Tris Extender with 1% or 2% Soya Bean Lecithin as a Replacement of Egg Yolk. Reprod. Domest. Anim. 2016, 51, 262–268. [Google Scholar] [CrossRef] [PubMed]
  149. Hermansson, U.; Johannisson, A.; Axner, E. Cryopreservation of dog semen in a Tris extender with two different 1% soybean preparations compared with a Tris egg yolk extender. Vet. Med. Sci. 2021, 7, 812–819. [Google Scholar] [CrossRef]
  150. Beccaglia, M.; Anastasi, P.; Luvoni, G.C. Freezing of canine semen in an animal-free protein extender. Vet. Res. Commun. 2009, 33 (Suppl. 1), 77–80. [Google Scholar] [CrossRef] [PubMed]
  151. Sun, L.; Fan, W.; Wu, C.; Zhang, S.; Dai, J.; Zhang, D. Effect of substituting different concentrations of soybean lecithin and egg yolk in tris-based extender on goat semen cryopreservation. Cryobiology 2020, 92, 146–150. [Google Scholar] [CrossRef]
  152. Vidal, A.H.; Batista, A.M.; da Silva, E.C.B.; Gomes, W.A.; Pelinca, M.A.; Silva, S.V.; Guerra, M.M.P. Soybean lecithin-based extender as an alternative for goat sperm cryopreservation. Small Ruminant. Res. 2013, 109, 47–51. [Google Scholar] [CrossRef] [Green Version]
  153. Lucio, C.F.; Regazzi, F.M.; Silva, L.C.G.; Angrimani, D.S.R.; Nichi, M.; Vannucchi, C.I. Oxidative stress at different stages of two-step semen cryopreservation procedures in dogs. Theriogenology 2016, 85, 1568–1575. [Google Scholar] [CrossRef]
  154. Nakagata, N. Cryopreservation of mouse spermatozoa. Mamm. Genome 2000, 11, 572–576. [Google Scholar] [CrossRef] [PubMed]
  155. Dorado, J.; Rodriguez, I.; Hidalgo, M. Cryopreservation of goat spermatozoa: Comparison of two freezing extenders based on post-thaw sperm quality and fertility rates after artificial insemination. Theriogenology 2007, 68, 168–177. [Google Scholar] [CrossRef] [PubMed]
  156. Manjunath, P. New insights into the understanding of the mechanism of sperm protection by extender components. Animal Reproduction 2018, 9, 809–815. [Google Scholar]
  157. Abe, Y.; Lee, D.S.; Sano, H.; Akiyama, K.; Yanagimoto-Ueta, Y.; Asano, T.; Suwa, Y.; Suzuki, H. Artificial insemination with canine spermatozoa frozen in a skim milk/glucose-based extender. J. Reprod. Dev. 2008, 54, 290–294. [Google Scholar] [CrossRef] [Green Version]
  158. Knight, C.A.; Wen, D.; Laursen, R.A. Nonequilibrium antifreeze peptides and the recrystallization of ice. Cryobiology 1995, 32, 23–34. [Google Scholar] [CrossRef]
  159. Nabeel, A.H.T.; Jeon, Y.; Yu, I.J. Use of polyvinyl alcohol as a chemically defined compound in egg yolk-free extender for dog sperm cryopreservation. Reprod. Domest. Anim. 2019, 54, 1449–1458. [Google Scholar] [CrossRef]
  160. Fan, X.L.; Zhang, Y.; Li, X.; Fu, Q.L. Mechanisms underlying the protective effects of mesenchymal stem cell-based therapy. Cell Mol. Life Sci. 2020, 77, 2771–2794. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  161. Sensebe, L.; Krampera, M.; Schrezenmeier, H.; Bourin, P.; Giordano, R. Mesenchymal stem cells for clinical application. Vox Sang. 2010, 98, 93–107. [Google Scholar] [CrossRef] [PubMed]
  162. Doorn, J.; Moll, G.; Le Blanc, K.; van Blitterswijk, C.; de Boer, J. Therapeutic applications of mesenchymal stromal cells: Paracrine effects and potential improvements. Tissue Eng. Part B Rev. 2012, 18, 101–115. [Google Scholar] [CrossRef]
  163. Chen, J.; Crawford, R.; Chen, C.; Xiao, Y. The key regulatory roles of the PI3K/Akt signaling pathway in the functionalities of mesenchymal stem cells and applications in tissue regeneration. Tissue Eng. Part B Rev. 2013, 19, 516–528. [Google Scholar] [CrossRef] [Green Version]
  164. Zhou, Y.; Xu, H.; Xu, W.; Wang, B.; Wu, H.; Tao, Y.; Zhang, B.; Wang, M.; Mao, F.; Yan, Y.; et al. Exosomes released by human umbilical cord mesenchymal stem cells protect against cisplatin-induced renal oxidative stress and apoptosis in vivo and in vitro. Stem Cell Res. Ther. 2013, 4, 34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Fazeli, Z.; Abedindo, A.; Omrani, M.D.; Ghaderian, S.M.H. Mesenchymal Stem Cells (MSCs) Therapy for Recovery of Fertility: A Systematic Review. Stem Cell Rev. Rep. 2018, 14, 1–12. [Google Scholar] [CrossRef]
  166. Cakici, C.; Buyrukcu, B.; Duruksu, G.; Haliloglu, A.H.; Aksoy, A.; Isik, A.; Uludag, O.; Ustun, H.; Subasi, C.; Karaoz, E. Recovery of fertility in azoospermia rats after injection of adipose-tissue-derived mesenchymal stem cells: The sperm generation. Biomed. Res. Int. 2013, 2013, 529589. [Google Scholar] [CrossRef]
  167. Malard, P.F.; Peixer, M.A.S.; Grazia, J.G.; Brunel, H.; Feres, L.F.; Villarroel, C.L.; Siqueira, L.G.B.; Dode, M.A.N.; Pogue, R.; Viana, J.H.M.; et al. Intraovarian injection of mesenchymal stem cells improves oocyte yield and in vitro embryo production in a bovine model of fertility loss. Sci. Rep. 2020, 10, 8018. [Google Scholar] [CrossRef]
  168. Liu, C.H.; Yin, H.Q.; Jiang, H.; Du, X.; Wang, C.L.; Liu, Y.C.; Li, Y.; Yang, Z.L. Extracellular Vesicles Derived from Mesenchymal Stem Cells Recover Fertility of Premature Ovarian Insufficiency Mice and the Effects on their Offspring. Cell Transplant. 2020, 29, 0963689720923575. [Google Scholar] [CrossRef] [PubMed]
  169. Vermeulen, M.; Giudice, M.G.; Del Vento, F.; Wyns, C. Role of stem cells in fertility preservation: Current insights. Stem Cells Cloning 2019, 12, 27–48. [Google Scholar] [CrossRef] [Green Version]
  170. Qamar, A.Y.; Fang, X.; Kim, M.J.; Cho, J. Improved Post-Thaw Quality of Canine Semen after Treatment with Exosomes from Conditioned Medium of Adipose-Derived Mesenchymal Stem Cells. Animals 2019, 9, 865. [Google Scholar] [CrossRef] [Green Version]
  171. Qamar, A.Y.; Fang, X.; Kim, M.J.; Cho, J. Improved viability and fertility of frozen-thawed dog sperm using adipose-derived mesenchymal stem cells. Sci. Rep. 2020, 10, 1–10. [Google Scholar] [CrossRef] [PubMed]
  172. Mahiddine, F.Y.; Qamar, A.Y.; Kim, M.J. Canine amniotic membrane derived mesenchymal stem cells exosomes addition in canine sperm freezing medium. J. Anim. Reprod. Biotechnol. 2020, 35, 268–272. [Google Scholar] [CrossRef]
  173. Mahiddine, F.Y.; Kim, J.W.; Qamar, A.Y.; Ra, J.C.; Kim, S.H.; Jung, E.J.; Kim, M.J. Conditioned Medium from Canine Amniotic Membrane-Derived Mesenchymal Stem Cells Improved Dog Sperm Post-Thaw Quality-Related Parameters. Animals 2020, 10, 1899. [Google Scholar] [CrossRef]
  174. Mokarizadeh, A.; Rezvanfar, M.A.; Dorostkar, K.; Abdollahi, M. Mesenchymal stem cell derived microvesicles: Trophic shuttles for enhancement of sperm quality parameters. Reprod. Toxicol. 2013, 42, 78–84. [Google Scholar] [CrossRef]
  175. Jafarzadeh, H.; Nazarian, H.; Ghaffari Novin, M.; Shams Mofarahe, Z.; Eini, F.; Piryaei, A. Improvement of oocyte in vitro maturation from mice with polycystic ovary syndrome by human mesenchymal stromal cell-conditioned media. J. Cell. Biochem. 2018, 119, 10365–10375. [Google Scholar] [CrossRef]
  176. Ling, B.; Feng, D.Q.; Zhou, Y.; Gao, T.; Wei, H.M.; Tian, Z.G. Effect of conditioned medium of mesenchymal stem cells on the in vitro maturation and subsequent development of mouse oocyte. Braz. J. Med. Biol. Res. 2008, 41, 978–985. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Ghiasi, M.; Fazaely, H.; Asaii, E.; Sheykhhasan, M.J.V. In Vitro Maturation of Human Oocytes using Conditioned Medium of Mesenchymal Stem Cells and Formation of Embryo by Use of ICSI. Vitro 2014, 41, 978–985. [Google Scholar]
  178. Maldonado, M.; Huang, T.; Chen, J.; Zhong, Y. Differentiation Potential of Human Wharton’s Jelly-Derived Mesenchymal Stem Cells and Paracrine Signaling Interaction Contribute to Improve the In Vitro Maturation of Mouse Cumulus Oocyte Complexes. Stem Cells Int. 2018, 2018, 7609284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Hsiao, C.H.; Ji, A.T.; Chang, C.C.; Chien, M.H.; Lee, L.M.; Ho, J.H. Mesenchymal stem cells restore the sperm motility from testicular torsion-detorsion injury by regulation of glucose metabolism in sperm. Stem Cell Res. Ther. 2019, 10, 270. [Google Scholar] [CrossRef]
  180. Zhang, D.; Liu, X.; Peng, J.; He, D.; Lin, T.; Zhu, J.; Li, X.; Zhang, Y.; Wei, G. Potential spermatogenesis recovery with bone marrow mesenchymal stem cells in an azoospermic rat model. Int. J. Mol. Sci. 2014, 15, 13151–13165. [Google Scholar] [CrossRef] [PubMed]
  181. Eilts, B.E. Theoretical aspects of canine semen cryopreservation. Theriogenology 2005, 64, 692–697. [Google Scholar] [CrossRef]
  182. Brito, M.M.; Lucio, C.F.; Angrimani, D.S.; Losano, J.D.; Dalmazzo, A.; Nichi, M.; Vannucchi, C.I. Comparison of Cryopreservation Protocols (Single and Two-steps) and Thawing (Fast and Slow) for Canine Sperm. Anim. Biotechnol. 2017, 28, 67–73. [Google Scholar] [CrossRef] [PubMed]
  183. Okano, T.; Murase, T.; Asano, M.; Tsubota, T. Effects of final dilution rate, sperm concentration and times for cooling and glycerol equilibration on post-thaw characteristics of canine spermatozoa. J. Vet. Med. Sci. 2004, 66, 1359–1364. [Google Scholar] [CrossRef] [Green Version]
  184. Kaur, P.; Schulz, K.; Heggland, I.; Aschner, M.; Syversen, T. The use of fluorescence for detecting MeHg-induced ROS in cell cultures. Toxicol. Vitr. 2008, 22, 1392–1398. [Google Scholar] [CrossRef]
  185. Saeki, K.; Yuo, A.; Kato, M.; Miyazono, K.; Yazaki, Y.; Takaku, F. Cell density-dependent apoptosis in HL-60 cells, which is mediated by an unknown soluble factor, is inhibited by transforming growth factor β1 and overexpression of Bcl-2. J. Biol. Chem. 1997, 272, 20003–20010. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. De Loecker, W.; Koptelov, V.A.; Grischenko, V.I.; De Loecker, P. Effects of cell concentration on viability and metabolic activity during cryopreservation. Cryobiology 1998, 37, 103–109. [Google Scholar] [CrossRef]
  187. Pegg, D.E. The effect of cell concentration on the recovery of human erythrocytes after freezing and thawing in the presence of glycerol. Cryobiology 1981, 18, 221–228. [Google Scholar] [CrossRef]
  188. Phelps, J.; Sanati-Nezhad, A.; Ungrin, M.; Duncan, N.A.; Sen, A. Bioprocessing of Mesenchymal Stem Cells and Their Derivatives: Toward Cell-Free Therapeutics. Stem Cells Int. 2018, 2018, 9415367. [Google Scholar] [CrossRef] [Green Version]
  189. Rani, S.; Ryan, A.E.; Griffin, M.D.; Ritter, T. Mesenchymal Stem Cell-derived Extracellular Vesicles: Toward Cell-free Therapeutic Applications. Mol. Ther. 2015, 23, 812–823. [Google Scholar] [CrossRef] [Green Version]
  190. Keshtkar, S.; Azarpira, N.; Ghahremani, M.H. Mesenchymal stem cell-derived extracellular vesicles: Novel frontiers in regenerative medicine. Stem Cell Res. Ther. 2018, 9, 63. [Google Scholar] [CrossRef]
  191. Katsuda, T.; Kosaka, N.; Takeshita, F.; Ochiya, T. The therapeutic potential of mesenchymal stem cell-derived extracellular vesicles. Proteomics 2013, 13, 1637–1653. [Google Scholar] [CrossRef] [PubMed]
  192. Diomede, F.; Gugliandolo, A.; Scionti, D.; Merciaro, I.; Cavalcanti, M.F.; Mazzon, E.; Trubiani, O. Biotherapeutic Effect of Gingival Stem Cells Conditioned Medium in Bone Tissue Restoration. Int. J. Mol. Sci. 2018, 19, 329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Thery, C.; Zitvogel, L.; Amigorena, S. Exosomes: Composition, biogenesis and function. Nat. Rev. Immunol. 2002, 2, 569–579. [Google Scholar] [CrossRef] [PubMed]
  194. Pegtel, D.M.; Gould, S.J. Exosomes. Annu. Rev. Biochem. 2019, 88, 487–514. [Google Scholar] [CrossRef]
  195. Cordeiro, L.; Lin, H.H.; Vitorino Carvalho, A.; Grasseau, I.; Uzbekov, R.; Blesbois, E. First insights on seminal extracellular vesicles in chickens of contrasted fertility. Reproduction 2021, 161, 489–498. [Google Scholar] [CrossRef]
  196. Murdica, V.; Giacomini, E.; Alteri, A.; Bartolacci, A.; Cermisoni, G.C.; Zarovni, N.; Papaleo, E.; Montorsi, F.; Salonia, A.; Viganò, P. Seminal plasma of men with severe asthenozoospermia contain exosomes that affect spermatozoa motility and capacitation. Fertil. Steril. 2019, 111, 897–908. [Google Scholar] [CrossRef]
  197. Chen, Y.X.; Zeng, Z.C.; Sun, J.; Zeng, H.Y.; Huang, Y.; Zhang, Z.Y. Mesenchymal stem cell-conditioned medium prevents radiation-induced liver injury by inhibiting inflammation and protecting sinusoidal endothelial cells. J. Radiat. Res. 2015, 56, 700–708. [Google Scholar] [CrossRef]
  198. Yamaguchi, S.; Shibata, R.; Yamamoto, N.; Nishikawa, M.; Hibi, H.; Tanigawa, T.; Ueda, M.; Murohara, T.; Yamamoto, A. Dental pulp-derived stem cell conditioned medium reduces cardiac injury following ischemia-reperfusion. Sci. Rep. 2015, 5, 16295. [Google Scholar] [CrossRef] [Green Version]
  199. Ra, K.; Oh, H.J.; Kim, G.A.; Kang, S.K.; Ra, J.C.; Lee, B.C. High Frequency of Intravenous Injection of Human Adipose Stem Cell Conditioned Medium Improved Embryo Development of Mice in Advanced Maternal Age through Antioxidant Effects. Animals 2020, 10, 978. [Google Scholar] [CrossRef] [PubMed]
  200. Ramirez, M.I.; Amorim, M.G.; Gadelha, C.; Milic, I.; Welsh, J.A.; Freitas, V.M.; Nawaz, M.; Akbar, N.; Couch, Y.; Makin, L.; et al. Technical challenges of working with extracellular vesicles. Nanoscale 2018, 10, 881–906. [Google Scholar] [CrossRef] [Green Version]
  201. Gunawardena, T.N.A.; Rahman, M.T.; Abdullah, B.J.J.; Abu Kasim, N.H. Conditioned media derived from mesenchymal stem cell cultures: The next generation for regenerative medicine. J. Tissue Eng. Regen. Med. 2019, 13, 569–586. [Google Scholar] [CrossRef] [PubMed]
  202. Jha, K.N.; Shumilin, I.A.; Digilio, L.C.; Chertihin, O.; Zheng, H.; Schmitz, G.; Visconti, P.E.; Flickinger, C.J.; Minor, W.; Herr, J.C. Biochemical and structural characterization of apolipoprotein AI binding protein, a novel phosphoprotein with a potential role in sperm capacitation. Endocrinology 2008, 149, 2108–2120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Bernardini, A.; Hozbor, F.; Sanchez, E.; Fornes, M.W.; Alberio, R.H.; Cesari, A. Conserved ram seminal plasma proteins bind to the sperm membrane and repair cryopreservation damage. Theriogenology 2011, 76, 436–447. [Google Scholar] [CrossRef] [PubMed]
  204. Milardi, D.; Grande, G.; Vincenzoni, F.; Messana, I.; Pontecorvi, A.; De Marinis, L.; Castagnola, M.; Marana, R. Proteomic approach in the identification of fertility pattern in seminal plasma of fertile men. Fertil. Steril. 2012, 97, 67–73. [Google Scholar] [CrossRef] [PubMed]
  205. Arner, E.S.J.; Holmgren, A. Physiological functions of thioredoxin and thioredoxin reductase. Eur. J. Biochem. 2000, 267, 6102–6109. [Google Scholar] [CrossRef]
  206. Gasdaska, J.R.; Berggren, M.; Powis, G. Cell growth stimulation by the redox protein thioredoxin occurs by a novel helper mechanism. Cell Growth Differ. 1995, 6, 1643–1650. [Google Scholar]
  207. Uysal, O.; Bucak, M.N. Effects of oxidized glutathione, bovine serum albumin, cysteine and lycopene on the quality of frozen-thawed ram semen. Acta Vet. Brno 2007, 76, 383–390. [Google Scholar] [CrossRef]
Table 1. List of antioxidants used for sperm cryopreservation in dogs and their effects in different species.
Table 1. List of antioxidants used for sperm cryopreservation in dogs and their effects in different species.
AntioxidantSpeciesDosageResults
AstaxanthinBoar2 μMProtected post-thaw sperm motility, membrane and acrosome integrity. Inhibited lipid peroxidation. Regulation of membrane fatty acid composiion. Enhanced IVF efficiency and embryonic development [28].
Supplementation of 0.5 μM decreased apoptotic-like changes.
Supplementation of 15 μM had a negative effect on sperm parameters [29].
Canine1 µMProtected post-thaw sperm kinematic parameters, viability, mitochondrial activity, plasma membrane, chromatin and acrosome integrity [30].
Miniature Pig500 µMProtected post-thaw sperm motility and progressive motility. Reduced ROS production [31].
Sheep2 µM, 4 µMProtected post-thaw sperm viability and plasma membrane integrity.
Decreased acrosome abnormalities and malondialdehyde formation [32].
ResveratrolBoar50 µMProtected post-thaw sperm progressive motility, membrane and acrosome integrity, mitochondrial activity, activities of enzymatic antioxidants, and phosphorylation of AMPK [33].
Bovine50 μMProtected membrane integrity and antioxidant capacity.
Decreased ROS production, and capacitation-like changes.
Improved in vitro fertilizing ability [34].
Canine200 µMProtected post-thaw sperm motility, viability, plasma membrane, acrosome and chromatin integrity, and mitochondrial activity [35].
Equine5 μMProtected post-thaw DNA and membrane integrity, total and progressive motility, and viability in subfertile stallions [36].
Human0.1 mM, 1.0 mM, 10.0 mMPrevented lipid damage in a non dose-dependent manner [37].
QuercetinBovine25 μg/mLProtected post-thaw DNA integrity. No effects on post-thaw sperm kinematic parameters, and plasma membrane integrity [38].
Canine5 μg/mLProtected post-thaw kinematic parameters and fertility [39].
Equine0.1 mMProtected post-thaw total and progressive motility. Reduced oxidative stress [40]
Human50 μMProtected post-thaw kinematic parameters, viability, and DNA integrity [41].
MyoinositolCanine1 mg/mLProtected sperm motility, kinematic parameters, and membrane integrity. Reduced chromatin damage and apoptosis-like changes [42].
Equine30 mMAmplitude of lateral head displacement increased [43].
Human2 mg/mLProgressive motility percentage protected in normozoospermic, oligo-astheno-teratozoospermic men. Mitochondrial function improvement in patients with impaired sperm parameters. Decreased DNA fragmentation and lipid peroxidation [44,45,46].
1 mg/mLIncreased cryosurvival rate [47].
Sheep5 mM, 10 mMNo significant enhancement [48].
CurcuminBoar0.25 mmol/L, 0.50 mmol/LProtected post-thaw progressive motility and acrosome integrity [49].
Bovine0.5 mMProtected post-thaw plasma membrane integrity and oxidative defense. Reduced percentage of abnormal sperm [50].
Canine2.50 mMProtected post-thaw DNA integrity and oxidative defense [51].
Human20 μMProtected post-thaw progressive motilty. Reduced DNA fragmentation and intracellular ROS [52].
IodixanolBovine2.50%Protected post-thaw motility, progressive motility, viability, and acrosome and membrane integrity [53,54].
Protection against oxidative stress [55].
Canine1.50%Protected frozen–thawed motility. Decreased capacitation, protamine deficiency, and apoptosis-like changes. Reduced mitochondrial reactive oxygen production [56].
Equine5%Protected post-thaw progressive motility, plasma membrane, and DNA integrity [57].
Rat1%, 2%Protected post thaw motility [58].
Sheep5%Protected post-thaw progressive motility, morphology, and acrosome and membrane integrity [59,60].
SpermineBovineAssociated with a nitric acid donor, 10 μM Protected frozen–thawed sperm motility, viability, membrane integrity, and decreased lipid peroxidation [61].
Canine5.0 mMProtected post-thaw kinematic parameters, and membrane integrity. Decreased reactive oxygen species production, and cryocapacitation [62].
Equine1 mg/mL, 2 mg/mLDecreased capacitation and DNA fragmentation index [63].
KinetinCanine50 μMReduced sperm post-thaw oxidative damages. Protected post-thaw motility, viability, and membrane integrity [64].
Sheep50 μM, 100 μM Protected cooled sperm antioxidant activity, kinematic parameters, viability, and plasma membrane. Decreased lipid peroxidation [65].
MelatoninBovine1 mM, 2mM, 3 mMProtected post-thaw motility, and antioxidant capacity. Reduced lipid peroxidation [66,67].
0.1 mMProtected post-thaw plasma membrane, acrosome region, and ultrastructure integrity [68].
Canine0.1 mM, 0.25 mMProtected post-thaw membrane and acrosome integrity [69].
Chicken10−3, 10−6 MDecreased lipid peroxidation, DNA fragmentation, and apoptosis-like changes. Protected post-thaw motility [70].
Equine1 μMHigher mitochondrial membrane potentials, and protected membrane integrity [71]. Reduced lipid peroxidation [72].
Goat20.0 µg Protected post-thaw motility [73].
Human0.01 mM, 3 mMProtected post-thaw motility, progressive motility, and viability. Decreased intracellular reactive oxygen species, malondialdehyde, and caspase-3 activity [74,75,76].
Mouse0.125 mg/mL [77]Protected post-thaw progressive motility, and anti-apoptotic gene expression [77].
Rabbits0.1 mMProtected post-thaw motility, membrane and acrosome integrity, and mitochondrial membrane potential [78].
Sheep1 mM Protected post-thaw motility, viability, intracellular ATP concentrations, and DNA integrity [79].
MetforminCanine50 µMProtected post-thaw motility, oxidative stress defense, and quality-related markers [80].
Mouse5000 µMEnhanced AMPK activity, and in vitro fertilization success [81].
Olive-derived antioxidantsCanine10 μg/mL, 30 μg/mL, 50 μg/mL and 70 μg/mLProtected post-thaw kinematic parameters, viability, plasma membrane integrity, and oxidative defense. Reduced DNA damage [82]
Sheep10 μg/mL, 30 μg/mL, 50 μg/mL and 70 μg/mLNo effects on post-thaw sperm kinematic parameters. Reduced lipid peroxidation [83].
Table 2. Use of mesenchymal stem cells in male reproduction and ART.
Table 2. Use of mesenchymal stem cells in male reproduction and ART.
Mesenchymal Stem CellsFormSpeciesTreatment TypeEffects on Sperm
Adipose-derived MSCsCellsCanineCryopreservationProtected post-thaw sperm motility, viability, membrane, and acrosome and chromatin integrity [171].
CellsRatsInfertilityReestablishment of spermatogenesis, and restoration of fertility [166].
CellsRatsTesticular injuryProtected progressive motility and vitality. Activated Akt/GSK3 axis and stimulated glucolysis [179].
ExosomesCanineCryopreservationProtected post-thaw sperm plasma membrane and chromatin integrity, motility, and viability [170].
Amniotic-membrane-derived MSCsConditioned mediumCanineCryopreservationProtected post-thaw sperm plasma membrane integrity, motility, mitochondrial activity, and viability [173].
ExosomesCanineCryopreservationNo effects on post-thaw sperm quality-related parameters [172].
Bone-marrow-derived MSCsCellsRatsInfertilityRestoration of fertility [180].
MicrovesiclesRatsCryopreservationProtected post-thaw sperm viability, progressive motility, and antioxidant capacity. Reduced levels of necrosis, and apoptosis. Increased expression of surface adhesion molecules [174].
MSC, mesenchymal stem cells; AKT, protein kinase B; GSK3, glycogen synthase kinase 3.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Mahiddine, F.Y.; Kim, M.-J. Overview on the Antioxidants, Egg Yolk Alternatives, and Mesenchymal Stem Cells and Derivatives Used in Canine Sperm Cryopreservation. Animals 2021, 11, 1930. https://doi.org/10.3390/ani11071930

AMA Style

Mahiddine FY, Kim M-J. Overview on the Antioxidants, Egg Yolk Alternatives, and Mesenchymal Stem Cells and Derivatives Used in Canine Sperm Cryopreservation. Animals. 2021; 11(7):1930. https://doi.org/10.3390/ani11071930

Chicago/Turabian Style

Mahiddine, Feriel Yasmine, and Min-Jung Kim. 2021. "Overview on the Antioxidants, Egg Yolk Alternatives, and Mesenchymal Stem Cells and Derivatives Used in Canine Sperm Cryopreservation" Animals 11, no. 7: 1930. https://doi.org/10.3390/ani11071930

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop