Next Article in Journal
Molecular Characterization of ASFV and Differential Diagnosis of Erysipelothrix in ASFV-Infected Pigs in Pig Production Regions in Cameroon
Previous Article in Journal
Use of Phages to Treat Antimicrobial-Resistant Salmonella Infections in Poultry
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Importance of Antioxidant Supplementation during In Vitro Maturation of Mammalian Oocytes

by
Shimaa I. Rakha
1,2,
Mohammed A. Elmetwally
1,2,
Hossam El-Sheikh Ali
1,2,
Ahmed Balboula
1,2,3,
Abdelmonem Montaser Mahmoud
1,2 and
Samy M. Zaabel
1,2,*
1
Department of Theriogenology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
2
Reproductive Biotechnology Research Laboratory, College of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
3
Animal Sciences Research Center, University of Missouri, Columbia, MO 65211, USA
*
Author to whom correspondence should be addressed.
Vet. Sci. 2022, 9(8), 439; https://doi.org/10.3390/vetsci9080439
Submission received: 30 July 2022 / Revised: 12 August 2022 / Accepted: 14 August 2022 / Published: 18 August 2022

Abstract

:

Simple Summary

In vitro embryo production (IVEP) is a technology that is widely used in the field of reproductive biology. The first and most important step of IVEP is termed in vitro maturation or IVM, in which the oocyte is allowed to mature in a synthetic medium under controlled laboratory settings. The quality of in vitro matured oocytes is still low compared to that of those matured inside an animal’s body. One of the key elements that was found to affect oocyte quality is the accumulation of large amounts of reactive oxygen species (ROS) within oocytes during IVM. The addition of antioxidants to oocyte IVM media is one of the most effective methods for preventing ROS accumulation. In this article, we highlight the latest events showing how ROS harms mammalian oocytes. We also discuss the possible impacts of antioxidant supplementation on the IVM rate and oocyte quality.

Abstract

The in vitro embryo production (IVEP) technique is widely used in the field of reproductive biology. In vitro maturation (IVM) is the first and most critical step of IVEP, during which, the oocyte is matured in an artificial maturation medium under strict laboratory conditions. Despite all of the progress in the field of IVEP, the quality of in vitro matured oocytes remains inferior to that of those matured in vivo. The accumulation of substantial amounts of reactive oxygen species (ROS) within oocytes during IVM has been regarded as one of the main factors altering oocyte quality. One of the most promising approaches to overcome ROS accumulation within oocytes is the supplementation of oocyte IVM medium with antioxidants. In this article, we discuss recent advancements depicting the adverse effects of ROS on mammalian oocytes. We also discuss the potential use of antioxidants and their effect on both oocyte quality and IVM rate.

1. Introduction

The in vitro embryo production (IVEP) technique is frequently employed to overcome problems associated with infertility in different mammalian species [1,2]. IVEP has been identified as a key technique for the production of a large number of offspring from animals with superior genetic traits [3].
The in vitro maturation (IVM) of oocytes is the first and most crucial phase in the IVEP process, during which, oocytes gain the potential to sustain continued embryonic development [4]. As a result, identifying the appropriate IVM environment is essential for effective IVEP procedures [4].
The success of IVM is dependent on various factors including the quality of collected oocytes and the setup of culture conditions [5]. Antioxidants supplied periodically from fluids of the female reproductive organs act to minimize the damage induced by reactive oxygen species, ROS, natural by-products of oocyte metabolism [6]. One of the challenges that oocytes face during IVM is oxidative stress caused by the increased generation of ROS by oocytes in the IVM media [7]. Excessive ROS production could ultimately result in oocyte death and embryonic loss [8,9].
Other forms of stress that oocytes may face include high ambient temperature [10], increased mother age [11], and fertilization delays caused by the failure of the sperm to reach and penetrate the oocyte membranes [12]. All these stress types represent a potential risk for female infertility since they predispose oocytes to oxidative damage and are associated with a series of physiological, metabolic, cellular, and molecular alterations in the reproductive tract [13].
Antioxidant supplementation during IVM has been suggested to protect oocytes from the injurious effects of oxidative stress via maintaining physiological levels of ROS [14,15]. In this article, current understandings of the sources of ROS during IVM of mammalian oocytes, their deleterious effects on oocyte biology, and the promising role of exogenously supplemented antioxidants are presented and discussed.

2. In Vitro Maturation of Mammalian Oocytes

Oocyte maturation involves a series of complex and diverse events of nuclear and cytoplasmic modifications that provide oocytes the inherent potential to promote embryo development and activate the embryonic genome [16]. Nuclear and cytoplasmic maturations are two components of oocyte maturation [17]. Cumulus expansion is also regarded as a sign of oocyte maturation [18].
Cytoplasmic maturation involves events related to the cytoplasmic capacitation of the oocyte [19]. It entails the accumulation of mRNA, proteins, and substrates necessary for fertilization and subsequent development [20]. Through the regulation of protein and ATP synthesis as well as chromosome segregation, cell organelles including mitochondria, endoplasmic reticulum (ER), and microtubules play key roles in both cytoplasmic and nuclear maturation [16,21].
The nuclear maturation of oocytes implies the resumption of meiosis and progression to the metaphase II stage. The mammalian oocyte passes through two successive cell divisions during its maturation process [22]. First, the oocyte is arrested at the diplotene stage. At this stage, the oocyte appears to contain a large nucleus, the germinal vesicle (GV). Germinal vesicle breakdown (GVBD) involves chromatin condensation and the disintegration of the nuclear membrane [23]. Following GVBD, the oocyte resumes meiosis and enters the metaphase I (MI) [24]. The first meiosis ends with the formation of a haploid oocyte and the extrusion of the first polar body. The former is kept at metaphase II (MII) until fertilization [25]. Inaccuracies of these meiotic events might prevent oocytes from reaching their proper maturation [26].
The process of oocyte maturation is a complex process that requires coordinated interactions between the oocyte and its surrounding cumulus cells as well as between oocyte cytoplasmic and nuclear compartments [27]. For instance, mixing of the GV contents during its breakdown with the cytoplasm has been assumed to activate critical aspects of cytoplasmic maturation and subsequent developmental competence [28]. In certain species, including equines and humans, the process of cytoplasmic maturation has been found to be deficient in several aspects, especially those related to microtubule patterning and cell division, with negative outcomes on rates of embryo formation [29,30]. Several approaches have been investigated to improve IVM in the aforementioned species by creating suitable conditions for better cytoplasmic maturation. In equines, the treatment of oocytes with the meiosis-inhibiting factor Roscovitine for a period of time prior to IVM improved microfilaments’ organization in matured oocytes [31]. Recently, co-culturing equine COCs with granulosa cells from small follicles (<15 mm) has been found to significantly improve the cytoplasmic maturation of oocytes on the basis of cortical granules’ distribution and mitochondrial function [32].

3. Normal Production and Elimination of Reactive Oxygen Species (ROS) by Mammalian Oocytes

ROS are oxygen-containing molecules produced as a by-product of cellular respiration and metabolism [33]. They primarily consist of superoxide anions (O2), hydrogen peroxide (H2O2), and hydroxyl radicals (OH) [33]. The oxidation of unsaturated fatty acids, also known as lipid peroxidation, is one of the most severe outcomes of free radical attack. One of the end products of lipid peroxidation is malondialdehyde (MDA). It can be used as a cumulative marker of lipid peroxidation because it is a stable end product [34]. In oocytes, MDA is thus regarded as an indicator of oxidative stress [35].
Physiological levels of ROS have been found to be beneficial to gamete function and development [36]. However, the excessive production of ROS at levels that exceed oocyte total antioxidant capacity could result in a state of oxidative stress (OS) [13]. OS is characterized by a wide range of cellular destruction including DNA damage, arrested growth and reduced oocyte quality [37].
Intracellular ROS are repeatedly removed by a group of intracellular antioxidants. These antioxidants include an extensive set of cooperating enzymatic and non-enzymatic factors [38]. These factors are abundantly present within the follicular fluid and act to protect oocytes from harmful effects [39]. Superoxide dismutases (SODs), catalase (CAT) and glutathione peroxidase (GPx) are examples of enzymatic antioxidants. SODs convert O2 into H2O2 via its mitochondrial, manganese-dependent superoxide dismutase (Mn-SOD) and cytoplasmic, copper, zinc superoxide dismutase (Cu/ZnSOD), while CAT and GPx mediate the breakdown of H2O2 into water and oxygen [40,41,42].
Non-enzymatic antioxidants include thiols [43], ascorbic acid, alpha-tocopherol [44,45], melatonin [46], l-carnitine [47,48], and lycopene [49]. They prevent oxidative damage by interrupting free radical chain reactions [14].

4. The Alteration in Oxidative Status of Mammalian Oocytes during Their IVM

The in vitro handling of gametes and embryos during ARTs is associated with the production of large amounts of ROS that exceed the typical antioxidant capacity of the cell [50]. Organelle failure, spindle abnormalities, DNA fragmentation, and apoptosis are all among the detrimental effects of high ROS on IVM oocytes [50].
IVM involves the culture of cumulus–oocyte complexes (COCs) in a synthetic medium. The media used for IVM usually contain fewer antioxidant enzymes than the in vivo milieu of follicular and oviduct fluids provided by the mother [6]. During IVM, OS is caused by an imbalance between ROS production and clearance induced by a shortage of maternal antioxidants. Such an alteration causes oocyte maturation and subsequent embryonic development to be disrupted [51].
The incubation of oocytes in high oxygen concentrations [52] and their exposure to visible light [53], pollutants [54], and certain components of IVM [50] all promote ROS generation during the in vitro maturation of mammalian oocytes. Increased ROS levels disrupted maturation-promoting factors and triggered programmed cell death in oocytes from several mammal species [9,55].
The lipids in the membranes of in vitro generated embryos are damaged by ROS [56]. Non-competent embryos had higher ROS levels than competent embryos, which was linked to mitochondrial damage [57]. In bovines, the presence of free radicals during IVEP reduced the blastocyst rate in a dose-dependent manner [58]. Even when the damage was not fatal, it had a detrimental impact on cellular development, metabolic activity, and embryo viability [59].
Oocytes are exposed to OS during IVEP not only as a result of their metabolic and respiratory processes, but also as a result of external stimuli including oxygen tension and light. The oxygen content in the oviduct and uterus is lower (2–8%) than that used in in vitro research (about 20%) [60,61]. In cows [62], sheep [62], goats [63], pigs [64], and mice [65], toxic impacts of atmospheric oxygen concentrations and beneficial impacts of lower ones on the outcome of IVEP were reported [66].
The normal development of oocytes occurs inside the female reproductive tract in complete darkness. On the other hand, they are rapidly exposed to light during their in vitro handling. This exposure has been shown to promote OS in embryos and somatic cells by either increasing pro-oxidant production or blocking endogenous antioxidant mechanisms [67]. One-cell hamster embryos exposed to 14,000 lux light for a brief time period (30 s) revealed a considerable rise in H2O2 levels [67].
The redox status of oocytes and embryos was found to be affected by the amounts of nutrients in the culture medium. Hashimoto, Minami [68] found that high glucose concentrations in the IVM medium hampered embryo development, which was accompanied with high ROS and low glutathione concentrations. On the other hand, bovine COCs exposed to 50–100 mM H2O2 for one hour during IVM showed some improvement in their subsequent development [69].
Several studies have been conducted on the role of cumulus cells in the redox status of COCs. Spermatozoa incubated with intact COCs produced more ROS than spermatozoa incubated with denuded oocytes [53]. The enhanced production of ROS in intact COCs was observed to improve the sperm penetration of oocyte membranes. Tatemoto, Sakurai [70] observed that after 44 h of culture under the hypoxanthine–xanthine oxidase system, denuded oocytes had more DNA and apoptotic damages than intact COCs. Furthermore, Luciano, Goudet [71] found that supplementing horse COCs with cysteamine boosted GPx mRNA expression in cumulus cells but not in oocytes in vivo and in vitro. Cetica, Pintos [72] also noted higher SODs, CAT, and GPx enzymatic levels in separated cumulus cells of bovine COCs during IVM compared to denuded oocytes, which decreased afterwards. In addition, removing cumulus cells before the in vitro maturation of bovine oocytes resulted in reductions in their maturation, fertilization and embryo development rates [73].
The role of reactive oxygen species (ROS) in the process of GVBD is controversial. Tamura, Takasaki [74] showed that high levels of H2O2 impaired GVBD during human oocyte maturation, which was reversed by melatonin. In swine oocytes, inhibiting SODs’ enzymatic activity resulted in a considerable decrease in meiotic development [75]. On the other hand, Takami, Preston [76] reported that the addition of a number of cell-permeable antioxidants to IVM medium for two hours blocked spontaneous GVBD in both intact COCs and denuded oocytes in rats. These antioxidants included nordihydroguaiaretic acid, 2(3)-tert-butyl-4-hydroxyanisole, octyl gallate, ethoxyquin, 2,6-di-tert-butyl-hydroxymethyl phenol, butylated hydroxytoluene, tert-butyl hydroquinone, propyl gallate, lauryl gallate, and 2,4,5-trihydroxybutrophenone. Tarin [77] also showed no variations in IVM rates in mouse oocytes when the oxidizing agent tertiary-butyl-hydroperoxide was added (tBH).
During oocyte development, a high risk of aneuploidy or chromosomal abnormalities has been reported, which becomes increasingly apparent during meiosis I [78]. Tarin [77] reported that OS induced by tBH treatment resulted in abnormal-shaped meiotic spindles associated with defects in the alignment of chromosomes. Tarin, Vendrell [79] also observed a modest reduction in oocytes’ aneuploidy from mice who received antioxidant supplementation.
Oocyte maturation is dependent on the proper assembly of the meiotic spindle. Several studies have related changes in the morphology of the meiotic spindle to gamete origin and culture circumstances during IVM [80,81,82]. During the IVM of mouse oocytes, Choi, Banerjee [83] reported that OS induced by H2O2 addition caused time-dependent changes in microtubule dynamics and chromosomal alignment. These changes were partially corrected by vitamin C supplementation via alleviating the harmful effect of H2O2 on chromosome alignment but not on microtubule alteration.

5. Antioxidant Supplementation during IVM of Mammalian Oocytes to Counteract ROS-Induced Damage

Currently, various antioxidant substances are being added during IVM to ensure a balanced intracellular redox status and good oocyte quality [84,85]. Adding antioxidants, such as thiols, polyphenolic compounds, melatonin, carotenoids, resveratrol, and vitamins such as C and E, to the IVM medium has been proven in several trials to improve oocyte quality and alleviate their damage induced by excessive ROS exposure [86,87]. Details of these antioxidants are listed in Table 1.
Thiols, or mercaptans, are organosulfur compounds similar to alcohols and phenols but have a sulfur atom in place of the oxygen atom [111]. Glutathione is a tripeptide thiol that naturally presents in either a reduced (GSH) or oxidized (GSSG) form [112]. It is made up of three amino acids: cysteine, glycine, and glutamate. It is a powerful reducing agent and also acts as a GPx electron donor. The presence of GSH in the oviductal fluid was reported [113]. Adding 0.6 mM cysteine, non-essential sulfur-containing amino acid, to bovine oocytes during IVM significantly increased embryo rates, regardless of exposure time [114].
Other thiols such as β-mercaptoethanol (β-ME) or cysteamine were added to boost intracellular GSH content and cell growth rates [115,116]. When these thiols were added to IVM medium in cows [117], sheep [118], pigs [119], dogs [120], and mice [98], growth-promoting effects were seen. Even at high oxygen concentrations, the addition of β-ME to the IVM medium was associated with increased embryo growth rates of mammalian embryos [119,121].
Polyphenolic compounds are plant metabolites that showed strong antioxidant activities [122]. Luteolin is a polyphenolic compound that revealed a major role in protecting several cell types against oxidative-stress-induced damage [123]. Luteolin improved the quality of porcine oocytes incubated under oxidative stress conditions and enhanced their subsequent embryonic development following IVF by alleviating oxidative damage to cell organelles [124].
Melatonin is a hormone that is produced naturally by the pineal gland. Melatonin receptors were found to be expressed in oocytes and cumulus cells of bovines [125,126] and mice [127]. The supplementation of IVM medium of bovine oocytes with exogenous melatonin significantly increased the rates of oocyte nuclear maturation, cleavage, and blastocyst formation [125,126]. Melatonin addition to the IVM medium also increased maturation and developmental competence in heat-stressed bovines [128] and pigs [129].
The supplementation of IVM medium with a mixture of 10 µM of alpha-tocopherol and 250 µM of l-ascorbic acid resulted in a larger proportion of denuded porcine oocytes progressing to the MII stage when compared with control groups [130]. A study by Choi, Banerjee [83] also suggested a beneficial role of ascorbic acid in protecting MII mouse oocytes from H2O2-induced damage.
Carotenoids are yellow- to red-colored naturally occurring pigments that are found mostly in plants, algae, marine organisms, and certain bacteria [131]. Over 1100 carotenoids have been reported [132]. They cannot be synthesized by mammals; thus, they must be obtained from the diet [133].
Carotenoids include α-carotene, β-carotene, and lycopene [134]. Xanthophylls are oxygenated derivatives of carotenes and represent the most abundant type of carotenoids [135]. Examples of xanthophylls are lutein, astaxanthin, and canthaxanthin. Carotenoids that contain one or more ketone groups are termed ketocarotenoids [136].
β-carotene supplementation to the IVM medium of mouse oocytes has been reported to reduce ROS levels, decrease cell apoptosis, and improve the overall structure of oocytes [137].
The supplementation of the IVM media of bovine and mouse oocytes with lycopene at a concentration of 0.2 μM resulted in a significant increase in the IVM rate compared to control oocytes, which was associated with reduced intracellular ROS levels and increased mitochondrial activities of oocytes [49,94].
Several keto-carotenoids have been employed in IVM experimental protocols. Canthaxanthin is a keto-carotenoid with significant antioxidant activity [138,139]. Canthaxanthin supplementation during IVM increased porcine oocyte maturation and subsequent developmental competence after parthenogenetic activation and somatic cell nuclear transfer [140].
Astaxanthin is another keto-carotenoid with potent antioxidant properties. Astaxanthin supplementation during IVM improved the maturation, fertilization, and development of pig oocytes [141].

6. Heat Stress and ROS Production by Mammalian Oocytes

6.1. Effect of Heat Stress on Oocyte ROS Production

Global warming is the steady rise in the Earth’s average temperature caused by increased rates of emissions of greenhouse gases, which trap heat and warm the Earth [142]. The rising global temperature exposes animals to stressful environmental circumstances, particularly in the summer, resulting in a reduction in domestic animal fertility due to an increase in body temperature exceeding physiological limitations, a condition known as heat stress [143].
Heat stress represents a potential risk for female infertility since it causes a series of physiological, metabolic, cellular, and molecular alterations in the reproductive tract [144]. Among the alterations induced by heat stress in different body tissues is the oxidative damage of intracellular components which results in structural and functional changes and causes apoptosis [145].
Heat stress has been reported to alter oocyte nuclear and cytoskeletal architecture and delay early embryonic development [146]. Heat stress stimulates excessive production of free radicals within maturing oocytes [38]. In cattle, the increased generation of free radicals within the oocyte following heat stress is associated with meiotic arrest, poor oocyte quality, and a lower rate of embryo development [147,148]. Moreover, the incubation of buffalo oocytes under heat stress condition led to an increase in the levels of ROS, lipid peroxide, and nitric oxide [149]. Additionally, mouse oocyte cytoplasmic maturation is more susceptible to thermal stress than nuclear maturation [150]. Furthermore, it was found that autophagy induction in pig oocytes is increased under heat stress conditions [151].
Several studies have shown that the process of oocyte maturation is disturbed at high temperatures [150,152,153,154]. The impact of elevated temperature on oocytes and embryos has been linked to a parallel increase in ROS production and a decrease in antioxidant defense enzyme activity [155,156,157,158]. In mouse oocytes and embryos, heat stress caused a decrease in the glutathione content and a rise in the H2O2 levels [156]. Culturing oocytes at high temperature during IVM has been also found to augment ROS production in bovines [159] and porcine [160].
It has been established that oocytes and embryos are the primary targets of the damaging effects of heat stress [161,162]. The cellular damage induced by heat stress involves diverse cellular organelles. Heat shock during the IVM of bovine oocytes has been reported to disturb spindle formation [163], mitochondrial function [164], microtubule and microfilament organization [165], and cortical granule distribution [166].
Breakdown of the germinal vesicle and expulsion of the first polar body are two significant characteristics of oocyte maturation [167]. Reduced rates of polar body extrusion and germinal vesicle breakdown were seen in oocytes matured at an elevated temperature (41.5 °C) for 22 h compared to oocytes matured at an optimum condition (38.5 °C) for the same time period in porcine [129,160], cattle [153,168] and buffalos [169]. On the other hand, heat stress (41 °C) increased the nuclear maturation kinetics of porcine oocytes, resulting in a larger proportion of MII oocytes after 16–18 h of IVM, according to Tseng, Tang [170].

6.2. Use of Antioxidants to Counteract ROS-Induced Damage of Heat-Stressed Oocytes

The possible involvement of antioxidant supplements in combating the negative effects of heat stress on oocyte developmental competence and IVEP is a subject of investigation [171]. The addition of retinol to the IVM medium of bovine oocytes prevented heat-induced reductions in oocyte maturation and improved the rate of blastocyst yield [172]. In mice, the administration of the antioxidant epigallocatechin gallate decreased the deleterious effects of maternal hyperthermia on follicle-enclosed oocytes via the suppression of ROS generation [173].
Porcine oocytes supplemented with 2 μM of resveratrol during their IVM under heat stress conditions revealed higher rates of polar body formation compared to heat-stressed oocytes with no resveratrol added [174].
The treatment of bovine COCs with astaxanthin at concentrations of 12.5 and 25 nM rescued the developmental competence of heat-shocked oocytes via enhancing SOD activity and decreasing ROS levels [175].
Melatonin has been shown to overcome the negative influences of heat stress on oocytes and preimplantation embryos [174,176,177,178]. Melatonin treatment upsurged SOD enzyme levels in heat-shocked mouse oocytes [156]. Melatonin addition at a 1 μM concentration decreased ROS levels of maturing bovine oocytes [179].
Coenzyme Q10 supplementation to IVM media improved mitochondrial properties and alleviated the effects of thermal stress on bovine oocytes’ developmental competence [180].

7. Maternal Aging and ROS Production by Mammalian Oocytes

7.1. Effect of Maternal Aging on Oocyte ROS Production

The aging of female mammals is widely established to be associated with decreased fertility [11]. The ovary is much more sensitive to the effects of aging than any other body tissue, and reproductive success has been shown to be inversely associated with age [181]. Ovarian senescence progressively reduces both follicle quantity and oocyte quality, resulting in a steady decrease in fertility which ends ultimately with sterility [182]. Mammalian oocytes display higher ROS levels with age, and thus, reproductive aging can be linked to the cumulative oxidative damage of oocytes [183,184,185].
Oxidative stress is one of the key processes underpinning aging. It arises as a result of the slow buildup of damage caused by free radicals created during normal metabolism [186]. The oocytes of most animals initiate meiosis in the fetal ovary. However, they enter a prolonged period of rest at the dictyate stage of the first meiotic prophase (prophase I) from fetal life until puberty. The duration of this period ranges from weeks in mice to months and years in domestic animals [182]. During this prolonged rest, physiological levels of generated ROS contribute to oocyte maturation within the follicle [187]. However, the cyclic production of these damaging agents negatively affects oocyte quality and may lead to an increased risk of ovarian pathologies, especially under circumstances of reduced antioxidant status [187].
Lipid metabolism is a powerful source of energy, and its relevance during oocyte maturation is becoming more apparent [188]. Lipid peroxidation is a hallmark of oxidative stress, which is known to increase as oocytes age [189]. Oocytes from aged mice were found to display significantly higher levels of lipid peroxides compared to those from adult mice [190].

7.2. Use of Antioxidants to Counteract ROS-Induced Damage of Maternally Aged Oocytes

In maternally aged oocytes, the disrupted balance between oxidant production and elimination leads to the development of oxidative stress [191]. The use of exogenous antioxidants is thus thought to antagonize the effects of maternal aging on oocytes via reducing oxidative stress. The oral administration of vitamins C and E reduced the negative effects of aging on ovarian reserve (number of both ovarian and ovulated oocytes) and oocyte quality (chromosomal aberrations and apoptotic changes) in a mouse model [192]. Melatonin and coenzyme Q10 exerted anti-aging effects on mouse oocytes through modulating mitochondrial activity and ROS levels during reproductive aging [193,194,195].

8. Postovulatory Aging and ROS Production by Mammalian Oocytes

8.1. Effect of Postovulatory Aging on Oocyte ROS Production

Ovulation is a complex process involving the rupture of the dominant ovarian follicle and the release of oocytes into the uterine tube [196]. The oocytes of domestic mammals are ovulated during the metaphase of the second meiotic (MII) division and stay in this state until fertilization [197]. Fertilization occurs when male and female gametes fuse within 10 h of ovulation in most domestic animals [198]; however, it can take up to 15 h in mice [199]. If an oocyte waits for a prolonged time without being fertilized by a viable sperm after ovulation, it will go through a series of deteriorating changes known as oocyte postovulatory aging [200].
The postovulatory aging of oocytes involves cellular and molecular alterations that reduce the developmental competence of oocytes [12]. The structural integrity of numerous oocyte components, including the zona pellucida, mitochondria, and meiotic spindle, has been shown to be adversely affected by postovulatory aging [201,202]. Furthermore, postovulatory aging has been linked to biochemical changes in oocytes, including an increase in the formation of ROS in mouse oocytes [203,204]. In postovulatory-aged mouse oocytes, a considerable increase in ROS levels has been linked to cell membrane disruption and DNA damage [205]. Increased ROS generation has been shown to lower intracellular ATP levels in aged bovine oocytes [206], decrease the glutathione (GSH)/glutathione disulfide (GSSG) ratio, and expedite oocyte fragmentation in mouse and pig oocytes [207,208].
Normally, there is a balance between the generation and removal of ROS. A growing body of research implies that the buildup of ROS in postovulatory-aged oocytes is progressively rising with time [209,210]. ROS accumulation is likely to be higher in in vitro matured oocytes than in in vivo matured ones due to laboratory conditions such as light exposure, high oxygen tension, and a lack of antioxidants derived from the surrounding ovarian and tubal microenvironments [8,211]. It was thought that the depletion of intracellular antioxidant defense system supplies in oocytes, such as glutathione, would exacerbate the oxidative damage caused by ROS buildup [212,213]. Consequently, incorporating antioxidant-rich compounds in programs used for both the in vivo and in vitro maturation of domestic mammal oocytes might reduce oxidative damage inflicted by postovulatory aging [202].

8.2. Use of Antioxidants to Counteract ROS-Induced Damage of Postovulatory Aged Oocytes

A number of studies have analyzed the effect of antioxidant supplementation on oocyte postovulatory aging in vitro. L-ascorbic acid and 6-methoxy-2,5,7,8 tetramethylchlormane-2-carboxylic acid (trolox/vitamin E) were shown to be ineffective in preventing oocyte fragmentation during postovulatory aging [214]. The reducing agent dithiothreitol (mda), on the other hand, increased oocyte quality, which resulted in higher rates of fertilization and blastocyst development [214]. Despite its protective effect on aging oocytes, the practical use of DTT is limited by its DNA damaging properties [215]. In vitro aged mouse oocytes treated with melatonin revealed a reduced rate of fragmented oocytes together with a decrease in ROS concentrations compared to counterparts aged without melatonin [210].
Mouse oocytes that were allowed to age in the presence of 200 nM lycopene showed significantly less fragmentation and degeneration, lower concentrations of H2O2 and MDA, and higher concentrations of TAC, GSH, and SOD than those aged without lycopene [216].

9. Conclusions

In vitro maturation (IVM) constitutes the basic foundation for several assisted reproductive techniques including IVEP. Subsequently, the successful practice of IVM holds great promise for the better conduction of these techniques. The disconnection of oocytes from their surrounding in vivo microenvironment during IVM makes them prone to very minor damaging insults. Additionally, the in vitro handling of oocytes during IVM exposes them to supraphysiological levels of stress that could result in their damage. Therefore, the success of an IVM protocol remains a great challenge to reproductive biologists.
The buildup of large amounts of ROS during IVM constitutes a startup point for oxidative stress that might increase the risk of IVEP failure. Among the several laboratory factors that contribute to ROS accumulation within maturing oocytes and IVM media are increased light intensities, high ambient temperatures, and the increased partial pressure of oxygen (PO2). Other factors include maternal senescence, oocyte aging caused by fertilization delays, and inadequate nutrients provided by IVM media (Figure 1). The latter factor is inevitable, as several components of the in vivo milieu cannot be simulated under standard laboratory conditions, e.g., blood circulation and signaling molecules.
The lack of sufficient antioxidant protection for oocytes that is usually achieved by their natural intrafollicular habitat highlights the importance of antioxidant supplementation during IVM. Although the selection of antioxidant(s) to be added during the IVM of mammalian oocytes is a subject of rigorous investigation, generally, these substances are tested and applied at very small doses to achieve physiological levels similar to those found in mammalian biological fluids. As the follicular fluid represents the ideal site for oocyte development, future studies should be aimed at surveying and using species-specific follicular fluid resident factors to help in suppressing ROS activity during IVM. In addition to these factors, the testing of natural antioxidants with previously uncharacterized effects on oocytes and/or the combinatorial use of antioxidants with well-characterized effects on oocytes will definitely help to maximize the success rate of the IVM of mammalian oocytes.

Author Contributions

Conceptualization, S.I.R. and S.M.Z.; software, S.I.R. and M.A.E.; validation, S.I.R. and M.A.E.; formal analysis, S.I.R. and M.A.E.; investigation, S.I.R.; resources, S.I.R. and A.M.M.; data curation, S.I.R. and M.A.E.; writing—original draft preparation, S.I.R.; writing—review and editing, S.I.R., A.B. and M.A.E.; visualization, S.I.R.; supervision, A.M.M. and S.M.Z.; project administration, H.E.-S.A., A.M.M. and S.M.Z.; funding acquisition, S.I.R., H.E.-S.A., A.M.M., and S.M.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors would like to thank all members of the department of Theriogenology, Faculty of Veterinary Medicine at Mansoura University for their help.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Baldassarre, H. Laparoscopic Ovum Pick-Up Followed by In Vitro Embryo Production and Transfer in Assisted Breeding Programs for Ruminants. Animals 2021, 11, 216. [Google Scholar] [CrossRef] [PubMed]
  2. Wani, N.A. In vitro embryo production (IVEP) in camelids, Present status and future perspectives. Reprod. Biol. 2021, 21, 100471. [Google Scholar] [CrossRef] [PubMed]
  3. Baldassarre, H. Practical aspects for implementing in vitro embryo production and cloning programs in sheep and goats. Anim. Reprod. 2018, 9, 188–194. [Google Scholar]
  4. Hatırnaz, Ş.; Ata, B.; Hatırnaz, E.S.; Dahan, M.H.; Tannus, S.; Tan, J.; Tan, S.L. Oocyte in vitro maturation, A sytematic review. Turk. J. Obstet. Gynecol. 2018, 15, 112–125. [Google Scholar] [CrossRef] [PubMed]
  5. Mikkelsen, A.L.; Smith, S.; Lindenberg, S. Possible factors affecting the development of oocytes in in-vitro maturation. Hum. Reprod. 2000, 15 (Suppl. 5), 11–17. [Google Scholar] [CrossRef]
  6. Ye, R.; Xu, S.; Liu, Y.; Pang, L.; Lian, X.; Zhong, Y.; Su, Y.; Wang, S. Protective Effect of Icariin on the Development of Preimplantation Mouse Embryos against Hydrogen Peroxide-Induced Oxidative Injury. Oxid. Med. Cell. Longev. 2017, 2017, 2704532. [Google Scholar] [CrossRef]
  7. Jiang, H.; Liang, S.; Yao, X.R.; Jin, Y.X.; Shen, X.H.; Yuan, B.; Zhang, J.B.; Kim, N.H. Laminarin improves developmental competence of porcine early stage embryos by inhibiting oxidative stress. Theriogenology 2018, 115, 38–44. [Google Scholar] [CrossRef]
  8. Guérin, P.; El Mouatassim, S.; Ménézo, Y. Oxidative stress and protection against reactive oxygen species in the pre-implantation embryo and its surroundings. Hum. Reprod. Update 2001, 7, 175–189. [Google Scholar] [CrossRef]
  9. Khazaei, M.; Aghaz, F. Reactive oxygen species generation and use of antioxidants during in vitro maturation of oocytes. Int. J. Fertil. Steril. 2017, 11, 63–70. [Google Scholar]
  10. Hansen, P.J. Effects of heat stress on mammalian reproduction. Philos. Trans. R. Soc. Lond. Ser. B Biol. Sci. 2009, 364, 3341–3350. [Google Scholar] [CrossRef]
  11. Sasaki, H.; Hamatani, T.; Kamijo, S.; Iwai, M.; Kobanawa, M.; Ogawa, S.; Miyado, K.; Tanaka, M. Impact of oxidative stress on age-associated decline in oocyte developmental competence. Front. Endocrinol. 2019, 10, 811. [Google Scholar] [CrossRef] [PubMed]
  12. McGinnis, L.K.; Pelech, S.; Kinsey, W.H. Post-ovulatory aging of oocytes disrupts kinase signaling pathways and lysosome biogenesis. Mol. Reprod. Dev. 2014, 81, 928–945. [Google Scholar] [CrossRef] [PubMed]
  13. Prasad, S.; Tiwari, M.; Pandey, A.N.; Shrivastav, T.G.; Chaube, S.K. Impact of stress on oocyte quality and reproductive outcome. J. Biomed. Sci. 2016, 23, 36. [Google Scholar] [CrossRef]
  14. Takahashi, M. Oxidative stress and redox regulation on in vitro development of mammalian embryos. J. Reprod. Dev. 2012, 58, 1–9. [Google Scholar] [CrossRef]
  15. Silva, E.; Greene, A.F.; Strauss, K.; Herrick, J.R.; Schoolcraft, W.B.; Krisher, R.L. Antioxidant supplementation during in vitro culture improves mitochondrial function and development of embryos from aged female mice. Reprod. Fertil. Dev. 2015, 27, 975–983. [Google Scholar] [CrossRef]
  16. Ferreira, E.; Vireque, A.; Adona, P.; Meirelles, F.; Ferriani, R.; Navarro, P. Cytoplasmic maturation of bovine oocytes: Structural and biochemical modifications and acquisition of developmental competence. Theriogenology 2009, 71, 836–848. [Google Scholar] [CrossRef]
  17. Rao, B.; Naidu, K.; Amarnath, D.; Vagdevi, R.; Rao, A.; Brahmaiah, K.; Rao, V. In vitro maturation of sheep oocytes in different media during breeding and non-breeding seasons. Small Rumin. Res. 2002, 43, 31–36. [Google Scholar] [CrossRef]
  18. Gupta, P.; Ravindra, J.; Kumar, V.G.; Raghu, H.; Nandi, S. Stimulation of in vitro ovine oocyte maturation with a novel peptide isolated from follicular fluid of the buffalo (Bubalus bubalis). Small Rumin. Res. 2005, 59, 33–40. [Google Scholar] [CrossRef]
  19. Miclea, I.; Hettig, A.; Zahan, M.; Roman, I.; Miclea, V. The effect of several α-tocopherol concentrations on swine oocyte maturation and embryo culture. Bull. UASVM Anim. Sci. Biotechnol. 2009, 66, 385–392. [Google Scholar]
  20. Watson, A. Oocyte cytoplasmic maturation: A key mediator of oocyte and embryo developmental competence. J. Anim. Sci. 2007, 85, E1–E3. [Google Scholar] [CrossRef]
  21. Mao, L.; Lou, H.; Lou, Y.; Wang, N.; Jin, F. Behaviour of cytoplasmic organelles and cytoskeleton during oocyte maturation. Reprod. BioMed. Online 2014, 28, 284–299. [Google Scholar] [CrossRef] [PubMed]
  22. Sun, S.-C.; Kim, N.-H. Molecular mechanisms of asymmetric division in oocytes. Microsc. Microanal. 2013, 19, 883–897. [Google Scholar] [CrossRef] [PubMed]
  23. Gosden, R.; Lee, B. Portrait of an oocyte: Our obscure origin. J. Clin. Investig. 2010, 120, 973–983. [Google Scholar] [CrossRef] [PubMed]
  24. Almonacid, M.; Al Jord, A.; El-Hayek, S.; Othmani, A.; Coulpier, F.; Lemoine, S.; Miyamoto, K.; Grosse, R.; Klein, C.; Piolot, T. Active fluctuations of the nuclear envelope shape the transcriptional dynamics in oocytes. Dev. Cell 2019, 51, 145–157.e10. [Google Scholar] [CrossRef]
  25. Sun, S.-C.; Wang, Z.-B.; Xu, Y.-N.; Lee, S.-E.; Cui, X.-S.; Kim, N.-H. Arp2/3 complex regulates asymmetric division and cytokinesis in mouse oocytes. PLoS ONE 2011, 6, e18392. [Google Scholar] [CrossRef]
  26. Zeng, J.; Jiang, M.; Wu, X.; Diao, F.; Qiu, D.; Hou, X.; Wang, H.; Li, L.; Li, C.; Ge, J. SIRT 4 is essential for metabolic control and meiotic structure during mouse oocyte maturation. Aging Cell 2018, 17, e12789. [Google Scholar] [CrossRef]
  27. Gilchrist, R.B.; Thompson, J.G. Oocyte maturation: Emerging concepts and technologies to improve developmental potential in vitro. Theriogenology 2007, 67, 6–15. [Google Scholar] [CrossRef]
  28. Eppig, J.J. Coordination of nuclear and cytoplasmic oocyte maturation in eutherian mammals. Reprod. Fertil. Dev. 1996, 8, 485–489. [Google Scholar] [CrossRef]
  29. Combelles, C.M.H.; Cekleniak, N.A.; Racowsky, C.; Albertini, D.F. Assessment of nuclear and cytoplasmic maturation in in-vitro matured human oocytes. Hum. Reprod. 2002, 17, 1006–1016. [Google Scholar] [CrossRef]
  30. Goudet, G.; Bézard, J.; Duchamp, G.; Gérard, N.; Palmer, E. Equine oocyte competence for nuclear and cytoplasmic in vitro maturation: Effect of follicle size and hormonal environment. Biol. Reprod. 1997, 57, 232–245. [Google Scholar] [CrossRef]
  31. Lange Consiglio, A.; Arrighi, S.; Cremonesi, F. Time course of in vitro maturation of compact cumulus horse oocytes after roscovitine-induced meiotic inhibition: Effects on the coordination between nuclear and cytoplasmic maturation. Reprod. Domest. Anim. 2010, 45, e313–e322. [Google Scholar] [CrossRef] [PubMed]
  32. Zhu, X.; Zhao, S.; Xu, S.; Zhang, D.; Zhu, M.; Pan, Q.; Huang, J. Granulosa Cells Improved Mare Oocyte Cytoplasmic Maturation by Providing Collagens. Front. Cell Dev. Biol. 2022, 10, 914735. [Google Scholar] [CrossRef] [PubMed]
  33. Halliwell, B.; Gutteridge, J.M. Free Radicals in Biology and Medicine; Oxford University Press: Oxford, UK, 2015. [Google Scholar]
  34. Oborna, I.; Wojewodka, G.; de Sanctis, J.B.; Fingerova, H.; Svobodova, M.; Brezinova, J.; Hajduch, M.; Novotny, J.; Radova, L.; Radzioch, D. Increased lipid peroxidation and abnormal fatty acid profiles in seminal and blood plasma of normozoospermic males from infertile couples. Hum. Reprod. 2010, 25, 308–316. [Google Scholar] [CrossRef] [PubMed]
  35. Yalçınkaya, E.; Çakıroğlu, Y.; Doğer, E.; Budak, Ö.; Çekmen, M.; Çalışkan, E. Effect of follicular fluid NO, MDA and GSH levels on in vitro fertilization outcomes. J. Turk. Ger. Gynecol. Assoc. 2013, 14, 136–141. [Google Scholar] [CrossRef]
  36. Wiener-Megnazi, Z.; Vardi, L.; Lissak, A.; Shnizer, S.; Reznick, A.Z.; Ishai, D.; Lahav-Baratz, S.; Shiloh, H.; Koifman, M.; Dirnfeld, M. Oxidative stress indices in follicular fluid as measured by the thermochemiluminescence assay correlate with outcome parameters in in vitro fertilization. Fertil. Steril. 2004, 82, 1171–1176. [Google Scholar] [CrossRef] [PubMed]
  37. Sugino, N. Roles of reactive oxygen species in the corpus luteum. Anim. Sci. J. 2006, 77, 556–565. [Google Scholar] [CrossRef]
  38. Combelles, C.M.; Gupta, S.; Agarwal, A. Could oxidative stress influence the in-vitro maturation of oocytes? Reprod. Biomed. Online 2009, 18, 864–880. [Google Scholar] [CrossRef]
  39. Kala, M.; Shaikh, M.V.; Nivsarkar, M. Equilibrium between anti-oxidants and reactive oxygen species: A requisite for oocyte development and maturation. Reprod. Med. Biol. 2017, 16, 28–35. [Google Scholar] [CrossRef]
  40. Chelikani, P.; Fita, I.; Loewen, P.C. Diversity of structures and properties among catalases. Cell. Mol. Life Sci. 2004, 61, 192–208. [Google Scholar] [CrossRef]
  41. Brigelius-Flohé, R. Tissue-specific functions of individual glutathione peroxidases. Free. Radic. Biol. Med. 1999, 27, 951–965. [Google Scholar] [CrossRef]
  42. Johnson, F.; Giulivi, C. Superoxide dismutases and their impact upon human health. Mol. Asp. Med. 2005, 26, 340–352. [Google Scholar] [CrossRef] [PubMed]
  43. Changyong, C.; Yong-Won, S.; Eun-Jin, K.; Sang-Rae, C.; Hyun-Jong, K.; Man-Hye, H.; Jaehee, H.; Dong-Soo, S.; Dawon, K. Synergistic effects of glutathione and β-mercaptoethanol treatment during in vitro maturation of porcine oocytes on early embryonic development in a culture system supplemented with L-cysteine. J. Reprod. Dev. 2010, 56, 575–582. [Google Scholar]
  44. Nishikimi, M.; Yagi, K. Biochemistry and molecular biology of ascorbic acid biosynthesis. Subcell. Biochem. Ascorbic Acid Biochem. Biomed. Cell Biol. 1996, 25, 17–39. [Google Scholar] [CrossRef]
  45. Schneider, C. Chemistry and biology of vitamin E. Mol. Nutr. Food Res. 2005, 49, 7–30. [Google Scholar] [CrossRef]
  46. Zhao, X.M.; Hao, H.S.; Du, W.H.; Zhao, S.J.; Wang, H.Y.; Wang, N.; Wang, D.; Liu, Y.; Qin, T.; Zhu, H.B. Melatonin inhibits apoptosis and improves the developmental potential of vitrified bovine oocytes. J. Pineal Res. 2016, 60, 132–141. [Google Scholar] [CrossRef]
  47. Wu, G.-Q.; Jia, B.-Y.; Li, J.-J.; Fu, X.-W.; Zhou, G.-B.; Hou, Y.-P.; Zhu, S.-E. L-carnitine enhances oocyte maturation and development of parthenogenetic embryos in pigs. Theriogenology 2011, 76, 785–793. [Google Scholar] [CrossRef]
  48. You, J.; Lee, J.; Hyun, S.-H.; Lee, E. L-carnitine treatment during oocyte maturation improves in vitro development of cloned pig embryos by influencing intracellular glutathione synthesis and embryonic gene expression. Theriogenology 2012, 78, 235–243. [Google Scholar] [CrossRef]
  49. Chowdhury, M.; Mesalam, A.; Khan, I.; Joo, M.D.; Lee, K.L.; Xu, L.; Afrin, F.; Kong, I.K. Improved developmental competence in embryos treated with lycopene during in vitro culture system. Mol. Reprod. Dev. 2018, 85, 46–61. [Google Scholar] [CrossRef]
  50. Agarwal, A.; Said, T.M.; Bedaiwy, M.A.; Banerjee, J.; Alvarez, J.G. Oxidative stress in an assisted reproductive techniques setting. Fertil. Steril. 2006, 86, 503–512. [Google Scholar] [CrossRef]
  51. Brunet, S.; Maro, B. Cytoskeleton and cell cycle control during meiotic maturation of the mouse oocyte: Integrating time and space. Reproduction 2005, 130, 801–811. [Google Scholar] [CrossRef]
  52. Leoni, G.G.; Rosati, I.; Succu, S.; Bogliolo, L.; Bebbere, D.; Berlinguer, F.; Ledda, S.; Naitana, S. A low oxygen atmosphere during IVF accelerates the kinetic of formation of in vitro produced ovine blastocysts. Reprod. Domest. Anim. 2007, 42, 299–304. [Google Scholar] [CrossRef] [PubMed]
  53. Liebel, F.; Kaur, S.; Ruvolo, E.; Kollias, N.; Southall, M.D. Irradiation of skin with visible light induces reactive oxygen species and matrix-degrading enzymes. J. Investig. Dermatol. 2012, 132, 1901–1907. [Google Scholar] [CrossRef] [PubMed]
  54. Khoudja, R.Y.; Xu, Y.; Li, T.; Zhou, C. Better IVF outcomes following improvements in laboratory air quality. J. Assist. Reprod. Genet. 2013, 30, 69–76. [Google Scholar] [CrossRef] [PubMed]
  55. Tiwari, M.; Prasad, S.; Tripathi, A.; Pandey, A.N.; Singh, A.K.; Shrivastav, T.G.; Chaube, S.K. Involvement of reactive oxygen species in meiotic cell cycle regulation and apoptosis in mammalian oocytes. React. Oxyg. Species 2016, 1, 110–116. [Google Scholar] [CrossRef]
  56. Johnson, M.H.; Nasresfahani, M.H. Radical solutions and cultural problems: Could free oxygen radicals be responsible for the impaired development of preimplantation mammalian embryos in vitro? Bioessays 1994, 16, 31–38. [Google Scholar] [CrossRef]
  57. Velez-Pardo, C.; Morales, A.T.; del Rio, M.J.; Olivera-Angel, M. Endogenously generated hydrogen peroxide induces apoptosis via mitochondrial damage independent of NF-κB and p53 activation in bovine embryos. Theriogenology 2007, 67, 1285–1296. [Google Scholar] [CrossRef]
  58. Fujitani, Y.; Kasai, K.; Ohtani, S.; Nishimura, K.; Yamada, M.; Utsumi, K. Effect of oxygen concentration and free radicals on in vitro development of in vitro-produced bovine embryos. J. Anim. Sci. 1997, 75, 483–489. [Google Scholar] [CrossRef] [PubMed]
  59. Olson, S.; Seidel, G. Culture of in vitro-produced bovine embryos with vitamin E improves development in vitro and after transfer to recipients. Biol. Reprod. 2000, 62, 248–252. [Google Scholar] [CrossRef]
  60. Fischer, B.; Bavister, B. Oxygen tension in the oviduct and uterus of rhesus monkeys, hamsters and rabbits. J. Reprod. Fertil. 1993, 99, 673–679. [Google Scholar] [CrossRef]
  61. Kaufman, D.L.; Mitchell, J.A. Intrauterine oxygen tension during the oestrous cycle in the hamster: Patterns of change. Comp. Biochem. Physiol. Part A Physiol. 1994, 107, 673–678. [Google Scholar] [CrossRef]
  62. Thompson, J.; Simpson, A.; Pugh, P.; Donnelly, P.; Tervit, H. Effect of oxygen concentration on in-vitro development of preimplantation sheep and cattle embryos. J. Reprod. Fertil. 1990, 89, 573–578. [Google Scholar] [CrossRef] [PubMed]
  63. Batt, P.; Gardner, D.; Cameron, A. Oxygen concentration and protein source affect the development of preimplantation goat embryos in vitro. Reprod. Fertil. Dev. 1991, 3, 601–607. [Google Scholar] [CrossRef] [PubMed]
  64. Booth, P.J.; Holm, P.; Callesen, H. The effect of oxygen tension on porcine embryonic development is dependent on embryo type. Theriogenology 2005, 63, 2040–2052. [Google Scholar] [CrossRef] [PubMed]
  65. Rinaudo, P.F.; Giritharan, G.; Talbi, S.; Dobson, A.T.; Schultz, R.M. Effects of oxygen tension on gene expression in preimplantation mouse embryos. Fertil. Steril. 2006, 86, 1252–1265. [Google Scholar] [CrossRef]
  66. Ciray, H.N.; Aksoy, T.; Yaramanci, K.; Karayaka, I.; Bahceci, M. In vitro culture under physiologic oxygen concentration improves blastocyst yield and quality: A prospective randomized survey on sibling oocytes. Fertil. Steril. 2009, 91, 1459–1461. [Google Scholar] [CrossRef]
  67. Nakayama, T.; Noda, Y.; Goto, Y.; Mori, T. Effects of visible light and other environmental factors on the production of oxygen radicals by hamster embryos. Theriogenology 1994, 41, 499–510. [Google Scholar] [CrossRef]
  68. Hashimoto, S.; Minami, N.; Yamada, M.; Imai, H. Excessive concentration of glucose during in vitro maturation impairs the developmental competence of bovine oocytes after in vitro fertilization: Relevance to intracellular reactive oxygen species and glutathione contents. Mol. Reprod. Dev. 2000, 56, 520–526. [Google Scholar] [CrossRef]
  69. Vandaele, L.; Thys, M.; Bijttebier, J.; van Langendonckt, A.; Donnay, I.; Maes, D.; Meyer, E.; van Soom, A. Short-term exposure to hydrogen peroxide during oocyte maturation improves bovine embryo development. Reproduction 2010, 139, 505–511. [Google Scholar] [CrossRef]
  70. Tatemoto, H.; Sakurai, N.; Muto, N. Protection of porcine oocytes against apoptotic cell death caused by oxidative stress during in vitro maturation: Role of cumulus cells. Biol. Reprod. 2000, 63, 805–810. [Google Scholar] [CrossRef]
  71. Luciano, A.M.; Goudet, G.; Perazzoli, F.; Lahuec, C.; Gérard, N. Glutathione content and glutathione peroxidase expression in in vivo and in vitro matured equine oocytes. Mol. Reprod. Dev. 2006, 73, 658–666. [Google Scholar] [CrossRef] [PubMed]
  72. Cetica, P.; Pintos, L.; Dalvit, G.; Beconi, M. Antioxidant enzyme activity and oxidative stress in bovine oocyte in vitro maturation. IUBMB Life 2001, 51, 57–64. [Google Scholar] [CrossRef] [PubMed]
  73. Zhang, L.; Jiang, S.; Wozniak, P.J.; Yang, X.; Godke, R.A. Cumulus cell function during bovine oocyte maturation, fertilization, and embryo development in vitro. Mol. Reprod. Dev. 1995, 40, 338–344. [Google Scholar] [CrossRef] [PubMed]
  74. Tamura, H.; Takasaki, A.; Miwa, I.; Taniguchi, K.; Maekawa, R.; Asada, H.; Taketani, T.; Matsuoka, A.; Yamagata, Y.; Shimamura, K. Oxidative stress impairs oocyte quality and melatonin protects oocytes from free radical damage and improves fertilization rate. J. Pineal Res. 2008, 44, 280–287. [Google Scholar] [CrossRef] [PubMed]
  75. Tatemoto, H.; Muto, N.; Sunagawa, I.; Shinjo, A.; Nakada, T. Protection of porcine oocytes against cell damage caused by oxidative stress during in vitro maturation: Role of superoxide dismutase activity in porcine follicular fluid. Biol. Reprod. 2004, 71, 1150–1157. [Google Scholar] [CrossRef]
  76. Takami, M.; Preston, S.; Toyloy, V.; Behrman, H.R. Antioxidants reversibly inhibit the spontaneous resumption of meiosis. Am. J. Physiol.-Endocrinol. Metab. 1999, 276, E684–E688. [Google Scholar] [CrossRef]
  77. Tarin, J.J. Potential effects of age-associated oxidative stress on mammalian oocytes/embryos. Mol. Hum. Reprod. 1996, 2, 717–724. [Google Scholar] [CrossRef]
  78. Hassold, T.; Hunt, P. To err (meiotically) is human: The genesis of human aneuploidy. Nat. Rev. Genet. 2001, 2, 280–291. [Google Scholar] [CrossRef] [PubMed]
  79. Tarin, J.; Vendrell, F.J.; Ten, J.; Cano, A. Antioxidant therapy counteracts the disturbing effects of diamide and maternal ageing on meiotic division and chromosomal segregation in mouse oocytes. Mol. Hum. Reprod. 1998, 4, 281–288. [Google Scholar] [CrossRef]
  80. Sanfins, A.; Plancha, C.E.; Overstrom, E.W.; Albertini, D.F. Meiotic spindle morphogenesis in in vivo and in vitro matured mouse oocytes: Insights into the relationship between nuclear and cytoplasmic quality. Hum. Reprod. 2004, 19, 2889–2899. [Google Scholar] [CrossRef]
  81. Li, Y.; Feng, H.-L.; Cao, Y.-J.; Zheng, G.-J.; Yang, Y.; Mullen, S.; Critser, J.K.; Chen, Z.-J. Confocal microscopic analysis of the spindle and chromosome configurations of human oocytes matured in vitro. Fertil. Steril. 2006, 85, 827–832. [Google Scholar] [CrossRef]
  82. Sanfins, A.; Lee, G.Y.; Plancha, C.E.; Overstrom, E.W.; Albertini, D.F. Distinctions in Meiotic Spindle Structure and Assembly During In Vitro and In Vivo Maturation of Mouse Oocytes. Biol. Reprod. 2003, 69, 2059–2067. [Google Scholar] [CrossRef] [PubMed]
  83. Choi, W.-J.; Banerjee, J.; Falcone, T.; Bena, J.; Agarwal, A.; Sharma, R.K. Oxidative stress and tumor necrosis factor–α–induced alterations in metaphase II mouse oocyte spindle structure. Fertil. Steril. 2007, 88, 1220–1231. [Google Scholar] [CrossRef] [PubMed]
  84. Morado, S.A.; Cetica, P.D.; Beconi, M.T.; Dalvit, G.C. Reactive oxygen species in bovine oocyte maturation in vitro. Reprod. Fertil. Dev. 2009, 21, 608–614. [Google Scholar] [CrossRef] [PubMed]
  85. Romero, S.; Pella, R.; Zorrilla, I.; Berrío, P.; Escudero, F.; Pérez, Y.; García, M.; Gonzalez, C.; Orihuela, P. Coenzyme Q10 improves the in vitro maturation of oocytes exposed to the intrafollicular environment of patients on fertility treatment. JBRA Assist. Reprod. 2020, 24, 283–288. [Google Scholar] [CrossRef] [PubMed]
  86. Sovernigo, T.; Adona, P.; Monzani, P.; Guemra, S.; Barros, F.; Lopes, F.; Leal, C. Effects of supplementation of medium with different antioxidants during in vitro maturation of bovine oocytes on subsequent embryo production. Reprod. Domest. Anim. 2017, 52, 561–569. [Google Scholar] [CrossRef] [PubMed]
  87. Budani, M.C.; Tiboni, G.M. Effects of Supplementation with Natural Antioxidants on Oocytes and Preimplantation Embryos. Antioxidants 2020, 9, 612. [Google Scholar] [CrossRef]
  88. Kang, J.T.; Koo, O.J.; Kwon, D.K.; Park, H.J.; Jang, G.; Kang, S.K.; Lee, B.C. Effects of melatonin on in vitro maturation of porcine oocyte and expression of melatonin receptor RNA in cumulus and granulosa cells. J. Pineal Res. 2009, 46, 22–28. [Google Scholar] [CrossRef]
  89. Zhao, X.-M.; Min, J.-T.; Du, W.-H.; Hao, H.-S.; Liu, Y.; Qin, T.; Wang, D.; Zhu, H.-B. Melatonin enhances the in vitro maturation and developmental potential of bovine oocytes denuded of the cumulus oophorus. Zygote 2015, 23, 525–536. [Google Scholar] [CrossRef]
  90. Nagina, G.; Asima, A.; Nemat, U.; Shamim, A. Effect of melatonin on maturation capacity and fertilization of Nili-Ravi buffalo (Bubalus bubalis) oocytes. Open Vet. J. 2016, 6, 128–134. [Google Scholar] [CrossRef]
  91. Tian, X.; Wang, F.; Zhang, L.; He, C.; Ji, P.; Wang, J.; Zhang, Z.; Lv, D.; Abulizi, W.; Wang, X.; et al. Beneficial Effects of Melatonin on the In Vitro Maturation of Sheep Oocytes and Its Relation to Melatonin Receptors. Int. J. Mol. Sci. 2017, 18, 834. [Google Scholar] [CrossRef]
  92. Keshavarzi, S.; Salehi, M.; Farifteh-Nobijari, F.; Hosseini, T.; Hosseini, S.; Ghazifard, A.; Novin, M.G.; Fallah-Omrani, V.; Nourozian, M.; Hosseini, A. Melatonin modifies histone acetylation during in vitro maturation of mouse oocytes. Cell J. 2018, 20, 244. [Google Scholar] [PubMed]
  93. Chowdhury, M.; Choi, B.-H.; Khan, I.; Lee, K.-L.; Mesalam, A.; Song, S.-H.; Xu, L.; Joo, M.-D.; Afrin, F.; Kong, I.-K. Supplementation of lycopene in maturation media improves bovine embryo quality in vitro. Theriogenology 2017, 103, 173–184. [Google Scholar] [CrossRef]
  94. Rakha, S.I.; Elmetwally, M.A.; Ali, H.E.; Balboula, A.Z.; Mahmoud, A.M.; Zaabel, S.M. Lycopene improves maturation rate and antioxidant status of in vitro matured mouse oocytes. Int. J. Vet. Sci. 2022; unpublished manuscript. [Google Scholar]
  95. Xiang, D.-C.; Jia, B.-Y.; Fu, X.-W.; Guo, J.-X.; Hong, Q.-H.; Quan, G.-B.; Wu, G.-Q. Role of astaxanthin as an efficient antioxidant on the in vitro maturation and vitrification of porcine oocytes. Theriogenology 2021, 167, 13–23. [Google Scholar] [CrossRef] [PubMed]
  96. Patel, P.A.; Chaudhary, S.S.; Puri, G.; Singh, V.K.; Odedara, A.B. Effects of β-mercaptoethanol on in vitro maturation and glutathione level of buffalo oocytes. Vet. World 2015, 8, 213–216. [Google Scholar] [CrossRef]
  97. Merlo, B.; Iacono, E.; Bucci, D.; Spinaci, M.; Galeati, G.; Mari, G. Beta-mercaptoethanol supplementation of in vitro maturation medium does not influence nuclear and cytoplasmic maturation of equine oocytes. Reprod. Domest. Anim. 2016, 51, 992–996. [Google Scholar] [CrossRef]
  98. Chen, N.; Liow, S.-L.; Yip, W.-Y.; Tan, L.-G.; Ng, S.-C. Influence of cysteamine supplementation and culture in portable dry-incubator on the in vitro maturation, fertilization and subsequent development of mouse oocytes. Theriogenology 2005, 63, 2300–2310. [Google Scholar] [CrossRef]
  99. Nadri, B.; Zeinoaldini, S.; Kohram, H. Ascorbic acid effects on in vitro maturation of mouse oocyte with or without cumulus cell. Afr. J. Biotechnol. 2009, 8, 5627–5631. [Google Scholar]
  100. Dalvit, G.; Llanes, S.; Descalzo, A.; Insani, M.; Beconi, M.; Cetica, P. Effect of alpha-tocopherol and ascorbic acid on bovine oocyte in vitro maturation. Reprod. Domest. Anim. 2005, 40, 93–97. [Google Scholar] [CrossRef]
  101. Tareq, K.; Akter, Q.S.; Mam, Y.; Tsujii, H. Selenium and vitamin E improve the in vitro maturation, fertilization and culture to blastocyst of porcine oocytes. J. Reprod. Dev. 2012, 58, 621–628. [Google Scholar] [CrossRef]
  102. Wang, F.; Tian, X.; Zhang, L.; He, C.; Ji, P.; Li, Y.; Tan, D.; Liu, G. Beneficial effect of resveratrol on bovine oocyte maturation and subsequent embryonic development after in vitro fertilization. Fertil. Steril. 2014, 101, 577–586.e1. [Google Scholar] [CrossRef] [PubMed]
  103. Wang, X.; Zhu, X.; Liang, X.; Xu, H.; Liao, Y.; Lu, K.; Lu, S. Effects of resveratrol on in vitro maturation of porcine oocytes and subsequent early embryonic development following somatic cell nuclear transfer. Reprod. Domest. Anim. 2019, 54, 1195–1205. [Google Scholar] [CrossRef] [PubMed]
  104. Cao, Y.; Zhao, H.; Wang, Z.; Zhang, C.; Bian, Y.; Liu, X.; Zhang, C.; Zhang, X.; Zhao, Y. Quercetin promotes in vitro maturation of oocytes from humans and aged mice. Cell Death Dis. 2020, 11, 965. [Google Scholar] [CrossRef] [PubMed]
  105. Fathi, M.; El-Shahat, K. L-carnitine enhances oocyte maturation and improves in vitro development of embryos in dromedary camels (Camelus dromedaries). Theriogenology 2017, 104, 18–22. [Google Scholar] [CrossRef] [PubMed]
  106. Moawad, A.R.; Salama, A.; Badr, M.R.; Fathi, M. Beneficial Effects of L-Carnitine Supplementation during IVM of Canine Oocytes on Their Nuclear Maturation and Development In Vitro. Animals 2021, 11, 581. [Google Scholar] [CrossRef]
  107. Pu, Y.; Wang, Z.; Bian, Y.; Zhang, F.; Yang, P.; Li, Y.; Zhang, Y.; Liu, Y.; Fang, F.; Cao, H. All-trans retinoic acid improves goat oocyte nuclear maturation and reduces apoptotic cumulus cells during in vitro maturation. Anim. Sci. J. 2014, 85, 833–839. [Google Scholar] [CrossRef]
  108. Saadeldin, I.M.; Swelum, A.A.; Elsafadi, M.; Mahmood, A.; Yaqoob, S.H.; Alfayez, M.; Alowaimer, A.N. Effects of all-trans retinoic acid on the in vitro maturation of camel (Camelus dromedarius) cumulus-oocyte complexes. J. Reprod. Dev. 2019, 65, 215–221. [Google Scholar] [CrossRef]
  109. Maside, C.; Martinez, C.A.; Cambra, J.M.; Lucas, X.; Martinez, E.A.; Gil, M.A.; Rodriguez-Martinez, H.; Parrilla, I.; Cuello, C. Supplementation with exogenous coenzyme Q10 to media for in vitro maturation and embryo culture fails to promote the developmental competence of porcine embryos. Reprod. Domest. Anim. 2019, 54, 72–77. [Google Scholar] [CrossRef]
  110. Ma, L.; Cai, L.; Hu, M.; Wang, J.; Xie, J.; Xing, Y.; Shen, J.; Cui, Y.; Liu, X.J.; Liu, J. Coenzyme Q10 supplementation of human oocyte in vitro maturation reduces postmeiotic aneuploidies. Fertil. Steril. 2020, 114, 331–337. [Google Scholar] [CrossRef]
  111. Click, R.E. A review: Alteration of in vitro reproduction processes by thiols—Emphasis on 2-mercaptoethanol. J. Reprod. Dev. 2014, 60, 399–405. [Google Scholar] [CrossRef]
  112. Deponte, M. The Incomplete Glutathione Puzzle, Just Guessing at Numbers and Figures? Antioxid. Redox Signal. 2017, 27, 1130–1161. [Google Scholar] [CrossRef] [PubMed]
  113. Gardiner, C.S.; Salmen, J.J.; Brandt, C.J.; Stover, S.K. Glutathione is present in reproductive tract secretions and improves development of mouse embryos after chemically induced glutathione depletion. Biol. Reprod. 1998, 59, 431–436. [Google Scholar] [CrossRef] [PubMed]
  114. Ali, A.; Bilodeau, J.; Sirard, M. Antioxidant requirements for bovine oocytes varies during in vitro maturation, fertilization and development. Theriogenology 2003, 59, 939–949. [Google Scholar] [CrossRef]
  115. Janjic, D.; Wollheim, C.B. Effect of 2-mercaptoethanol on glutathione levels, cystine uptake and insulin secretion in insulin-secreting cells. Eur. J. Biochem. 1992, 210, 297–304. [Google Scholar] [CrossRef] [PubMed]
  116. Ishii, T.; Bannai, S.; Sugita, Y. Mechanism of growth stimulation of L1210 cells by 2-mercaptoethanol in vitro. Role of the mixed disulfide of 2-mercaptoethanol and cysteine. J. Biol. Chem. 1981, 256, 12387–12392. [Google Scholar]
  117. de Matos, D.G.; Furnus, C.C.; Moses, D.F.; Baldassarre, H. Effect of cysteamine on glutathione level and developmental capacity of bovine oocyte matured in vitro. Mol. Reprod. Dev. 1995, 42, 432–436. [Google Scholar] [CrossRef]
  118. de Matos, D.G.; Gasparrini, B.; Pasqualini, S.R.; Thompson, J.G. Effect of glutathione synthesis stimulation during in vitro maturation of ovine oocytes on embryo development and intracellular peroxide content. Theriogenology 2002, 57, 1443–1451. [Google Scholar] [CrossRef]
  119. Kitagawa, Y.; Suzuki, K.; Yoneda, A.; Watanabe, T. Effects of oxygen concentration and antioxidants on the in vitro developmental ability, production of reactive oxygen species (ROS), and DNA fragmentation in porcine embryos. Theriogenology 2004, 62, 1186–1197. [Google Scholar] [CrossRef]
  120. Kim, M.K.; Fibrianto, Y.H.; Oh, H.J.; Jang, G.; Kim, H.J.; Lee, K.S.; Kang, S.K.; Lee, B.C.; Hwang, W.S. Effect of beta-mercaptoethanol or epidermal growth factor supplementation on in vitro maturation of canine oocytes collected from dogs with different stages of the estrus cycle. J. Vet. Sci. 2004, 5, 253–258. [Google Scholar] [CrossRef]
  121. Takahashi, M.; Nagai, T.; Okamura, N.; Takahashi, H.; Okano, A. Promoting Effect of β-Mercaptoethanol on In Vitro Development under Oxidative Stress and Cystine Uptake of Bovine Embryos. Biol. Reprod. 2002, 66, 562–567. [Google Scholar] [CrossRef]
  122. Pandey, K.B.; Rizvi, S.I. Plant polyphenols as dietary antioxidants in human health and disease. Oxid. Med. Cell. Longev. 2009, 2, 270–278. [Google Scholar] [CrossRef] [PubMed]
  123. An, F.; Wang, S.; Yuan, D.; Gong, Y.; Wang, S. Attenuation of oxidative stress of erythrocytes by plant-derived flavonoids, orientin and luteolin. Evid.-Based Complementary Altern. Med. 2016, 2016, 3401269. [Google Scholar] [CrossRef] [PubMed]
  124. Park, S.H.; Jeong, P.-S.; Joo, Y.E.; Kang, H.-G.; Kim, M.J.; Lee, S.; Song, B.-S.; Kim, S.-U.; Cho, S.-K.; Sim, B.-W. Luteolin orchestrates porcine oocyte meiotic progression by maintaining organelle dynamics under oxidative stress. Front. Cell Dev. Biol. 2021, 9, 1510. [Google Scholar] [CrossRef] [PubMed]
  125. El-Raey, M.; Geshi, M.; Somfai, T.; Kaneda, M.; Hirako, M.; Abdel-Ghaffar, A.E.; Sosa, G.A.; El-Roos, M.E.; Nagai, T. Evidence of melatonin synthesis in the cumulus oocyte complexes and its role in enhancing oocyte maturation in vitro in cattle. Mol. Reprod. Dev. 2011, 78, 250–262. [Google Scholar] [CrossRef] [PubMed]
  126. Sampaio, R.V.; Conceição, D.S.B.; Miranda, M.S.; de Fatima, S.S.L.; Ohashi, O.M. MT3 melatonin binding site, MT1 and MT2 melatonin receptors are present in oocyte, but only MT1 is present in bovine blastocyst produced in vitro. Reprod. Biol. Endocrinol. 2012, 10, 103. [Google Scholar] [CrossRef]
  127. Wang, J.; Zhuo, Z.; Ma, X.; Liu, Y.; Xu, J.; He, C.; Fu, Y.; Wang, F.; Ji, P.; Zhang, L.; et al. Melatonin Alleviates the Suppressive Effect of Hypoxanthine on Oocyte Nuclear Maturation and Restores Meiosis via the Melatonin Receptor 1 (MT1)-Mediated Pathway. Front. Cell Dev. Biol. 2021, 9, 648148. [Google Scholar] [CrossRef]
  128. Cebrian-Serrano, A.; Salvador, I.; Raga, E.; Dinnyes, A.; Silvestre, M. Beneficial Effect of Melatonin on Blastocyst In Vitro Production from Heat-Stressed Bovine Oocytes. Reprod. Domest. Anim. 2013, 48, 738–746. [Google Scholar] [CrossRef]
  129. Li, Y.; Zhang, Z.; He, C.; Zhu, K.; Xu, Z.; Ma, T.; Tao, J.; Liu, G. Melatonin protects porcine oocyte in vitro maturation from heat stress. J. Pineal Res. 2015, 59, 365–375. [Google Scholar] [CrossRef]
  130. Tao, Y.; Zhou, B.; Xia, G.; Wang, F.; Wu, Z.; Fu, M. Exposure to L-Ascorbic Acid or α-Tocopherol Facilitates the Development of Porcine Denuded Oocytes from Metaphase I to Metaphase II and Prevents Cumulus Cells from Fragmentation. Reprod. Domest. Anim. 2004, 39, 52–57. [Google Scholar] [CrossRef]
  131. Goodwin, T. The Biochemistry of the Carotenoids: Volume I Plants; Springer Science & Business Media: Berlin/Heidelberg, Germany, 2012. [Google Scholar]
  132. Yabuzaki, J. Carotenoids Database: Structures, chemical fingerprints and distribution among organisms. Database 2017, 2017, bax004. [Google Scholar] [CrossRef]
  133. Nagao, A. Metabolism of Carotenoids in Mammals. In Carotenoids, Biosynthetic and Biofunctional Approaches; Springer: Berlin/Heidelberg, Germany, 2021; pp. 67–78. [Google Scholar]
  134. Stahl, W.; Sies, H. Antioxidant activity of carotenoids. Mol. Asp. Med. 2003, 24, 345–351. [Google Scholar] [CrossRef]
  135. Jackson, H.; Braun, C.L.; Ernst, H. The chemistry of novel xanthophyll carotenoids. Am. J. Cardiol. 2008, 101, S50–S57. [Google Scholar] [CrossRef] [PubMed]
  136. Langi, P.; Kiokias, S.; Varzakas, T.; Proestos, C. Carotenoids, From Plants to Food and Feed Industries. Methods Mol. Biol. 2018, 1852, 57–71. [Google Scholar] [PubMed]
  137. Yu, S.; Zhao, Y.; Feng, Y.; Zhang, H.; Li, L.; Shen, W.; Zhao, M.; Min, L. β-carotene improves oocyte development and maturation under oxidative stress in vitro. In Vitro Cell. Dev. Biol. Anim. 2019, 55, 548–558. [Google Scholar] [CrossRef] [PubMed]
  138. Surai, P. The antioxidant properties of canthaxanthin and its potential effects in the poultry eggs and on embryonic development of the chick. Part 1. World’s Poult. Sci. J. 2012, 68, 465–476. [Google Scholar] [CrossRef]
  139. Baker, R.; Günther, C. The role of carotenoids in consumer choice and the likely benefits from their inclusion into products for human consumption. Trends Food Sci. Technol. 2004, 15, 484–488. [Google Scholar] [CrossRef]
  140. Taweechaipaisankul, A.; Jin, J.X.; Lee, S.; Kim, G.A.; Lee, B.C. The effects of canthaxanthin on porcine oocyte maturation and embryo development in vitro after parthenogenetic activation and somatic cell nuclear transfer. Reprod. Domest. Anim. 2016, 51, 870–876. [Google Scholar] [CrossRef]
  141. Do, L.T.K.; Luu, V.V.; Morita, Y.; Taniguchi, M.; Nii, M.; Peter, A.T.; Otoi, T. Astaxanthin present in the maturation medium reduces negative effects of heat shock on the developmental competence of porcine oocytes. Reprod. Biol. 2015, 15, 86–93. [Google Scholar] [CrossRef]
  142. Bose, B.K. Global warming, Energy, environmental pollution, and the impact of power electronics. IEEE Ind. Electron. Mag. 2010, 4, 6–17. [Google Scholar] [CrossRef]
  143. Boni, R. Heat stress, a serious threat to reproductive function in animals and humans. Mol. Reprod. Dev. 2019, 86, 1307–1323. [Google Scholar] [CrossRef]
  144. Roth, Z. Effect of Heat Stress on Reproduction in Dairy Cows, Insights into the Cellular and Molecular Responses of the Oocyte. Annu. Rev. Anim. Biosci. 2017, 5, 151–170. [Google Scholar] [CrossRef] [PubMed]
  145. Ahmed, J.A.; Nashiruddullah, N.; Dutta, D.; Biswas, R.K.; Borah, P. Cumulus cell expansion and ultrastructural changes in in vitro matured bovine oocytes under heat stress. Iran. J. Vet. Res. 2017, 18, 203–207. [Google Scholar]
  146. Andreu-Vázquez, C.; López-Gatius, F.; García-Ispierto, I.; Maya-Soriano, M.J.; Hunter, R.H.; López-Béjar, M. Does heat stress provoke the loss of a continuous layer of cortical granules beneath the plasma membrane during oocyte maturation? Zygote 2010, 18, 293–299. [Google Scholar] [CrossRef] [PubMed]
  147. Sakatani, M. Effects of heat stress on bovine preimplantation embryos produced in vitro. J. Reprod. Dev. 2017, 63, 347–352. [Google Scholar] [CrossRef] [PubMed]
  148. Diaz, F.A.; Gutierrez-Castillo, E.J.; Foster, B.A.; Hardin, P.T.; Bondioli, K.R.; Jiang, Z. Evaluation of Seasonal Heat Stress on Transcriptomic Profiles and Global DNA Methylation of Bovine Oocytes. Front. Genet. 2021, 12, 699920. [Google Scholar] [CrossRef] [PubMed]
  149. Waiz, S.A.; Raies-Ul-Haq, M.; Dhanda, S.; Kumar, A.; Goud, T.S.; Chauhan, M.S.; Upadhyay, R.C. Heat stress and antioxidant enzyme activity in bubaline (Bubalus bubalis) oocytes during in vitro maturation. Int. J. Biometeorol. 2016, 60, 1357–1366. [Google Scholar] [CrossRef]
  150. Wang, J.Z.; Sui, H.S.; Miao, D.Q.; Liu, N.; Zhou, P.; Ge, L.; Tan, J.H. Effects of heat stress during in vitro maturation on cytoplasmic versus nuclear components of mouse oocytes. Reproduction 2009, 137, 181–189. [Google Scholar] [CrossRef]
  151. Hale, B.J.; Li, Y.; Adur, M.K.; Keating, A.F.; Baumgard, L.H.; Ross, J.W. Characterization of the effects of heat stress on autophagy induction in the pig oocyte. Reprod. Biol. Endocrinol. 2021, 19, 107. [Google Scholar] [CrossRef]
  152. Ju, J.C.; Tseng, J.K. Nuclear and cytoskeletal alterations of in vitro matured porcine oocytes under hyperthermia. Mol. Reprod. Dev. 2004, 68, 125–133. [Google Scholar] [CrossRef]
  153. Payton, R.R.; Romar, R.; Coy, P.; Saxton, A.M.; Lawrence, J.L.; Edwards, J.L. Susceptibility of bovine germinal vesicle-stage oocytes from antral follicles to direct effects of heat stress in vitro. Biol. Reprod. 2004, 71, 1303–1308. [Google Scholar] [CrossRef]
  154. Hansen, P.J. Physiological and cellular adaptations of zebu cattle to thermal stress. Anim. Reprod. Sci. 2004, 82–83, 349–360. [Google Scholar] [CrossRef] [PubMed]
  155. Sakatani, M.; Yamanaka, K.; Kobayashi, S.; Takahashi, M. Heat shock-derived reactive oxygen species induce embryonic mortality in in vitro early stage bovine embryos. J. Reprod. Dev. 2008, 54, 496–501. [Google Scholar] [CrossRef] [PubMed]
  156. Ozawa, M.; Hirabayashi, M.; Kanai, Y. Developmental competence and oxidative state of mouse zygotes heat-stressed maternally or in vitro. Reproduction 2002, 124, 683–689. [Google Scholar] [CrossRef] [PubMed]
  157. Nabenishi, H.; Ohta, H.; Nishimoto, T.; Morita, T.; Ashizawa, K.; Tsuzuki, Y. The effects of cysteine addition during in vitro maturation on the developmental competence, ROS, GSH and apoptosis level of bovine oocytes exposed to heat stress. Zygote 2012, 20, 249–259. [Google Scholar] [CrossRef]
  158. Sakatani, M.; Kobayashi, S.; Takahashi, M. Effects of heat shock on in vitro development and intracellular oxidative state of bovine preimplantation embryos. Mol. Reprod. Dev. 2004, 67, 77–82. [Google Scholar] [CrossRef]
  159. Paes, V.M.; Vieira, L.A.; Correia, H.H.V.; Sa, N.A.R.; Moura, A.A.A.; Sales, A.D.; Rodrigues, A.P.R.; Magalhães-Padilha, D.M.; Santos, F.W.; Apgar, G.A.; et al. Effect of heat stress on the survival and development of in vitro cultured bovine preantral follicles and on in vitro maturation of cumulus-oocyte complex. Theriogenology 2016, 86, 994–1003. [Google Scholar] [CrossRef]
  160. Yin, C.; Liu, J.; He, B.; Jia, L.; Gong, Y.; Guo, H.; Zhao, R. Heat stress induces distinct responses in porcine cumulus cells and oocytes associated with disrupted gap junction and trans-zonal projection colocalization. J. Cell. Physiol. 2019, 234, 4787–4798. [Google Scholar] [CrossRef]
  161. Gendelman, M.; Aroyo, A.; Yavin, S.; Roth, Z. Seasonal effects on gene expression, cleavage timing, and developmental competence of bovine preimplantation embryos. Reproduction 2010, 140, 73–82. [Google Scholar] [CrossRef]
  162. Edwards, J.L.; Bogart, A.N.; Rispoli, L.A.; Saxton, A.M.; Schrick, F.N. Developmental competence of bovine embryos from heat-stressed ova. J. Dairy Sci. 2009, 92, 563–570. [Google Scholar] [CrossRef]
  163. Ju, J.C.; Jiang, S.; Tseng, J.K.; Parks, J.E.; Yang, X. Heat shock reduces developmental competence and alters spindle configuration of bovine oocytes. Theriogenology 2005, 64, 1677–1689. [Google Scholar] [CrossRef]
  164. Nabenishi, H.; Takagi, S.; Kamata, H.; Nishimoto, T.; Morita, T.; Ashizawa, K.; Tsuzuki, Y. The role of mitochondrial transition pores on bovine oocyte competence after heat stress, as determined by effects of cyclosporin A. Mol. Reprod. Dev. 2012, 79, 31–40. [Google Scholar] [CrossRef] [PubMed]
  165. Rodrigues, T.A.; Ispada, J.; Risolia, P.H.; Rodrigues, M.T.; Lima, R.S.; Assumpção, M.E.; Visintin, J.A.; Paula-Lopes, F.F. Thermoprotective effect of insulin-like growth factor 1 on in vitro matured bovine oocyte exposed to heat shock. Theriogenology 2016, 86, 2028–2039. [Google Scholar] [CrossRef] [PubMed]
  166. Maya-Soriano, M.J.; Taberner, E.; López-Béjar, M. Retinol improves in vitro oocyte nuclear maturation under heat stress in heifers. Zygote 2013, 21, 377–384. [Google Scholar] [CrossRef] [PubMed]
  167. Kidder, B.L. In vitro maturation and in vitro fertilization of mouse oocytes and preimplantation embryo culture. Methods Mol. Biol. 2014, 1150, 191–199. [Google Scholar] [PubMed]
  168. Paula-Lopes, F.F.; Lima, R.S.; Satrapa, R.A.; Barros, C.M. Physiology and Endocrinology Symposium: Influence of cattle genotype (Bos indicus vs. Bos taurus) on oocyte and preimplantation embryo resistance to increased temperature. J. Anim. Sci. 2013, 91, 1143–1153. [Google Scholar] [PubMed]
  169. El-Sayed, A.; Nagy, R.; El-Asheeri, A.K.; Eid, L.N. Developmental and molecular responses of buffalo (Bubalus bubalis) cumulus-oocyte complex matured in vitro under heat shock conditions. Zygote 2018, 26, 177–190. [Google Scholar] [CrossRef]
  170. Tseng, J.K.; Tang, P.C.; Ju, J.C. In vitro thermal stress induces apoptosis and reduces development of porcine parthenotes. Theriogenology 2006, 66, 1073–1082. [Google Scholar] [CrossRef]
  171. Abdelnour, S.A.; Yang, C.Y.; Swelum, A.A.; El-Hack, M.E.A.; Khafaga, A.F.; Abdo, M.; Shang, J.H.; Lu, Y.Q. Molecular functional, and cellular alterations of oocytes and cumulus cells induced by heat stress and shock in animals. Environ. Sci. Pollut. Res. Int. 2020, 27, 38472–38490. [Google Scholar] [CrossRef]
  172. Lawrence, J.L.; Payton, R.R.; Godkin, J.D.; Saxton, A.M.; Schrick, F.N.; Edwards, J.L. Retinol improves development of bovine oocytes compromised by heat stress during maturation. J. Dairy Sci. 2004, 87, 2449–2454. [Google Scholar] [CrossRef]
  173. Roth, Z.; Aroyo, A.; Yavin, S.; Arav, A. The antioxidant epigallocatechin gallate (EGCG) moderates the deleterious effects of maternal hyperthermia on follicle-enclosed oocytes in mice. Theriogenology 2008, 70, 887–897. [Google Scholar] [CrossRef]
  174. Li, Y.; Wang, J.; Zhang, Z.; Yi, J.; He, C.; Wang, F.; Tian, X.; Yang, M.; Song, Y.; He, P.; et al. Resveratrol compares with melatonin in improving in vitro porcine oocyte maturation under heat stress. J. Anim. Sci. Biotechnol. 2016, 7, 33. [Google Scholar] [CrossRef] [PubMed]
  175. Ispada, J.; Rodrigues, T.A.; Risolia, P.H.B.; Lima, R.S.; Gonçalves, D.R.; Rettori, D.; Nichi, M.; Feitosa, W.B.; Paula-Lopes, F.F. Astaxanthin counteracts the effects of heat shock on the maturation of bovine oocytes. Reprod. Fertil. Dev. 2018, 30, 1169–1179. [Google Scholar] [CrossRef] [PubMed]
  176. Wang, F.; Tian, X.; Zhang, L.; Gao, C.; He, C.; Fu, Y.; Ji, P.; Li, Y.; Li, N.; Liu, G. Beneficial effects of melatonin on in vitro bovine embryonic development are mediated by melatonin receptor 1. J. Pineal Res. 2014, 56, 333–342. [Google Scholar] [CrossRef] [PubMed]
  177. Fernandes, H.; Castro, F.; Schefer, L.; Paschoal, D.; Leal, C. 163 Effect of melatonin and its receptors on bovine oocyte maturation and cumulus cell gene expression after heat shock in vitro: Preliminary results. Reprod. Fertil. Dev. 2019, 31, 206–207. [Google Scholar] [CrossRef]
  178. Garcia-Ispierto, I.; Abdelfatah, A.; López-Gatius, F. Melatonin treatment at dry-off improves reproductive performance postpartum in high-producing dairy cows under heat stress conditions. Reprod. Domest. Anim. 2013, 48, 577–583. [Google Scholar] [CrossRef]
  179. Yaacobi-Artzi, S.; Shimoni, C.; Kalo, D.; Hansen, P.J.; Roth, Z. Melatonin slightly alleviates the effect of heat shock on bovine oocytes and resulting blastocysts. Theriogenology 2020, 158, 477–489. [Google Scholar] [CrossRef]
  180. Gendelman, M.; Roth, Z. Incorporation of coenzyme Q10 into bovine oocytes improves mitochondrial features and alleviates the effects of summer thermal stress on developmental competence. Biol. Reprod. 2012, 87, 118. [Google Scholar]
  181. Templeton, A.; Morris, J.K.; Parslow, W. Factors that affect outcome of in-vitro fertilisation treatment. Lancet 1996, 348, 1402–1406. [Google Scholar] [CrossRef]
  182. Li, Q.; Geng, X.; Zheng, W.; Tang, J.; Xu, B.; Shi, Q. Current understanding of ovarian aging. Sci. China Life Sci. 2012, 55, 659–669. [Google Scholar] [CrossRef]
  183. Igarashi, H.; Takahashi, T.; Nagase, S. Oocyte aging underlies female reproductive aging: Biological mechanisms and therapeutic strategies. Reprod. Med. Biol. 2015, 14, 159–169. [Google Scholar] [CrossRef]
  184. Li, Y.-J.; Han, Z.; Ge, L.; Zhou, C.-J.; Zhao, Y.-F.; Wang, D.-H.; Ren, J.; Niu, X.-X.; Liang, C.-G. C-phycocyanin protects against low fertility by inhibiting reactive oxygen species in aging mice. Oncotarget 2016, 7, 17393. [Google Scholar] [CrossRef] [PubMed]
  185. Yamada-Fukunaga, T.; Yamada, M.; Hamatani, T.; Chikazawa, N.; Ogawa, S.; Akutsu, H.; Miura, T.; Miyado, K.; Tarín, J.J.; Kuji, N. Age-associated telomere shortening in mouse oocytes. Reprod. Biol. Endocrinol. 2013, 11, 108. [Google Scholar] [CrossRef] [PubMed]
  186. Salmon, A.B.; Richardson, A.; Pérez, V.I. Update on the oxidative stress theory of aging: Does oxidative stress play a role in aging or healthy aging? Free. Radic. Biol. Med. 2010, 48, 642–655. [Google Scholar] [CrossRef]
  187. Behrman, H.R.; Kodaman, P.H.; Preston, S.L.; Gao, S. Oxidative stress and the ovary. J. Soc. Gynecol. Investig. 2001, 8, S40–S42. [Google Scholar] [PubMed]
  188. Dunning, K.R.; Russell, D.L.; Robker, R.L. Lipids and oocyte developmental competence: The role of fatty acids and β-oxidation. Reproduction 2014, 148, R15–R27. [Google Scholar] [CrossRef] [PubMed]
  189. Mihalas, B.P.; de Iuliis, G.N.; Redgrove, K.A.; McLaughlin, E.A.; Nixon, B. The lipid peroxidation product 4-hydroxynonenal contributes to oxidative stress-mediated deterioration of the ageing oocyte. Sci. Rep. 2017, 7, 6247. [Google Scholar] [CrossRef]
  190. Simsek-Duran, F.; Li, F.; Ford, W.; Swanson, R.J.; Jones, H.W., Jr.; Castora, F.J. Age-associated metabolic and morphologic changes in mitochondria of individual mouse and hamster oocytes. PLoS ONE 2013, 8, e64955. [Google Scholar] [CrossRef]
  191. Thouas, G.A.; Trounson, A.O.; Jones, G.M. Effect of female age on mouse oocyte developmental competence following mitochondrial injury. Biol. Reprod. 2005, 73, 366–373. [Google Scholar] [CrossRef]
  192. Tarín, J.J.; Pérez-Albalá, S.; Cano, A. Oral antioxidants counteract the negative effects of female aging on oocyte quantity and quality in the mouse. Mol. Reprod. Dev. 2002, 61, 385–397. [Google Scholar] [CrossRef]
  193. Tamura, H.; Kawamoto, M.; Sato, S.; Tamura, I.; Maekawa, R.; Taketani, T.; Aasada, H.; Takaki, E.; Nakai, A.; Reiter, R.J.; et al. Long-term melatonin treatment delays ovarian aging. J. Pineal Res. 2017, 62, e12381. [Google Scholar] [CrossRef]
  194. Ben-Meir, A.; Burstein, E.; Borrego-Alvarez, A.; Chong, J.; Wong, E.; Yavorska, T.; Naranian, T.; Chi, M.; Wang, Y.; Bentov, Y.; et al. Coenzyme Q10 restores oocyte mitochondrial function and fertility during reproductive aging. Aging Cell 2015, 14, 887–895. [Google Scholar] [CrossRef] [PubMed]
  195. Zhang, L.; Zhang, Z.; Wang, J.; Lv, D.; Zhu, T.; Wang, F.; Tian, X.; Yao, Y.; Ji, P.; Liu, G. Melatonin regulates the activities of ovary and delays the fertility decline in female animals via MT1/AMPK pathway. J. Pineal Res. 2019, 66, e12550. [Google Scholar] [CrossRef] [PubMed]
  196. Irianni, F.; Hodgen, G.D. Mechanism of ovulation. Endocrinol. Metab. Clin. N. Am. 1992, 21, 19–38. [Google Scholar] [CrossRef]
  197. Florman, H.M.; Ducibella, T. Fertilization in mammals. Knobil Neill’s Physiol. Reprod. 2006, 3, 55–112. [Google Scholar]
  198. Okabe, M. The cell biology of mammalian fertilization. Development 2013, 140, 4471–4479. [Google Scholar] [CrossRef]
  199. Marston, J.H.; Chang, M.C. The fertilizable life of ova and their morphology following delayed insemination in mature and immature mice. J. Exp. Zool. 1964, 155, 237–251. [Google Scholar] [CrossRef]
  200. Miao, Y.L.; Kikuchi, K.; Sun, Q.Y.; Schatten, H. Oocyte aging: Cellular and molecular changes, developmental potential and reversal possibility. Hum. Reprod. Update 2009, 15, 573–585. [Google Scholar] [CrossRef]
  201. Takahashi, T.; Takahashi, E.; Igarashi, H.; Tezuka, N.; Kurachi, H. Impact of oxidative stress in aged mouse oocytes on calcium oscillations at fertilization. Mol. Reprod. Dev. 2003, 66, 143–152. [Google Scholar] [CrossRef]
  202. Lord, T.; Aitken, R.J. Oxidative stress and ageing of the post-ovulatory oocyte. Reproduction 2013, 146, R217–R227. [Google Scholar] [CrossRef]
  203. Tatone, C.; Carbone, M.C.; Gallo, R.; Monache, S.D.; di Cola, M.; Alesse, E.; Amicarelli, F. Age-associated changes in mouse oocytes during postovulatory in vitro culture: Possible role for meiotic kinases and survival factor BCL2. Biol. Reprod. 2006, 74, 395–402. [Google Scholar] [CrossRef]
  204. Zhang, T.; Zhou, Y.; Li, L.; Wang, H.H.; Ma, X.S.; Qian, W.P.; Shen, W.; Schatten, H.; Sun, Q.Y. SIRT1, 2, 3 protect mouse oocytes from postovulatory aging. Aging 2016, 8, 685–696. [Google Scholar] [CrossRef] [PubMed]
  205. Takahashi, T.; Igarashi, H.; Kawagoe, J.; Amita, M.; Hara, S.; Kurachi, H. Poor embryo development in mouse oocytes aged in vitro is associated with impaired calcium homeostasis. Biol. Reprod. 2009, 80, 493–502. [Google Scholar] [CrossRef] [PubMed]
  206. Koyama, K.; Kang, S.S.; Huang, W.; Yanagawa, Y.; Takahashi, Y.; Nagano, M. Aging-related changes in in vitro-matured bovine oocytes: Oxidative stress, mitochondrial activity and ATP content after nuclear maturation. J. Reprod. Dev. 2014, 60, 136–142. [Google Scholar] [CrossRef]
  207. Tang, D.W.; Fang, Y.; Liu, Z.X.; Wu, Y.; Wang, X.L.; Zhao, S.; Han, G.C.; Zeng, S.M. The disturbances of endoplasmic reticulum calcium homeostasis caused by increased intracellular reactive oxygen species contributes to fragmentation in aged porcine oocytes. Biol. Reprod. 2013, 89, 124. [Google Scholar] [CrossRef] [PubMed]
  208. Asano, Y. Age-related accumulation of non-heme ferric and ferrous iron in mouse ovarian stroma visualized by sensitive non-heme iron histochemistry. J. Histochem. Cytochem. Off. J. Histochem. Soc. 2012, 60, 229–242. [Google Scholar] [CrossRef]
  209. Tatone, C.; di Emidio, G.; Barbaro, R.; Vento, M.; Ciriminna, R.; Artini, P.G. Effects of reproductive aging and postovulatory aging on the maintenance of biological competence after oocyte vitrification: Insights from the mouse model. Theriogenology 2011, 76, 864–873. [Google Scholar] [CrossRef]
  210. Lord, T.; Nixon, B.; Jones, K.T.; Aitken, R.J. Melatonin prevents postovulatory oocyte aging in the mouse and extends the window for optimal fertilization in vitro. Biol. Reprod. 2013, 88, 67. [Google Scholar] [CrossRef] [PubMed]
  211. Goto, Y.; Noda, Y.; Mori, T.; Nakano, M. Increased generation of reactive oxygen species in embryos cultured in vitro. Free Radic. Biol. Med. 1993, 15, 69–75. [Google Scholar] [CrossRef]
  212. Boerjan, M.L.; de Boer, P. First cell cycle of zygotes of the mouse derived from oocytes aged postovulation in vivo and fertilized in vivo. Mol. Reprod. Dev. 1990, 25, 155–163. [Google Scholar] [CrossRef]
  213. Yoshida, M.; Ishigaki, K.; Nagai, T.; Chikyu, M.; Pursel, V.G. Glutathione concentration during maturation and after fertilization in pig oocytes: Relevance to the ability of oocytes to form male pronucleus. Biol. Reprod. 1993, 49, 89–94. [Google Scholar] [CrossRef]
  214. Tarín, J.J.; Ten, J.; Vendrell, F.J.; Cano, A. Dithiothreitol prevents age-associated decrease in oocyte/conceptus viability in vitro. Hum. Reprod. 1998, 13, 381–386. [Google Scholar] [CrossRef] [PubMed]
  215. Oikawa, S.; Hiraku, Y.; Fujiwara, T.; Saito, I.; Kawanishi, S. Site-specific hydroxylation at polyguanosine in double-stranded DNA by nickel(II) in the presence of SH compounds: Comparison with singlet oxygen-induced DNA damage. Chem. Res. Toxicol. 2002, 15, 1017–1022. [Google Scholar] [CrossRef] [PubMed]
  216. Rakha, S.I.; Elmetwally, M.A.; Ali, H.E.; Balboula, A.Z.; Mahmoud, A.M.; Zaabel, S.M. Lycopene Reduces the In Vitro Aging Phenotypes of Mouse Oocytes by Improving Their Oxidative Status. Vet. Sci. 2022, 9, 336. [Google Scholar] [CrossRef] [PubMed]
  217. BioRender. Available online: https://biorender.com/ (accessed on 29 July 2022).
Figure 1. Schematic diagram summarizing the main factors causing oxidative stress of mammalian oocytes during their in vitro maturation. Consequences of oxidative stress on oocyte components are also shown. PO2, partial pressure of oxygen; ROS, reactive oxygen species. The diagram was created with BioRender.com [217].
Figure 1. Schematic diagram summarizing the main factors causing oxidative stress of mammalian oocytes during their in vitro maturation. Consequences of oxidative stress on oocyte components are also shown. PO2, partial pressure of oxygen; ROS, reactive oxygen species. The diagram was created with BioRender.com [217].
Vetsci 09 00439 g001
Table 1. List, concentrations, and effects of antioxidants used to improve in vitro maturation of oocytes in different animals.
Table 1. List, concentrations, and effects of antioxidants used to improve in vitro maturation of oocytes in different animals.
AntioxidantTypeDoseSpeciesMaturation Rate vs. (Control) References
MelatoninHormone4.3 × 10−8 M (10 ng/mL)porcine84.6 (75.6) *[88]
10−9 Mbovine82.3 (65.7) *[89]
2.5 × 10−4 M buffalo 42.8 (33)ns [90]
10−7 M sheep 85.3 (75.3) * [91]
10−6 Mmouse85 (64) *[92]
LycopeneCarotenoid2 × 10−7 Mbovine76 (66.3) *[93]
2 × 10−7 Mmouse89.9 (66.7) *[94]
AstaxanthinCarotenoid2.5 × 10−6 Mporcine89.5 (87.1)ns[95]
Beta-Mercaptoethanol (β-ME)Thiol2 × 10−5 Mbuffalo76.2 (66.7)ns[96]
10−5 Mequine55.6 (51.9)ns[97]
CystamineThiol10−5 Mmouse80.1 (57.7) *[98]
Vitamin CVitamin2.5 × 10−4 Mmouse29.7 (70.3) *[99]
2.3 × 10−3 M (1 mg/mL)bovine~80 (~80)ns[100]
Vitamin EVitamin2.3 × 10−3 M (1 mg/mL)bovine~80 (~80)ns[100]
10−3 Mporcine72.2 (67.6)ns[101]
Selenium (SeMet)Trace element2.5 × 10−8 Mporcine80.2 (67.6) *[101]
Vitamin E; Selenium
(SeMet)
Vitamin; trace element10−3 M; 2.5 × 10−8 Mporcine85.1 (67.6) *[101]
ResveratrolPolyphenolic compound10−6 Mbovine93.4 (87.9) *[102]
5 × 10−6 Mporcine84.5 (72.6) *[103]
QuercetinPolyphenolic compound10−5 Mmouse86.6 (79.7) *[104]
human92.3 (87.5)ns
L-CarnitineAmino acid derivative3.1 × 10−3 M (0.5 mg/mL)porcine60.7 (56.4) *[47]
3.1 × 10−3 M (0.5 mg/mL)camel74.7 (60.2) *[105]
3.7 × 10−3 M (0.6 mg/mL)canine41.4 (23.4) *[106]
Retinoic acidVitamin A metabolite10−8 Mgoat78.7 (65.1) *[107]
2 × 10−5 Mcamel69.4 (52.9) *[108]
Coenzyme Q10Coenzyme10−5 Mporcine76.4 (66)ns[109]
5 × 10−5 Mhuman82.6 (63.0) *[110]
* Maturation rate significantly changed (p < 0.05); ns maturation rate non-significantly changed; SeMet, Seleon-L-methionine.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Rakha, S.I.; Elmetwally, M.A.; El-Sheikh Ali, H.; Balboula, A.; Mahmoud, A.M.; Zaabel, S.M. Importance of Antioxidant Supplementation during In Vitro Maturation of Mammalian Oocytes. Vet. Sci. 2022, 9, 439. https://doi.org/10.3390/vetsci9080439

AMA Style

Rakha SI, Elmetwally MA, El-Sheikh Ali H, Balboula A, Mahmoud AM, Zaabel SM. Importance of Antioxidant Supplementation during In Vitro Maturation of Mammalian Oocytes. Veterinary Sciences. 2022; 9(8):439. https://doi.org/10.3390/vetsci9080439

Chicago/Turabian Style

Rakha, Shimaa I., Mohammed A. Elmetwally, Hossam El-Sheikh Ali, Ahmed Balboula, Abdelmonem Montaser Mahmoud, and Samy M. Zaabel. 2022. "Importance of Antioxidant Supplementation during In Vitro Maturation of Mammalian Oocytes" Veterinary Sciences 9, no. 8: 439. https://doi.org/10.3390/vetsci9080439

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