Next Article in Journal
An Isoform of Nedd4-2 Plays a Pivotal Role in Electrophysiological Cardiac Abnormalities
Next Article in Special Issue
Melatonin as a Novel Interventional Candidate for Fragile X Syndrome with Autism Spectrum Disorder in Humans
Previous Article in Journal
Expression of Salivary miR-203a-3p Was Related with Oral Health-Related Quality of Life in Healthy Volunteers
Previous Article in Special Issue
AA-NAT, MT1 and MT2 Correlates with Cancer Stem-Like Cell Markers in Colorectal Cancer: Study of the Influence of Stage and p53 Status of Tumors
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Melatonin Scavenger Properties against Oxidative and Nitrosative Stress: Impact on Gamete Handling and In Vitro Embryo Production in Humans and Other Mammals

1
Centro de Biotecnología de la Reproducción (BIOREN-CEBIOR), Facultad de Medicina, Universidad de La Frontera, Temuco 4811230, Chile
2
Departamento de Ciencias Preclínicas, Facultad de Medicina, Universidad de La Frontera, Temuco 4811230, Chile
3
Departamento de Producción Agropecuaria, Facultad de Ciencias Agropecuarias y Forestales, Universidad de La Frontera, Temuco 4811230, Chile
4
Departamento de Ciencias Agronómicas y Recursos Naturales, Facultad de Ciencias Agropecuarias y Forestales, Universidad de La Frontera, Temuco 4811230, Chile
5
Departamento de Ciencias Básicas, Universidad de La Frontera, Temuco 4811230, Chile
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2017, 18(6), 1119; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18061119
Submission received: 24 March 2017 / Revised: 19 May 2017 / Accepted: 21 May 2017 / Published: 14 June 2017
(This article belongs to the Special Issue Melatonin and Its Analogues: Experimental and Clinical Aspects)

Abstract

:
Oxidative and nitrosative stress are common problems when handling gametes in vitro. In vitro development in mammalian embryos is highly affected by culture conditions, especially by reactive oxygen species (ROS) and reactive nitrogen species (RNS), because their absence or overproduction causes embryo arrest and changes in gene expression. Melatonin in gamete co-incubation during in vitro fertilization (IVF) has deleterious or positive effects, depending on the concentration used in the culture medium, demonstrating the delicate balance between antioxidant and pro-oxidant activity. Further research is needed to better understand the possible impact of melatonin on the different IVP steps in humans and other mammals, especially in seasonal breeds where this neuro-hormone system highly regulates its reproduction physiology.

Graphical Abstract

1. Introduction

1.1. Free Radicals on Reproduction (ROS/RNS)

The protocols for in vitro maturation, fertilization, and embryo culture in assisted reproductive techniques (ART) have been greatly improved during the last decade. However, only a few embryos produced by ARTs are capable of carrying out development to full term. This is mainly due to the lack of optimal in vitro conditions that cannot mimic the in vivo conditions, leading to several differences between both conditions such as increased levels of ROS or RNS [1]. Both free radicals are generated as sub-products in physiological processes where the oxygen consumption is produced in the electron transport chain during cellular respiration in the mitochondria [2].
There is a duality in the role of ROS and RNS. Physiological levels are needed in several contexts: ROS are necessary in human follicles to establish pregnancy [3], as potential markers in patients for predicting the success of in vitro fertilization (IVF) [4], during the in vitro maturation of oocytes [5], in the resumption of meiosis from diplotene arrested oocytes [6], for stimulating the release of intracellular Ca+2 in oocytes [7], and for stimulating mitogen-activated protein kinases (MAPKs) [8]. In sperm physiology, ROS participate in hyperactivation [9], sperm capacitation [10,11,12,13,14], tyrosine phosphorylation [15], and the acrosome reaction [16].
RNS, on the other side, are necessary for the development of large antral follicles [17,18], to stimulate meiotic maturation in oocytes [19,20], in the ovulatory process [21], in early folliculogenesis up to the maturation step [22] and in preimplantation embryonic development [23,24]. Additionally, RNS participate in sperm capacitation [25,26,27] and the acrosome reaction [28].
When an imbalance between pro-oxidant molecules occurs due to the increase of ROS/RNS levels or the reduction of the antioxidant defense mechanisms, the phenomenon called oxidative or nitrosative stress is triggered [29,30].

1.2. Oxidative Stress

Oxygen (O2) is an essential element for aerobic organisms because oxidative metabolism represents the main energy source. The partial reduction of O2 results in ROS formation; these are molecules that contain one oxygen atom in their structure and possess at least one highly reactive unpaired electron in an outer orbital [31]. These molecules include two major groups: free radicals such as the superoxide anion (O2) and hydroxyl radical (OH), and molecules such as hydrogen peroxide (H2O2) [32]. The production of O2 is the initial step for the formation of ROS, which is generated by the acceptance of an electron by O2, catalyzed by NADPH oxidase or xanthine oxidase. This radical can be converted into H2O2 by the action of the superoxide dismutase (SOD) enzyme, and then degraded to H2O and O2 by catalase or glutathione peroxidase [33]. OH is generated during the Haber-Weiss reaction which produces more toxic free radicals through the interaction between O2 and H2O2 [29]. Alternatively, two reactions using iron ions (Fe+3 and Fe+2), the Fenton reaction, can also generate OH [29].
In pathological events, ROS have been involved in patients with endometriosis [34]. The total amount of ROS in culture medium is negatively related with embryo implantation potential [35] or pregnancy [36]. High levels are correlated with poor oocyte quality [37] and cell meiotic arrest [6]. Previously, we investigated the induction of stress tolerance in bovine cumulus oocyte complexes (COCs) to generate oxidative stress resistance by incubation with H2O2 during in vitro embryo production [38]. We observed that exposing COCs to low H2O2 levels could induce stress tolerance in these embryos, determined by the embryo development, quality, and gene expression pattern [39].

1.3. Nitrosative Stress

Like ROS, RNS such as nitric oxide (NO) act as signaling molecules modulating various aspects of the reproductive physiology [40]; they influence and mediate the gametes and crucial reproductive processes such as sperm–oocyte interaction, implantation, and early embryo development [41]. Nitric oxide (NO) is generated either by enzymes, including neuronal nitric oxide synthase (nNOS), endothelial nitric oxide synthase (eNOS), and inducible nitric oxide synthase (iNOS) [40,42], or by a non-enzymatic pathway from nitrite involving hydrogen peroxide and d-or l-arginine [40]. High and sustained levels of RNS result in nitrosative stress with negative consequences for cells [40], leading to different pathologies [43]. The chemical reactivity of NO is rather low, but it reacts with O2 yielding peroxynitrite (ONOO), which is a potent oxidant inducing protein, lipid, and DNA damage [44].
Previously, we investigated nitrosative stress tolerance in oocytes by the in vitro incubation of oocytes adding NO donors during in vitro embryo production in bovine. However, no differences in the embryo quality or resistance to nitrosative stress were observed for incubation with either added 3-morpholinosidnonimine (SIN-1) [45] or added sodium nitroprusside (SNP) [46].
Currently, the administration of antioxidants is recommended for counteracting oxidative and nitrosative stress in cells. This review synthesises the experimental data that has been published and the advances in the knowledge on the effects of melatonin on gametes and the different steps of in vitro reproduction. Here, we argue that the application of melatonin should be considered for improving the efficiency and outcomes of reproductive biotechnologies in humans and domestic animals due to its capacity as a powerful antioxidant counteracting ROS/NOS induced damage.

1.4. Melatonin and ROS/RNS

Melatonin (N-acetyl-5-methoxytryptamine) is a multifunctional molecule secreted by the pineal gland in response to changes in light levels and other tissues [43,47]. Melatonin is produced at higher amounts by different tissues including Leydig cells, spermatocytes, and spermatids in testes [48]; in extrapineal organs, tissues, and fluids of mammals and humans [49,50]; and mast cells [51]. However, only melatonin secreted in the pineal gland plays an important role in the circadian sleep regulation [52] and reproductive function in seasonally breeding animals [53,54,55]. In humans, melatonin administration to IVF patients with sleep disorders improves the oocyte and embryo quality [56]. Additionally, the melatonin pattern secretion influences endocrine effects of the photoperiod, resulting in physiological alterations in reproduction [57]. It regulates the complex embryo-fetal developmental processes [58]. For example, the cold and dark winter periods in Norway may suppress the ovarian activity and estrus expression in cows, showing a higher reproductive performance during the summer months compared to the winter season [59], agreeing with the low in vitro embryo production rates we observed during the winter season in our geographic zone [60]. Circadian genes such as CLOCK, BMAL1, CRY1, CRY2, PER1, and PER2 are expressed and function as maternal mRNA regulating events in the oocytes and preimplantation embryos [61], and are involved in physiological processes, such as meiosis [62].
Melatonin is a potent free radical scavenger [52,63], directly reducing the ROS concentration and preventing the depletion of endogenous antioxidant enzymes [64]. The scavenging potential stems from it´s antioxidant reactions against ROS, including singlet oxygen, nitric oxide, hydrogen peroxide, and hydroxyl radical [65], and against RNS including nitric acid, peroxynitrite, and peroxynitrous acid [66]. Melatonin up-regulates the gene expression and activity of several antioxidant proteins [64,67,68,69], preserves optimal the mitochondrial function, and contributes to maintaining homeostasis against oxidative stress [70]. Its metabolites exhibit a powerful antioxidant capacity [44,71,72,73,74]. Melatonin readily combines with a superoxide releasing NO, thus preventing the formation of peroxynitrite, a free radical even more harmful than NO [75]. It has been described as a direct peroxynitrite scavenger [76].
The effects of melatonin on gametes and in vitro production (Figure 1) for humans are summarized in Table 1 and for other mammals in Table 2, for data published between 2012 and 2016.

2. Melatonin Modulates Oxidative Stress on Gametes and In Vitro Embryo Production (IVP)

Its amphiphilic nature allows melatonin to pass biological barriers. This feature makes it an effective antioxidant, resulting in protections embryos of macromolecules against ROS [74,120]. In mammalian oocytes, melatonin can prevent damage generated by hypochlorous acid (HOCl) on spindle microtubules and chromosome alterations in metaphase-II mouse oocytes [121], upregulate MnSOD [90,122] and Cu-ZnSOD transcripts in cumulus cells [90], decrease ROS levels in oocytes [90], suppress Bax protein expression and decrease the Bax/Bcl-2 ratio in the ovaries [122], and prevent DNA damage [123] and nuclear fragmentation in cumulus cells [90]. Long term treatments with melatonin in humans have shown reductions of ovarian ageing, and increases in the litter size, pools of follicles, and in the telomere length [122]. Melatonin has also been shown to protect oocytes against the inhibitory effect of oxidative stress generated by H2O2 [123]. The protection results in an increased in vitro maturation rate [124], reduced oxidative damage in oocytes during in vitro maturation, and decreased mitochondrial activity [124]. The optimal mitochondrial membrane potential can be maintained by activating uncoupling proteins or by inhibiting the mitochondrial permeability transition pore [125]. The mechanism by which melatonin promotes oocyte maturation is not yet clear, but it is believed to be mediated via melatonin membrane receptors such as the melatonin receptor agonist IIK7 [109].
In human, intrafollicular concentrations of 8-OHdG and hexanoyl-lysine were significantly reduced by melatonin (3 mg/day) and vitamin E (600 mg/day) treatments [123]. The fertilization rate was improved by melatonin treatment compared to the previous IVF-ET cycle [123] and melatonin levels are associated with oocyte quantity and quality [122,126]. Melatonin improves progesterone production by corpus luteum in infertile women with a luteal phase defect [127].
Spermatozoa are sensitive to oxidative stress, leading to an apoptosis-like process. Melatonin can decrease mitochondrial ROS production when sperm is exposed to oxidative stress [128]. Thus, it is a powerful antioxidant and anti-apoptotic agent in ejaculated human spermatozoa by the inhibition of caspase-3 and caspase-9 activities [80,128,129]. Melatonin can prevent mitochondrial ROS formation under basal conditions and at an early time point upon oxidative stress induced by H2O2 exposure [130], increasing MnSOD expression [85], glutathione peroxidase [131], and glutathione reductase [132]; preventing DNA fragmentation [129], and therefore, improving sperm quality [130]. Additionally, the melatonin supplementation of semen extenders increases sperm motility and viability, and decreases ROS levels and lipid peroxidation [80], which improves the sperm quality after the freezing-thawing processes [133]. This antioxidant can protect from testicular injury induced by oxidative stress after cadmium (Cd) exposure [96]. Melatonin helps to protect sperm from ROS induced by cell sorting, a widely used technique for in vitro fertilization or artificial insemination [133].
Our experience during the supplementation of IVF medium with melatonin shows that this antioxidant has a dual effect over sperm function and embryo development in bovine [105]: lower concentrations (10–1000 nM) modulate the sperm quality by inducing changes in the sperm motility, increasing the Wobbler coefficient. On the other hand, a high melatonin concentration during sperm incubation (1000 nM) decreased the number of viable sperm with an intact acrosome membrane and induced high levels of DNA fragmentation and DNA oxidation. Similarly, high melatonin concentrations in IVF (0.01–1 mM) generated decreases in the blastocyst production rate, without affecting the embryo quality. During embryo culture, cells are exposed to higher oxygen concentrations, resulting in increased ROS production. Melatonin supplementation has a beneficial effect on in vitro fertilization in human patients [82,123] by improving the blastocyst formation rate and decreasing the DNA fragmentation of blastomeres [104].
Cryopreservation is a highly stressful process that significantly reduces the potential for embryo development. Melatonin added to the culture medium increases cleavage, blastocyst, and hatching rates [100,134]. It increases the total number of cells (TCN) [113] and trophectoderm (TE) cells, and the inner cell mass (ICM) ratio in vitrified embryos [134]. Melatonin reduces the apoptotic index [113,134], promotes the activation of antioxidant enzymes such as GST and SOD [100], decreases the level of oxidative substrates [100], and ameliorates the down-regulation of genes including NANOG and POU5F1, which are important for early embryo development [100].

3. Potential Use of Melatonin against Nitrosative Stress during ART

Melatonin has been described to reduce nitrosative/oxidative stress in many different tissues and organelles [74]. These data corroborate its protective effect against drugs, toxins, metals, and herbicides [135]. Melatonin acts on the NO/NOS system by reducing peroxynitrite formation in the brain during the first steps of the ischemic cascade, influencing the NO/NOS pathway and reducing oxidative and nitrosative stress [42]. High levels of NO are produced during acute renal failure by iNOS due to ROS/RNS activation, but can be counteracted by melatonin, attenuating lipid peroxidation and protein oxidation in the kidneys [136]. Similarly, melatonin administration counteracted iNOS activation and mitochondrial damage in the liver during sepsis [137]. Melatonin preserves fetal growth in rats through protection against ischemia/reperfusion-induced oxidative/nitrosative stress by preventing the oxidative damage in placental DNA and mitochondria [70]. Is has a neuroprotective due to counteracting i-mtNOS induction, oxidative stress, and mitochondrial dysfunction [138].
Melatonin has been shown to be protective for gamete handling in vitro. It has been proposed that melatonin inhibits the activity of the pro-oxidative enzyme nitric oxide synthase (NOS) in the Graafian follicle [55]. Melatonin delays ovarian ageing by multiple mechanisms, including an antioxidant action and by reducing the decline in oocyte quantity and quality in mice [139]. Therefore, melatonin could be useful against nitrosative stress due to the in vitro maturation of the oocytes.
A beneficial effect on male fertility has been described for humans and domestic animals: melatonin induces a significant decrease in the intracellular NO concentration in human sperm, increasing sperm motility and viability [43]. The NO concentration changes during the annual reproductive cycle in male adult buffalo, whereby NO is mainly present in the caput epididymis during short photoperiods coinciding with maximum gonadal activity [140]. According to this, and considering the influence of melatonin on the seasonal reproduction in these animals, we can suggest the potential use of melatonin to modulate NO levels to increase buffalo fertility or in other seasonal breeds, both during semen storage or IVF.
ARTs can induce vascular dysfunction and arterial hypertension related to epigenetic alterations of the regulation of the eNOS gene. However, this can be prevented by the addition of melatonin during the in vitro culture of embryos, which has doubled the success rate of IVF [141].
Melatonin has been found to protect the fetus and placenta from oxidative stress due to ROS and RNS [55]. Clinical melatonin treatment could be useful to increase or maintain umbilical blood flow by NO-dependent mechanisms in complicated pregnancies [75], as after embryo transfer of in vitro produced embryos (ET/IVP) in domestic animals.
Despite the fact that more specific studies regarding the melatonin effect against nitrosative stress in reproductive biotechnologies are scarce, melatonin has demonstrated direct and indirect beneficial effects against ROS. Therefore, considering that ROS can generate RNS, we can deduct that melatonin could have a protective action over nitrosative stress during gamete and embryo handling in the laboratory, as has been demonstrated in other tissues.

4. Conclusions

There is a long list of studies that support the use of melatonin against oxidative stress; however, much remains to be investigated regarding the role that melatonin might have on nitrosative stress during the in vitro manipulation and cryopreservation of gametes and embryos. Therefore, the evidence is clear that melatonin is involved in the protection against oxidative/nitrosative stress by scavenging free radicals, inducing the activity of antioxidant enzymes and preventing the induction of the mitochondrial pathway of apoptosis, improving gamete and embryo quality, both in human and domestic animals during ARTs.

Acknowledgments

This work was supported by a FONDECYT Grant (No. 1130888), Carolina Cheuquemán’s Post-doctoral Grant (FONDECYT No. 3150320), and Pía Loren’s doctoral scholarship from CONICYT, Chile. The authors would like to thank Stephan M. Funk for his review of the English and for constructive suggestions.

Author Contributions

Raúl Sánchez conceived this paper; Pía Loren and Carolina Cheuquemán wrote this paper; María Elena Arias, Ricardo Felmer, and Jennie Risopatrón revised the paper.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. 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] [PubMed]
  2. Liu, Y.; Fiskum, G.; Schubert, D. Generation of reactive oxygen species by the mitochondrial electron transport chain. J. Neurochem. 2002, 80, 780–787. [Google Scholar] [CrossRef] [PubMed]
  3. Pasqualotto, E.B.; Agarwal, A.; Sharma, R.K.; Izzo, V.M.; Pinotti, J.A.; Joshi, N.J.; Rose, B.I. Effect of oxidative stress in follicular fluid on the outcome of assisted reproductive procedures. Fertil. Steril. 2004, 81, 973–976. [Google Scholar] [CrossRef] [PubMed]
  4. Attaran, M.; Pasqualotto, E.; Falcone, T.; Goldberg, J.M.; Miller, K.F.; Agarwal, A.; Sharma, R.K. The effect of follicular fluid reactive oxygen species on the outcome of in vitro fertilization. Int. J. Fertil. Womens Med. 2000, 45, 314–320. [Google Scholar] [PubMed]
  5. 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]
  6. Tripathi, A.; Khatun, S.; Pandey, A.N.; Mishra, S.K.; Chaube, R.; Shrivastav, T.G.; Chaube, S.K. Intracellular levels of hydrogen peroxide and nitric oxide in oocytes at various stages of meiotic cell cycle and apoptosis. Free Radic. Res. 2009, 43, 287–294. [Google Scholar] [CrossRef] [PubMed]
  7. Chaube, S.K.; Khatun, S.; Misra, S.K.; Shrivastav, T.G. Calcium ionophore-induced egg activation and apoptosis are associated with the generation of intracellular hydrogen peroxide. Free Radic. Res. 2008, 42, 212–220. [Google Scholar] [CrossRef] [PubMed]
  8. Shkolnik, K.; Tadmor, A.; Ben-Dor, S.; Nevo, N.; Galiani, D.; Dekel, N. Reactive oxygen species are indispensable in ovulation. Proc. Natl. Acad. Sci. USA 2011, 108, 1462–1467. [Google Scholar] [CrossRef] [PubMed]
  9. De Lamirande, E.; Gagnon, C. A positive role for the superoxide anion in triggering hyperactivation and capacitation of human spermatozoa. Int. J. Androl. 1993, 16, 21–25. [Google Scholar] [CrossRef] [PubMed]
  10. Bize, I.; Santander, G.; Cabello, P.; Driscoll, D.; Sharpe, C. Hydrogen peroxide is involved in hamster sperm capacitation in vitro. Biol. Reprod. 1991, 44, 398–403. [Google Scholar] [CrossRef] [PubMed]
  11. De Lamirande, E.; Gagnon, C. Human sperm hyperactivation and capacitation as parts of an oxidative process. Free Radic. Biol. Med. 1993, 14, 157–166. [Google Scholar] [CrossRef]
  12. Leclerc, P.; de Lamirande, E.; Gagnon, C. Interaction between Ca2+, cyclic 3′,5′ adenosine monophosphate, the superoxide anion, and tyrosine phosphorylation pathways in the regulation of human sperm capacitation. J. Androl. 1998, 19, 434–443. [Google Scholar] [PubMed]
  13. Leclerc, P.; de Lamirande, E.; Gagnon, C. Regulation of protein-tyrosine phosphorylation and human sperm capacitation by reactive oxygen derivatives. Free Radic. Biol. Med. 1997, 22, 643–656. [Google Scholar] [CrossRef]
  14. Roy, S.C.; Atreja, S.K. Effect of reactive oxygen species on capacitation and associated protein tyrosine phosphorylation in buffalo (Bubalus bubalis) spermatozoa. Anim. Reprod. Sci. 2008, 107, 68–84. [Google Scholar] [CrossRef] [PubMed]
  15. Dona, G.; Fiore, C.; Tibaldi, E.; Frezzato, F.; Andrisani, A.; Ambrosini, G.; Fiorentin, D.; Armanini, D.; Bordin, L.; Clari, G. Endogenous reactive oxygen species content and modulation of tyrosine phosphorylation during sperm capacitation. Int. J. Androl. 2011, 34, 411–419. [Google Scholar] [CrossRef] [PubMed]
  16. Rivlin, J.; Mendel, J.; Rubinstein, S.; Etkovitz, N.; Breitbart, H. Role of hydrogen peroxide in sperm capacitation and acrosome reaction. Biol. Reprod. 2004, 70, 518–522. [Google Scholar] [CrossRef] [PubMed]
  17. Zheng, K.; Sulieman, F.J.; Li, J.; Wei, Q.; Xu, M.; Shi, F. Nitric oxide and thyroid hormone receptor alpha 1 contribute to ovarian follicular development in immature hyper- and hypo-thyroid rats. Reprod. Biol. 2015, 15, 27–33. [Google Scholar] [CrossRef] [PubMed]
  18. Dubey, P.K.; Tripathi, V.; Singh, R.P.; Saikumar, G.; Nath, A.; Pratheesh, M.D.; Gade, N.; Sharma, G.T. Expression of nitric oxide synthase isoforms in different stages of buffalo (Bubalus bubalis) ovarian follicles: Effect of nitric oxide on in vitro development of preantral follicle. Theriogenology 2012, 77, 280–291. [Google Scholar] [CrossRef] [PubMed]
  19. Bu, S.; Xia, G.; Tao, Y.; Lei, L.; Zhou, B. Dual effects of nitric oxide on meiotic maturation of mouse cumulus cell-enclosed oocytes in vitro. Mol. Cell. Endocrinol. 2003, 207, 21–30. [Google Scholar] [CrossRef]
  20. Viana, K.S.; Caldas-Bussiere, M.C.; Matta, S.G.; Faes, M.R.; de Carvalho, C.S.; Quirino, C.R. Effect of sodium nitroprusside, a nitric oxide donor, on the in vitro maturation of bovine oocytes. Anim. Reprod. Sci. 2007, 102, 217–227. [Google Scholar] [CrossRef] [PubMed]
  21. Jablonka-Shariff, A.; Olson, L.M. The role of nitric oxide in oocyte meiotic maturation and ovulation: Meiotic abnormalities of endothelial nitric oxide synthase knock-out mouse oocytes. Endocrinology 1998, 139, 2944–2954. [Google Scholar] [CrossRef] [PubMed]
  22. Pires, P.R.L.; Santos, N.P.; Adona, P.R.; Natori, M.M.; Schwarz, K.R.L.; de Bem, T.H.C.; Leal, C.L.V. Endothelial and inducible nitric oxide synthases in oocytes of cattle. Anim. Reprod. Sci. 2009, 116, 233–243. [Google Scholar] [CrossRef] [PubMed]
  23. Tranguch, S.; Steuerwald, N.; Huet-Hudson, Y.M. Nitric oxide synthase production and nitric oxide regulation of preimplantation embryo development. Biol. Reprod. 2003, 68, 1538–1544. [Google Scholar] [CrossRef] [PubMed]
  24. Tesfaye, D.; Kadanga, A.; Rings, F.; Bauch, K.; Jennen, D.; Nganvongpanit, K.; Holker, M.; Tholen, E.; Ponsuksili, S.; Wimmers, K.; et al. The effect of nitric oxide inhibition and temporal expression patterns of the mRNA and protein products of nitric oxide synthase genes during in vitro development of bovine pre-implantation embryos. Reprod. Domest. Anim. 2006, 41, 501–509. [Google Scholar] [CrossRef] [PubMed]
  25. Roessner, C.; Paasch, U.; Glander, H.J.; Grunewald, S. Activity of nitric oxide synthase in mature and immature human spermatozoa. Andrologia 2010, 42, 132–137. [Google Scholar] [CrossRef] [PubMed]
  26. Rodriguez, P.C.; Valdez, L.B.; Zaobornyj, T.; Boveris, A.; Beconi, M.T. Nitric oxide and superoxide anion production during heparin-induced capacitation in cryopreserved bovine spermatozoa. Reprod. Domest. Anim. 2011, 46, 74–81. [Google Scholar] [CrossRef] [PubMed]
  27. De Lamirande, E.; Lamothe, G. Reactive oxygen-induced reactive oxygen formation during human sperm capacitation. Free Radic. Biol. Med. 2009, 46, 502–510. [Google Scholar] [CrossRef] [PubMed]
  28. Herrero, M.B.; de Lamirande, E.; Gagnon, C. Nitric oxide is a signaling molecule in spermatozoa. Curr. Pharm. Des. 2003, 9, 419–425. [Google Scholar] [CrossRef] [PubMed]
  29. Agarwal, A.; Aponte-Mellado, A.; Premkumar, B.J.; Shaman, A.; Gupta, S. The effects of oxidative stress on female reproduction: A review. Reprod. Biol. Endocrinol. 2012, 10, 49. [Google Scholar] [CrossRef] [PubMed]
  30. Ma, Q. Transcriptional responses to oxidative stress: Pathological and toxicological implications. Pharmacol. Ther. 2010, 125, 376–393. [Google Scholar] [CrossRef] [PubMed]
  31. Devlin, T.M. Bioquímica: Libro de Texto con Aplicaciones Clínicas, 3rd ed.; Revert: Barcelona, Spain, 2004; Volume 1, pp. 217–266. [Google Scholar]
  32. Pourova, J.; Kottova, M.; Voprsalova, M.; Pour, M. Reactive oxygen and nitrogen species in normal physiological processes. Acta Physiol. 2010, 198, 15–35. [Google Scholar] [CrossRef] [PubMed]
  33. Takahashi, M. Oxidative Stress and Redox Regulation on In Vitro development of mammalian embryos. J. Reprod. Dev. 2012, 58, 1–9. [Google Scholar] [CrossRef] [PubMed]
  34. Liu, F.; He, L.; Liu, Y.; Shi, Y.; Du, H. The expression and role of oxidative stress markers in the serum and follicular fluid of patients with endometriosis. Clin. Exp. Obstet. Gynecol. 2013, 40, 372–376. [Google Scholar] [PubMed]
  35. Lee, T.H.; Lee, M.S.; Liu, C.H.; Tsao, H.M.; Huang, C.C.; Yang, Y.S. The association between microenvironmental reactive oxygen species and embryo development in assisted reproduction technology cycles. Reprod. Sci. 2012, 19, 725–732. [Google Scholar] [CrossRef] [PubMed]
  36. Bedaiwy, M.A.; Elnashar, S.A.; Goldberg, J.M.; Sharma, R.; Mascha, E.J.; Arrigain, S.; Agarwal, A.; Falcone, T. Effect of follicular fluid oxidative stress parameters on intracytoplasmic sperm injection outcome. Gynecol. Endocrinol. 2012, 28, 51–55. [Google Scholar] [CrossRef] [PubMed]
  37. Rajani, S.; Chattopadhyay, R.; Goswami, S.K.; Ghosh, S.; Sharma, S.; Chakravarty, B. Assessment of oocyte quality in polycystic ovarian syndrome and endometriosis by spindle imaging and reactive oxygen species levels in follicular fluid and its relationship with IVF-ET outcome. J. Hum. Reprod. Sci. 2012, 5, 187–193. [Google Scholar] [CrossRef] [PubMed]
  38. Loren, P.; Cheuquemán, C.; Risopatrón, J.; Felmer, R.; Arias, M.E.; Sánchez, R. Modulación del Estado de Óxido-Reducción por Peróxido de Hidrógeno en la Etapa de Maduración Ovocitaria: Efecto sobre el Desarrollo Embrionario en Bovinos. Int. J. Morphol. 2016, 34, 431–435. [Google Scholar] [CrossRef]
  39. Loren, P.; Sanchez, E.; Risopatrón, J.; Arias, M.; Felmer, R.; Sánchez, R. Efecto de estrés oxidativo in vitro en complejos ovocito-cumulus: Evaluación del potencial de desarrollo embrionario, calidad y perfil de expresión génica en bovinos. In Proceedings of the VII Congreso de la Asociación Iberoamericana de Sociedades de Andrología, Lisboa, Portugal, 3–5 December 2016. [Google Scholar]
  40. Pandey, A.N.; Tripathi, A.; PremKumar, K.V.; Shrivastav, T.G.; Chaube, S.K. Reactive oxygen and nitrogen species during meiotic resumption from diplotene arrest in mammalian oocytes. J. Cell. Biochem. 2010, 111, 521–528. [Google Scholar] [CrossRef] [PubMed]
  41. Baker, M.A.; Aitken, R.J. The importance of redox regulated pathways in sperm cell biology. Mol. Cell. Endocrinol. 2004, 216, 47–54. [Google Scholar] [CrossRef] [PubMed]
  42. Blanco, S.; Hernández, R.; Franchelli, G.; Ramos-Álvarez, M.M.; Peinado, M.Á. Melatonin influences NO/NOS pathway and reduces oxidative and nitrosative stress in a model of hypoxic-ischemic brain damage. Nitric Oxide 2017, 62, 32–43. [Google Scholar] [CrossRef] [PubMed]
  43. Du Plessis, S.S.; Hagenaar, K.; Lampiao, F. The in vitro effects of melatonin on human sperm function and its scavenging activities on NO and ROS. Andrologia 2010, 42, 112–116. [Google Scholar] [CrossRef] [PubMed]
  44. Galano, A.; Tan, D.X.; Reiter, R.J. Melatonin as a natural ally against oxidative stress: A physicochemical examination. J. Pineal Res. 2011, 51, 1–16. [Google Scholar] [CrossRef] [PubMed]
  45. Loren, P.; Cheuqueman, C.; Sanchez, E.; Risopatron, J.; Arias, M.E.; Felmer, R.; Sanchez, R. Effect of short-term exposure of cumulus-oocyte complex to 3-morpholinosydnonimine on in vitro embryo development and gene expression in cattle. Reprod. Domest. Anim. 2016, 51, 1010–1019. [Google Scholar] [CrossRef] [PubMed]
  46. Cheuqueman, C.; Loren, P.; Arias, M.; Risopatron, J.; Felmer, R.; Alvarez, J.; Mogas, T.; Sanchez, R. Effects of short-term exposure of mature oocytes to sodium nitroprusside on in vitro embryo production and gene expression in bovine. Theriogenology 2015, 84, 1431–1437. [Google Scholar] [CrossRef] [PubMed]
  47. Reiter, R.J.; Tan, D.X.; Fuentes-Broto, L. Melatonin: A multitasking molecule. Prog. Brain Res. 2010, 181, 127–151. [Google Scholar] [PubMed]
  48. Gonzalez-Arto, M.; Hamilton, T.R.D.S.; Gallego, M.; Gaspar-Torrubia, E.; Aguilar, D.; Serrano-Blesa, E.; Abecia, J.A.; Pérez-Pé, R.; Muiño-Blanco, T.; Cebrián-Pérez, J.A.; et al. Evidence of melatonin synthesis in the ram reproductive tract. Andrology 2016, 4, 163–171. [Google Scholar] [CrossRef] [PubMed]
  49. Acuña-Castroviejo, D.; Escames, G.; Venegas, C.; Díaz-Casado, M.E.; Lima-Cabello, E.; López, L.C.; Rosales-Corral, S.; Tan, D.-X.; Reiter, R.J. Extrapineal melatonin: Sources, regulation, and potential functions. Cell. Mol. Life Sci. 2014, 71, 2997–3025. [Google Scholar] [CrossRef] [PubMed]
  50. Venegas, C.; Garcia, J.A.; Escames, G.; Ortiz, F.; Lopez, A.; Doerrier, C.; Garcia-Corzo, L.; Lopez, L.C.; Reiter, R.J.; Acuna-Castroviejo, D. Extrapineal melatonin: Analysis of its subcellular distribution and daily fluctuations. J. Pineal Res. 2012, 52, 217–227. [Google Scholar] [CrossRef] [PubMed]
  51. Maldonado, M.D.; Mora-Santos, M.; Naji, L.; Carrascosa-Salmoral, M.P.; Naranjo, M.C.; Calvo, J.R. Evidence of melatonin synthesis and release by mast cells. Possible modulatory role on inflammation. Pharmacol. Res. 2010, 62, 282–287. [Google Scholar] [CrossRef] [PubMed]
  52. Reiter, R.J.; Tan, D.X.; Galano, A. Melatonin: Exceeding expectations. Physiology 2014, 29, 325–333. [Google Scholar] [CrossRef] [PubMed]
  53. Barrett, P.; Bolborea, M. Molecular pathways involved in seasonal body weight and reproductive responses governed by melatonin. J. Pineal Res. 2012, 52, 376–388. [Google Scholar] [CrossRef] [PubMed]
  54. Zarazaga, L.A.; Celi, I.; Guzman, J.L.; Malpaux, B. Melatonin concentrations in the two jugular veins, and relationship with the seasonal reproductive activity in goats. Theriogenology 2010, 74, 221–228. [Google Scholar] [CrossRef] [PubMed]
  55. Reiter, R.J.; Tan, D.X.; Manchester, L.C.; Paredes, S.D.; Mayo, J.C.; Sainz, R.M. Melatonin and reproduction revisited. Biol. Reprod. 2009, 81, 445–456. [Google Scholar] [CrossRef] [PubMed]
  56. Eryilmaz, O.G.; Devran, A.; Sarikaya, E.; Aksakal, F.N.; Mollamahmutoğlu, L.; Cicek, N. Melatonin improves the oocyte and the embryo in IVF patients with sleep disturbances, but does not improve the sleeping problems. J. Assist. Reprod. Genet. 2011, 28, 815–820. [Google Scholar] [CrossRef] [PubMed]
  57. Dahl, G.E.; Buchanan, B.A.; Tucker, H.A. Photoperiodic effects on dairy cattle: A review. J. Dairy Sci. 2000, 83, 885–893. [Google Scholar] [CrossRef]
  58. Voiculescu, S.E.; Zygouropoulos, N.; Zahiu, C.D.; Zagrean, A.M. Role of melatonin in embryo fetal development. J. Med. Life 2014, 7, 488–492. [Google Scholar] [PubMed]
  59. Refsdal, A.O. Reproductive performance of Norwegian cattle from 1985 to 2005: Trends and seasonality. Acta Vet. Scand. 2007, 49, 5. [Google Scholar] [CrossRef] [PubMed]
  60. Cheuquemán, C.; Loren, P.; Arias, M.; Risopatrón, J.; Felmer, R.; Álvarez, J.; Mogas, T.; Sánchez, R. Decrease in bovine in vitro embryo production efficiency during winter season in a warm-summer Mediterranean climate. Andrologia 2016. [Google Scholar] [CrossRef] [PubMed]
  61. Amano, T.; Tokunaga, K.; Kakegawa, R.; Yanagisawa, A.; Takemoto, A.; Tatemizo, A.; Watanabe, T.; Hatanaka, Y.; Matsushita, A.; Kishi, M.; et al. Expression analysis of circadian genes in oocytes and preimplantation embryos of cattle and rabbits. Anim. Reprod. Sci. 2010, 121, 225–235. [Google Scholar] [CrossRef] [PubMed]
  62. Amano, T.; Matsushita, A.; Hatanaka, Y.; Watanabe, T.; Oishi, K.; Ishida, N.; Anzai, M.; Mitani, T.; Kato, H.; Kishigami, S.; et al. Expression and functional analyses of circadian genes in mouse oocytes and preimplantation embryos: Cry1 is involved in the meiotic process independently of circadian clock regulation. Biol. Reprod. 2009, 80, 473–483. [Google Scholar] [CrossRef] [PubMed]
  63. Reiter, R.J.; Tan, D.X.; Manchester, L.C.; Qi, W. Biochemical reactivity of melatonin with reactive oxygen and nitrogen species: A review of the evidence. Cell Biochem. Biophys. 2001, 34, 237–256. [Google Scholar] [CrossRef]
  64. Antolin, I.; Rodriguez, C.; Sainz, R.M.; Mayo, J.C.; Uria, H.; Kotler, M.L.; Rodriguez-Colunga, M.J.; Tolivia, D.; Menendez-Pelaez, A. Neurohormone melatonin prevents cell damage: Effect on gene expression for antioxidant enzymes. FASEB J. 1996, 10, 882–890. [Google Scholar] [PubMed]
  65. Poeggeler, B.; Reiter, R.J.; Hardeland, R.; Tan, D.X.; Barlow-Walden, L.R. Melatonin and structurally-related, endogenous indoles act as potent electron donors and radical scavengers in vitro. Redox Rep. Commun. Free Radic. Res. 1996, 2, 179–184. [Google Scholar] [CrossRef] [PubMed]
  66. Zhang, H.; Squadrito, G.L.; Pryor, W.A. The reaction of melatonin with peroxynitrite: Formation of melatonin radical cation and absence of stable nitrated products. Biochem. Biophys. Res. Commun. 1998, 251, 83–87. [Google Scholar] [CrossRef] [PubMed]
  67. Barlow-Walden, L.R.; Reiter, R.J.; Abe, M.; Pablos, M.; Menendez-Pelaez, A.; Chen, L.D.; Poeggeler, B. Melatonin stimulates brain glutathione peroxidase activity. Neurochem. Int. 1995, 26, 497–502. [Google Scholar] [CrossRef]
  68. Mayo, J.C.; Sainz, R.M.; Antoli, I.; Herrera, F.; Martin, V.; Rodriguez, C. Melatonin regulation of antioxidant enzyme gene expression. Cell. Mol. Life Sci. 2002, 59, 1706–1713. [Google Scholar] [CrossRef] [PubMed]
  69. Rodriguez, C.; Mayo, J.C.; Sainz, R.M.; Antolin, I.; Herrera, F.; Martin, V.; Reiter, R.J. Regulation of antioxidant enzymes: A significant role for melatonin. J. Pineal Res. 2004, 36, 1–9. [Google Scholar] [CrossRef] [PubMed]
  70. Nagai, R.; Watanabe, K.; Wakatsuki, A.; Hamada, F.; Shinohara, K.; Hayashi, Y.; Imamura, R.; Fukaya, T. Melatonin preserves fetal growth in rats by protecting against ischemia/reperfusion-induced oxidative/nitrosative mitochondrial damage in the placenta. J. Pineal Res. 2008, 45, 271–276. [Google Scholar] [CrossRef] [PubMed]
  71. Tan, D.X.; Manchester, L.C.; Terron, M.P.; Flores, L.J.; Reiter, R.J. One molecule, many derivatives: A never-ending interaction of melatonin with reactive oxygen and nitrogen species? J. Pineal Res. 2007, 42, 28–42. [Google Scholar] [CrossRef] [PubMed]
  72. Ressmeyer, A.R.; Mayo, J.C.; Zelosko, V.; Sainz, R.M.; Tan, D.X.; Poeggeler, B.; Antolin, I.; Zsizsik, B.K.; Reiter, R.J.; Hardeland, R. Antioxidant properties of the melatonin metabolite N1-acetyl-5-methoxykynuramine (AMK): Scavenging of free radicals and prevention of protein destruction. Redox Rep. Commun. Free Radic. Res. 2003, 8, 205–213. [Google Scholar] [CrossRef] [PubMed]
  73. Reiter, R.J.; Tan, D.X.; Terron, M.P.; Flores, L.J.; Czarnocki, Z. Melatonin and its metabolites: New findings regarding their production and their radical scavenging actions. Acta Biochim. Pol. 2007, 54, 1–9. [Google Scholar] [PubMed]
  74. Reiter, R.J.; Paredes, S.D.; Korkmaz, A.; Jou, M.-J.; Tan, D.-X. Melatonin combats molecular terrorism at the mitochondrial level. Interdiscip. Toxicol. 2008, 1, 137–149. [Google Scholar] [CrossRef] [PubMed]
  75. Thakor, A.S.; Herrera, E.A.; Seron-Ferre, M.; Giussani, D.A. Melatonin and vitamin C increase umbilical blood flow via nitric oxide-dependent mechanisms. J. Pineal Res. 2010, 49, 399–406. [Google Scholar] [CrossRef] [PubMed]
  76. Singh, I.N.; Sullivan, P.G.; Hall, E.D. Peroxynitrite-mediated oxidative damage to brain mitochondria: Protective effects of peroxynitrite scavengers. J. Neurosci. Res. 2007, 85, 2216–2223. [Google Scholar] [CrossRef] [PubMed]
  77. Jahromi, B.N.; Sadeghi, S.; Alipour, S.; Parsanezhad, M.E.; Alamdarloo, S.M. Effect of melatonin on the outcome of assisted reproductive technique cycles in women with diminished ovarian reserve: A double-blinded randomized clinical trial. Iran. J. Med. Sci. 2017, 42, 73–78. [Google Scholar] [PubMed]
  78. Arjmand, F.; Khanmohammadi, M.; Arasteh, S.; Mohammadzadeh, A.; Kazemnejad, S.; Akhondi, M.-M. Extended culture of encapsulated human blastocysts in alginate hydrogel containing decidualized endometrial stromal cells in the presence of melatonin. Mol. Biotechnol. 2016, 58, 684–694. [Google Scholar] [CrossRef] [PubMed]
  79. Pacchiarotti, A.; Carlomagno, G.; Antonini, G.; Pacchiarotti, A. Effect of myo-inositol and melatonin versus myo-inositol, in a randomized controlled trial, for improving in vitro fertilization of patients with polycystic ovarian syndrome. Gynecol. Endocrinol. 2016, 32, 69–73. [Google Scholar] [CrossRef] [PubMed]
  80. 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]
  81. Bejarano, I.; Monllor, F.; Marchena, A.M.; Ortiz, A.; Lozano, G.; Jimenez, M.I.; Gaspar, P.; Garcia, J.F.; Pariente, J.A.; Rodriguez, A.B.; et al. Exogenous melatonin supplementation prevents oxidative stress-evoked DNA damage in human spermatozoa. J. Pineal Res. 2014, 57, 333–339. [Google Scholar] [CrossRef] [PubMed]
  82. Nishihara, T.; Hashimoto, S.; Ito, K.; Nakaoka, Y.; Matsumoto, K.; Hosoi, Y.; Morimoto, Y. Oral melatonin supplementation improves oocyte and embryo quality in women undergoing in vitro fertilization-embryo transfer. Gynecol. Endocrinol. 2014, 30, 359–362. [Google Scholar] [CrossRef] [PubMed]
  83. Jin, J.X.; Lee, S.; Taweechaipaisankul, A.; Kim, G.A.; Lee, B.C. Melatonin regulates lipid metabolism in porcine oocytes. J. Pineal Res. 2017, 62. [Google Scholar] [CrossRef] [PubMed]
  84. Majrashi, K.A.; Barakat, I.A.; Al-Himaidi, A.R.; Adham, K.A. Effect of exogenous melatonin treatment on the reproductive characteristics and progeny of male rats exposed to different periods from light and darkness. Physiol. Res. 2017, 62, e12388. [Google Scholar]
  85. Li, B.; He, X.; Zhuang, M.; Niu, B.; Wu, C.; Mu, H.; Tang, F.; Cui, Y.; Liu, W.; Zhao, B.; et al. Melatonin ameliorates busulfan-induced spermatogonial stem cell oxidative apoptosis in mouse testes. Antioxid. Redox Signal. 2017. [Google Scholar] [CrossRef] [PubMed]
  86. Dai, X.; Lu, Y.; Zhang, M.; Miao, Y.; Zhou, C.; Cui, Z.; Xiong, B. Melatonin improves the fertilization ability of post-ovulatory aged mouse oocytes by stabilizing ovastacin and Juno to promote sperm binding and fusion. Hum. Reprod. 2017, 32, 596–606. [Google Scholar] [CrossRef] [PubMed]
  87. Ortega, M.S.; Rocha-Frigoni, N.A.; Mingoti, G.Z.; Roth, Z.; Hansen, P.J. Modification of embryonic resistance to heat shock in cattle by melatonin and genetic variation in HSPA1L. J. Dairy Sci. 2016, 99, 9152–9164. [Google Scholar] [CrossRef] [PubMed]
  88. Zhang, Y.; Li, W.; Ma, Y.; Wang, D.; Zhao, X.; Zeng, C.; Zhang, M.; Zeng, X.; Meng, Q.; Zhou, G. Improved development by melatonin treatment after vitrification of mouse metaphase II oocytes. Cryobiology 2016, 73, 335–342. [Google Scholar] [CrossRef] [PubMed]
  89. Pang, Y.W.; Sun, Y.Q.; Jiang, X.L.; Huang, Z.Q.; Zhao, S.J.; Du, W.H.; Hao, H.S.; Zhao, X.M.; Zhu, H.B. Protective effects of melatonin on bovine sperm characteristics and subsequent in vitro embryo development. Mol. Reprod. Dev. 2016, 83, 993–1002. [Google Scholar] [CrossRef] [PubMed]
  90. Rodrigues-Cunha, M.C.; Mesquita, L.G.; Bressan, F.; Collado, M.D.; Balieiro, J.C.; Schwarz, K.R.; de Castro, F.C.; Watanabe, O.Y.; Watanabe, Y.F.; de Alencar Coelho, L.; et al. Effects of melatonin during IVM in defined medium on oocyte meiosis, oxidative stress, and subsequent embryo development. Theriogenology 2016, 86, 1685–1694. [Google Scholar] [CrossRef] [PubMed]
  91. Pang, Y.W.; Sun, Y.Q.; Sun, W.J.; Du, W.H.; Hao, H.S.; Zhao, S.J.; Zhu, H.B. Melatonin inhibits paraquat-induced cell death in bovine preimplantation embryos. J. Pineal Res. 2016, 60, 155–166. [Google Scholar] [CrossRef] [PubMed]
  92. Park, J.Y.; Park, H.J.; Kim, J.W.; Park, S.Y.; Yang, S.G.; Jung, J.M.; Kim, M.J.; Koo, D.B. 173 Melatonin alleviates the endoplasmic reticulum stress through the regulating of unfolding protein response signaling during porcine oocyte maturation in vitro. Reprod. Fertil. Dev. 2016, 29, 195. [Google Scholar] [CrossRef] [PubMed]
  93. Remiao, M.H.; Lucas, C.G.; Domingues, W.B.; Silveira, T.; Barther, N.N.; Komninou, E.R.; Basso, A.C.; Jornada, D.S.; Beck, R.C.; Pohlmann, A.R.; et al. Melatonin delivery by nanocapsules during in vitro bovine oocyte maturation decreased the reactive oxygen species of oocytes and embryos. Reprod. Toxicol. 2016, 63, 70–81. [Google Scholar] [CrossRef] [PubMed]
  94. Komninou, E.R.; Remiao, M.H.; Lucas, C.G.; Domingues, W.B.; Basso, A.C.; Jornada, D.S.; Deschamps, J.C.; Beck, R.C.; Pohlmann, A.R.; Bordignon, V.; et al. Effects of two types of melatonin-loaded nanocapsules with distinct supramolecular structures: Polymeric (nc) and lipid-core nanocapsules (lnc) on bovine embryo culture model. PLoS ONE 2016, 11, e0157561. [Google Scholar] [CrossRef] [PubMed]
  95. He, C.; Wang, J.; Zhang, Z.; Yang, M.; Li, Y.; Tian, X.; Ma, T.; Tao, J.; Zhu, K.; Song, Y.; et al. Mitochondria synthesize melatonin to ameliorate its function and improve mice oocyte's quality under in vitro conditions. Int. J. Mol. Sci. 2016, 17. [Google Scholar] [CrossRef] [PubMed]
  96. Li, R.; Luo, X.; Li, L.; Peng, Q.; Yang, Y.; Zhao, L.; Ma, M.; Hou, Z. The protective effects of melatonin against oxidative stress and inflammation induced by acute cadmium exposure in mice testis. Biol. Trace Element Res. 2016, 170, 152–164. [Google Scholar] [CrossRef] [PubMed]
  97. 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]
  98. 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] [PubMed]
  99. Liang, S.; Guo, J.; Choi, J.W.; Kim, N.H.; Cui, X.S. Effect and possible mechanisms of melatonin treatment on the quality and developmental potential of aged bovine oocytes. Reprod. Fertil. Dev. 2016. [Google Scholar] [CrossRef] [PubMed]
  100. Mehaisen, G.M.; Saeed, A.M.; Gad, A.; Abass, A.O.; Arafa, M.; El-Sayed, A. Antioxidant capacity of melatonin on preimplantation development of fresh and vitrified rabbit embryos: Morphological and molecular aspects. PLoS ONE 2015, 10, e0139814. [Google Scholar] [CrossRef] [PubMed]
  101. Ganji, R.; Nabiuni, M.; Faraji, R. Development of mouse preantral follicle after in vitro culture in a medium containing melatonin. Cell J. 2015, 16, 546–553. [Google Scholar] [PubMed]
  102. Su, J.; Wang, Y.; Xing, X.; Zhang, L.; Sun, H.; Zhang, Y. Melatonin significantly improves the developmental competence of bovine somatic cell nuclear transfer embryos. J. Pineal Res. 2015, 59, 455–468. [Google Scholar] [CrossRef] [PubMed]
  103. 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] [PubMed]
  104. Do, L.T.; Shibata, Y.; Taniguchi, M.; Nii, M.; Nguyen, T.V.; Tanihara, F.; Takagi, M.; Otoi, T. Melatonin supplementation during in vitro maturation and development supports the development of porcine embryos. Reprod. Domest. Anim. 2015, 50, 1054–1058. [Google Scholar] [CrossRef] [PubMed]
  105. Cheuqueman, C.; Arias, M.E.; Risopatron, J.; Felmer, R.; Alvarez, J.; Mogas, T.; Sanchez, R. Supplementation of IVF medium with melatonin: Effect on sperm functionality and in vitro produced bovine embryos. Andrologia 2015, 47, 604–615. [Google Scholar] [CrossRef] [PubMed]
  106. Mehaisen, G.M.; Saeed, A.M. In vitro development rate of preimplantation rabbit embryos cultured with different levels of melatonin. Zygote 2015, 23, 111–115. [Google Scholar] [CrossRef] [PubMed]
  107. Wang, F.; Tian, X.; Zhou, Y.; Tan, D.; Zhu, S.; Dai, Y.; Liu, G. Melatonin improves the quality of in vitro produced (IVP) bovine embryos: Implications for blastocyst development, cryotolerance, and modifications of relevant gene expression. PLoS ONE 2014, 9, e93641. [Google Scholar] [CrossRef] [PubMed]
  108. 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]
  109. Tian, X.; Wang, F.; He, C.; Zhang, L.; Tan, D.; Reiter, R.J.; Xu, J.; Ji, P.; Liu, G. Beneficial effects of melatonin on bovine oocytes maturation: A mechanistic approach. J. Pineal Res. 2014, 57, 239–247. [Google Scholar] [CrossRef] [PubMed]
  110. Niknafs, B.; Mehdipour, A.; Mohammadi Roushandeh, A. Melatonin improves development of early mouse embryos impaired by actinomycin-D and TNF-alpha. Iran. J. Reprod. Med. 2014, 12, 799–804. [Google Scholar] [PubMed]
  111. Minaii, B.; Moayeri, A.; Shokri, S.; Habibi Roudkenar, M.; Golmohammadi, T.; Malek, F.; Barbarestani, M. Melatonin improve the sperm quality in forced swimming test induced oxidative stress in nandrolone treated Wistar rats. Acta Med. Iran. 2014, 52, 496–504. [Google Scholar] [PubMed]
  112. Salehi, M.; Kato, Y.; Tsunoda, Y. Effect of melatonin treatment on developmental potential of somatic cell nuclear-transferred mouse oocytes in vitro. Zygote 2014, 22, 213–217. [Google Scholar] [CrossRef] [PubMed]
  113. Succu, S.; Pasciu, V.; Manca, M.E.; Chelucci, S.; Torres-Rovira, L.; Leoni, G.G.; Zinellu, A.; Carru, C.; Naitana, S.; Berlinguer, F. Dose-dependent effect of melatonin on postwarming development of vitrified ovine embryos. Theriogenology 2014, 81, 1058–1066. [Google Scholar] [CrossRef] [PubMed]
  114. Wang, L.; Zhuo, Z.Y.; Shi, W.Q.; Tan, D.X.; Gao, C.; Tian, X.Z.; Zhang, L.; Zhou, G.B.; Zhu, S.E.; Yun, P.; et al. Melatonin promotes superovulation in sika deer (Cervus nippon). Int. J. Mol. Sci. 2014, 15, 12107–12118. [Google Scholar] [CrossRef] [PubMed]
  115. Zhang, L.; Chai, M.; Tian, X.; Wang, F.; Fu, Y.; He, C.; Deng, S.; Lian, Z.; Feng, J.; Tan, D.X.; et al. Effects of melatonin on superovulation and transgenic embryo transplantation in small-tailed han sheep (Ovis aries). Neuro Endocrinol. Lett. 2013, 34, 294–301. [Google Scholar] [PubMed]
  116. Pang, Y.W.; An, L.; Wang, P.; Yu, Y.; Yin, Q.D.; Wang, X.H.; Xin, Z.; Qian, Z.; Yang, M.L.; Min, G.; et al. Treatment of porcine donor cells and reconstructed embryos with the antioxidant melatonin enhances cloning efficiency. J. Pineal Res. 2013, 54, 389–397. [Google Scholar] [CrossRef] [PubMed]
  117. Bahadori, M.H.; Ghasemian, F.; Ramezani, M.; Asgari, Z. Melatonin effect during different maturation stages of oocyte and subsequent embryo development in mice. Iran. J. Reprod. Med. 2013, 11, 11–18. [Google Scholar] [PubMed]
  118. Cebrian-Serrano, A.; Salvador, I.; Raga, E.; Dinnyes, A.; Silvestre, M.A. Beneficial effect of melatonin on blastocyst in vitro production from heat-stressed bovine oocytes. Reprod. Domest. Anim. 2013, 48, 738–746. [Google Scholar] [CrossRef] [PubMed]
  119. Wang, F.; Tian, X.; Zhang, L.; Tan, D.; Reiter, R.J.; Liu, G. Melatonin promotes the in vitro development of pronuclear embryos and increases the efficiency of blastocyst implantation in murine. J. Pineal Res. 2013, 55, 267–274. [Google Scholar] [CrossRef] [PubMed]
  120. Cruz, M.H.; Leal, C.L.; Cruz, J.F.; Tan, D.X.; Reiter, R.J. Essential actions of melatonin in protecting the ovary from oxidative damage. Theriogenology 2014, 82, 925–932. [Google Scholar] [CrossRef] [PubMed]
  121. Banerjee, J.; Maitra, D.; Diamond, M.P.; Abu-Soud, H.M. Melatonin prevents hypochlorous acid-induced alterations in microtubule and chromosomal structure in metaphase-II mouse oocytes. J. Pineal Res. 2012, 53, 122–128. [Google Scholar] [CrossRef] [PubMed]
  122. Song, C.; Peng, W.; Yin, S.; Zhao, J.; Fu, B.; Zhang, J.; Mao, T.; Wu, H.; Zhang, Y. Melatonin improves age-induced fertility decline and attenuates ovarian mitochondrial oxidative stress in mice. Sci. Rep. 2016, 6, 35165. [Google Scholar] [CrossRef] [PubMed]
  123. Tamura, H.; Takasaki, A.; Miwa, I.; Taniguchi, K.; Maekawa, R.; Asada, H.; Taketani, T.; Matsuoka, A.; Yamagata, Y.; Shimamura, K.; et al. 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]
  124. He, B.; Yin, C.; Gong, Y.; Liu, J.; Guo, H.; Zhao, R. Melatonin-induced increase of lipid droplets accumulation and in vitro maturation in porcine oocytes is mediated by mitochondrial quiescence. J. Cell Physiol. 2017. [Google Scholar] [CrossRef] [PubMed]
  125. Tan, D.-X.; Manchester, L.; Qin, L.; Reiter, R. Melatonin: A Mitochondrial Targeting Molecule Involving Mitochondrial Protection and Dynamics. Int. J. Mol. Sci. 2016, 17, 2124. [Google Scholar] [CrossRef] [PubMed]
  126. Tong, J.; Sheng, S.; Sun, Y.; Li, H.; Li, W.P.; Zhang, C.; Chen, Z.J. Melatonin levels in follicular fluid as markers for IVF outcomes and predicting ovarian reserve. Reproduction 2017, 153, 443–451. [Google Scholar] [CrossRef] [PubMed]
  127. Tamura, H.; Takasaki, A.; Taketani, T.; Tanabe, M.; Kizuka, F.; Lee, L.; Tamura, I.; Maekawa, R.; Asada, H.; Yamagata, Y.; et al. Melatonin as a free radical scavenger in the ovarian follicle. Endocr. J. 2013, 60, 1–13. [Google Scholar] [CrossRef] [PubMed]
  128. Espino, J.; Bejarano, I.; Ortiz, A.; Lozano, G.M.; Garcia, J.F.; Pariente, J.A.; Rodriguez, A.B. Melatonin as a potential tool against oxidative damage and apoptosis in ejaculated human spermatozoa. Fertil. Steril. 2010, 94, 1915–1917. [Google Scholar] [CrossRef] [PubMed]
  129. Espino, J.; Ortiz, A.; Bejarano, I.; Lozano, G.M.; Monllor, F.; Garcia, J.F.; Rodriguez, A.B.; Pariente, J.A. Melatonin protects human spermatozoa from apoptosis via melatonin receptor- and extracellular signal-regulated kinase-mediated pathways. Fertil. Steril. 2011, 95, 2290–2296. [Google Scholar] [CrossRef] [PubMed]
  130. Jang, H.Y.; Kim, Y.H.; Kim, B.W.; Park, I.C.; Cheong, H.T.; Kim, J.T.; Park, C.K.; Kong, H.S.; Lee, H.K.; Yang, B.K. Ameliorative effects of melatonin against hydrogen peroxide-induced oxidative stress on boar sperm characteristics and subsequent in vitro embryo development. Reprod. Domest. Anim. 2010, 45, 943–950. [Google Scholar] [CrossRef] [PubMed]
  131. Okatani, Y.; Wakatsuki, A.; Kaneda, C. Melatonin increases activities of glutathione peroxidase and superoxide dismutase in fetal rat brain. J. Pineal Res. 2000, 28, 89–96. [Google Scholar] [CrossRef] [PubMed]
  132. Kaur, H.; Bhatla, S.C. Melatonin and nitric oxide modulate glutathione content and glutathione reductase activity in sunflower seedling cotyledons accompanying salt stress. Nitric Oxide 2016, 59, 42–53. [Google Scholar] [CrossRef] [PubMed]
  133. Li, X.X.; Yang, X.G.; Lu, Y.Q.; Lu, S.S.; Zhang, M.; Yao, H.I.; Meng, L.J.; Lu, K.H. Protective effects of melatonin against oxidative stress in flow cytometry-sorted buffalo sperm. Reprod. Domest. Anim. 2012, 47, 299–307. [Google Scholar] [CrossRef] [PubMed]
  134. Dehghani-Mohammadabadi, M.; Salehi, M.; Farifteh, F.; Nematollahi, S.; Arefian, E.; Hajjarizadeh, A.; Parivar, K.; Nourmohammadi, Z. Melatonin modulates the expression of BCL-xl and improve the development of vitrified embryos obtained by IVF in mice. J. Assist. Reprod. Genet. 2014, 31, 453–461. [Google Scholar] [CrossRef] [PubMed]
  135. Reiter, R.J.; Korkmaz, A.; Paredes, S.D.; Manchester, L.C.; Tan, D.X. Melatonin reduces oxidative/nitrosative stress due to drugs, toxins, metals, and herbicides. Neuro Endocrinol. Lett. 2008, 29, 609–613. [Google Scholar] [PubMed]
  136. Ersoz, N.; Guven, A.; Cayci, T.; Uysal, B.; Turk, E.; Oztas, E.; Akgul, E.O.; Korkmaz, A.; Cetiner, S. Comparison of the efficacy of melatonin and 1400W on renal ischemia/reperfusion injury: A role for inhibiting iNOS. Ren. Fail. 2009, 31, 704–710. [Google Scholar] [CrossRef] [PubMed]
  137. Garcia, J.A.; Ortiz, F.; Miana, J.; Doerrier, C.; Fernandez-Ortiz, M.; Rusanova, I.; Escames, G.; Garcia, J.J.; Acuna-Castroviejo, D. Contribution of inducible and neuronal nitric oxide synthases to mitochondrial damage and melatonin rescue in LPS-treated mice. J. Physiol. Biochem. 2017, 73, 235–244. [Google Scholar] [CrossRef] [PubMed]
  138. Tapias, V.; Escames, G.; Lopez, L.C.; Lopez, A.; Camacho, E.; Carrion, M.D.; Entrena, A.; Gallo, M.A.; Espinosa, A.; Acuna-Castroviejo, D. Melatonin and its brain metabolite N(1)-acetyl-5-methoxykynuramine prevent mitochondrial nitric oxide synthase induction in parkinsonian mice. J. Neurosci. Res. 2009, 87, 3002–3010. [Google Scholar] [CrossRef] [PubMed]
  139. 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. [Google Scholar] [CrossRef] [PubMed]
  140. Scala, G.; Maruccio, L. Nitric oxide (NO) expression during annual reproductive activity in buffalo epididymis: A histochemical and immunocytochemical study. Theriogenology 2012, 78, 49–56. [Google Scholar] [CrossRef] [PubMed]
  141. Rexhaj, E.; Pireva, A.; Paoloni-Giacobino, A.; Allemann, Y.; Cerny, D.; Dessen, P.; Sartori, C.; Scherrer, U.; Rimoldi, S.F. Prevention of vascular dysfunction and arterial hypertension in mice generated by assisted reproductive technologies by addition of melatonin to culture media. Am. J. Physiol. Heart Circ. Physiol. 2015, 309, H1151–H1156. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Effects of melatonin application in different steps on gametes and in vitro embryo production.
Figure 1. Effects of melatonin application in different steps on gametes and in vitro embryo production.
Ijms 18 01119 g001
Table 1. Effect of melatonin on different steps of assisted reproductive techniques in humans.
Table 1. Effect of melatonin on different steps of assisted reproductive techniques in humans.
SpecieTissueTreatmentResultsReference
HumanPatient3 mg per day from the fifth day onwards of one cycle in women with diminished ovarian reserveIncreases the mean number of M-II oocytes, top-quality embryos with grade 1 and 2[77]
HumanBlastocyst10−7 M in culture system in 3D (Encapsulation)Increases the survival time of encapsulated embryos[78]
HumanPatient3 mg for 14 days in patients with polycystic ovarian syndromeEnhances the oocyte and embryo quality[79]
HumanSperm0.01 mM in freezing extender before cryopreservation of sperm from infertile menIncreases motility and viability, decreases ROS and MDA levels[80]
HumanPatient6 mg for 45 daysIncreases the antioxidant capacity in seminal plasma, reduces the oxidative damage caused in sperm DNA, increases the quality of embryos[81]
HumanPatient3 mg/day for 2 weeksIncreases the fertilization rate I the second cycle, improves the fertilization and embryos quality rate[82]
Table 2. Effect of melatonin in different steps of assisted reproductive techniques in other mammals.
Table 2. Effect of melatonin in different steps of assisted reproductive techniques in other mammals.
SpecieTissueTreatmentResultsReference
PorcineOocytes10−9 M during in vitro maturationIncreases cleavage and blastocyst rate and the total cell number of blastocyst; promotes lipid metabolis, providing energy for oocyte maturation and embryo development[83]
RatAnimalIntraperitoneal injection of 20 mg/kg for 4 weeksIncreases testosterone hormone in blood serum and body weigh[84]
MouseSpermatogonial stem cells10 mg/kg for 2 weeks after busulfan treatmentRelieves the loss and apoptosis in mouse testes; upregulates MnSOD[85]
MouseOocytes10−9 to 10−3 M after in vitro maturationIncreases in vitro fertilization rate, reduces ROS and inhibits apoptosis[86]
BovineZygotes1 µM for 3 h after insemination and at 40 °CReduces ROS levels in embryos[87]
MouseOocyte M-II10−9 mol/L during vitrification/warming and PAIncreases blastocyst rate after warming compared with control group[88]
BovineSperm10−3 M for 3 h before in vitro fertilizationImproves plasma membrane and acrosome integrity, mitochondrial activity; decreases intracellular ROS levels; increases the blastocyst rate and it decreases apoptosis rate[89]
BovineOocytes10−6 or 10−9 M for 24 h during in vitro maturationUp-regulates MnSOD and Cu-ZnSOD in cumulus cells; decreases fragmentation. Decreases ROS levels In oocytes[90]
BovineEmbryos10−7 M melatonin for 24 h prior to exposure to 250 µM Paraquat (herbicide)Decreases the incidence of apoptotic nuclei induced by Paraquat[91]
PorcineOocytes0.1 μM for 22–44 h after endoplasmic reticulum stress during in vitro maturationImproves oocyte maturation and cumulus cells expansion induced by endoplasmic reticulum stress[92]
BovineOocytesMelatonin-loaded lipid-core nanocapsules at 10−6 M, 10−9 M and 10−12 M during in vitro maturationEnhances in vitro embryo production, decreases ROS levels and the apoptotic nuclei, upregulates GPX1 and SOD2 and downregulates CASP3 and BAX[93]
BovineZygotesMelatonin-loaded lipid-core nanocapsules at 10−9 M during in vitro cultureIncreases hatching rate and embryo cell number, decreases cell apoptosis and ROS levels; downregulates BAX, CASP3, and SHC1 genes, and upregulates CAT and SOD2 genes[94]
MouseOocytes10−7 M during in vitro maturationImproves blastocyst rate and cell number of blastocysts[95]
MouseSperm10 mg/kg body weight for 7 days during cadmium exposureReduces oxidative stress and inflammation induced by cadmium in male reproductive system[96]
MouseSperm0.125 mg/mL in freezing extender during cryopreservationIncreases progressive motility, decreases ROS levels and upregulates BCL-XL[97]
BuffaloOocytes250 µM during in vitro maturationImproves fertilization rate[98]
BovineOocytes1 µM during in vitro maturation of aged oocytesDecreased aberrant spindle organization, increases ATP production, increases the development of bovine oocytes and reduces apoptotic rat; downregulates BAX and CASP3 and increases BCL2[99]
RabbitMorula10−3 M prior in vitro culture, prior vitrificationPromotes blastocyst rate, increases SOD activity and decreases LPO and NO levels[100]
MousePreantral follicles10 pM after vitrification, during cultureIncreases diameter of follicles and their survival[101]
BovineEmbryos produced by SCNT10−11 to 10−2 M during in vitro cultureIncreases total cell number, ICM and the development of bovine SCNT embryos; suppresses the expression of p53 and Bax, and upregulates SOD1, Gpx4, BCL2L1 and SOX2[102]
PorcineOocyte10−7 M during in vitro maturation under heat stressImproves polar body and blastocyst rate impaired by heat stress.; preserves normal levels of steroid hormone, reduces ROS, enhances GSH production and inhibits apoptosis[103]
PorcineOocyte and embryos25 ng/mL during in vitro maturation and cultureIncreases blastocyst rate and decrease apoptotic nuclei in embryos[104]
BovineSperm1000 nMIncreases higher wobbler coefficient, decreases sperm with intact acrosome and viable spermatozoa with ROS[105]
RabbitEmbryos10−9 to 10−3 M during in vitro cultureIncreases in vitro development and improves hatching rate[106]
BovineZygotes10−7 M during in vitro culturePromotes the cleavage and blastocyst rate, accelerates the development of in vitro embryos and improves the quality of blastocysts[107]
BovineZygotes10−7 M for 2 days at the beginning of in vitro cultureIncreases the blastocysts and hatched blastocyst rate[108]
BovineZygotes10−9 M for after 2 days of pre-culture and for the remaining 6 days of cultureIncreases the blastocysts and hatched blastocysts rate[108]
BovineGV oocytes10−9 or 10−7 M during in vitro maturationImproves embryo development and the total cell number after in vitro fertilization; upregulates genes associated during in vitro maturation: GDF9, MARF1 and DNMT1α[109]
Mouse2-cell embryos10 µM during in vitro cultureImproves quality and developmental rate of embryos; can prevent cell death[110]
RatSperm10 mg/kg weekly for 8 weeksImproves sperm motility[111]
MouseEmbryos10−12 M during in vitro culture of embryos produced by SCNTIncreases embryo development[112]
OvineBlastocysts10−9 M during thawing after cryopreservationImproves embryo development after post warming culture[113]
DeerAnimalSubcutaneous implantation of 40 mgElevates serum FSH and LH levels, increases number of corpora luteal and the number of embryos[114]
SheepAnimalSubcutaneous implantation of 40 or 80 mgIncreases corpus lutea, the number of recovered embryos, pregnancy and birth rates, and the number of lambs born per embryo[115]
PorcineDonor cell and embryos10−10 M in the medium for donor cell and 10−9 M during in vitro culture of embryos produced by SCNTIncreases proliferation of fetal fibroblasts and the blastocysts rate,; reduces the apoptotic nuclei.; upregulates BCL2L1 and downregulates BAX and p53[116]
MouseOocytes10 to 100 nM during in vitro maturationIncreases expansion, maturation, fertilization and blastocyst rate in a dose dependent manner[117]
BovineOocytes10−12 to 10−3 M during in vitro maturation under heat stressIncreases blastocyst rate of embryos submitted to heat stress[118]
MurinePronuclear embryos10−7 M during in vitro culturePromotes embryo development, blastocyst rate, hatching rate and blastocyst cell number; upregulates SOD and BCL2 and downregulates CAS3 and p53[119]

Share and Cite

MDPI and ACS Style

Loren, P.; Sánchez, R.; Arias, M.-E.; Felmer, R.; Risopatrón, J.; Cheuquemán, C. Melatonin Scavenger Properties against Oxidative and Nitrosative Stress: Impact on Gamete Handling and In Vitro Embryo Production in Humans and Other Mammals. Int. J. Mol. Sci. 2017, 18, 1119. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18061119

AMA Style

Loren P, Sánchez R, Arias M-E, Felmer R, Risopatrón J, Cheuquemán C. Melatonin Scavenger Properties against Oxidative and Nitrosative Stress: Impact on Gamete Handling and In Vitro Embryo Production in Humans and Other Mammals. International Journal of Molecular Sciences. 2017; 18(6):1119. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18061119

Chicago/Turabian Style

Loren, Pía, Raúl Sánchez, María-Elena Arias, Ricardo Felmer, Jennie Risopatrón, and Carolina Cheuquemán. 2017. "Melatonin Scavenger Properties against Oxidative and Nitrosative Stress: Impact on Gamete Handling and In Vitro Embryo Production in Humans and Other Mammals" International Journal of Molecular Sciences 18, no. 6: 1119. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms18061119

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