Please wait a minute...
Journal of Integrative Agriculture  2018, Vol. 17 Issue (10): 2320-2335    DOI: 10.1016/S2095-3119(18)61942-2
Animal Science · Veterinary Medicine Advanced Online Publication | Current Issue | Archive | Adv Search |
Beneficial role of melatonin in protecting mammalian gametes and embryos from oxidative damage
PANG Yun-wei, JIANG Xiao-long, ZHAO Shan-jiang, HUANG Zi-qiang, ZHU Hua-bin
Embryo Biotechnology and Reproduction Laboratory, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R.China
Download:  PDF in ScienceDirect  
Export:  BibTeX | EndNote (RIS)      
Abstract  

Mammalian gametes and embryos are particularly vulnerable to oxidative stress-induced damage, which is mainly caused by reactive oxygen species (ROS) originating from normal metabolism and/or the external environment.  Several researchers have implicated the role of oxidative stress in the activation of apoptosis, causing peroxidative damage to sperms/oocytes and inducing embryo fragmentation, arrest, or demise.  Melatonin is a tryptophan derivative that is known for its powerful free radical-scavenging activity and broad-spectrum antioxidant property.  Numerous studies have shown that melatonin and its metabolic derivatives can sequentially detoxify ROS in an antioxidant cascade, and modulate various antioxidant enzymes via its receptors to prevent radical-mediated damage.  The identification of melatonin receptors in cumulus/granulosa cells, oocytes, and epididymal tissues implies that melatonin has protective actions on gametes and embryos.  Enriching the semen extender or culture medium with melatonin significantly benefits sperm characteristics, improves oocyte maturation potential and quality, and enhances the developmental competence of preimplantation embryos.  Certainly, further comparative studies are needed to show the unique antioxidant role and the advantage of melatonin in this field.  This review summarizes the harmful effects of ROS and the beneficial role of melatonin against oxidative damage of gametes and embryos. 

Keywords:  reactive oxygen species        oxidative stress        melatonin        sperm        oocyte        embryo development  
Received: 09 November 2017   Accepted:
Fund: This work was supported by the grants from the Agricultural Science and Technology Innovation Program, China (ASTIP-IAS06) and the earmarked fund for China Agriculture Research System (CARS-36).
Corresponding Authors:  Correspondence ZHU Hua-bin, Tel: +86-10-62815892, Fax: +86-10-62815351, E-mail: zhuhuabin@caas.cn   
About author:  PANG Yun-wei, Tel: +86-10-62815892, E-mail: pangyunwei@caas.cn;

Cite this article: 

PANG Yun-wei, JIANG Xiao-long, ZHAO Shan-jiang, HUANG Zi-qiang, ZHU Hua-bin. 2018. Beneficial role of melatonin in protecting mammalian gametes and embryos from oxidative damage. Journal of Integrative Agriculture, 17(10): 2320-2335.

Abecia J A, Forcada F, Zuniga O. 2002. The effect of melatonin on the secretion of progesterone in sheep and on the development of ovine embryos in vitro. Veterinary Research Communications, 26, 151–158.
Agarwal A, Gupta S, Sekhon L, Shah R. 2008. Redox considerations in female reproductive function and assisted reproduction: From molecular mechanisms to health implications. Antioxidants & Redox Signaling, 10, 1375–1403.
Agarwal A, Gupta S, Sharma R K. 2005. Role of oxidative stress in female reproduction. Reproductive Biology and Endocrinology, 3, 28.
Agarwal A, Said T M, Bedaiwy M A, Banerjee J, Alvarez J G. 2006. Oxidative stress in an assisted reproductive techniques setting. Fertility and Sterility, 86, 503–512.
Aitken J, Fisher H. 1994. Reactive oxygen species generation and human spermatozoa: The balance of benefit and risk. BioEssays, 16, 259–267.
Alvarez G M, Morado S A, Soto M P, Dalvit G C, Cetica P D. 2015. The control of reactive oxygen species influences porcine oocyte in vitro maturation. Reproduction in Domestic Animals, 50, 200–205.
Arendt J. 2000. Melatonin, circadian rhythms, and sleep. The New England Journal of Medicine, 343, 1114–1116.
Asgari Z, Ghasemian F, Ramezani M, Bahadori M H. 2012. The effect of melatonin on the developmental potential and implantation rate of mouse embryos. Cell Journal, 14, 203–208.
Ashrafi I, Kohram H, Ardabili F F. 2013. Antioxidative effects of melatonin on kinetics, microscopic and oxidative parameters of cryopreserved bull spermatozoa. Animal Reproduction Science, 139, 25–30.
Bain N T, Madan P, Betts D H. 2011. The early embryo response to intracellular reactive oxygen species is developmentally regulated. Reproduction Fertility and Development, 23, 561–575.
Bain N T, Madan P, Betts D H. 2013. Elevated p66Shc is associated with intracellular redox imbalance in developmentally compromised bovine embryos. Molecular Reproduction and Development, 80, 22–34.
Baker M A, Aitken R J. 2004. The importance of redox regulated pathways in sperm cell biology. Molecular and Cellular Endocrinology, 216, 47–54.
Balaban R S, Nemoto S, Finkel T. 2005. Mitochondria, oxidants, and aging. Cell, 120, 483–495.
Batioglu A S, Sahin U, Gurlek B, Ozturk N, Unsal E. 2012. The efficacy of melatonin administration on oocyte quality. Gynecological Endocrinology, 28, 91–93.
Benkhalifa M, Ferreira Y J, Chahine H, Louanjli N, Miron P, Merviel P, Copin H. 2014. Mitochondria: Participation to infertility as source of energy and cause of senescence. International Journal of Biochemistry & Cell Biology, 55, 60–64.
Berlinguer F, Leoni G G, Succu S, Spezzigu A, Madeddu M, Satta V, Bebbere D, Contreras-Solis I, Gonzalez-Bulnes A, Naitana S. 2009. Exogenous melatonin positively influences follicular dynamics, oocyte developmental competence and blastocyst output in a goat model. Journal of Pineal Research, 46, 383–391.
Betts D H, Bain N T, Madan P. 2014. The p66(Shc) adaptor protein controls oxidative stress response in early bovine embryos. PLoS ONE, 9, e86978.
Betts D H, Madan P. 2008. Permanent embryo arrest: Molecular and cellular concepts. Molecular Human Reproduction, 14, 445–453.
Bilodeau J F, Chatterjee S, Sirard M A, Gagnon C. 2000. Levels of antioxidant defenses are decreased in bovine spermatozoa after a cycle of freezing and thawing. Molecular Reproduction and Development, 55, 282–288.
Bornman M S, Oosthuizen J M, Barnard H C, Schulenburg G W, Boomker D, Reif S. 1989. Melatonin and sperm motility. Andrologia, 21, 483–485.
Burruel V, Klooster K, Barker C M, Pera R R, Meyers S. 2014. Abnormal early cleavage events predict early embryo demise: sperm oxidative stress and early abnormal cleavage. Scientific Reports, 4, 6598.
Carrillo-Vico A, Reiter R J, Lardone P J, Herrera J L, Fernandez-Montesinos R, Guerrero J M, Pozo D. 2006. The modulatory role of melatonin on immune responsiveness. Current Opinion in Investigational Drugs, 7, 423–431.
Casao A, Gallego M, Abecia J A, Forcada F, Perez-Pe R, Muino-Blanco T, Cebrian-Perez J A. 2012. Identification and immunolocalisation of melatonin MT(1) and MT(2) receptors in Rasa Aragonesa ram spermatozoa. Reproduction Fertility and Development, 24, 953–961.
Casao A, Mendoza N, Perez-Pe R, Grasa P, Abecia J A, Forcada F, Cebrian-Perez J A, Muino-Blanco T. 2010a. Melatonin prevents capacitation and apoptotic-like changes of ram spermatozoa and increases fertility rate. Journal of Pineal Research, 48, 39–46.
Casao A, Perez-Pe R, Abecia J A, Forcada F, Muino-Blanco T, Cebrian-Perez J A. 2013. The effect of exogenous melatonin during the non-reproductive season on the seminal plasma hormonal profile and the antioxidant defence system of Rasa Aragonesa rams. Animal Reproduction Science, 138, 168–174.
Casao A, Vega S, Palacin I, Perez-Pe R, Lavina A, Quintin F J, Sevilla E, Abecia J A, Cebrian-Perez J A, Forcada F, Muino-Blanco T. 2010b. Effects of melatonin implants during non-breeding season on sperm motility and reproductive parameters in Rasa Aragonesa rams. Reproduction in Domestic Animals, 45, 425–432.
Cassina A, Silveira P, Cantu L, Montes J M, Radi R, Sapiro R. 2015. Defective human sperm cells are associated with mitochondrial dysfunction and oxidant production. Biology of Reproduction, 93, 119.
de Castro L S, de Assis P M, Siqueira A F, Hamilton T R, Mendes C M, Losano J D, Nichi M, Visintin J A, Assumpcao M E. 2016. Sperm oxidative stress is detrimental to embryo development: A dose-dependent study model and a new and more sensitive oxidative status evaluation. Oxidative Medicine and Cellular Longevity, 2016, 8213071.
Cebral E, Carrasco I, Vantman D, Smith R. 2007. Preimplantation embryotoxicity after mouse embryo exposition to reactive oxygen species. Biocell, 31, 51–59.
Cebrian-Perez J A, Casao A, Gonzalez-Arto M, dos Santos Hamilton T R, Perez-Pe R, Muino-Blanco T. 2014. Melatonin in sperm biology: Breaking paradigms. Reproduction in Domestic Animals, 49(Suppl. 4), 11–21.
Chan K H, Wong Y H. 2013. A molecular and chemical perspective in defining melatonin receptor subtype selectivity. International Journal of Molecular Sciences, 14, 18385–18406.
Chen Y C, Sheen J M, Tiao M M, Tain Y L, Huang L T. 2013. Roles of melatonin in fetal programming in compromised pregnancies. International Journal of Molecular Sciences, 14, 5380–5401.
Cheuqueman C, Arias M E, Risopatron J, Felmer R, Alvarez J, Mogas T, Sanchez R. 2015. Supplementation of IVF medium with melatonin: Effect on sperm functionality and in vitro produced bovine embryos. Andrologia, 47, 604–615.
Choi J, Park S M, Lee E, Kim J H, Jeong Y I, Lee J Y, Park S W, Kim H S, Hossein M S, Jeong Y W, Kim S, Hyun S H, Hwang W S. 2008. Anti-apoptotic effect of melatonin on preimplantation development of porcine parthenogenetic embryos. Molecular Reproduction and Development, 75, 1127–1135.
Cruz M H, Leal C L, Cruz J F, Tan D X, Reiter R J. 2014a. Essential actions of melatonin in protecting the ovary from oxidative damage. Theriogenology, 82, 925–932.
Cruz M H, Leal C L, da Cruz J F, Tan D X, Reiter R J. 2014b. Role of melatonin on production and preservation of gametes and embryos: A brief review. Animal Reproduction Science, 145, 150–160.
Cutando A, Aneiros-Fernandez J, Lopez-Valverde A, Arias-Santiago S, Aneiros-Cachaza J, Reiter R J. 2011. A new perspective in oral health: Potential importance and actions of melatonin receptors MT1, MT2, MT3, and RZR/ROR in the oral cavity. Archives of Oral Biology, 56, 944–950.
Dumollard R, Carroll J, Duchen M R, Campbell K, Swann K. 2009. Mitochondrial function and redox state in mammalian embryos. Seminars in Cell & Development Biology, 20, 346–353.
Dumollard R, Ward Z, Carroll J, Duchen M R. 2007. Regulation of redox metabolism in the mouse oocyte and embryo. Development, 134, 455–465.
El-Raey M, Geshi M, Somfai T, Kaneda M, Hirako M, Abdel-Ghaffar A E, Sosa G A, El-Roos M E, Nagai T. 2011. Evidence of melatonin synthesis in the cumulus oocyte complexes and its role in enhancing oocyte maturation
in vitro in cattle. Molecular Reproduction and Development, 78, 250–262.
Favetta L A, Madan P, Mastromonaco G F, John St E J, King W A, Betts D H. 2007a. The oxidative stress adaptor p66Shc is required for permanent embryo arrest in vitro. BMC Developmental Biology, 7, 132.
Favetta L A, Robert C, St John E J, Betts D H, King W A. 2004. p66shc, but not p53, is involved in early arrest of in vitro-produced bovine embryos. Molecular Human Reproduction, 10, 383–392.
Favetta L A, St John E J, King W A, Betts D H. 2007b. High levels of p66shc and intracellular ROS in permanently arrested early embryos. Free Radical Biology and Medicine, 42, 1201–1210.
Feng R, Han J, Ziegler J, Yang M, Castranova V. 2012. Apaf-1
deficiency confers resistance to ultraviolet-induced apoptosis in mouse embryonic fibroblasts by disrupting reactive oxygen species amplification production and mitochondrial pathway. Free Radical Biology and Medicine, 52, 889–897.
Fischer T W, Kleszczynski K, Hardkop L H, Kruse N, Zillikens D. 2013. Melatonin enhances antioxidative enzyme gene expression (CAT, GPx, SOD), prevents their UVR-induced depletion, and protects against the formation of DNA damage (8-hydroxy-2´-deoxyguanosine) in ex vivo human skin. Journal of Pineal Research, 54, 303–312.
Fisher S P, Davidson K, Kulla A, Sugden D. 2008. Acute sleep-promoting action of the melatonin agonist, ramelteon, in the rat. Journal of Pineal Research, 45, 125–132.
Forcada F, Abecia J A, Cebrian-Perez J A, Muino-Blanco T, Valares J A, Palacin I, Casao A. 2006. The effect of melatonin implants during the seasonal anestrus on embryo production after superovulation in aged high-prolificacy Rasa Aragonesa ewes. Theriogenology, 65, 356–365.
Frungieri M B, Mayerhofer A, Zitta K, Pignataro O P, Calandra R S, Gonzalez-Calvar S I. 2005. Direct effect of melatonin on Syrian hamster testes: Melatonin subtype 1a receptors, inhibition of androgen production, and interaction with the local corticotropin-releasing hormone system. Endocrinology, 146, 1541–1552.
Fujii J, Tsunoda S. 2011. Redox regulation of fertilisation and the spermatogenic process. Asian Journal of Andrology, 13, 420–423.
Fujinoki M. 2008. Melatonin-enhanced hyperactivation of hamster sperm. Reproduction, 136, 533–541.
Galano A, Tan D X, Reiter R J. 2011. Melatonin as a natural ally against oxidative stress: A physicochemical examination. Journal of Pineal Research, 51, 1–16.
Galano A, Tan D X, Reiter R J. 2013. On the free radical scavenging activities of melatonin’s metabolites, AFMK and AMK. Journal of Pineal Research, 54, 245–257.
Ghaleno L R, Valojerdi M R, Hassani F, Chehrazi M, Janzamin E. 2014. High level of intracellular sperm oxidative stress negatively influences embryo pronuclear formation after intracytoplasmic sperm injection treatment. Andrologia, 46, 1118–1127.
Giorgio M, Migliaccio E, Orsini F, Paolucci D, Moroni M, Contursi C, Pelliccia G, Luzi L, Minucci S, Marcaccio M, Pinton P, Rizzuto R, Bernardi P, Paolucci F, Pelicci P G. 2005. Electron transfer between cytochrome c and p66Shc generates reactive oxygen species that trigger mitochondrial apoptosis. Cell, 122, 221–233.
Gonzalez-Arto M, Luna C, Perez-Pe R, Muino-Blanco T, Cebrian-Perez J A, Casao A. 2014. New evidence of melatonin receptor contribution to ram sperm functionality. Reproduction Fertility and Development, 28, 924–935.
Gonzalez-Arto M, Vicente-Carrillo A, Martinez-Pastor F, Fernandez-Alegre E, Roca J, Miro J, Rigau T, Rodriguez-Gil J E, Perez-Pe R, Muino-Blanco T, Cebrian-Perez J A, Casao A. 2016. Melatonin receptors MT1 and MT2 are expressed in spermatozoa from several seasonal and nonseasonal breeder species. Theriogenology, 86, 1958–1968.
Guerin P, El Mouatassim S, Menezo Y. 2001. Oxidative stress and protection against reactive oxygen species in the pre-implantation embryo and its surroundings. Human Reproduction Update, 7, 175–189.
Hashimoto S, Minami N, Yamada M, Imai H. 2000. 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. Molecular Reproduction and Development, 56, 520–526.
He C, Wang J, Li Y, Zhu K, Xu Z, Song Y, Song Y, Liu G. 2015. Melatonin-related genes expressed in the mouse uterus during early gestation promote embryo implantation. Journal of Pineal Research, 58, 300–309.
He C, Wang J, Zhang Z, Yang M, Li Y, Tian X, Ma T, Tao J, Zhu K, Song Y, Ji P, Liu G. 2016. Mitochondria synthesize melatonin to ameliorate its function and improve mice oocyte’s quality under in vitro conditions. International Journal of Molecular Sciences, 17, E939.
He Y, Deng H, Jiang Z, Li Q, Shi M, Chen H, Han Z. 2016. Effects of melatonin on follicular atresia and granulosa cell apoptosis in the porcine. Molecular Reproduction and Development, 83, 692–700.
Ishizuka B, Kuribayashi Y, Murai K, Amemiya A, Itoh M T. 2000. The effect of melatonin on in vitro fertilization and embryo development in mice. Journal of Pineal Research, 28, 48–51.
Itoh M T, Ishizuka B, Kuribayashi Y, Amemiya A, Sumi Y. 1999. Melatonin, its precursors, and synthesizing enzyme activities in the human ovary. Molecular Human Reproduction, 5, 402–408.
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. 2010. Ameliorative effects of melatonin against hydrogen peroxide-induced oxidative stress on boar sperm characteristics and subsequent in vitro embryo development. Reproduction in Domestic Animals, 45, 943–950.
Jin J X, Lee S, Taweechaipaisankul A, Kim G A, Lee B C. 2017. Melatonin regulates lipid metabolism in porcine oocytes. Journal of Pineal Research, 62, e12388.
Jockers R, Delagrange P, Dubocovich M L, Markus R P, Renault N, Tosini G, Cecon E, Zlotos D P. 2016. Update on melatonin receptors: IUPHAR Review 20. British Journal of Pharmacology, 173, 2702–2725.
Kang J T, Koo O J, Kwon D K, Park H J, Jang G, Kang S K, Lee B C. 2009. Effects of melatonin on in vitro maturation of porcine oocyte and expression of melatonin receptor RNA in cumulus and granulosa cells. Journal of Pineal Research, 46, 22–28.
Kim J H, Lee S H, Kim S, Jeong Y W, Koo O J, Hashem M D, Park S M, Lee E G, Hossein M S, Kang S K, Lee B C, Hwang W S. 2006. Embryotrophic effects of ethylenediaminetetraacetic acid and hemoglobin on
in vitro porcine embryos development. Theriogenology, 66, 449–455.
Kitagawa Y, Suzuki K, Yoneda A, Watanabe T. 2004. 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, 62, 1186–1197.
Koppers A J, De Iuliis G N, Finnie J M, McLaughlin E A, Aitken R J. 2008. Significance of mitochondrial reactive oxygen species in the generation of oxidative stress in spermatozoa. The Journal of Clinical Endocrinology and Metabolism, 93, 3199–3207.
Koppers A J, Garg M L, Aitken R J. 2010. Stimulation of mitochondrial reactive oxygen species production by unesterified, unsaturated fatty acids in defective human spermatozoa. Free Radical Biology and Medicine, 48, 112–119.
Kurcer Z, Hekimoglu A, Aral F, Baba F, Sahna E. 2010. Effect of melatonin on epididymal sperm quality after testicular ischemia/reperfusion in rats. Fertility and Sterility, 93, 1545–1549.
Lane M, McPherson N O, Fullston T, Spillane M, Sandeman L, Kang W X, Zander-Fox D L. 2014. Oxidative stress in mouse sperm impairs embryo development, fetal growth and alters adiposity and glucose regulation in female offspring. PLoS ONE, 9, e100832.
Lanoix D, Lacasse A A, Reiter R J, Vaillancourt C. 2012. Melatonin: The smart killer: The human trophoblast as a model. Molecular and Cellular Endocrinology, 348, 1–11.
Li R, Luo X, Li L, Peng Q, Yang Y, Zhao L, Ma M, Hou Z. 2016. The protective effects of melatonin against oxidative stress and inflammation induced by acute cadmium exposure in mice testis. Biological Trace Element Research, 170, 152–164.
Li X X, Yang X G, Lu Y Q, Lu S S, Zhang M, Yao H I, Meng L J, Lu K H. 2012. Protective effects of melatonin against oxidative stress in flow cytometry-sorted buffalo sperm. Reproduction in Domestic Animals, 47, 299–307.
Li Y, Zhang Z, He C, Zhu K, Xu Z, Ma T, Tao J, Liu G. 2015. Melatonin protects porcine oocyte in vitro maturation from heat stress. Journal of Pineal Research, 59, 365–375.
Lima G N, Maganhin C C, Simoes R S, Baracat M C, Sasso G R, Fuchs L F, Simoes Mde J, Baracat E C, Soares Jr. J M. 2015. Steroidogenesis-related gene expression in the rat ovary exposed to melatonin supplementation. Clinics, 70, 144–151.
Lopes A S, Lane M, Thompson J G. 2010. Oxygen consumption and ROS production are increased at the time of fertilization and cell cleavage in bovine zygotes. Human Reproduction, 25, 2762–2773.
Lord T, Aitken R J. 2013. Oxidative stress and ageing of the post-ovulatory oocyte. Reproduction, 146, R217–R227.
Lord T, Nixon B, Jones K T, Aitken R J. 2013. Melatonin prevents postovulatory oocyte aging in the mouse and extends the window for optimal fertilization in vitro. Biology of Reproduction, 88, 67.
Manjunatha B M, Devaraj M, Gupta P S, Ravindra J P, Nandi S. 2009. Effect of taurine and melatonin in the culture medium on buffalo in vitro embryo development. Reproduction in Domestic Animals, 44, 12–16.
Manka S, Majewska E. 2016. Immunoregulatory action of melatonin. The mechanism of action and the effect on inflammatory cells. Postepy Higieny I Medycyny Doswiadczalnej, 70, 1059–1067.
Martin-Hidalgo D, Baron F J, Bragado M J, Carmona P, Robina A, Garcia-Marin L J, Gil M C. 2011. The effect of melatonin on the quality of extended boar semen after long-term storage at 17 degrees C. Theriogenology, 75, 1550–1560.
McCormack A L, Atienza J G, Johnston L C, Andersen J K, Vu S, Di Monte D A. 2005. Role of oxidative stress in paraquat-induced dopaminergic cell degeneration. Journal of Neurochemistry, 93, 1030–1037.
Mehaisen G M, Saeed A M. 2015. In vitro development rate of preimplantation rabbit embryos cultured with different levels of melatonin. Zygote, 23, 111–115.
Mehaisen G M, Saeed A M, Gad A, Abass A O, Arafa M, El-Sayed A. 2015. Antioxidant capacity of melatonin on preimplantation development of fresh and vitrified rabbit embryos: Morphological and molecular aspects. PLoS ONE, 10, e0139814.
Nakano M, Kato Y, Tsunoda Y. 2012. Effect of melatonin treatment on the developmental potential of parthenogenetic and somatic cell nuclear-transferred porcine oocytes in vitro. Zygote, 20, 199–207.
Nikmard F, Hosseini E, Bakhtiyari M, Ashrafi M, Amidi F, Aflatoonian R. 2017. Effects of melatonin on oocyte maturation in PCOS mouse model. Animal Science Journal, 88, 586–592.
Niknafs B, Mehdipour A, Mohammadi Roushandeh A. 2014. Melatonin improves development of early mouse embryos impaired by actinomycin-D and TNF-alpha. Iranian Journal of Reproductive Medicine, 12, 799–804.
Niles L P, Wang J, Shen L, Lobb D K, Younglai E V. 1999. Melatonin receptor mRNA expression in human granulosa cells. Molecular and Cellular Endocrinology, 156, 107–110.
Niso-Santano M, Bravo-San Pedro J M, Gomez-Sanchez R, Climent V, Soler G, Fuentes J M, Gonzalez-Polo R A. 2011. ASK1 overexpression accelerates paraquat-induced autophagy via endoplasmic reticulum stress. Toxicological Sciences, 119, 156–168.
Ortiz A, Espino J, Bejarano I, Lozano G M, Monllor F, Garcia J F, Pariente J A, Rodriguez A B. 2011. High endogenous melatonin concentrations enhance sperm quality and short-term in vitro exposure to melatonin improves aspects of sperm motility. Journal of Pineal Research, 50, 132–139.
Othman A I, Edrees G M, El-Missiry M A, Ali D A, Aboel-Nour M, Dabdoub B R. 2016. Melatonin controlled apoptosis and protected the testes and sperm quality against bisphenol A-induced oxidative toxicity. Toxicology and Industrial Health, 32, 1537–1549.
Ou X H, Li S, Wang Z B, Li M, Quan S, Xing F, Guo L, Chao S B, Chen Z, Liang X W, Hou Y, Schatten H, Sun Q Y. 2012. Maternal insulin resistance causes oxidative stress and mitochondrial dysfunction in mouse oocytes. Human Reproduction, 27, 2130–2145.
Pacini N, Borziani F. 2016. Oncostatic-cytoprotective effect of melatonin and other bioactive molecules: A common target in mitochondrial respiration. International Journal of Molecular Sciences, 17, 341.
Pandi-Perumal S R, Trakht I, Srinivasan V, Spence D W, Maestroni G J, Zisapel N, Cardinali D P. 2008. Physiological effects of melatonin: Role of melatonin receptors and signal transduction pathways. Progress in Neurobiology, 85, 335–353.
Pang Y W, An L, Wang P, Yu Y, Yin Q D, Wang X H, Xin Z, Qian Z, Yang M L, Min G, Wu Z H, Tian J H. 2013. Treatment of porcine donor cells and reconstructed embryos with the antioxidant melatonin enhances cloning efficiency. Journal of Pineal Research, 54, 389–397.
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. 2016a. Protective effects of melatonin on bovine sperm characteristics and subsequent in vitro embryo development. Molecular Reproduction and Development, 83, 993–1002.
Pang Y W, Sun Y Q, Sun W J, Du W H, Hao H S, Zhao S J, Zhu H B. 2016b. Melatonin inhibits paraquat-induced cell death in bovine preimplantation embryos. Journal of Pineal Research, 60, 155–166.
Papis K, Poleszczuk O, Wenta-Muchalska E, Modlinski J A. 2007. Melatonin effect on bovine embryo development
in vitro in relation to oxygen concentration. Journal of Pineal Research, 43, 321–326.
du Plessis S S, Hagenaar K, Lampiao F. 2010. The in vitro effects of melatonin on human sperm function and its scavenging activities on NO and ROS. Andrologia, 42, 112–116.
Poeggeler B, Reiter R J, Tan D X, Chen L D, Manchester L C. 1993. Melatonin, hydroxyl radical-mediated oxidative damage, and aging: A hypothesis. Journal of Pineal Research, 14, 151–168.
Qian D, Li Z, Zhang Y, Huang Y, Wu Q, Ru G, Chen M, Wang B. 2016. Response of mouse zygotes treated with mild hydrogen peroxide as a model to reveal novel mechanisms of oxidative stress-induced injury in early embryos. Oxidative Medicine and Cellular Longevity, 2016, 1521428.
Ramalho-Santos J, Varum S, Amaral S, Mota P C, Sousa A P, Amaral A. 2009. Mitochondrial functionality in reproduction: From gonads and gametes to embryos and embryonic stem cells. Human Reproduction Update, 15, 553–572.
Ray P D, Huang B W, Tsuji Y. 2012. Reactive oxygen species (ROS) homeostasis and redox regulation in cellular signaling. Cellular Signalling, 24, 981–990.
Reiter R J, Rosales-Corral S A, Manchester L C, Tan D X. 2013. Peripheral reproductive organ health and melatonin: Ready for prime time. International Journal of Molecular Sciences, 14, 7231–7272.
Reiter R J, Tan D X, Fuentes-Broto L. 2010. Melatonin: A multitasking molecule. Progress in Brain Research, 181, 127–151.
Reiter R J, Tan D X, Korkmaz A. 2009a. The circadian melatonin rhythm and its modulation: Possible impact on hypertension. Journal of Hypertension Supplement, 27, S17–S20.
Reiter R J, Tan D X, Korkmaz A, Rosales-Corral S A. 2014. Melatonin and stable circadian rhythms optimize maternal, placental and fetal physiology. Human Reproduction Update, 20, 293–307.
Reiter R J, Tan D X, Maldonado M D. 2005. Melatonin as an antioxidant: Physiology versus pharmacology. Journal of Pineal Research, 39, 215–216.
Reiter R J, Tan D X, Manchester L C, Paredes S D, Mayo J C, Sainz R M. 2009b. Melatonin and reproduction revisited. Biology of Reproduction, 81, 445–456.
Reiter R J, Tan D X, Poeggeler B, Menendez-Pelaez A, Chen L D, Saarela S. 1994. Melatonin as a free radical scavenger: Implications for aging and age-related diseases. Annals of the New York Academy Sciences, 719, 1–12.
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, Junior A S, Seixas F K, Campos V F, Guterres S S, Collares T. 2016. Melatonin delivery by nanocapsules during in vitro bovine oocyte maturation decreased the reactive oxygen species of oocytes and embryos. Reproductive Toxicology, 63, 70–81.
Ren L, Wang Z, An L, Zhang Z, Tan K, Miao K, Tao L, Cheng L, Zhang Z, Yang M, Wu Z, Tian J. 2015. Dynamic comparisons of high-resolution expression profiles highlighting mitochondria-related genes between in vivo and in vitro fertilized early mouse embryos. Human Reproduction, 30, 2892–2911.
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, Leal C L. 2016. Effects of melatonin during IVM in defined medium on oocyte meiosis, oxidative stress, and subsequent embryo development. Theriogenology, 86, 1685–1694.
Rodriguez C, Mayo J C, Sainz R M, Antolin I, Herrera F, Martin V, Reiter R J. 2004. Regulation of antioxidant enzymes: A significant role for melatonin. Journal of Pineal Research, 36, 1–9.
Rodriguez-Osorio N, Kim I J, Wang H, Kaya A, Memili E. 2007. Melatonin increases cleavage rate of porcine preimplantation embryos in vitro. Journal of Pineal Research, 43, 283–288.
Roychoudhury S, Nath S, Massanyi P, Stawarz R, Kacaniova M, Kolesarova A. 2016. Copper-induced changes in reproductive functions: In vivo and in vitro effects. Physiological Research, 65, 11–22.
Sakaguchi K, Itoh M T, Takahashi N, Tarumi W, Ishizuka B. 2013. The rat oocyte synthesises melatonin. Reproduction Fertility and Development, 25, 674–682.
Salehi M, Kato Y, Tsunoda Y. 2014. Effect of melatonin treatment on developmental potential of somatic cell nuclear-transferred mouse oocytes in vitro. Zygote, 22, 213–217.
Sapanidou V, Taitzoglou I, Tsakmakidis I, Kourtzelis I, Fletouris D, Theodoridis A, Zervos I, Tsantarliotou M. 2015. Antioxidant effect of crocin on bovine sperm quality and
in vitro fertilization. Theriogenology, 84, 1273–1282.
Sarabia L, Maurer I, Bustos-Obregon E. 2009. Melatonin prevents damage elicited by the organophosphorous pesticide diazinon on mouse sperm DNA. Ecotoxicology and Environmental Safety, 72, 663–668.
Sariozkan S, Bucak M N, Tuncer P B, Tasdemir U, Kinet H, Ulutas P A. 2010. Effects of different extenders and centrifugation/washing on postthaw microscopic-oxidative stress parameters and fertilizing ability of Angora buck sperm. Theriogenology, 73, 316–323.
Seidler E A, Moley K H. 2015. Metabolic determinants of mitochondrial function in oocytes. Seminars in Reproductive Medicine, 33, 396–400.
Shaeib F, Banerjee J, Maitra D, Diamond M P, Abu-Soud H M. 2013. Impact of hydrogen peroxide-driven Fenton reaction on mouse oocyte quality. Free Radical Biology and Medicine, 58, 154–159.
Shi J M, Tian X Z, Zhou G B, Wang L, Gao C, Zhu S E, Zeng S M, Tian J H, Liu G S. 2009. Melatonin exists in porcine follicular fluid and improves in vitro maturation and parthenogenetic development of porcine oocytes. Journal of Pineal Research, 47, 318–323.
Shiu S Y, Li L, Siu S W, Xi S C, Fong S W, Pang S F. 2000. Biological basis and possible physiological implications of melatonin receptor-mediated signaling in the rat epididymis. Biological Signals and Receptors, 9, 172–187.
da Silva C M, Macias-Garcia B, Miro-Moran A, Gonzalez-Fernandez L, Morillo-Rodriguez A, Ortega-Ferrusola C, Gallardo-Bolanos J M, Stilwell G, Tapia J A, Pena F J. 2011. Melatonin reduces lipid peroxidation and apoptotic-like changes in stallion spermatozoa. Journal of Pineal Research, 51, 172–179.
Simoes R, Feitosa W B, Siqueira A F, Nichi M, Paula-Lopes F F, Marques M G, Peres M A, Barnabe V H, Visintin J A, Assumpcao M E. 2013. Influence of bovine sperm DNA fragmentation and oxidative stress on early embryo in vitro development outcome. Reproduction, 146, 433–441.
Soares Jr J M, Masana M I, Ersahin C, Dubocovich M L. 2003. Functional melatonin receptors in rat ovaries at various stages of the estrous cycle. The Journal of Pharmacology and Experimental Therapeutics, 306, 694–702.
Song C, Peng W, Yin S, Zhao J, Fu B, Zhang J, Mao T, Wu H, Zhang Y. 2016. Melatonin improves age-induced fertility decline and attenuates ovarian mitochondrial oxidative stress in mice. Scientific Reports, 6, 35165.
Sonmez M, Yuce A, Turk G. 2007. The protective effects of melatonin and vitamin E on antioxidant enzyme activities and epididymal sperm characteristics of homocysteine treated male rats. Reproductive Toxicology, 23, 226–231.
Su J, Wang Y, Xing X, Zhang L, Sun H, Zhang Y. 2015. Melatonin significantly improves the developmental competence of bovine somatic cell nuclear transfer embryos. Journal of Pineal Research, 59, 455–468.
Succu S, Berlinguer F, Pasciu V, Satta V, Leoni G G, Naitana S. 2011. Melatonin protects ram spermatozoa from cryopreservation injuries in a dose-dependent manner. Journal of Pineal Research, 50, 310–318.
Succu S, Pasciu V, Manca M E, Chelucci S, Torres-Rovira L, Leoni G G, Zinellu A, Carru C, Naitana S, Berlinguer F. 2014. Dose-dependent effect of melatonin on postwarming development of vitrified ovine embryos. Theriogenology, 81, 1058–1066.
Takada L, Junior A M, Mingoti G Z, Balieiro J C, Cipolla-Neto J, Coelho L A. 2012. Effect of melatonin on DNA damage of bovine cumulus cells during in vitro maturation (IVM) and on in vitro embryo development. Research in Veterinary Science, 92, 124–127.
Taketani T, Tamura H, Takasaki A, Lee L, Kizuka F, Tamura I, Taniguchi K, Maekawa R, Asada H, Shimamura K, Reiter R J, Sugino N. 2011. Protective role of melatonin in progesterone production by human luteal cells. Journal of Pineal Research, 51, 207–213.
Tamura H, Kawamoto M, Sato S, Tamura I, Maekawa R, Taketani T, Aasada H, Takaki E, Nakai A, Reiter R J, Sugino N. 2016. Long term melatonin treatment delays ovarian aging. Journal of Pineal Research, 62, e12381.
Tamura H, Nakamura Y, Korkmaz A, Manchester L C, Tan D X, Sugino N, Reiter R J. 2009. Melatonin and the ovary: Physiological and pathophysiological implications. Fertility and Sterility, 92, 328–343.
Tamura H, Takasaki A, Miwa I, Taniguchi K, Maekawa R, Asada H, Taketani T, Matsuoka A, Yamagata Y, Shimamura K, Morioka H, Ishikawa H, Reiter R J, Sugino N. 2008. Oxidative stress impairs oocyte quality and melatonin protects oocytes from free radical damage and improves fertilization rate. Journal of Pineal Research, 44, 280–287.
Tamura H, Takasaki A, Taketani T, Tanabe M, Kizuka F, Lee L, Tamura I, Maekawa R, Aasada H, Yamagata Y, Sugino N. 2012. The role of melatonin as an antioxidant in the follicle. Journal of Ovarian Research, 5, 5.
Tamura H, Takasaki A, Taketani T, Tanabe M, Kizuka F, Lee L, Tamura I, Maekawa R, Asada H, Yamagata Y, Sugino N. 2013. Melatonin as a free radical scavenger in the ovarian follicle. Endocrine Journal, 60, 1–13.
Tan K, An L, Wang S M, Wang X D, Zhang Z N, Miao K, Sui L L, He S Z, Nie J Z, Wu Z H, Tian J H. 2015. Actin disorganization plays a vital role in impaired embryonic development of in vitro-produced mouse preimplantation embryos. PLoS ONE, 10, e0130382.
Tanavde V S, Maitra A. 2003. In vitro modulation of steroidogenesis and gene expression by melatonin: A study with porcine antral follicles. Endocrine Research, 29, 399–410.
Tasdemir U, Buyukleblebici S, Tuncer P B, Coskun E, Ozgurtas T, Aydin F N, Buyukleblebici O, Gurcan I S. 2013. Effects of various cryoprotectants on bull sperm quality, DNA integrity and oxidative stress parameters. Cryobiology, 66, 38–42.
Tian X, Wang F, He C, Zhang L, Tan D, Reiter R J, Xu J, Ji P, Liu G. 2014. Beneficial effects of melatonin on bovine oocytes maturation: A mechanistic approach. Journal of Pineal Research, 57, 239–247.
Tomas-Zapico C, Coto-Montes A. 2005. A proposed mechanism to explain the stimulatory effect of melatonin on antioxidative enzymes. Journal of Pineal Research, 39, 99–104.
Tripathi A, Shrivastav T G, Chaube S K. 2013. An increase of granulosa cell apoptosis mediates aqueous neem (Azadirachta indica) leaf extract-induced oocyte apoptosis in rat. International Journal of Applied & Basic Medical Research, 3, 27–36.
Vargas A, Bustos-Obregon E, Hartley R. 2011. Effects of hypoxia on epididymal sperm parameters and protective role of ibuprofen and melatonin. Biological Research, 44, 161–167.
Vazquez M I, Abecia J A, Forcada F, Casao A. 2010. Effects of exogenous melatonin on in vivo embryo viability and oocyte competence of undernourished ewes after weaning during the seasonal anestrus. Theriogenology, 74, 618–626.
Velez-Pardo C, Morales A T, Del Rio M J, Olivera-Angel M. 2007. Endogenously generated hydrogen peroxide induces apoptosis via mitochondrial damage independent of NF-kappaB and p53 activation in bovine embryos. Theriogenology, 67, 1285–1296.
van Vuuren R J, Pitout M J, van Aswegen C H, Theron J J. 1992. Putative melatonin receptor in human spermatozoa. Clinical Biochemistry, 25, 125–127.
Wang F, Tian X, Zhang L, Gao C, He C, Fu Y, Ji P, Li Y, Li N, Liu G. 2014a. Beneficial effects of melatonin on in vitro bovine embryonic development are mediated by melatonin receptor 1. Journal of Pineal Research, 56, 333–342.
Wang F, Tian X, Zhang L, Tan D, Reiter R J, Liu G. 2013. Melatonin promotes the in vitro development of pronuclear embryos and increases the efficiency of blastocyst implantation in murine. Journal of Pineal Research, 55, 267–274.
Wang F, Tian X, Zhou Y, Tan D, Zhu S, Dai Y, Liu G. 2014b. Melatonin improves the quality of in vitro produced (IVP) bovine embryos: Implications for blastocyst development, cryotolerance, and modifications of relevant gene expression. PLoS ONE, 9, e93641.
Wang Q, Ratchford A M, Chi M M, Schoeller E, Frolova A, Schedl T, Moley K H. 2009. Maternal diabetes causes mitochondrial dysfunction and meiotic defects in murine oocytes. Molecular Endocrinology, 23, 1603–1612.
Wang S J, Liu W J, Wu C J, Ma F H, Ahmad S, Liu B R, Han L, Jiang X P, Zhang S J, Yang L G. 2012. Melatonin suppresses apoptosis and stimulates progesterone production by bovine granulosa cells via its receptors (MT1 and MT2). Theriogenology, 78, 1517–1526.
Winiarska K, Fraczyk T, Malinska D, Drozak J, Bryla J. 2006. Melatonin attenuates diabetes-induced oxidative stress in rabbits. Journal of Pineal Research, 40, 168–176.
Woo M M, Tai C J, Kang S K, Nathwani P S, Pang S F, Leung P C. 2001. Direct action of melatonin in human granulosa-luteal cells. The Journal of Clinical Endocrinology and Metabolism, 86, 4789–4797.
Yang H W, Hwang K J, Kwon H C, Kim H S, Choi K W, Oh K S. 1998. Detection of reactive oxygen species (ROS) and apoptosis in human fragmented embryos. Human Reproduction, 13, 998–1002.
Yu S, Long H, Lyu Q F, Zhang Q H, Yan Z G, Liang H X, Chai W R, Yan Z, Kuang Y P, Qi C. 2014. Protective effect of quercetin on the development of preimplantation mouse embryos against hydrogen peroxide-induced oxidative injury. PLoS ONE, 9, e89520.
Zhang H M, Zhang Y. 2014. Melatonin: A well-documented antioxidant with conditional pro-oxidant actions. Journal of Pineal Research, 57, 131–146.
Zhang L, Chai M, Tian X, Wang F, Fu Y, He C, Deng S, Lian Z, Feng J, Tan D X, Liu G. 2013. Effects of melatonin on superovulation and transgenic embryo transplantation in small-tailed han sheep (Ovis aries). Neuroendocrinology Letters, 34, 294–301.
Zhao M H, Liang S, Kim S H, Cui X S, Kim N H. 2015. Fe(III) is essential for porcine embryonic development via mitochondrial function maintenance. PLoS ONE, 10, e0130791.
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. 2016. Melatonin inhibits apoptosis and improves the developmental potential of vitrified bovine oocytes. Journal of Pineal Research, 60, 132–141.

[1] XU Yan-xia, ZHANG Jing, WAN Zi-yun, HUANG Shan-xia, DI Hao-chen, HE Ying, JIN Song-heng. Physiological and transcriptome analyses provide new insights into the mechanism mediating the enhanced tolerance of melatonin-treated rhododendron plants to heat stress[J]. >Journal of Integrative Agriculture, 2023, 22(8): 2397-2411.
[2] WANG Pei-hong, WANG Sai, NIE Wen-han, WU Yan, Iftikhar AHMAD, Ayizekeranmu YIMING, HUANG Jin, CHEN Gong-you, ZHU Bo. A transferred regulator that contributes to Xanthomonas oryzae pv. oryzicola oxidative stress adaptation and virulence by regulating the expression of cytochrome bd oxidase genes[J]. >Journal of Integrative Agriculture, 2022, 21(6): 1673-1682.
[3] YIN Jun-jie, XIONG Jun, XU Li-ting, CHEN Xue-wei, LI Wei-tao. Recent advances in plant immunity with cell death: A review[J]. >Journal of Integrative Agriculture, 2022, 21(3): 610-620.
[4] ZHAO Ying-jia, ZHANG Yan-yang, BAI Xin-yang, LIN Rui-ze, SHI Gui-qing, DU Ping-ping, XIAO Kai. TaNF-YB11, a gene of NF-Y transcription factor family in Triticum aestivum, confers drought tolerance on plants via modulating osmolyte accumulation and reactive oxygen species homeostasis[J]. >Journal of Integrative Agriculture, 2022, 21(11): 3114-3130.
[5] Muhammad Ahsan ASGHAR, JIANG Heng-ke, SHUI Zhao-wei, CAO Xi-yu, HUANG Xi-yu, Shakeel IMRAN, Bushra AHMAD, ZHANG Hao, YANG Yue-ning, SHANG Jing, YANG Hui, YU Liang, LIU Chun-yan, YANG Wen-yu, SUN Xin, DU Jun-bo. Interactive effect of shade and PEG-induced osmotic stress on physiological responses of soybean seedlings[J]. >Journal of Integrative Agriculture, 2021, 20(9): 2382-2394.
[6] Miilion P MADEBO, LUO Si-ming, WANG Li, ZHENG Yong-hua, JIN Peng. Melatonin treatment induces chilling tolerance by regulating the contents of polyamine, γ-aminobutyric acid, and proline in cucumber fruit[J]. >Journal of Integrative Agriculture, 2021, 20(11): 3060-3074.
[7] Iram SHAFIQ, Sajad HUSSAIN, Muhammad Ali RAZA, Nasir IQBAL, Muhammad Ahsan ASGHAR, Ali RAZA, FAN Yuan-fang, Maryam MUMTAZ, Muhammad SHOAIB, Muhammad ANSAR, Abdul MANAF, YANG Wen-yu, YANG Feng. Crop photosynthetic response to light quality and light intensity[J]. >Journal of Integrative Agriculture, 2021, 20(1): 4-23.
[8] WANG Yan, YAN Hao, WANG Qi, ZHENG Ran, XIA Kai, LIU Yang. Regulation of the phytotoxic response of Arabidopsis thaliana to the Fusarium mycotoxin deoxynivalenol[J]. >Journal of Integrative Agriculture, 2020, 19(3): 759-767.
[9] HUANG Cheng-zhen, XU Lei, Sun Jin-jing, ZHANG Zhong-hua, FU Mei-lan, TENG Hui-ying, YI Ke-ke.
Allelochemical p-hydroxybenzoic acid inhibits root growth via regulating ROS accumulation in cucumber (Cucumis sativus L.)
[J]. >Journal of Integrative Agriculture, 2020, 19(2): 518-527.
[10] WU Tian-ci, ZHU Xiu-liang, LÜ Liang-jie, CHEN Xi-yong, XU Gang-biao, ZHANG Zeng-yan. The wheat receptor-like cytoplasmic kinase TaRLCK1B is required for host immune response to the necrotrophic pathogen Rhizoctonia cerealis[J]. >Journal of Integrative Agriculture, 2020, 19(11): 2616-2627.
[11] LI Cheng-yang, ZHANG Nan, GUAN Bin, ZHOU Zhu-qing, MEI Fang-zhu . Reactive oxygen species are involved in cell death in wheat roots against powdery mildew[J]. >Journal of Integrative Agriculture, 2019, 18(9): 1961-1970.
[12] ZHAO Ying, LIU Meng, WANG Feng, DING Dong, ZHAO Chang-jiang, HE Lin, LI Zuo-tong, XU Jing-yu. The role of AtGPDHc2 in regulating cellular redox homeostasis of Arabidopsis under salt stress[J]. >Journal of Integrative Agriculture, 2019, 18(6): 1266-1279.
[13] ZHANG Bin, CUI Guo-bing, CHANG Chang-qing, WANG Yi-xu, ZHANG Hao-yang, CHEN Bao-shan, DENG Yi-zhen, JIANG Zi-de. The autophagy gene ATG8 affects morphogenesis and oxidative stress tolerance in Sporisorium scitamineum[J]. >Journal of Integrative Agriculture, 2019, 18(5): 1024-1034.
[14] XIAO Qian, ZHAO Chang-zhi, LIN Rui-yi, LI Guang-lei, LI Chang-chu, WANG Hai-yan, XU Jing, XIE Sheng-song, YU Mei, ZHAO Shu-hong. Production of homeobox A10 gene transgenic pigs by somatic cell nuclear transfer[J]. >Journal of Integrative Agriculture, 2019, 18(5): 1072-1079.
[15] Lü Jia-le, ZHANG Bao-he, JIANG Xiao-huan, WANG En-dong, XU Xue-nong. Quantitative impact of mating duration on reproduction and offspring sex ratio of Phytoseiulus persimilis (Acari: Phytoseiidae)[J]. >Journal of Integrative Agriculture, 2019, 18(4): 884-892.
No Suggested Reading articles found!