Next Article in Journal
Structural Optimization of BIPPO Analogs as Potent Antimalarials
Previous Article in Journal
Naphthalene-Based Polymers as Catalytic Supports for Suzuki Cross-Coupling
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of Melatonin in Bovine Reproductive Biotechnology

1
Joint Laboratory of the Modern Agricultural Technology International Cooperation, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
2
Key Lab of Animal Production, Product Quality, and Security, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
3
College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China
*
Authors to whom correspondence should be addressed.
Molecules 2023, 28(13), 4940; https://doi.org/10.3390/molecules28134940
Submission received: 15 May 2023 / Revised: 7 June 2023 / Accepted: 21 June 2023 / Published: 23 June 2023

Abstract

:
Melatonin has profound antioxidant activity and numerous functions in humans as well as in livestock and poultry. Additionally, melatonin plays an important role in regulating the biological rhythms of animals. Combining melatonin with scientific breeding management has considerable potential for optimizing animal physiological functions, but this idea still faces significant challenges. In this review, we summarized the beneficial effects of melatonin supplementation on physiology and reproductive processes in cattle, including granulosa cells, oocytes, circadian rhythm, stress, inflammation, testicular function, spermatogenesis, and semen cryopreservation. There is much emerging evidence that melatonin can profoundly affect cattle. In the future, we hope that melatonin can not only be applied to cattle, but can also be used to safely and effectively improve the efficiency of animal husbandry.

1. Introduction

Melatonin was discovered by Aaron Lerner in 1958 in the bovine pineal gland, and was determined to have an important role in the regulation of the circadian rhythm [1]. Consequently, melatonin became the “gold standard” drug for modulating the circadian rhythm [2] and was used to regulate sleep quality and body rhythms. Over the past 60 years, melatonin has been implicated in various tissues and cells in several animals [3,4,5,6], including the testes [7,8], ovaries [9], placenta [10], granulosa cells [11], and oocytes [12]. Melatonin has many roles in these tissues and cells, including scavenging reactive oxygen species (ROS) [13,14,15] and antioxidant [16,17], anti-apoptosis [18,19,20], anti-inflammatory [20,21,22], and anti-aging [23,24,25] activities.
The economic viability of the cattle industry depends heavily on good reproductive performance, which is affected by genetics, age, parity, body weight, nutrition [26], stress [27], endometritis [28], embryo quality [29], and conception rate [30]. Furthermore, bull breed and semen quality are also important factors affecting reproductive performance [31]. These factors can have causal relationships or may directly affect reproductive performance.
Although studies of melatonin in cattle are rapidly accumulating, there has not been a systematic review of its effects on cattle reproductive performance. Therefore, the main objective of this review was to focus on various factors that can influence bovine reproduction and explore how melatonin can mediate these factors. This includes examining the effects of melatonin on granulosa cells, oocytes, circadian rhythms, stress, inflammation, testicular function, spermatogenesis, and semen cryopreservation (Figure 1). We aim to provide insights to increase the use of exogenous melatonin in livestock reproduction.

2. Application of Melatonin in Bovine Granulosa Cells

Follicle atresia is important in the decline in bovine reproductive performance, and the development of ovarian follicles relies heavily on granulosa cells. Any alteration in the state of these cells, including apoptosis, autophagy, cell cycle arrest, or an accumulation of ROS, can trigger follicular atresia [32,33,34]. In addition, changes in steroid hormone synthesis also affect granulosa cell states [35].
Melatonin is widely recognized for its ability to scavenge ROS and regulate cellular physiology; in granulosa cells, it can reduce ROS and inhibit apoptosis through various mechanisms [33,35]. During the early stage of follicular atresia, the inner granulosa cell layer undergoes apoptosis, whereas the cumulus–oocyte and outer granulosa cells do not undergo apoptosis [36]. Therefore, granulosa cell variation may be an important first step in follicular atresia. Granulosa cells have critical roles in maintaining and supporting the growth of follicles in vivo, with various physiological states determining follicle fate [37]. Alterations in key cellular functions, including those of mitochondria, have important consequences, including ROS release, which triggers apoptosis and autophagy. Mitochondria are one of the main sources of ROS. Whereas some O2 is used to produce ATP to maintain mitochondrial function, some will generate ROS, and in excess concentrations, this will cause extracellular Ca2+ to enter cells, causing mitochondrial swelling and even rupture. Melatonin can sustain antioxidant enzymes and eliminate active oxygen by regulating ER oxidoreductin1 (ERO1) to improve the activity of superoxide dismutase (SOD) and catalase (CAT) [18,38]. Furthermore, melatonin can counteract β-zearalenol-mediated oxidative stress and apoptosis in bovine ovarian granulosa cells, along with significant increases in SOD and CAT proteins [33]. Inhibition of the melatonin receptors MT1 and MT2 can abate the effects of melatonin and block changes in the cell cycle [32]. In addition to scavenging ROS, melatonin has different effects depending on temperature and O2 concentration. At 37.5 °C and 5% O2, which approximates in vivo conditions, a low melatonin concentration promoted cell proliferation, but at 40 °C, a high melatonin concentration promoted cell proliferation [39]. Therefore, melatonin interacts with temperature in a dose-dependent manner, with the potential for melatonin to reduce stress caused by high temperatures. However, the body’s physiological status and melatonin secretion vary, posing challenges for safe and effective application of exogenous melatonin, emphasizing the need for reliable data to make evidence-based decisions.

3. Application of Melatonin in Bovine Oocyte Cells

Oocyte quality is a crucial factor affecting the reproductive performance of female animals and a key limiting factor in ruminant embryo transfer. Live birth rates were significantly higher for the transfer of fresh versus cryopreserved embryos [40]. Oocyte cryopreservation has been a longstanding challenge due to factors such as rates of survival, fertilization, and development [41]. Therefore, it is necessary to improve the quality of oocytes cultured in vitro prior to cryopreservation, with melatonin having considerable promise.
Numerous studies have demonstrated that melatonin can improve the developmental capacity of oocytes both in vitro and in vivo [42]. For instance, it enhances oocyte developmental competence and embryonic development in prepubertal and adult cattle by mitigating ROS [43]. Melatonin reduces the ROS content of heat-shocked oocytes, increases oocyte maturation rate and the proportion of embryos that develop into blastocysts, and increases the transcriptional abundance of genes related to mitochondrial function [44]. Additionally, melatonin also protects bovine oocytes from other harmful substances, e.g., preventing paraquat-induced oocyte damage and preserving embryonic developmental capacity [45]. There is abundant evidence for the ability of melatonin to promote bovine oocyte development [46]. In this regard, melatonin promotes the synthesis of antioxidant enzymes via specific membrane or nuclear receptors to remove ROS. Acetylserotonin O-methyltransferase (ASMT) in cumulus–oocyte complexes (COCs) may be involved in melatonin synthesis [47]. Melatonin reduces the oxidative stress of oocytes through the MT1 membrane receptor, protecting the spindle body function to maintain oocyte development. In Holstein cows, 20 mg of melatonin on days 190–262 of gestation increased uterine blood flow, possibly due to its effect on steroid metabolism [48]. Melatonin also altered estradiol metabolism to improve uteroplacental development in heifers [49]. In our studies of estrus and artificial insemination in cattle, exogenous melatonin significantly increased progesterone concentrations, enhanced SOD, CAT, and glutathione peroxidase (GSH-Px) activity, and decreased MDA concentrations in cattle blood, with a significant increase in pregnancy rate [50]. Melatonin could be a valuable tool for improving oocyte and embryonic development in vitro and a means to enhance in vivo fertility.

4. Melatonin Regulates Circadian Rhythms in Cattle

Melatonin can regulate behavior and reproduction by controlling the expression of genes involved in the circadian rhythm [51]. In addition, circadian rhythms can also affect cow physiology [52]. For example, in one study, luteinizing hormone (LH) secretion peaks before ovulation appeared 2–3 times more often at night than during daylight hours, whereas melatonin secretion peaked at night [53]. Interestingly, in luteal cells, melatonin increases the secretion of gonadotropin-releasing hormone (GnRH) and LH, thus enhancing progesterone secretion in a dose-dependent manner. Whereas granulosa cells are critical for estrogen secretion, luteal cells secrete more progesterone [54]. Perhaps melatonin directly affects ovulation in animals. For example, the influence of melatonin on GnRH and LH is blocked by luzindole, an inhibitor of the melatonin receptors MT1 and MT2. However, further investigation is needed to confirm the effects of melatonin on ovulation.
Melatonin also affects cow performance, as inhibiting melatonin secretion with light increases milk production in high-producing cows [55]. Long-day exposure reduces blood melatonin concentrations and increases blood prolactin concentrations in cattle [56,57], whereas exogenous melatonin suppresses prolactin [58]. Consequently, melatonin has profound effects on dairy cow performance, with the potential to modulate cow physiology and performance with light control and exogenous melatonin. Photoperiod has been used to improve animal productivity, including in chickens [59], sheep [60], and horses [61].
Melatonin is considered the “switch” that regulates circadian rhythms. One of the core genes of the circadian rhythm is BMAL1, which is considered indispensable. BMAL1 is in various cells and tissues, including the liver, testes, oocytes, and granulosa cells [62]. If damaged, it will cause the body to have a series of physiological abnormalities, including abnormal sleep, abnormal ovulation, and a shortened lifespan [63]. Melatonin restores sleep disorders in Parkinson’s disease (PD) patients by promoting BMAL1 gene expression. [64]. Furthermore, melatonin not only enhances autophagy through BMAL1 but also improves cerebral ischemia–reperfusion in diabetic mice [65]. It can also increase the expression of clock proteins to affect endocrine status and improve obesity in mice [66]. Blocking the BMAL1 gene with siRNA reduced the effects of melatonin on rooster circadian rhythms, implying that melatonin primarily functions through BMAL1 [67], with BMAL1 at the apex of the circadian clock feedback pathway in the avian retina [68]. Melatonin can regulate the secretion of testosterone and progesterone in male and female animals, which may be achieved through the core gene BMAL1. Despite no direct evidence to support this conjecture, reducing the expression of BMAL1 reduced testosterone secretion [69], and BMAL1 knockout mice had delayed genital development and puberty [70], perhaps due to the effects of melatonin on testosterone and progesterone secretion. Although most effects of circadian rhythms on cattle are centered on photoperiod, there is also evidence that BMAL1 has an important role in cattle. Knockdown of BMAL1 decreased prostaglandin F2α (PGF2α) in bovine uterine stromal cells (USCs) [71]. There is also evidence that BMAL1 functions as a core gene in bovine oocytes and preimplantation embryos to perform the same function as maternal mRNA [72]. In addition, circadian rhythm genes are involved in regulating neutrophil functions, helpful for assessing perinatal disease susceptibility in cattle [73]. Clearly, photoperiod and circadian rhythms affect cattle reproduction, with further studies needed to determine the specific roles of melatonin and mechanisms of action.

5. Effects of Melatonin on Inflammation in Cattle

Melatonin is a “regulator” of the immune system with both pro- and anti-inflammatory effects [74]. However, melatonin’s role in inflammation is not innate, but it depends on specific conditions. Melatonin generally has antioxidant and anti-inflammatory functions in most cells to maintain homeostasis [75,76]. In contrast, in tumor and cancer cells, it has potent pro-oxidative and pro-apoptotic therapeutic effects [77,78].
Exogenous melatonin can be given to treat various diseases by reducing inflammation and oxidative stress [74]. For example, it attenuated metabolic inflammation in mice by increasing exosomal α-ketoglutarate (αKG) [79]. It can also alleviate secondary brain injury caused by cerebral hemorrhage in rats by inhibiting inflammation [21]. Additionally, exogenous melatonin inhibited the release of inflammatory factors IL-6, IL-1β, and TNF-α in human nucleus pulposus cells (NPC), thereby suppressing inflammation [22]. Although less research has been conducted on the role of melatonin in bovine inflammation, it enhanced endometrial receptivity by alleviating ammonia-induced inflammation and apoptosis through the TLR4/NF-κB/IL-6 signaling pathway [80]. Rumen bypass membrane feeding (RBMF) can inhibit stress response and inflammation in dairy cows [81]. Melatonin also improved Staphylococcus aureus-induced mastitis by acting on the Microrna-16B/YAP1 pathway [82]. In addition, exogenous melatonin improved the efficiency of the bovine viral diarrhea virus vaccine [83], and based on in vitro studies, it inhibited NF-κB activity and reduced IL-1β and IL-6 mRNA levels [83]. Furthermore, melatonin also enhanced the immune response of sheep inoculated with Dichelobacter nodosus and increased serum IgG concentrations [84]. Therefore, melatonin has some anti-inflammatory and antiviral effects, with the potential to prevent or treat cattle diseases.

6. Effects of Melatonin on Testicular Function, Spermatogenesis, and Semen Cryopreservation in Bulls

Cryopreservation is an important assisted reproductive technology for long-term gamete preservation in ruminants such as cattle and sheep [85]. In addition to the importance of oocytes, high-quality semen also has an important role in accelerating genetic improvement, and is important for sustaining endangered animals. Therefore, there is much impetus to improve semen cryopreservation technology.
Semen contains a certain concentration of reactive oxygen species that are necessary for sperm capacitation [85]. However, high concentrations of ROS can cause oxidative stress to damage sperm physiological functions, including morphology and DNA integrity [86]. Melatonin reduces ROS-induced oxidative stress during sperm freezing and maximizes the quality of frozen-thawed sperm due to its ROS scavenging ability [10,87,88].
Adding 1 mM melatonin to semen extender improved the quality of swamp buffalo sperm [89]. Adding melatonin to semen extender mitigated reductions in the quality of frozen-thawed sperm from heat-stressed rams [90]. In Murrah buffalo bulls, a melatonin implant (18 mg/50 kg of body weight) lasted for 2 months and significantly reduced morphologically abnormal sperm and increased sperm motility, both in terms of curve and linear velocity. In addition, melatonin increased the concentration of total protein and cholesterol in seminal plasma and improved the semen quality of Murrah buffalo bulls during the non-breeding season under tropical conditions [91].
Su et al. [92] systematically studied effects of melatonin on semen cryopreservation, oocyte maturation, and embryonic development. During these processes, the optimal concentration of melatonin was not consistent. During semen freezing, 10−3 M was optimal, whereas 10−7 M significantly increased the oocyte maturation rate and also increased the total number of blastocysts in in vitro fertilization (IVF). In our previous work, various concentrations of melatonin had different effects on sperm motility and antioxidant indicators. Both low (0.125 mg/mL) and high (0.5 mg/mL) melatonin concentrations reduced ROS content. In addition, a medium concentration (0.25 mg/mL) of melatonin reduced MDA content. Although all three concentrations of melatonin improved antioxidant indicators, the medium concentration had the best advantage [87]. In conclusion, adding an appropriate concentration of melatonin to semen extender and sperm preparation for in vitro fertilization can improve the quality of frozen-thawed sperm, embryonic development, and success of in vitro fertilization.
In addition to exogenous melatonin in extender improving semen quality, melatonin is a key factor in spermatogenesis, which regulates testicular function through the hypothalamic–pituitary–gonadal axis [93,94]. The hypothalamic–pituitary–gonadal (HPG) axis is key to regulating reproductive hormones. Puberty was delayed in pups in pregnant female rats given melatonin, attributed to decreased LH and prolactin [95,96]. In addition, melatonin inhibits GnRH-induced LH release, thereby inhibiting testosterone production, whereas luzidole, an inhibitor of melatonin receptor MT1, essentially counteracts the effects of melatonin [96,97,98]. During testicular growth and development, melatonin has a critical role in several testicular cell types and hormone secretion. Melatonin protects the testes from local inflammation and reactive oxygen species [93,99,100]. Male reproductive function depends on the HPG axis, and melatonin can affect hormone synthesis, e.g., by modulating the growth and development of several testicular cells through its receptor [101]. Melatonin released by the pineal gland can also be absorbed by the testes through blood circulation, thereby modulating testicular activity [93]. In addition, melatonin can act through its unique receptors to regulate testosterone secretion, apoptosis, and autophagy [7,102,103]. Melatonin treatment of bovine Sertoli cells in vitro increased the expression of genes related to spermatogenesis, including Cyclin D1, Cyclin E, Pdgfa, Dhh, Occludin, and Claudin [104]. After healthy men took melatonin for 6-month, there were changes in some aspects of their semen, perhaps due to melatonin-induced inhibition of aromatase [105]. In a testicular ischemia–reperfusion model, melatonin significantly reduced morphologically abnormal sperm [106]. In vitro, melatonin increased the percentage of motile sperm and increased mitochondrial activity, implying that melatonin affected spermatogenesis and development through the blood–testis barrier [107]. These findings confirmed that melatonin regulated spermatogenesis via the development of testicular cells.

7. Positive Effects of Melatonin in Livestock Cells

Many positive effects of melatonin have been described above. However, optimal concentrations of melatonin seem to differ according to tissues or cells of various species. Due to relatively long estrous cycles (~21 days) and pregnancy (~285 days), it can be technically challenging to study the effects of melatonin in cattle. To the best of our knowledge, we were the first to report that melatonin improved fertility in cows [50]. Moreover, melatonin regulates the circadian rhythm and has a vital role in the milk production and metabolism of lactating cows [108]. The HPA axis can also promote gonadal atrophy of dairy cows in winter by increasing the secretion of melatonin [109]. There are limited studies on melatonin in domestic livestock, with a variety of doses and outcomes (Table 1). Melatonin can directly remove free radicals and ROS, and improve the antioxidant capacity by regulating cosmodal peroxidase and glutathione reduction [110].
Melatonin can also mitigate body damage nitrogen peroxide compound enzymes and nitrogen-based poison [111]. Melatonin can interact with heavy metals [112], combining with iron and copper. In hemoglobin, melatonin can restore iron (Feiv-O) to Iron (III) [113]. Melatonin can also be combined with CU (II) and CU (I) to reduce the lipid peroxidation of copper mediated in the liver [114]. Furthermore, melatonin can also assist mitochondria to clear free radicals and ROS [115]. Mitochondria are the site of ATP and ROS production [116]. The core theory of aging is mitochondrial damage, and melatonin can target mitochondria to generate ATP, remove ROS, and delay aging [117].
Melatonin can improve human sleep quality [118], regulate cardiovascular [119], central system [120], nervous system [121], and immune system function [122], and prevent the occurrence of some diseases to a certain extent, including Parkinson’s disease [123], Alzheimer’s disease [124], depression [125], and ischemic brain injury [126]. These human studies demonstrate the safety and effectiveness of melatonin, which lays the foundation for its use in animal husbandry, where there is much potential.
Table 1. Positive effects of melatonin on cattle, sheep, and pigs.
Table 1. Positive effects of melatonin on cattle, sheep, and pigs.
SpeciesPositive Effects of MelatoninConcentrationReferences
CattleMelatonin promoted diameter of bovine follicles and growth of secondary oocytes10−7 M[127]
Melatonin in cattle feed changed the β diversity of vaginal microorganisms20 mg[128]
Melatonin promoted proliferation of bovine theca cells and inhibited steroid production1 μM[129]
Melatonin promoted bovine oocyte development and maturation10−7 M[12]
Melatonin inhibited oxidative stress and apoptosis of bovine granulosa cells100 μM[33]
Melatonin increased conception rates in cattle0.24 mg/kg[50]
Melatonin decreased ROS production in bovine sperm and increased sperm viability, plasma membrane integrity, mitochondrial integrity, and acrosome integrity10−3 M[92]
Melatonin promoted development and function of bovine Sertoli cells320 pg/mL[104]
PigMelatonin regulated lipid metabolism in porcine oocytes10−9 M[130]
Melatonin reduced ROS production in porcine oocytes and promoted mitochondrial function and embryonic development500 nmol/L[131]
Melatonin improved the quality of porcine embryos1 nM[132]
Melatonin improved semen viability and acrosome integrity in pigs1 μM[133]
Melatonin regulated ATP metabolism and antioxidant enzyme activity of boar sperm1 μM[134]
SheepMelatonin inhibited LPS-induced inflammation of sheep epididymal epithelial cells10−7 M[135]
Melatonin was involved in activation of primordial follicles in sheep ovaries100 pg/mL[136]
Melatonin promoted development of transgenic sheep embryos and improved transgenic efficiency10−7 M[137]
Melatonin reduced ROS accumulation in sheep testicular interstitial cells and promoted testosterone synthesis10 ng/mL[138]
Dietary supplementation of melatonin increased activities of glucose amylase, isomaltase, and maltase in small intestine of sheep5 mg/d[139]
Melatonin reduced ROS and improved sperm quality10−7 M[140]
Melatonin enhanced DNA integrity and fertilization ability of sheep sperm1 mM[141]

8. Conclusions and Prospects

The physiological states of domestic animals will affect their reproductive function. Melatonin has important roles in the growth, development, and metabolism of various cells, but is rarely studied in livestock. This review emphasizes how melatonin works and its potential for use in animal, but much remains unknown.
In female livestock, melatonin can promote ovulation, enhance ovarian cell development and embryonic development, enhance placenta development, and increase pregnancy rate. In male livestock, melatonin can enhance testicular function, improve sperm morphology and motility, improve protein content in sperm, and enhance mitochondrial activity (Figure 2).
Improving the efficiency of livestock reproduction is critical for improving the sustainability of cattle reproduction. However, there are various factors affecting the breeding of domestic livestock. The physiological state of livestock and its interaction with the environment are complicated. As expected, melatonin has become a human health product, and its safety has been recognized. However, it is limited to basic research on domestic animals, which is even more rare for improving reproduction efficiency and has certain limitations. Melatonin as an endogenous hormone is important in the field of animal science and progress should be made in large livestock animals. We expect that in the future, scientific researchers can use melatonin to change domestic animal physiology (e.g., endocrinology, metabolism, and estrus cycle) and pathology. The combination of melatonin and scientific breeding management may have the best effect, thereby improving the reproductive performance of domestic animals.

Author Contributions

Conceptualization, Z.L. Literature collection and review, K.Z. and Y.Z.; Writing—original draft. Z.L., Revision and editing of original draft. J.W., Discussion and proofreading, J.Z.; Supervision and project administration, W.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Wenfa Lu and Jun Wang, grant number [2021YFF1000701-4], [31861143014], [CARS-37]. And The APC was funded by the Key R&D special subproject of the Ministry of Science and Technology in the 14th Five-Year Plan: Genetic basis and molecular regulatory Network Analysis of reproductive Traits in dairy cows; the National Natural Science Foundation of China International Cooperative Research and Exchange Program; and the China Agriculture Research System of MOF and MARA (CARS-37).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors thank the funds provided by the Key R&D special subproject of the Ministry of Science and Technology in the 14th Five-Year Plan: Genetic basis and molecular regulatory Network Analysis of reproductive Traits in dairy cows (Grant No. 2021YFF1000701-4), the National Natural Science Foundation of China International Cooperative Research and Exchange Program (Grant No. 31861143014), and the China Agriculture Research System of MOF and MARA (CARS-37).

Conflicts of Interest

The authors declare that they have no conflicts of interest.

References

  1. Yasmin, F.; Sutradhar, S.; Das, P.; Mukherjee, S. Gut melatonin: A potent candidate in the diversified journey of melatonin research. Gen. Comp. Endocrinol. 2021, 303, 113693. [Google Scholar] [CrossRef]
  2. Herxheimer, A.; Petrie, K.J. Melatonin for preventing and treating jet lag. Cochrane Database Syst. Rev. 2002, 2002, CD001520. [Google Scholar]
  3. Olcese, J.M. Melatonin and Female Reproduction: An Expanding Universe. Front. Endocrinol. 2020, 11, 85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Auld, F.; Maschauer, E.L.; Morrison, I.; Skene, D.J.; Riha, R.L. Evidence for the efficacy of melatonin in the treatment of primary adult sleep disorders. Sleep Med. Rev. 2017, 34, 10–22. [Google Scholar] [CrossRef]
  5. Paterson, A.M.; Foldes, A. Melatonin and farm animals: Endogenous rhythms and exogenous applications. J. Pineal Res. 1994, 16, 167–177. [Google Scholar] [CrossRef]
  6. Tosini, G.; Owino, S.; Guillaume, J.-L.; Jockers, R. Understanding melatonin receptor pharmacology: Latest insights from mouse models, and their relevance to human disease. Bioessays 2014, 36, 778–787. [Google Scholar] [CrossRef] [Green Version]
  7. Li, Z.; Zhao, J.; Liu, H.; Wang, J.; Lu, W. Melatonin inhibits apoptosis in mouse Leydig cells via the retinoic acid-related orphan nuclear receptor α/p53 pathway. Life Sci. 2020, 246, 117431. [Google Scholar] [CrossRef] [PubMed]
  8. Frungieri, M.B.; Mayerhofer, A.; Zitta, K.; Pignataro, O.P.; Calandra, R.S.; Gonzalez-Calvar, S.I. 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 2005, 146, 1541–1552. [Google Scholar] [CrossRef] [PubMed]
  9. Sundaresan, N.R.; Marcus Leo, M.D.; Subramani, J.; Anish, D.; Sudhagar, M.; Ahmed, K.A.; Saxena, M.; Tyagi, J.S.; Sastry, K.V.H.; Saxena, V.K. Expression analysis of melatonin receptor subtypes in the ovary of domestic chicken. Vet. Res. Commun. 2009, 33, 49–56. [Google Scholar] [CrossRef] [PubMed]
  10. Reiter, R.J.; Tan, D.X.; Korkmaz, A.; Rosales-Corral, S.A. Melatonin and stable circadian rhythms optimize maternal, placental and fetal physiology. Hum. Reprod. Update 2014, 20, 293–307. [Google Scholar] [CrossRef] [Green Version]
  11. Shen, M.; Cao, Y.; Jiang, Y.; Wei, Y.; Liu, H. Melatonin protects mouse granulosa cells against oxidative damage by inhibiting FOXO1-mediated autophagy: Implication of an antioxidation-independent mechanism. Redox Biol. 2018, 18, 138–157. [Google Scholar] [CrossRef] [PubMed]
  12. 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]
  13. Moniruzzaman, M.; Ghosal, I.; Das, D.; Chakraborty, S.B. Melatonin ameliorates HO-induced oxidative stress through modulation of Erk/Akt/NFkB pathway. Biol. Res. 2018, 51, 17. [Google Scholar] [CrossRef] [PubMed]
  14. He, R.; Cui, M.; Lin, H.; Zhao, L.; Wang, J.; Chen, S.; Shao, Z. Melatonin resists oxidative stress-induced apoptosis in nucleus pulposus cells. Life Sci. 2018, 199, 122–130. [Google Scholar] [CrossRef] [PubMed]
  15. Li, Z.; Wang, H.; Zhang, K.; Zhao, J.; Liu, H.; Ma, X.; Guo, J.; Wang, J.; Lu, W. Melatonin inhibits autophagy in TM3 cells via AKT/FOXO1 pathway. Mol. Biol. Rep. 2022, 49, 2925–2932. [Google Scholar] [CrossRef]
  16. Tamura, H.; Takasaki, A.; Taketani, T.; Tanabe, M.; Kizuka, F.; Lee, L.; Tamura, I.; Maekawa, R.; Aasada, H.; Yamagata, Y.; et al. The role of melatonin as an antioxidant in the follicle. J. Ovarian Res. 2012, 5, 5. [Google Scholar] [CrossRef] [Green Version]
  17. Reiter, R.J.; Paredes, S.D.; Manchester, L.C.; Tan, D.-X. Reducing oxidative/nitrosative stress: A newly-discovered genre for melatonin. Crit. Rev. Biochem. Mol. Biol. 2009, 44, 175–200. [Google Scholar] [CrossRef]
  18. Fernández, A.; Ordóñez, R.; Reiter, R.J.; González-Gallego, J.; Mauriz, J.L. Melatonin and endoplasmic reticulum stress: Relation to autophagy and apoptosis. J. Pineal Res. 2015, 59, 292–307. [Google Scholar] [CrossRef]
  19. Zhai, M.; Li, B.; Duan, W.; Jing, L.; Zhang, B.; Zhang, M.; Yu, L.; Liu, Z.; Yu, B.; Ren, K.; et al. Melatonin ameliorates myocardial ischemia reperfusion injury through SIRT3-dependent regulation of oxidative stress and apoptosis. J. Pineal Res. 2017, 63, e12419. [Google Scholar] [CrossRef]
  20. Zhi, S.M.; Fang, G.X.; Xie, X.M.; Liu, L.H.; Yan, J.; Liu, D.B.; Yu, H.Y. Melatonin reduces OGD/R-induced neuron injury by regulating redox/inflammation/apoptosis signaling. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 1524–1536. [Google Scholar] [CrossRef]
  21. Wang, Z.; Zhou, F.; Dou, Y.; Tian, X.; Liu, C.; Li, H.; Shen, H.; Chen, G. Melatonin Alleviates Intracerebral Hemorrhage-Induced Secondary Brain Injury in Rats via Suppressing Apoptosis, Inflammation, Oxidative Stress, DNA Damage, and Mitochondria Injury. Transl. Stroke Res. 2018, 9, 74–91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Zhang, Y.; He, F.; Chen, Z.; Su, Q.; Yan, M.; Zhang, Q.; Tan, J.; Qian, L.; Han, Y. Melatonin modulates IL-1β-induced extracellular matrix remodeling in human nucleus pulposus cells and attenuates rat intervertebral disc degeneration and inflammation. Aging 2019, 11, 10499–10512. [Google Scholar] [CrossRef]
  23. Tamura, H.; Kawamoto, M.; Sato, S.; Tamura, I.; Maekawa, R.; Taketani, T.; Aasada, H.; Takaki, E.; Nakai, A.; Reiter, R.J.; et al. Long-term melatonin treatment delays ovarian aging. J. Pineal Res. 2017, 62, e12381. [Google Scholar] [CrossRef]
  24. Lee, J.H.; Yoon, Y.M.; Song, K.-H.; Noh, H.; Lee, S.H. Melatonin suppresses senescence-derived mitochondrial dysfunction in mesenchymal stem cells via the HSPA1L-mitophagy pathway. Aging Cell 2020, 19, e13111. [Google Scholar] [CrossRef] [Green Version]
  25. Hardeland, R. Aging, Melatonin, and the Pro- and Anti-Inflammatory Networks. Int. J. Mol. Sci. 2019, 20, 1223. [Google Scholar] [CrossRef] [Green Version]
  26. D’Occhio, M.J.; Baruselli, P.S.; Campanile, G. Influence of nutrition, body condition, and metabolic status on reproduction in female beef cattle: A review. Theriogenology 2019, 125, 277–284. [Google Scholar] [CrossRef] [PubMed]
  27. Ciampi, F.; Sordillo, L.M.; Gandy, J.C.; Caroprese, M.; Sevi, A.; Albenzio, M.; Santillo, A. Evaluation of natural plant extracts as antioxidants in a bovine in vitro model of oxidative stress. J. Dairy Sci. 2020, 103, 8938–8947. [Google Scholar] [CrossRef]
  28. Dickson, M.J.; Piersanti, R.L.; Ramirez-Hernandez, R.; de Oliveira, E.B.; Bishop, J.V.; Hansen, T.R.; Ma, Z.; Jeong, K.C.C.; Santos, J.E.P.; Sheldon, M.I.; et al. Experimentally Induced Endometritis Impairs the Developmental Capacity of Bovine Oocytes. Biol. Reprod. 2020, 103, 508–520. [Google Scholar] [CrossRef]
  29. Fonseca Balvís, N.; Garcia-Martinez, S.; Pérez-Cerezales, S.; Ivanova, E.; Gomez-Redondo, I.; Hamdi, M.; Rizos, D.; Coy, P.; Kelsey, G.; Gutierrez-Adan, A. Cultured bovine embryo biopsy conserves methylation marks from original embryo. Biol. Reprod. 2017, 97, 189–196. [Google Scholar] [CrossRef] [PubMed]
  30. Shan, S.; Xu, F.; Bleyer, M.; Becker, S.; Melbaum, T.; Wemheuer, W.; Hirschfeld, M.; Wacker, C.; Zhao, S.; Schütz, E.; et al. Association of α/β-Hydrolase D16B with Bovine Conception Rate and Sperm Plasma Membrane Lipid Composition. Int. J. Mol. Sci. 2020, 21, 627. [Google Scholar] [CrossRef]
  31. Moore, S.G.; Hasler, J.F. A 100-Year Review: Reproductive technologies in dairy science. J. Dairy Sci. 2017, 100, 10314–10331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Wang, S.; Liu, W.; Wen, A.; Yang, B.; Pang, X. Luzindole and 4P-PDOT block the effect of melatonin on bovine granulosa cell apoptosis and cell cycle depending on its concentration. PeerJ 2021, 9, e10627. [Google Scholar] [CrossRef] [PubMed]
  33. Yang, F.; Li, L.; Chen, K.; Li, C.; Wang, Y.; Wang, G. Melatonin alleviates β-zearalenol and HT-2 toxin-induced apoptosis and oxidative stress in bovine ovarian granulosa cells. Environ. Toxicol. Pharmacol. 2019, 68, 52–60. [Google Scholar] [CrossRef] [PubMed]
  34. Ma, L.; Zheng, Y.; Tang, X.; Gao, H.; Liu, N.; Gao, Y.; Hao, L.; Liu, S.; Jiang, Z. miR-21-3p inhibits autophagy of bovine granulosa cells by targeting VEGFA via PI3K/AKT signaling. Reproduction 2019, 158, 441–452. [Google Scholar] [CrossRef] [PubMed]
  35. Wang, S.; Liu, W.; Pang, X.; Dai, S.; Liu, G. The Mechanism of Melatonin and Its Receptor MT2 Involved in the Development of Bovine Granulosa Cells. Int. J. Mol. Sci. 2018, 19, 2028. [Google Scholar] [CrossRef] [Green Version]
  36. Nakayama, M.; Manabe, N.; Nishihara, S.; Miyamoto, H. Species-specific Differences in Apoptotic Cell Localization in Granulosa and Theca Interna Cells during Follicular Atresia in Porcine and Bovine Ovaries. J. Reprod. Dev. 2000, 46, 147–156. [Google Scholar] [CrossRef] [Green Version]
  37. Yuan, X.-H.; Yang, C.-R.; Wang, X.-N.; Zhang, L.-L.; Gao, X.-R.; Shi, Z.-Y. Progesterone maintains the status of granulosa cells and slows follicle development partly through PGRMC1. J. Cell. Physiol. 2018, 234, 709–720. [Google Scholar] [CrossRef] [Green Version]
  38. Tamura, H.; Jozaki, M.; Tanabe, M.; Shirafuta, Y.; Mihara, Y.; Shinagawa, M.; Tamura, I.; Maekawa, R.; Sato, S.; Taketani, T.; et al. Importance of Melatonin in Assisted Reproductive Technology and Ovarian Aging. Int. J. Mol. Sci. 2020, 21, 1135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Zeebaree, B.K.; Kwong, W.Y.; Mann, G.E.; Gutierrez, C.G.; Sinclair, K.D. Physiological responses of cultured bovine granulosa cells to elevated temperatures under low and high oxygen in the presence of different concentrations of melatonin. Theriogenology 2018, 105, 107–114. [Google Scholar] [CrossRef]
  40. Insogna, I.G.; Lanes, A.; Lee, M.S.; Ginsburg, E.S.; Fox, J.H. Association of Fresh Embryo Transfers Compared With Cryopreserved-Thawed Embryo Transfers With Live Birth Rate Among Women Undergoing Assisted Reproduction Using Freshly Retrieved Donor Oocytes. JAMA 2021, 325, 156–163. [Google Scholar] [CrossRef]
  41. Chen, S.U.; Lien, Y.R.; Chao, K.H.; Ho, H.N.; Yang, Y.S.; Lee, T.Y. Effects of cryopreservation on meiotic spindles of oocytes and its dynamics after thawing: Clinical implications in oocyte freezing--a review article. Mol. Cell. Endocrinol. 2003, 202, 101–107. [Google Scholar] [CrossRef]
  42. Sananmuang, T.; Puthier, D.; Nguyen, C.; Chokeshaiusaha, K. Novel classifier orthologs of bovine and human oocytes matured in different melatonin environments. Theriogenology 2020, 156, 82–89. [Google Scholar] [CrossRef]
  43. Gutiérrez-Añez, J.C.; Lucas-Hahn, A.; Hadeler, K.-G.; Aldag, P.; Niemann, H. Melatonin enhances in vitro developmental competence of cumulus-oocyte complexes collected by ovum pick-up in prepubertal and adult dairy cattle. Theriogenology 2021, 161, 285–293. [Google Scholar] [CrossRef]
  44. Yaacobi-Artzi, S.; Shimoni, C.; Kalo, D.; Hansen, P.J.; Roth, Z. Melatonin slightly alleviates the effect of heat shock on bovine oocytes and resulting blastocysts. Theriogenology 2020, 158, 477–489. [Google Scholar] [CrossRef]
  45. Pang, Y.-W.; Jiang, X.-L.; Wang, Y.-C.; Wang, Y.-Y.; Hao, H.-S.; Zhao, S.-J.; Du, W.-H.; Zhao, X.-M.; Wang, L.; Zhu, H.-B. Melatonin protects against paraquat-induced damage during in vitro maturation of bovine oocytes. J. Pineal Res. 2019, 66, e12532. [Google Scholar] [CrossRef] [PubMed]
  46. Lan, M.; Han, J.; Pan, M.-H.; Wan, X.; Pan, Z.-N.; Sun, S.-C. Melatonin protects against defects induced by deoxynivalenol during mouse oocyte maturation. J. Pineal Res. 2018, 65, e12477. [Google Scholar] [CrossRef] [PubMed]
  47. El-Raey, M.; Geshi, M.; Somfai, T.; Kaneda, M.; Hirako, M.; Abdel-Ghaffar, A.E.; Sosa, G.A.; El-Roos, M.E.A.A.; Nagai, T. Evidence of melatonin synthesis in the cumulus oocyte complexes and its role in enhancing oocyte maturation in vitro in cattle. Mol. Reprod. Dev. 2011, 78, 250–262. [Google Scholar] [CrossRef] [PubMed]
  48. Brockus, K.E.; Hart, C.G.; Gilfeather, C.L.; Fleming, B.O.; Lemley, C.O. Dietary melatonin alters uterine artery hemodynamics in pregnant Holstein heifers. Domest. Anim. Endocrinol. 2016, 55, 1–10. [Google Scholar] [CrossRef] [PubMed]
  49. Lemley, C.O.; Vonnahme, K.A. Physiology and endocrinology symposium: Alterations in uteroplacental hemodynamics during melatonin supplementation in sheep and cattle. J. Anim. Sci. 2017, 95, 2211–2221. [Google Scholar] [CrossRef] [PubMed]
  50. Guo, L.; Li, M.; Gao, X.; Yang, Y.; Zhao, J.; Wang, J.; Lu, W. Two melatonin treatments improve the conception rate after fixed-time artificial insemination in beef heifers following synchronisation of oestrous cycles using the CoSynch-56 protocol. Aust. Vet. J. 2021, 99, 449–455. [Google Scholar] [CrossRef]
  51. Zhang, C.; Clough, S.J.; Adamah-Biassi, E.B.; Sveinsson, M.H.; Hutchinson, A.J.; Miura, I.; Furuse, T.; Wakana, S.; Matsumoto, Y.K.; Okanoya, K.; et al. Impact of endogenous melatonin on rhythmic behaviors, reproduction, and survival revealed in melatonin-proficient C57BL/6J congenic mice. J. Pineal Res. 2021, 71, e12748. [Google Scholar] [CrossRef] [PubMed]
  52. Shimizu, T.; Hirai, Y.; Murayama, C.; Miyamoto, A.; Miyazaki, H.; Miyazaki, K. Circadian Clock genes Per2 and clock regulate steroid production, cell proliferation, and luteinizing hormone receptor transcription in ovarian granulosa cells. Biochem. Biophys. Res. Commun. 2011, 412, 132–135. [Google Scholar] [CrossRef] [PubMed]
  53. Ginther, O.J.; Pinaffi, F.L.V.; Khan, F.A.; Duarte, L.F.; Beg, M.A. Circadian influence on the preovulatory LH surge, ovulation, and prolactin concentrations in heifers. Theriogenology 2013, 79, 528–533. [Google Scholar] [CrossRef]
  54. Zhang, W.; Zhang, Z.; Peng, J.; Yang, S.; Tong, D. Effects of melatonin on the production of GnRH and LH in luteal cells of pregnant sows. J. Mol. Endocrinol. 2022, 68, 111–123. [Google Scholar] [CrossRef] [PubMed]
  55. Murphy, B.A.; Herlihy, M.M.; Nolan, M.B.; O’Brien, C.; Furlong, J.G.; Butler, S.T. Identification of the blue light intensity administered to one eye required to suppress bovine plasma melatonin and investigation into effects on milk production in grazing dairy cows. J. Dairy Sci. 2021, 104, 12127–12138. [Google Scholar] [CrossRef] [PubMed]
  56. Peters, R.R.; Chapin, L.T.; Leining, K.B.; Tucker, H.A. Supplemental lighting stimulates growth and lactation in cattle. Science 1978, 199, 911–912. [Google Scholar] [CrossRef] [PubMed]
  57. Peters, R.R.; Chapin, L.T.; Emery, R.S.; Tucker, H.A. Milk yield, feed intake, prolactin, growth hormone, and glucocorticoid response of cows to supplemented light. J. Dairy Sci. 1981, 64, 1671–1678. [Google Scholar] [CrossRef] [PubMed]
  58. Sanchez-Barcelo, E.J.; Mediavilla, M.D.; Zinn, S.A.; Buchanan, B.A.; Chapin, L.T.; Tucker, H.A. Melatonin suppression of mammary growth in heifers. Biol. Reprod. 1991, 44, 875–879. [Google Scholar] [CrossRef]
  59. Tucker, H.A.; Ringer, R.K. Controlled photoperiodic environments for food animals. Science 1982, 216, 1381–1386. [Google Scholar] [CrossRef]
  60. Wyse, C.A.; Zhang, X.; McLaughlin, M.; Biello, S.M.; Hough, D.; Bellingham, M.; Curtis, A.M.; Robinson, J.E.; Evans, N.P. Circadian rhythms of melatonin and behaviour in juvenile sheep in field conditions: Effects of photoperiod, environment and weaning. Physiol. Behav. 2018, 194, 362–370. [Google Scholar] [CrossRef]
  61. O’Brien, C.; Darcy-Dunne, M.R.; Murphy, B.A. The effects of extended photoperiod and warmth on hair growth in ponies and horses at different times of year. PLoS ONE 2020, 15, e0227115. [Google Scholar] [CrossRef] [Green Version]
  62. Ray, S.; Valekunja, U.K.; Stangherlin, A.; Howell, S.A.; Snijders, A.P.; Damodaran, G.; Reddy, A.B. Circadian rhythms in the absence of the clock gene. Science 2020, 367, 800–806. [Google Scholar] [CrossRef] [PubMed]
  63. Wang, Y.; Chen, M.; Xu, J.; Liu, X.; Duan, Y.; Zhou, C.; Xu, Y. Core clock gene Bmal1 deprivation impairs steroidogenesis in mice luteinized follicle cells. Reproduction 2020, 160, 955–967. [Google Scholar] [CrossRef] [PubMed]
  64. Delgado-Lara, D.L.; González-Enríquez, G.V.; Torres-Mendoza, B.M.; González-Usigli, H.; Cárdenas-Bedoya, J.; Macías-Islas, M.A.; de la Rosa, A.C.; Jiménez-Delgado, A.; Pacheco-Moisés, F.; Cruz-Serrano, J.A.; et al. Effect of melatonin administration on the PER1 and BMAL1 clock genes in patients with Parkinson’s disease. Biomed. Pharmacother. 2020, 129, 110485. [Google Scholar] [CrossRef]
  65. Liu, L.; Cao, Q.; Gao, W.; Li, B.-Y.; Zeng, C.; Xia, Z.; Zhao, B. Melatonin ameliorates cerebral ischemia-reperfusion injury in diabetic mice by enhancing autophagy via the SIRT1-BMAL1 pathway. FASEB J. 2021, 35, e22040. [Google Scholar] [CrossRef]
  66. Arellanes-Licea, E.D.C.; Pérez-Mendoza, M.; Carmona-Castro, A.; Díaz-Muñoz, M.; Miranda-Anaya, M. Obese mice exhibit sexual dimorphism in the daily profile of circulating melatonin and clock proteins PER1 and BMAL1 in the hypothalamus and peripheral oscillators. Chronobiol. Int. 2021, 38, 584–597. [Google Scholar] [CrossRef] [PubMed]
  67. Ma, S.; Wang, Z.; Cao, J.; Dong, Y.; Chen, Y. BMAL1 but not CLOCK is associated with monochromatic green light-induced circadian rhythm of melatonin in chick pinealocytes. Endocr. Connect. 2019, 8, 57–68. [Google Scholar] [CrossRef] [Green Version]
  68. Bian, J.; Wang, Z.; Dong, Y.; Cao, J.; Chen, Y. Role of BMAL1 and CLOCK in regulating the secretion of melatonin in chick retina under monochromatic green light. Chronobiol. Int. 2020, 37, 1677–1692. [Google Scholar] [CrossRef]
  69. Ding, H.; Zhao, J.; Liu, H.; Wang, J.; Lu, W. BMAL1 knockdown promoted apoptosis and reduced testosterone secretion in TM3 Leydig cell line. Gene 2020, 747, 144672. [Google Scholar] [CrossRef]
  70. Boden, M.J.; Kennaway, D.J. 297. Reproduction in the arrhythmic Bmal1 knockout mouse. Reprod. Fertil. Dev. 2005, 17, 126. [Google Scholar] [CrossRef]
  71. Isayama, K.; Chen, H.; Yamauchi, N.; Hattori, M.-A. REV-ERBα inhibits the PTGS2 expression in bovine uterus endometrium stromal and epithelial cells exposed to ovarian steroids. J. Reprod. Dev. 2014, 60, 362–370. [Google Scholar] [CrossRef] [Green Version]
  72. 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]
  73. Nebzydoski, S.J.; Pozzo, S.; Nemec, L.; Rankin, M.K.; Gressley, T.F. The effect of dexamethasone on clock gene mRNA levels in bovine neutrophils and lymphocytes. Vet. Immunol. Immunopathol. 2010, 138, 183–192. [Google Scholar] [CrossRef]
  74. Hardeland, R. Melatonin and inflammation-Story of a double-edged blade. J. Pineal Res. 2018, 65, e12525. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Zhang, Z.; Lin, J.; Tian, N.; Wu, Y.; Zhou, Y.; Wang, C.; Wang, Q.; Jin, H.; Chen, T.; Nisar, M.; et al. Melatonin protects vertebral endplate chondrocytes against apoptosis and calcification via the Sirt1-autophagy pathway. J. Cell. Mol. Med. 2019, 23, 177–193. [Google Scholar] [CrossRef]
  76. Zhang, W.-X.; He, B.-M.; Wu, Y.; Qiao, J.-F.; Peng, Z.-Y. Melatonin protects against sepsis-induced cardiac dysfunction by regulating apoptosis and autophagy via activation of SIRT1 in mice. Life Sci. 2019, 21, 8–15. [Google Scholar] [CrossRef] [PubMed]
  77. Hardeland, R. Melatonin, noncoding RNAs, messenger RNA stability and epigenetics--evidence, hints, gaps and perspectives. Int. J. Mol. Sci. 2014, 15, 18221–18252. [Google Scholar] [CrossRef]
  78. Hardeland, R. Melatonin and the pathologies of weakened or dysregulated circadian oscillators. J. Pineal Res. 2017, 62, e12377. [Google Scholar] [CrossRef]
  79. Liu, Z.; Gan, L.; Zhang, T.; Ren, Q.; Sun, C. Melatonin alleviates adipose inflammation through elevating α-ketoglutarate and diverting adipose-derived exosomes to macrophages in mice. J. Pineal Res. 2018, 64, e12455. [Google Scholar] [CrossRef] [PubMed]
  80. Zheng, P.; Qin, X.; Feng, R.; Li, Q.; Huang, F.; Li, Y.; Zhao, Q.; Huang, H. Alleviative effect of melatonin on the decrease of uterine receptivity caused by blood ammonia through ROS/NF-κB pathway in dairy cow. Ecotoxicol. Environ. Saf. 2022, 231, 113166. [Google Scholar] [CrossRef] [PubMed]
  81. Yao, S.; Wu, H.; Ma, H.; Fu, Y.; Wei, W.; Wang, T.; Guan, S.; Yang, H.; Li, X.; Guo, J.; et al. Effects of rumen bypass melatonin feeding (RBMF) on milk quality and mastitis of Holstein cows. PeerJ 2020, 8, e9147. [Google Scholar] [CrossRef]
  82. Chen, Z.; Wang, K.; Guo, J.; Zhou, J.; Loor, J.J.; Yang, Z.; Yang, Y. Melatonin Maintains Homeostasis and Potentiates the Anti-inflammatory Response in Staphylococcus aureus-Induced Mastitis through microRNA-16b/YAP1. J. Agric. Food Chem. 2022, 70, 15255–15270. [Google Scholar] [CrossRef]
  83. Wang, Y.-X.; Yang, G.-H.; Zhang, L.-L.; Wang, J.; Wang, J.-F. Melatonin as Immune Potentiator for Enhancing Subunit Vaccine Efficacy against Bovine Viral Diarrhea Virus. Vaccines 2021, 9, 1039. [Google Scholar] [CrossRef]
  84. Regodón, S.; Ramos, A.; Morgado, S.; Tarazona, R.; Martín-Palomino, P.; Rosado, J.A.; Míguez, M.D.P. Melatonin enhances the immune response to vaccination against A1 and C strains of Dichelobacter nodosus. Vaccine 2009, 27, 1566–1570. [Google Scholar] [CrossRef]
  85. Ofosu, J.; Qazi, I.H.; Fang, Y.; Zhou, G. Use of melatonin in sperm cryopreservation of farm animals: A brief review. Anim. Reprod. Sci. 2021, 233, 106850. [Google Scholar] [CrossRef]
  86. Medrano, A.; Contreras, C.; Herrera, F.; Alcantar-Rodriguez, A. Melatonin as an antioxidant preserving sperm from domestic animals. Asian Pac. J. Reprod. 2017, 6, 241–246. [Google Scholar] [CrossRef]
  87. Appiah, M.O.; He, B.; Lu, W.; Wang, J. Antioxidative effect of melatonin on cryopreserved chicken semen. Cryobiology 2019, 89, 90–95. [Google Scholar] [CrossRef]
  88. Shahat, A.M.; Thundathil, J.C.; Kastelic, J.P. Melatonin or L-arginine in semen extender mitigate reductions in quality of frozen-thawed sperm from heat-stressed rams. Anim. Reprod. Sci. 2022, 238, 106934. [Google Scholar] [CrossRef] [PubMed]
  89. Inyawilert, W.; Rungruangsak, J.; Liao, Y.-J.; Tang, P.-C.; Paungsukpaibool, V. Melatonin supplementation improved cryopreserved Thai swamp buffalo semen. Reprod. Domest. Anim. 2021, 56, 83–88. [Google Scholar] [CrossRef] [PubMed]
  90. Shahat, A.M.; Thundathil, J.C.; Kastelic, J.P. Melatonin improves post-thaw sperm quality after mild testicular heat stress in rams. Reprod. Domest. Anim. 2023, 58, 423–430. [Google Scholar] [CrossRef]
  91. Ramadan, T.A.; Kumar, D.; Ghuman, S.S.; Singh, I. Melatonin-improved buffalo semen quality during nonbreeding season under tropical condition. Domest. Anim. Endocrinol. 2019, 68, 119–125. [Google Scholar] [CrossRef]
  92. Su, G.; Wu, S.; Wu, M.; Wang, L.; Yang, L.; Du, M.; Zhao, X.; Su, X.; Liu, X.; Bai, C.; et al. Melatonin improves the quality of frozen bull semen and influences gene expression related to embryo genome activation. Theriogenology 2021, 176, 54–62. [Google Scholar] [CrossRef] [PubMed]
  93. Frungieri, M.B.; Calandra, R.S.; Rossi, S.P. Local Actions of Melatonin in Somatic Cells of the Testis. Int. J. Mol. Sci. 2017, 18, 1170. [Google Scholar] [CrossRef] [Green Version]
  94. Shahat, A.M.; Thundathil, J.C.; Kastelic, J.P. Melatonin improves testicular hemodynamics and sperm quality in rams subjected to mild testicular heat stress. Theriogenology 2022, 188, 163–169. [Google Scholar] [CrossRef]
  95. Batmanabane, M.; Ramesh, K.G. Effect of exogenous melatonin on the onset of puberty in female albino rats. Anat. Rec. 1996, 245, 519–524. [Google Scholar] [CrossRef]
  96. Li, C.; Zhou, X. Melatonin and male reproduction. Clin. Chim. Acta 2015, 446, 175–180. [Google Scholar] [CrossRef]
  97. Vanecek, J. Melatonin inhibits release of luteinizing hormone (LH) via decrease of [Ca2+]i and cyclic AMP. Physiol. Res. 1998, 47, 329–335. [Google Scholar] [PubMed]
  98. Wu, C.S.; Leu, S.F.; Yang, H.Y.; Huang, B.M. Melatonin inhibits the expression of steroidogenic acute regulatory protein and steroidogenesis in MA-10 cells. J. Androl. 2001, 22, 245–254. [Google Scholar] [PubMed]
  99. Xu, K.; Wang, J.; Liu, H.; Zhao, J.; Lu, W. Melatonin Promotes the Proliferation of Chicken Sertoli Cells by Activating the ERK/Inhibin Alpha Subunit Signaling Pathway. Molecules 2020, 25, 1230. [Google Scholar] [CrossRef] [Green Version]
  100. El-Shalofy, A.; Hedia, M.; Kastelic, J. Melatonin improves testicular haemodynamics, echotexture and testosterone production in Ossimi rams during the breeding season. Reprod. Domest. Anim. 2021, 56, 1456–1463. [Google Scholar] [CrossRef]
  101. Heidarizadi, S.; Rashidi, Z.; Jalili, C.; Gholami, M. Overview of biological effects of melatonin on testis: A review. Andrologia 2022, 54, e14597. [Google Scholar] [CrossRef]
  102. Wang, M.; Zhu, C.-Q.; Zeng, L.; Cheng, L.; Ma, L.; Zhang, M.; Zhang, Y.-Z. Melatonin regulates the cross-talk between autophagy and apoptosis by SIRT3 in testicular Leydig cells. Biochem. Biophys. Res. Commun. 2021, 555, 182–189. [Google Scholar] [CrossRef]
  103. Qingyu, Z. Melatonin inhibits testosterone synthesis in Roosters Leydig cells by regulating lipolysis of lipid droplets. Theriogenology 2022, 189, 118–126. [Google Scholar]
  104. Yang, W.-C.; Tang, K.-Q.; Fu, C.-Z.; Riaz, H.; Zhang, Q.; Zan, L.-S. Melatonin regulates the development and function of bovine Sertoli cells via its receptors MT1 and MT2. Anim. Reprod. Sci. 2014, 147, 10–16. [Google Scholar] [CrossRef] [PubMed]
  105. Luboshitzky, R.; Shen-Orr, Z.; Nave, R.; Lavi, S.; Lavie, P. Melatonin administration alters semen quality in healthy men. J. Androl. 2002, 23, 572–578. [Google Scholar] [PubMed]
  106. Koksal, M.; Oğuz, E.; Baba, F.; Eren, M.A.; Ciftci, H.; Demir, M.E.; Kurcer, Z.; Take, G.; Aral, F.; Ocak, A.R.; et al. Effects of melatonin on testis histology, oxidative stress and spermatogenesis after experimental testis ischemia-reperfusion in rats. Eur. Rev. Med. Pharmacol. Sci. 2012, 16, 582–588. [Google Scholar] [PubMed]
  107. 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]
  108. Casey, T.M.; Plaut, K. Lactation Biology Symposium: Circadian clocks as mediators of the homeorhetic response to lactation. J. Anim. Sci. 2012, 90, 744–754. [Google Scholar] [CrossRef]
  109. Turek, F.W.; McMillan, J.P.; Menaker, M. Melatonin: Effects on the circadian locomotor rhythm of sparrows. Science 1976, 194, 1441–1443. [Google Scholar] [CrossRef]
  110. Poeggeler, B.; Reiter, R.J.; Tan, D.X.; Chen, L.D.; Manchester, L.C. Melatonin, hydroxyl radical-mediated oxidative damage, and aging: A hypothesis. J. Pineal Res. 1993, 14, 151–168. [Google Scholar] [CrossRef]
  111. Harasimowicz, J.; Marques, K.L.; Silva, A.F.T.; Costa, R.C.B.; Prior, J.A.V.; Rodrigues, S.S.M.; Santos, J.L.M. Chemiluminometric evaluation of melatonin and selected melatonin precursors’ interaction with reactive oxygen and nitrogen species. Anal. Biochem. 2012, 420, 1–6. [Google Scholar] [CrossRef]
  112. Gilad, E.; Cuzzocrea, S.; Zingarelli, B.; Salzman, A.L.; Szabó, C. Melatonin is a scavenger of peroxynitrite. Life Sci. 1997, 60, PL169–PL174. [Google Scholar] [CrossRef]
  113. Limson, J.; Nyokong, T.; Daya, S. The interaction of melatonin and its precursors with aluminium, cadmium, copper, iron, lead, and zinc: An adsorptive voltammetric study. J. Pineal Res. 1998, 24, 15–21. [Google Scholar] [CrossRef] [PubMed]
  114. Parmar, P.; Limson, J.; Nyokong, T.; Daya, S. Melatonin protects against copper-mediated free radical damage. J. Pineal Res. 2002, 32, 237–242. [Google Scholar] [CrossRef] [PubMed]
  115. Murphy, M.P. How mitochondria produce reactive oxygen species. Biochem. J. 2009, 417, 1–13. [Google Scholar] [CrossRef] [Green Version]
  116. Bratic, A.; Larsson, N.-G. The role of mitochondria in aging. J. Clin. Invest. 2013, 123, 951–957. [Google Scholar] [CrossRef] [Green Version]
  117. Poljsak, B. Strategies for reducing or preventing the generation of oxidative stress. Oxid. Med. Cell. Longev. 2011, 2011, 194586. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Zisapel, N. New perspectives on the role of melatonin in human sleep, circadian rhythms and their regulation. Br. J. Pharmacol. 2018, 175, 3190–3199. [Google Scholar] [CrossRef] [Green Version]
  119. Gombert, M.; Codoñer-Franch, P. Melatonin in Early Nutrition: Long-Term Effects on Cardiovascular System. Int. J. Mol. Sci. 2021, 22, 6809. [Google Scholar] [CrossRef]
  120. Claustrat, B.; Leston, J. Melatonin: Physiological effects in humans. Neurochirurgie 2015, 61, 77–84. [Google Scholar] [CrossRef]
  121. Tamtaji, O.R.; Mirhosseini, N.; Reiter, R.J.; Azami, A.; Asemi, Z. Melatonin, a calpain inhibitor in the central nervous system: Current status and future perspectives. J. Cell. Physiol. 2019, 234, 1001–1007. [Google Scholar] [CrossRef]
  122. Ma, N.; Zhang, J.; Reiter, R.J.; Ma, X. Melatonin mediates mucosal immune cells, microbial metabolism, and rhythm crosstalk: A therapeutic target to reduce intestinal inflammation. Med. Res. Rev. 2020, 40, 606–632. [Google Scholar] [CrossRef]
  123. Gilat, M.; Coeytaux Jackson, A.; Marshall, N.S.; Hammond, D.; Mullins, A.E.; Hall, J.M.; Fang, B.A.M.; Yee, B.J.; Wong, K.K.H.; Grunstein, R.R.; et al. Melatonin for rapid eye movement sleep behavior disorder in Parkinson’s disease: A randomised controlled trial. Mov. Disord. 2020, 35, 344–349. [Google Scholar] [CrossRef] [Green Version]
  124. Chen, C.; Yang, C.; Wang, J.; Huang, X.; Yu, H.; Li, S.; Li, S.; Zhang, Z.; Liu, J.; Yang, X.; et al. Melatonin ameliorates cognitive deficits through improving mitophagy in a mouse model of Alzheimer’s disease. J. Pineal Res. 2021, 71, e12774. [Google Scholar] [CrossRef]
  125. Ali, T.; Rahman, S.U.; Hao, Q.; Li, W.; Liu, Z.; Ali Shah, F.; Murtaza, I.; Zhang, Z.; Yang, X.; Liu, G.; et al. Melatonin prevents neuroinflammation and relieves depression by attenuating autophagy impairment through FOXO3a regulation. J. Pineal Res. 2020, 69, e12667. [Google Scholar] [CrossRef]
  126. Ramos, E.; Patiño, P.; Reiter, R.J.; Gil-Martín, E.; Marco-Contelles, J.; Parada, E.; de Los Rios, C.; Romero, A.; Egea, J. Ischemic brain injury: New insights on the protective role of melatonin. Free Radic. Biol. Med. 2017, 104, 32–53. [Google Scholar] [CrossRef]
  127. Paulino, L.R.F.M.; Barroso, P.A.A.; Silva, B.R.; Barroso, L.G.; Barbalho, E.C.; Bezerra, F.T.G.; Souza, A.L.P.; Monte, A.P.O.; Silva, A.W.B.; Matos, M.H.T.; et al. Immunolocalization of melatonin receptors in bovine ovarian follicles and in vitro effects of melatonin on growth, viability and gene expression in secondary follicles. Domest. Anim. Endocrinol. 2022, 81, 106750. [Google Scholar] [CrossRef] [PubMed]
  128. Messman, R.D.; Contreras-Correa, Z.E.; Paz, H.A.; Lemley, C.O. Melatonin-induced changes in the bovine vaginal microbiota during maternal nutrient restriction. J. Anim. Sci. 2021, 99, skab098. [Google Scholar] [CrossRef] [PubMed]
  129. Feng, T.; Schutz, L.F.; Morrell, B.C.; Perego, M.C.; Spicer, L.J. Effect of melatonin on bovine theca cells in vitro. Reprod. Fertil. Dev. 2018, 30, 643–650. [Google Scholar] [CrossRef] [PubMed]
  130. 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, e12388. [Google Scholar] [CrossRef]
  131. Niu, Y.-J.; Zhou, W.; Nie, Z.-W.; Shin, K.-T.; Cui, X.-S. Melatonin enhances mitochondrial biogenesis and protects against rotenone-induced mitochondrial deficiency in early porcine embryos. J. Pineal Res. 2020, 68, e12627. [Google Scholar] [CrossRef]
  132. Martinez, C.A.; Cuello, C.; Parrilla, I.; Maside, C.; Ramis, G.; Cambra, J.M.; Vazquez, J.M.; Rodriguez-Martinez, H.; Gil, M.A.; Martinez, E.A. Exogenous Melatonin in the Culture Medium Does Not Affect the Development of In Vivo-Derived Pig Embryos but Substantially Improves the Quality of In Vitro-Produced Embryos. Antioxidants 2022, 11, 1177. [Google Scholar] [CrossRef] [PubMed]
  133. Pezo, F.; Zambrano, F.; Uribe, P.; Moya, C.; de Andrade, A.F.C.; Risopatron, J.; Yeste, M.; Burgos, R.A.; Sánchez, R. Oxidative and nitrosative stress in frozen-thawed pig spermatozoa. I: Protective effect of melatonin and butylhydroxytoluene on sperm function. Res. Vet. Sci. 2021, 136, 143–150. [Google Scholar] [CrossRef] [PubMed]
  134. Lu, N.; Jiang, X.; Zhang, C.; Li, B.; Tu, W.; Lei, H.; Yao, W.; Xia, D. Melatonin mediates via melatonin receptor 1 in a temperature-dependent manner regulating ATP metabolism and antioxidative enzyme activity of boar spermatozoa in vitro. Theriogenology 2022, 188, 1–12. [Google Scholar] [CrossRef] [PubMed]
  135. Ge, W.-B.; Xiao, L.-F.; Duan, H.-W.; Li, Z.-S.; Jiang, Y.-T.; Yang, S.-S.; Hu, J.-J.; Zhang, Y.; Zhao, X.-X. Melatonin protects against lipopolysaccharide-induced epididymitis in sheep epididymal epithelial cells in vitro. Immunol. Lett. 2019, 214, 45–51. [Google Scholar] [CrossRef]
  136. Barberino, R.S.; Macedo, T.J.S.; Lins, T.L.B.G.; Menezes, V.G.; Silva, R.L.S.; Monte, A.P.O.; Palheta, R.C.; Smitz, J.E.J.; Matos, M.H.T. Immunolocalization of melatonin receptor type 1 in the sheep ovary and involvement of the PI3K/Akt/FOXO3a signaling pathway in the effects of melatonin on survival and in vitro activation of primordial follicles. Mol. Reprod. Dev. 2022, 89, 485–497. [Google Scholar] [CrossRef]
  137. Yao, Y.; Yang, A.; Li, G.; Wu, H.; Deng, S.; Yang, H.; Ma, W.; Lv, D.; Fu, Y.; Ji, P.; et al. Melatonin promotes the development of sheep transgenic cloned embryos by protecting donor and recipient cells. Cell Cycle 2022, 21, 1360–1375. [Google Scholar] [CrossRef]
  138. Ma, J.; Yang, H.; Liu, L.; Wan, Y.; Wang, F. Melatonin alleviated oxidative stress induced by energy restriction on sheep Leydig cells through Sirt1/Sod2 pathway. Theriogenology 2021, 173, 83–92. [Google Scholar] [CrossRef]
  139. Trotta, R.J.; Lemley, C.O.; Vonnahme, K.A.; Swanson, K.C. Effects of nutrient restriction and melatonin supplementation from mid-to-late gestation on maternal and fetal small intestinal carbohydrase activities in sheep. Domest. Anim. Endocrinol. 2021, 74, 106555. [Google Scholar] [CrossRef]
  140. Fang, Y.; Zhao, C.; Xiang, H.; Jia, G.; Zhong, R. Melatonin improves cryopreservation of ram sperm by inhibiting mitochondrial permeability transition pore opening. Reprod. Domest. Anim. 2020, 55, 1240–1249. [Google Scholar] [CrossRef]
  141. Succu, S.; Berlinguer, F.; Pasciu, V.; Satta, V.; Leoni, G.G.; Naitana, S. Melatonin protects ram spermatozoa from cryopreservation injuries in a dose-dependent manner. J. Pineal Res. 2011, 50, 310–318. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Effects of melatonin on physiological status of cattle.
Figure 1. Effects of melatonin on physiological status of cattle.
Molecules 28 04940 g001
Figure 2. Effects of melatonin on bovine reproduction.
Figure 2. Effects of melatonin on bovine reproduction.
Molecules 28 04940 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Li, Z.; Zhang, K.; Zhou, Y.; Zhao, J.; Wang, J.; Lu, W. Role of Melatonin in Bovine Reproductive Biotechnology. Molecules 2023, 28, 4940. https://doi.org/10.3390/molecules28134940

AMA Style

Li Z, Zhang K, Zhou Y, Zhao J, Wang J, Lu W. Role of Melatonin in Bovine Reproductive Biotechnology. Molecules. 2023; 28(13):4940. https://doi.org/10.3390/molecules28134940

Chicago/Turabian Style

Li, Zhiqiang, Kaiyan Zhang, Yuming Zhou, Jing Zhao, Jun Wang, and Wenfa Lu. 2023. "Role of Melatonin in Bovine Reproductive Biotechnology" Molecules 28, no. 13: 4940. https://doi.org/10.3390/molecules28134940

Article Metrics

Back to TopTop