Open access peer-reviewed chapter

An Overview of Effects on Reproductive Physiology of Melatonin

Written By

Volkan Gelen, Emin Şengül and Abdulsamed Kükürt

Submitted: 20 June 2022 Reviewed: 15 September 2022 Published: 23 October 2022

DOI: 10.5772/intechopen.108101

From the Edited Volume

Melatonin - Recent Updates

Edited by Volkan Gelen, Emin Şengül and Abdulsamed Kükürt

Chapter metrics overview

172 Chapter Downloads

View Full Metrics

Abstract

Melatonin is a neurotransmitter released from the pineal gland. The presence of receptor sites in the hypothalamus, pituitary gland, ovaries, and testicles and secretion of pituitary hormones (FSH and LH) are some of the effects of this hormone on reproduction. In addition to its systemic effect, it also showed an effect on ovarian physiology with the detection of high levels in the follicular fluid and the presence of melatonin receptors in the ovarian cells. In addition, it has been determined that melatonin affects follicular growth, oocyte maturation, ovulation, and luteal function. It has been stated that the effects of melatonin on the male reproductive system are indirectly effective through the gonads and indirectly by affecting the hormones. Again, some studies have expressed that melatonin has strong antioxidant properties and affects reproductive physiology due to this effect. This section discusses the effect of melatonin on male and female reproductive physiology.

Keywords

  • melatonin
  • reproduction
  • physiology
  • FSH
  • LH
  • ovarium
  • testes
  • oxidative stress

1. Introduction

Melatonin is a hormone secreted by the pineal gland and is known to be involved in the regulation of many body functions and the regulation of circadian and seasonal rhythms [1]. This hormone is rhythmically released from the epiphysis at night [2]. Since its release is regulated according to the nighttime, its secretion occurs with the internal reflection of the external photoperiod [3]. Melatonin is also called the “hormone of darkness”. It has been determined that melatonin is synthesized in the lacrimal gland, retina, erythrocytes, platelets, and some cells in the gastrointestinal tract other than the pineal gland, but this synthesis has little effect on the plasma melatonin level [4, 5, 6, 7]. The main synthesis site of melatonin is pinealocytes, and it is synthesized from tryptophan in these cells. Tryptophan is first converted to 5-hydroxytryptophan by the enzyme tryptophan hydroxylase. Then 5-hydroxytryptophan is converted to serotonin by amino acid decarboxylase. Serotonin is converted to N-acetyl serotonin by the enzyme N-acetyl transferase. N-acetyl serotonin is converted to melatonin by the enzyme methyltransferase. Thus, the synthesized melatonin is released into the blood circulation [8, 9]. Around 19.00–20.00, melatonin secretion begins to increase, reaches its highest point between 02.00 and 04.00 at night, and decreases with the increase of daylight in the morning. It starts to decrease between 07.00 and 09.00 in the morning [10, 11, 12]. Melatonin takes part in many biological and physiological regulations in the body. It is effective on biorhythm (circadian rhythm) and also has direct and indirect effects on the reproductive system, regenerating our cells, regulating the immune system, anticarcinogen, antioxidant, antiaging, and reproductive system [13]. This chapter, it is aimed to explain the physiological effects of melatonin, which has important bodily effects in the body, on the female and male reproductive systems.

Advertisement

2. Melatonin synthesis

Melatonin is synthesized from tryptophan in pinealocytes in the pineal gland. Synthesis stages are, respectively: Tryptophan is converted to 5-hydroxytryptophan by the enzyme tryptophan hydroxylase. Then 5-hydroxytryptophan is converted to serotonin by the amino acid decarboxylase enzyme. Serotonin is converted to N-acetyl serotonin by the enzyme N-acetyl transferase. N-acetyl serotonin is converted to melatonin by the enzyme methyltransferase. Thus, the synthesized melatonin is released into the blood circulation [8, 9]. The regulation of melatonin synthesis is controlled by the retinohypothalamic pathway and a system that surrounds the suprachiasmatic nuclei and contains multiple synapses within this pathway. Stimuli from the retina reach the hypothalamus and then this information reaches the pineal gland via peripheral postganglionic sympathetic fibers and melatonin is synthesized [9]. It activates protein kinase C (PKC) via β- and α1-adrenergic receptors on pinealocytes with increased norepinephrine stimulation at night. PKC activation increases the Ca2+ movement in the cell, resulting in an increase in the concentration of intracellular cyclic adenosine monophosphate (cAMP). This increase activates protein kinase A (PKA) (Figures 1 and 2). Activated PKA increases AANAT activity [14]. Once synthesized, it is directly secreted into the cerebrospinal fluid and blood of the third ventricle and reaches all body tissues in a very short time. After melatonin is released, it is transported by diffusion and some carrier molecules [15]. Melatonin can regulate a variety of physiological functions, from the well-known modulation of sleep/wake cycles and circadian rhythms to the maintenance and regulation of neural development and immune system and endocrine functions [16]. These effects of the melatonin are mediated by G protein-linked melatonin receptors MT1 and MT2. In addition, activation of a putative cytoplasmic melatonin receptor MT3 is also effective [17]. When melatonin binds to these receptors, due to the connection between the receptors and Gai/o proteins, it causes activity in the target tissue by regulating the levels of cAMP and calcium, the second messenger, as well as by regulating the activation of PKC subtypes (Figure 3) [19]. As a result of various studies, it has been determined that these receptors of Melatonin are in various structures of the brain belonging to the nervous system [20] as well as in nonneural tissues such as the immune system, endocrine system, bone, gastrointestinal system, cardiovascular system and reproductive organs, which we will focus on [21]. Again, as a result of a study, it was determined that melatonin causes various activities in various tissues through a mechanism independent of its receptors [22].

Advertisement

3. Melatonin receptors and their functions

It has been reported that melatonin synthesized by the pineal gland has 3 groups of receptors. These are MT1 receptors with high affinity. They are MT2 and MT3 receptors with low affinity [23, 24, 25]. MT1 stimulation from these receptors suppresses adenylate cyclase activity in the target tissue. It has been determined that these receptors are involved in retinal function, cerebral artery contraction, circadian rhythm, renal function, and reproduction. As a result of the stimulation of MT2 and MT3 receptors, phosphoinositol hydrolysis takes place in the target tissue [24, 25]. These receptors are located in the body of the brain, cardiovascular system, coronary and cerebral arteries, retina, ventricular wall, aorta, liver, and gall bladder, enterocytes, cecum, skin, colon, and appendix vermiformis, parotid gland, immune system cells, exocrine pancreas, kidney, platelets, ovary/granulosa cells, myometrium, placenta, and fetal kidney.

Advertisement

4. Effects of melatonin on the female reproductive system

Various studies have shown the effect of melatonin on the female reproductive system. It has been determined that this effect is by binding directly to the receptors in the cells in the ovary and through the hypothalamic/pituitary axis.

4.1 Direct effects of melatonin on the female reproductive system

Melatonin is considered to have an inhibitory effect on the hypothalamus-pituitary-gonads system. Demonstration of melatonin receptors in the ovary and structures such as mammary glands supports this effect. Studies have shown that melatonin receptors in rat ovarian tissue are more in the proestrus cycle than in the metestrus cycle and that granulosa cells are the main site for the MT1 receptor [26, 27]. The concentration of melatonin in follicular fluid was found to be ten times higher than in plasma. The source of the melatonin in the follicular fluid in circulation. The follicles receive this hormone from the bloodstream. The amount of hormone intake varies depending on the follicular growth period. The larger the follicles, the more melatonin is taken from the blood. Studies have reported that melatonin acts as an antioxidant in follicles and contributes to progesterone production by luteinizing granulosa cells [28]. Melatonin levels in humans during pregnancy and delivery are higher compared to the postpartum period [29]. Increased melatonin levels before birth may serve as an important circadian signal for the time of birth. Maternal melatonin originates from the maternal pineal gland and increases with the activities of placental hormones [28]. Melatonin, which is present in human follicular fluid in conjunction with plasma and does not affect granulosa cells’ steroidogenesis and follicular function, directly affects ovarian function [30]. Periodic fluctuations are seen in the follicular fluid that fills the antral cavity, and there is more melatonin in the preovulatory follicle than in the serum level [31]. As a result of these findings, it is concluded that melatonin is synthesized in the ovary and released into the follicular fluid [32]. Studies have shown that LH receptors increase in granulosa cells as a result of melatonin administration [30]. Similarly, melatonin has been reported to affect sex steroid hormone production in follicular maturation during ovulation [33]. During follicular growth, locally produced insulin-like growth factors and transforming growth factor-β together with gonadotropins show efficacy [34]. In one study, it was reported that Melatonin stimulated the production of IGF-I in human granulosa cells [34]. Melatonin is also known to induce the IGFI receptor and activate the P13K/AKT signaling pathway, which is associated with cell metabolism, and the MEK/ERK signaling pathway, which is involved in cell growth, proliferation, and differentiation [35]. The TGF-β superfamily is found in ovarian cells and acts as an intraovarian regulator of follicle development [36]. As a result of studies, it has been determined that TGF-β is produced by both theca and granulosa cells in humans [37, 38]. Studies have shown that melatonin treatment increases TGF-β gene expression in mice [39]. Recent studies have shown that melatonin causes induction of Bcl2 expression and a decrease in Caspase-3 activity, thus protecting tissue from induction of the mitochondrial pathway of apoptosis [40]. The increase in follicular melatonin is an important factor for escaping from growing follicle deatresia, and the amount of intrafollicular melatonin is directly related to atresia [40]. It is stated that the increased amount of melatonin in the follicle may be associated with increased progesterone production after luteinization and ovulation. Local productions of ovarian progesterone, angiotensin-II, and nitric oxide synthetase (NOS) also increase with ovulation [41]. These vasoactive molecules have a fundamental role in the control of follicular blood flow [40]. An increase in progesterone and estradiol levels is required for successful ovulation [42]. Although there is no clear information about the relationship between melatonin, prostaglandin, and estradiol in anovulation, a study on rats showed that melatonin treatment in the gastric mucosa significantly increased the concentration of prostaglandin and estradiol [42]. ROS emerging in folliculogenesis suppresses the production of prostaglandin, which stimulates the synthesis of melatonin from granulosa cells, and induces corpus luteum transformation [43]. In addition to these properties, melatonin also protects the corpus luteum from damage by reactive oxygen species that inhibit progesterone production in human luteal cells.

4.2 Indirect effects of melatonin on the female reproductive system

Considering the indirect effects of melatonin on the female reproductive system. Melatonin exerts its indirect inhibitory effect on the gonads by suppressing the production and secretion of GnRH at the hypothalamus level, inhibiting the release of LH (Luteinizing hormone) from the pituitary, as well as suppressing LH secretion by acting directly on the pituitary by changing the levels of intracellular second messengers such as Ca 2+ and cAMP. Melatonin also increases the secretion of opioid substances that reduce GnRH secretion, such as endorphins [44].

Advertisement

5. Effects of melatonin on the male reproductive system

Various studies have shown that melatonin has some effects on the male reproductive system as well as the female reproductive system. It has been determined that this effect is by binding to the receptors of Sertoli and Leydig cells in the testicles and via the hypothalamus/pituitary axis.

5.1 Direct effects of melatonin on the male reproductive system

When the direct effects of melatonin on testicular tissue are examined, it has been stated that melatonin affects testicular development by binding to the relevant receptors in the testis [45] and in a study, exogenous melatonin administration in rats caused a decrease in testicular size [46, 47]. Various studies have shown that MT1 and MT2 are expressed in the testes of young and adult rats [45]. In a study, it was observed that the external application of Melatonin had negative effects on the seminiferous tubules in the testicles of old male mice [48]. It has also been reported that the administration of melatonin causes shrinkage in testicular tissue and low spermatid count in rats [49]. In another study, significant morphological changes occurred in the tubular and interstitial compartments of the testicles in hamsters exposed to short-term sunlight for a long time [50]. In addition, it was determined that melatonin significantly reduced the volume of mitochondria and nongranular endoplasmic reticulum, which are organelles containing enzymes that have an important role in androgen biosynthesis in mice Leydig cells [51]. Looking at the mechanism of testosterone production, it was determined that the LH hormone released from the adenohypophysis stimulates the cAMP signal in Sertoli cells [52]. It was also stated that melatonin did not suppress the activity of P450scc and thus decreased steroid synthesis. It was determined that StAR protein expression regulated by melatonin LH or cAMP was significantly reduced [53]. Another pathway that is effective in the testosterone release mechanism is that GnRH does so by increasing cytosolic Ca2+ concentrations and activating protein kinase C [52]. In some studies using the fluorescent Ca2+ indicator, it has been reported that melatonin suppresses the release of GnRH-dependent Ca2+ from intracellular stores, thereby reducing cellular Ca2+ levels and suppressing testosterone release [54]. In addition to the aforementioned mechanisms, melatonin has been reported to regulate testosterone production by interacting with the CRH system in the testicles [55, 56]. It has been determined that the CRH hormone is produced in the testicles as well as the hypothalamus. CRH released from the testicles acts as an important negative autocrine regulator of LH-induced testosterone production [57]. In a study, it was reported that Melatonin administration significantly increased the levels of mRNA related to CRH in Leydig cells [58]. Another hormone involved in testicular function is Estrogen. In immature males, the main production site of estrogen is Sertoli cells. Estrogen receptor-alpha (ERα), one of the estrogen receptors, was found in Leydig cells, while ERβ, another estrogen receptor subtype, was found in Sertoli and germ cells [59, 60]. Studies show that ERβ plays a role in the regulation of Leydig cell proliferation and testosterone production in adult mouse testicles. The cytochrome P450 aromatase (P450arom) enzyme is a key enzyme found in the endoplasmic reticulum of various tissues and is responsible for the production of estrogen from androgens. P450arom enzyme has been identified in Leydig cells of several species [61]. Melatonin has been reported to reduce estrogen biosynthesis by inhibiting the activity and expression of aromatase [62, 63, 64]. It has been determined that the administration of melatonin in Leydig and Sertoli cells obtained from rams increases testosterone production by increasing the expression of stem cell factor and insulin-like growth factor-1 [65]. In addition, it was determined that melatonin administration regulated lactate metabolism in Sertoli cells. Lactate released from Sertoli cells provides nutritional support to cells and prevents apoptosis [66]. As a result of the studies, it has been determined that the level of melatonin in the semen is related to infertility [67, 68, 69, 70]. Especially in rams, melatonin level is directly related to sperm quality. Although rams can produce semen throughout the year, the quality of sperm outside the breeding season has been found to be low [71]. In the studies, it was determined that the application of melatonin outside of the breeding season increased sperm volume in the semen [72, 73, 74]. It was also determined that the dividing ability of oocytes fertilized by spermatozoa treated with melatonin increased. This increase is mediated by the increase of hyaluronidase enzyme activity by the administration of melatonin [75, 76].

5.2 Indirect effects of melatonin on the male reproductive system

When the indirect effects of melatonin on the male reproductive system are examined, it has been determined that there is an effect, especially on the hypothalamus-pituitary axis. In a study, it was determined that the application of melatonin to the hypothalamus significantly reduced testicular weight [77, 78, 79]. It was concluded that it showed this effect by suppressing GnRH secretion. In another study, it was determined that the administration of melatonin to mice caused a decrease in testicular and seminal vesicle mass, thus causing a decrease in sperm count [80]. GnIH, a hormone that suppresses the release of GnRH, was determined in a study conducted on quails in 2000 [81, 82, 83]. It has been determined that the effect of melatonin on GnRH secretion is mediated by this hormone. It has been determined that melatonin stimulates the release of GnIH by binding to the MT1 receptor, thus suppressing the release of GnRH [84, 85]. It has been determined that melatonin hormone androgen receptor and ABP levels are decreased in animals with seasonal reproduction. In some animals, it has been determined that tonin has positive effects on the male reproductive system. For example, it has been determined that it has indirect effects as well as direct effects on rams. Melatonin administration has been found to increase the testosterone concentration of somatic cells in ram testis tissue [86, 87]. Kispeptins, which are stimulators of GnRH neurons, have been found to be highly effective in transmitting the melatonin message [88, 89, 90]. In a study, it was determined kisspeptin expression decreased and atrophy occurred in the gonaon in winter days when the daylight decreased in Syrian hamsters. It has been determined that the level of testosterone increases with the injection of kisspeptin [91, 92]. In a study with zebrafish, it was determined that melatonin administration could induce the expression of Kiss1 and Kiss2 and GnRH3 genes in brain tissue and an increase in LH-β in the pituitary gland [93]. This induced gonad development. However, in a study in rats, it was reported that the administration of melatonin suppressed the release of FSH and LH, thus suppressing spermatogenesis in Sertoli cells and delaying sexual maturation [94]. Again, it was determined that the administration of melatonin to rats in the fetal period suppressed LHRH release and significantly reduced the LH level [95, 96]. Melatonin administration is thought to inhibit LH release by reducing Ca2+ flow and cAMP concentrations in the pituitary gland [97].

Advertisement

6. The relationship between melatonin and oxidative stress

Oxidative stress is a cell-damaging condition that can result from decreased antioxidants and/or increased production of reactive free radicals such as reactive oxygen species and/or reactive nitrogen species (ROS/RSN) [98, 99]. Increased free radical production causes cell damage, leading to an increase in various damage markers (MDA, 8-OhDH, TNF-alpha) [100, 101]. In many cases, the body protects the cell against the increase in oxidative stress by the regulation of antioxidant defense systems (GSH, SOD, CAT, GPx) [102, 103, 104, 105, 106, 107]. If oxidative stress can be neutralized, there is usually no adverse contribution to disease pathology. If antioxidant defense induction is inadequate or absent, concomitant cellular and tissue damage often occurs. Some diseases can be directly caused by oxidative stress, but in most diseases, oxidative stress is a consequence and can often only be a secondary event [108, 109, 110]. However, it plays an important role in promoting additional tissue damage in most diseases. When oxidative stress is excessive, it can be prevented by using various substances that have antioxidant properties or suppressing oxidative stress to prevent cell damage [111, 112]. One of the substances used for this purpose is melatonin. As a result of various studies, it has been determined that melatonin reduces oxidative stress and protects the cell against oxidative damage. Some studies have stated that it prevents cell damage by improving MDA levels [113, 114, 115, 116]. The effect of melatonin on reducing MDA occurs through several mechanisms. For example; Melatonin detoxifies a large number of free radicals such as peroxynitrite anion [117] and hydroxyl radical [118] detoxifies hydroxyl radical [119]. It has also been determined that peroxynitrous acid [120] cleans oxidizing particles such as nitric oxide [121] and thus prevents lipid peroxidation. Due to these effects, It has been reported that the effects of melatonin on oxidative stress are direct free radical scavenger and indirect antioxidant [122]. Again, as a result of studies, it was determined that Melatonin increased the level of antioxidant GSH, and also caused an increase in antioxidant enzyme activities such as GPX, SOD, and CAT. It has been determined that these effects are caused by an increase in mRNA expression by stimulating the melatonin receptors on the cell membrane [123]. It has also been determined that melatonin inhibits the nuclear translocation of NF-κB [124]. It has been determined that melatonin inhibits oxidative stress-induced DNA damage by suppressing oxidative stress [125]. Studies have shown that melatonin inhibits apoptosis by decreasing caspase-3 activity and activating the PI3K/AKT pathway, and it preserves membrane integrity. Activation of the PI3K/AKT pathway also increases gene expression such as Nrf2 [126], which plays an important role in the antioxidant defense system. These findings explain the cell-protective mechanism of melatonin against oxidative stress.

Advertisement

7. Conclusion

Melatonin is a hormone mainly synthesized from the pineal gland, and it has been shown by some studies that it has various effects on the immune system, oxidative stress, and reproduction, as well as taking part in the regulation of circadian rhythm. In this study, we focused on the direct, indirect, and antioxidant effects of melatonin on the female and male reproductive systems and the molecular mechanisms of these effects. As a result, melatonin affects the male and female reproductive systems directly via the gonads via secondary messengers and indirectly by stimulating various receptors/molecules via the hypothalamus-pituitary axis. These effects are in the direction of supporting reproduction in some living things and suppressing in others. In addition, melatonin prevents cell damage by increasing antioxidant enzyme activity and scavenging free radicals, especially in female and male gonads, thus preventing cell damage.

Figure 1.

Biosynthetic pathway of the melatonin.

Figure 2.

Synthesis of melatonin from tryptophan under the influence of light in the pineal gland [9]. SCG: Superior cervical ganglion; SCN: Suprachiasmatic nucleus; NA: Noradrenaline; PVN: Paraventricular nucleus; AADC: Aromatic-L-amino-acid decarboxylase AANAT: Arylalkylamine N-acetyltransferase, TPH: Hydroxytryptophan 5-hydroxylase. HIOMT: Hydroxy indole-O-methyltransferase NAS: N-asetlyserotonin.

Figure 3.

MT 1 and MT 2 melatonin receptor signaling. PKA, protein kinase a; cAMP response element-binding protein; MT, melatonin receptor; Akt, threonine protein kinase B (PKB; also known as Akt); cyclic adenosine monophosphate; cGMP, cyclic guanosine monophosphate; CREB, IP3, inositol trisphosphate; MAPK, mitogen-activated protein kinase [18].

References

  1. 1. Cipolla-Neto J, Amaral FGD. Melatonin as a hormone: New physiological and clinical insights. Endocrine Reviews. 2018;39(6):990-1028
  2. 2. Pevet P. Melatonin. Dialogues in Clinical Neuroscience. 2002;4(1):57-72
  3. 3. Pevet P, Challet E. Melatonin: Both master clock output and internal time-giver in the circadian clocks network. Journal of Physiology, Paris. 2011;105(4-6):170-182
  4. 4. Bubenik GA. Gastrointestinal melatonin: Localization, function, and clinical relevance. Digestive Diseases and Sciences. 2002;47:2336-2348
  5. 5. Slominski A, Fischer TW, Zmijewski MA, Wortsman J, Semak I, Zbytek B, et al. On the role of melatonin in skin physiology and pathology. Endocrine. 2005;27:137-148
  6. 6. Carrillo-Vico A, Calvo JR, Abreu P, Lardone PJ, GarcíaMauriño S, Reiter RJ, et al. Evidence of melatonin synthesis by human lymphocytes and its physiological significance: Possible role as intracrine, autocrine, and/or paracrine substance. The FASEB Journal. 2004;18:537-539
  7. 7. Cardinali DP, Rosner JM. Metabolism of serotonin by the rat retina in vitro. Journal of Neurochemistry. 1971;18:1769-1770
  8. 8. Berson DM, Dunn FA, Takao M. Phototransduction by retinal ganglion cells that set the circadian clock. Science. 2002;295:1070-1073. DOI: 10.1126/science.1067262
  9. 9. Simoneaux V, Ribelayga C. Generation of the melatonin endocrine message in mammals: A review of the complex regulation of melatonin synthesis by norepinephrine, peptides, and other pineal transmitters. Pharmacological Reviews. 2003;55(2):325-395
  10. 10. Ölmez E, Şahna E, Ağkadir M, Acet A. Melatonin: emeklilik yaşı 80 olur mu? Turgut Özal Tıp Dergisi. 2000;7:177-187
  11. 11. Sack RL, Lewy AJ, Hughes RJ. Use of melatonin for sleep and circadian rhythm disorders. Annals of Medicine. 1998;30:115-121
  12. 12. Çam A, Erdoğan MF. Melatonin. Ankara Üniversitesi Tıp Fakültesi. Mecmuası. 2003;56:103-112
  13. 13. Cipolla-Neto J, Amaral FG, Soares JM Jr, Gallo CC, Furtado A, Cavaco JE, et al. The crosstalk between melatonin and sex steroid hormones. Neuroendocrinology. 2022;112(2):115-129
  14. 14. Skwarlo-Sonta K, Majewski P, Markowska M, Oblap R, Olszanska B. Bidirectional communication between the pineal gland and the imimune system. Canadian Journal of Physiology and Pharmacology. 2003;81(4):342-349
  15. 15. Mayo JC, Aguado A, Cernuda-Cernuda R, Álvarez-Artime A, Cepas V, Quirós-González I, et al. Melatonin uptake by cells: an answer to its relationship with glucose? Molecules. 10 Aug 2018;23(8):1999
  16. 16. Amaral FGD, Cipolla-Neto J. A brief review about melatonin, a pineal hormone. Archives of Endocrinology and Metabolism. 2018;62(4):472-479
  17. 17. Liu J, Clough SJ, Hutchinson AJ, AdamahBiassi EB, Popovska-Gorevski M, Dubocovich ML. MT1 and MT2 melatonin receptors: A therapeutic perspective. Annual Review of Pharmacology and Toxicology. 2016;56:361-383
  18. 18. Masana MI, Dubocovich ML. Melatonin receptor signaling: finding the path through the dark. Science STKE. 2001;6(107):39
  19. 19. Pandi-Perumal SR, Trakht I, Srinivasan V, Spence DW, Maestroni GJ, Zisapel N, et al. Physiological effects of melatonin: Role of melatonin receptors and signal transduction pathways. Progress in Neurobiology. 2008;85(3):335-353
  20. 20. Ng KY, Leong MK, Liang H, Paxinos G. Melatonin receptors: Distribution in the mammalian brain and their respective putative functions. Brain Structure & Function. 2017;222(7):2921-2939
  21. 21. Slominski RM, Reiter RJ, Schlabritz-Loutsevitch N, Ostrom RS, Slominski AT. Melatonin membrane receptors in peripheral tissues: Distribution and functions. Molecular and Cellular Endocrinology. 2012;351(2):152-166
  22. 22. Reiter RJ, Tan DX, Galano A. Melatonin: Exceedance meditations. Physiology. 2014;29(5):325-333
  23. 23. Conway S, Canning SJ, Howell HE, Mowat ES, Barrett P, Drew JE, et al. Characterisation of human melatonin mt(1) and MT(2) receptors by CRE-luciferase reporter assay. European Journal of Pharmacology. 2000;390:15-24
  24. 24. Pintor J, Martin L, Pelaez T, Hoyle CH, Peral A. Involvement of melatonin MT(3) receptors in the regulation of intraocular pressure in rabbits. European Journal of Pharmacology. 2001;416:251-254
  25. 25. Grosjean O, Ferro M, Coge F, Beauverger P, Henlin JM, Lefoulon F, et al. Identification of the melatonin binding site MT3 as the quinone reductase 2. The Journal of Biological Chemistry. 2000;275:31311-31317
  26. 26. Conway S, Canning SJ, Howell HE, Mowat ES, Barrett P, Drew JE, et al. Characterisation of human melatonin mt(1) and MT(2) receptors by CRE-luciferase reporter assay. European Journal of Pharmacology. 2000;390:15-24
  27. 27. Pintor J, Martin L, Pelaez T, Hoyle CH, Peral A. Involvement of melatonin MT(3) receptors in the regulation of intraocular pressure in rabbits. European Journal of Pharmacology. 2001;416:251-254
  28. 28. Şahin D. İn vitro koşullarda sirkadien melatonin etkisine maruz bırakılan embriyolarda SOD ve HMGB1 genlerinin ekspresyonları ile melatonin etkisinin takibi. istanbul: istanbul Bilim Üniversitesi; 2014
  29. 29. Wierrani F, Grin W, Hlawka B, Kroiss A, Grünberger W. Elevated serum melatonin levels during human late pregnancy and labour. Journal of Obstetrics and Gynaecology. 1997;17:449-451
  30. 30. Woo MMM, Tai CJ, Kang SK, et al. Direct action of melatonin in human granulosa-luteal cells. The Journal of Clinical Endocrinology and Metabolism. 2001;86:4789-4797
  31. 31. Maitra SK, Chattoraj A, Mukherjee S, et al. Melatonin: A potent candidate in the regulation of fish oocyte growth and maturation. General and Comparative Endocrinology. 2013;181:215-222
  32. 32. Sakaguchi K, Itoh MT, Takahashi N, et al. The rat oocyte synthesizes melatonin. Reproduction, Fertility, and Development. 2013;25:674-682
  33. 33. Danforth DR. Endocrine and paracrine control of oocyte development. American Journal of Obstetrics & Gynecology. 1995;172:747-752
  34. 34. Poretsky L, Cataldo NA, Rosenwaks Z, et al. The insulin-related ovarian regülatöry system in health and disease. Endocrinology Reviews. 1999;20:535-582
  35. 35. Chun SY, Billig H, Tilly JL, et al. Gonadotropin suppression of apoptosis in cultured preovulatory follicles: Mediatory role of endogenous insulin-like growth factor I. Endocrinology. 1994;135:1845-1853
  36. 36. Tilly JL. Tilly KI inhibitors of oxidative stress mimic the ability of follicle- stimulating hormone to suppress apoptosis in cultured rat ovarian follicles. Endocrinology. 1995;136:242-252
  37. 37. Knight PG, Glister C. TGF-beta süper family members and ovarian follicle development. Reproduction. 2006;132:191-206
  38. 38. Tatemoto H, Muto N, Sunagawa I, et al. Protection of porcine oocytes against cell damage caused by oxidative stress during In vitro maturation: Role of superoxide dismutase activity in porcine follicular fluid. Biology of Reproduction. 2004;71:1150-1157
  39. 39. Ratts VS, Flaws JA, Kolp R, et al. Ablation of bcl-2 gene expression decreases the numbers of oocyte sand primordial follicles established in the post-natal female mouse gonad. Endocrinology. 1995;136:3665-3668
  40. 40. Tamura H, Takasaki A, Ichiro M, et al. Oxidative stress impairs oocyte quality and melatonin protects oocytes from free radical damage and improves fertilization rate. Journal of Pineal Research. 2008;44:280-287
  41. 41. Matikainen T, Perez GI, Zheng TS, et al. Caspase-3 gene knockout defines cell lineage specificity for programmed cell death signaling in the ovary. Endocrinology. 2001;142:2468-2480
  42. 42. Nakamura Y, Tamura H, Takayama H, et al. Increased endogenous level of melatonin in preovulatory human follicles does not directly influence progesterone production. Fertility and Sterility. 2003;80:1012-1016
  43. 43. Wetterberg L, Arendt J, Paunier L, et al. Human serum melatonin changes during the menstrual cycle. The Journal of Clinical Endocrinology and Metabolism. 1976;42:185-188
  44. 44. Aleandri V, Spina V, Morini A. The pineal gland and reproduction. Human Reproduction Update. 1996;2:225-223
  45. 45. Izzo G, Francesco A, Ferrara D, et al. Expression of melatonin (MT1, MT2) and melatonin-related receptors in the adult rat testes and during development. Zygote. 2010;18:257-264
  46. 46. Edmonds KE, StetsonMH. Photoperiod and melatonin affect testicular growth in the marsh rice rat (Oryzomys palustris). Journal of Pineal Research. 1994;17:86-93
  47. 47. Edmonds KE, Stetson MH. Pineal gland and melatonin affect testicular status in the adult marsh rice rat (Oryzomys palustris). General and Comparative Endocrinology. 1995;99:265-274
  48. 48. Mehraein F, Negahdar F. Morphometric evaluation of seminiferous tubules in aged mice testes after melatonin administration. Cell Journal. 2011;13:1-4
  49. 49. Rashed RA, Mohamed IK, Ei-Alfy SH. Effects of two different doses of melatonin on the spermatogenic cells of rat testes: A light and electron microscopic study. Egypt Journal of Histology. 2010;33:819-835
  50. 50. Mason AO, Duffy S, Zhao S, et al. Photoperiod and reproductive condition are associated with changes in RFamide-related peptide (RFRP) expression in Syrian hamsters (Mesocricetus auratus). Journal of Biological Rhythms. 2010;25:176-185
  51. 51. Redins CA, Redins GM, Novaes JC. The effects of treatment with melatonin on the ultrastructure ofmouse Leydig cells: A quantitative study. Brazilian Journal of Biology. 2002;62:517-523
  52. 52. Stojilković SS, Chang JP, Ngo D, Tasaka K, Izumi S, Catt KJ. Mechanism of action of GnRH: The participation of calcium mobilization and activation of protein kinase C in gonadotropin secretion. Journal of Steroid Biochemistry. 1989;33:693-703
  53. 53. Wu CS, Leu SF, Yang HY, Huang BM. Melatonin inhibits the expression of steroidogenic acute regulatory protein and steroidogenesis in MA-10 cells. Journal of Andrology. 2001;22:245-254
  54. 54. Valenti S, Thellung S, Florio T, Giusti M, Schettini G, Giordano G. A novel mechanism for the melatonin inhibition of testosterone secretion by rat Leydig cells: Reduction of GnRH-induced increase in cytosolic Ca2+. Journal of Molecular Endocrinology. 1999;23:299-306
  55. 55. Frungieri MB, Mayerhofer A, Zitta K, Pignataro OP, Calandra RS, Gonzalez-Calvar SI. Direct effect ofmelatonin on Syrian hamster testes: Melatonin subtype 1a receptors, inhibition of androgen production, and interaction with the local corticotropinreleasing hormone system. Endocrinology. 2005;146:1541-1552
  56. 56. Yao M, Schulkin J, Denver RJ. Evolutionarily conserved glucocorticoid regulation of corticotropin-releasing factor expression. Endocrinology. 2008;149:2352-2360
  57. 57. Dufau ML, Tinajero JC, Fabbri A. Corticotropin-releasing factor: An antireproductive hormone of the testis. The FASEB Journal. 1993;7:299-307
  58. 58. Rossi SP, Matzkin ME, Terradas C, et al. New insights into melatonin/CRH signaling in hamster Leydig cells. General and Comparative Endocrinology. 2012;178:153-163
  59. 59. Bili ´ nska B, Schmalz-Fraczek B, Kotula M, Carreau S. Photoperiod-dependent capability of androgen aromatization and the role of estrogens in the bank vole testis visualized by means of immunohistochemistry. Molecular and Cellular Endocrinology. 2001;178:189-198
  60. 60. Gould ML, Hurst PR, Nichol HD. The effects of oestrogen receptors alpha and beta on testicular cell number and steroidogenesis in mice. Reproduction. 2007;134:271-279
  61. 61. Carreau S. Germ cells: A new source of estrogens in the male gonad. Molecular and Cellular Endocrinology. 2001;178:65-72
  62. 62. Letellier K, Azeddine B, Parent S, Labelle H, Rompré PH, Moreau A, et al. Estrogen cross-talk with the melatonin signaling pathway in human osteoblasts derived from adolescent idiopathic scoliosis patients. Journal of Pineal Research. 2008;45:383-393
  63. 63. Yoshitane H, Honma S, Imamura K, Nakajima H, Nishide SY, Ono D, et al. JNK regulates the photic response of the mammalian circadian clock. EMBO Reports. 2012;13:455-461
  64. 64. Vriend J, Reiter RJ. Breast cancer: Modulation by melatonin and the ubiquitin-proteasome system–a review. Molecular and Cellular Endocrinology. 2015;417:1-9. [Cross Ref] [PubMed]
  65. 65. Deng SL, Wang ZP, Jin C, Kang XL, Batool A, Zhang Y, et al. Melatonin promotes sheep Leydig cell testosterone secretion in a co-culture with Sertoli cells. Theriogenology. 2018;106:170-177
  66. 66. Rocha CS, Martins AD, Rato L, Silva BM, Oliveira PF, Alves MG. Melatonin alters the glycolytic profile of Sertoli cells: Implications for male fertility. Molecular Human Reproduction. 2014;20:1067-1076
  67. 67. Awad H, Halawa F, Mostafa T, Atta H. Melatonin hormone profile in infertile males. International Journal of Andrology. 2006;29:409-413
  68. 68. Yie SM, Daya S, Brown GM, Deys L, YoungLai EV. Melatonin and aromatase stimulating activity of human seminal plasma. Andrologia. 1999;23:227-231
  69. 69. van Vuuren RJ, Pitout MJ, van Aswegen CH, Theron JJ. Putativemelatonin receptor in human spermatozoa. Clinical Biochemistry. 1992;25:125-127
  70. 70. Casao A, Gallego M, Abecia JA, et al. Identification and immunolocalisation of melatonin MT(1) and MT(2) receptors in rasa Aragonesa ram spermatozoa. Reproduction, Fertility, and Development. 2012;24:953-961
  71. 71. Azawi OI, Al-Khashab ANTM, Al-Kadoo NN. Effect of gonadotropin releasing hormone treatment on semen characteristics and enzymatic activities of Awassi rams in breeding and non-breeding seasons. Iranian Journal of Applied Animal Science. 2012;2:13-19
  72. 72. Ramadan TA, Taha TA, Samak MA, Hassan A. Effectiveness of exposure to longday followed by melatonin treatment on semen characteristics of Damascus male goats during breeding and non-breeding seasons. Theriogenology. 2009;71:458-468
  73. 73. Casao A, Vega S, Palacín I, et al. Effects of melatonin implants during non-breeding season on sperm motility and reproductive parameters in rasa Aragonesa rams. Reproduction in Domestic Animals. 2010;45:425-432
  74. 74. Casao A, Mendoza N, Pérez-Pé R, et al. Melatonin prevents capacitation and apoptotic-like changes of ram spermatozoa and increases fertility rate. Journal of Pineal Research. 2010;48:39-46
  75. 75. Tanyiladizi S, Bozkurt T, ÇİftÇİ O, SAKİN F. In vitro effects ofmelatonin on hyaluronidase activity and sperm motility in bull semen. Turk J vet. Animal Science. 2006;30:89-93
  76. 76. Luboshitzky R, Shen-Orr Z, Nave R, Lavi S, Lavie P. Melatonin administration alters semen quality in healthy men. Journal of Andrology. 2002;23:572-578
  77. 77. Wierman ME, Bruder JM, Kepa JK. Regulation of gonadotropin-releasing hormone (GnRH) gene expression in hypothalamic neuronal cells. Cellular and Molecular Neurobiology. 1995;15:79-88
  78. 78. Weaver DR, Stehle JH, Stopa EG, Reppert SM. Melatonin receptors in human hypothalamus and pituitary: Implications for circadian and reproductive responses to melatonin. The Journal of Clinical Endocrinology and Metabolism. 1993;76:295-301
  79. 79. Glass JD, Knotts LK. A brain site for the antigonadal action of melatonin in the whitefooted mouse (Peromyscus leucopus): Involvement of the immunoreactive GnRH neuronal system. Neuroendocrinology. 1987;46:48-55
  80. 80. Forger NG, Zucker I. Photoperiodic regulation of reproductive development in male white-footed mice (Peromyscus leucopus) born at different phases of the breeding season. Journal of Reproduction and Fertility. 1985;73:271-278
  81. 81. Tsutsui K, Saigoh E, Ukena K, et al. A novel avian hypothalamic peptide inhibiting gonadotropin release. Biochemical and Biophysical Research Communications. 2000;275:661-667
  82. 82. Ubuka T, Son YL, Bentley GE, Millar RP, Tsutsui K. Gonadotropin-inhibitory hormone (GnIH), GnIH receptor and cell signaling. General and Comparative Endocrinology. 2013;190:10-17
  83. 83. Tsutsui K, Ubuka T, Bentley GE, Kriegsfeld LJ. Review: Regulatory mechanisms of gonadotropin-inhibitory hormone (GnIH) synthesis and release in photoperiodic animals. Frontiers in Neuroscience. 2013;7:60
  84. 84. Tsutsui K, Ubuka T, Yin H, et al. Mode of action and functional significance of avian gonadotropin-inhibitory hormone (GnIH): A review. Journal of Experimental Zoology. Part A, Comparative Experimental Biology. 2006;305:801-806
  85. 85. Reiter RJ, Tan DX, Rosales-Corral S, Manchester LC. The universal nature, unequal distribution and antioxidant functions of melatonin and its derivatives. Mini Reviews in Medicinal Chemistry. 2013;13:373-384
  86. 86. Viguie C, Caraty A, Locatelli A, Malpaux B. Regulation of luteinizing hormone-releasing hormone (LHRH) secretion by melatonin in the ewe. II. Changes in N-methyl-DL-aspartic acid-induced LHRH release during the stimulation of luteinizing hormone secretion by melatonin. Biology of Reproduction. 1995;52:1156-1161
  87. 87. Deng SL, Chen SR, Wang ZP, Zhang Y, Tang JX, Li J, et al. Melatonin promotes development of haploid germ cells from early developing spermatogenic cells of Suffolk sheep under in vitro condition. Journal of Pineal Research. 2016;60:435-447
  88. 88. Buchanan KL, Yellon SM. Delayed puberty in the male Djungarian hamster: Effect of short photoperiod or melatonin treatment on the GnRH neuronal system. Neuroendocrinology. 1991;54:96-102
  89. 89. Ancel C, Bentsen AH, Sébert ME, Tena-Sempere M, Mikkelsen JD, Simonneaux V. Stimulatory effect of RFRP-3 on the gonadotrophic axis in the male Syrian hamster: The exception proves the rule. Endocrinology. 2012;153:1352-1363
  90. 90. Irwig MS, Fraley GS, Smith JT, Acohido BV, Popa SM, Cunningham MJ, et al. Kisspeptin activation of gonadotropin releasing hormone neurons and regulation of KiSS-1 mRNA in the male rat. Neuroendocrinology. 2004;80:264-272
  91. 91. Ansel L, Bentsen AH, Ancel C, Bolborea M, Klosen P, Mikkelsen JD, et al. Peripheral kisspeptin reverses short photoperiod-induced gonadal regression in Syrian hamsters by promoting GNRH release. Reproduction. 2011;142:417-425
  92. 92. Henningsen JB, Poirel VJ, Mikkelsen JD, Tsutsui K, Simonneaux V, Gauer F. Sex differences in the photoperiodic regulation of RF-amide related peptide (RFRP) and its receptor GPR147 in the Syrian hamster. The Journal of Comparative Neurology. 2016;524:1825-1838
  93. 93. Carnevali O, Gioacchini G, Maradonna F, Olivotto I, Migliarini B. Melatonin induces follicle maturation in Danio rerio. PLoS One. 2011;6:e19978
  94. 94. Lang U, Aubert ML, Rivest RW, Vinas-Bradtke JC, Sizonenko PC. Daily afternoon administration of melatonin does not irreversibly inhibit sexual maturation in the male rat. Endocrinology. 1984;115:2303-2310
  95. 95. Martin JE, Sattler C. Selectivity of melatonin pituitary inhibition for luteinizing hormone-releasing hormone. Neuroendocrinology. 1982;34:112-116
  96. 96. Hattori A, Herbert DC, Vaughan MK, Yaga K, Reiter RJ. Melatonin inhibits luteinizing hormone releasing hormone (LHRH) induction of LH release from fetal rat pituitary cells. Neuroscience Letters. 1995;184:109-112
  97. 97. Vanecek J. Melatonin inhibits release of luteinizing hormone (LH) via decrease of [Ca2+] and cyclic AMP. Physiological Research. 1998;47:329-335
  98. 98. Kükürt A, Gelen V, Başer ÖF, Deveci HA, Karapehlivan M. Thiols: Role in oxidative stress-related disorders. In: Lipid Peroxidation [Working Title]. London, UK: IntechOpen; 2021. DOI: 10.5772/intechopen.96682
  99. 99. Kara A, Gedikli S, Sengul E, Gelen V, Ozkanlar S. Oxidative Stress and Autophagy. 1st ed. London: InTechOpen, Free Radicals and Diseases; 2016. pp. 69-86
  100. 100. Gelen V, Yıldırım S, Şengül E, Çınar A, Çelebi F, Küçükkalem M, et al. Naringin attenuates oxidative stress, inflammation, apoptosis, and oxidative DNA damage in acrylamide-induced nephrotoxicity in rats. Asian Pacific Journal of Tropical Biomedicine. 2022;12(5):223-232
  101. 101. Gelen V, Şengül E, Yıldırım S, Atila G. The protective effects of naringin against 5-fluorouracil-induced hepatotoxicity and nephrotoxicity in rats. Iranian Journal of Basic Medical Sciences. 2018;21(4):404-410. DOI: 10.22038/IJBMS.2018.27510.6714
  102. 102. Karamese M, Guvendi B, Karamese SA, Cinar I, Can S, Erol HS, et al. The protective effects of epigallocatechin gallate on lipopolysaccharide-induced hepatotoxicity: An in vitro study on Hep3B cells. Iranian Journal of Basic Medical Sciences. 2016;19:483-489
  103. 103. Gelen V, Sengul E. Antioxidant, anti-inflammatory, and antiapoptotic effects of naringin on cardiac damage induced by cisplatin. IJTK. 2020;19(2):459-465
  104. 104. Sengul E, Gelen V, Yildirim S, et al. The effects of selenium in acrylamide-induced nephrotoxicity in rats: Roles of oxidative stress, inflammation, apoptosis, and DNA damage. Biological Trace Element Research. 2021;199:173-184
  105. 105. Şengül E, Gelen V, Gedikli S, Özkanlar S, Gür G, Çelebi F. The protective effect of quercetin on cyclophosphamide-induced lung toxicity in rats. Biomedicine & Pharmacotherapy. 2017;92:303-307
  106. 106. Sengul E, Gelen V, Gedikli S. Cardioprotective activities of quercetin and Rutin in Sprague Dawley rats treated with 5-fluorouracil. Journal of Animal and Plant Sciences. 2021;31(2):423-431
  107. 107. Kükürt A, Karapehlivan M. Protective effect of astaxanthin on experimental ovarian damage in rats. Journal of Biochemical and Molecular Toxicology. 2022;36(3):e22966. DOI: 10.1002/jbt.22966
  108. 108. Gelen V, Kükürt A, Şengül E, Devecı HA. Leptin and its role in oxidative stress and apoptosis: An overview. In: Role of Obesity in Human Health and Disease. [Working Title],. London, UK: IntechOpen; 2021. DOI: 10.5772/intechopen.101237
  109. 109. Gelen V, Kükürt A, Şengül E, Başer ÖF, Karapehlivan M. Can polyphenols be used as anti-inflammatory agents against Covid-19 (SARS-CoV-2)- induced inflammation? In: Badria FA, editor. Phenolic Compd. [Working Title]. Rijeka: IntechOpen; 2021. pp. 1-21. DOI: 10.5772/intechopen.98684
  110. 110. Gelen V, Kükürt A, Şengül E. Role of the renin-angiotensin-aldosterone system in various disease processes: An overview. In: Renin-Angiotensin Aldosterone Syst. [Working Title]. London, UK: IntechOpen; 2021. DOI: 10.5772/intechopen.97354
  111. 111. Gelen V, Şengül E, Yıldırım S, Senturk E, Tekin S, Kükürt A. The protective effects of hesperidin and curcumin on 5-fluorouracil–induced nephrotoxicity in mice. Environmental Science and Pollution Research. 2021. DOI: 10.1007/s11356-021-13969-5
  112. 112. Gedikli S, Ozkanlar S, Gur C, Sengul E, Gelen V. Preventive effects of quercetin on liver damages in high-fat diet-induced obesity. Journal of Histology & Histopathology. 2017;4:7. DOI: 10.7243/2055-091X-4-7
  113. 113. Fulia F, Gitto E, Cuzzocrea S, Reiter RJ, Dugo L, Gitto P, et al. Increased levels of malondialdehyde and nitrite/nitrate in the blood of asphyxiated newborns: Reduction by melatonin. Journal of Pineal Research. 2001;31(4):343-349
  114. 114. Allegra M, Gentile C, Tesoriere L, Livrea M. Protective effect of melatonin against cytotoxic actions of malondialdehyde: An in vitro study on human erythrocytes. Journal of Pineal Research. 2002;32(3):187-193
  115. 115. Ziaadini F, Aminae M, Rastegar MM, Abbasian S, Memari AH. Melatonin supplementation decreases aerobic exercise training induced-lipid peroxidation and malondialdehyde in sedentary young women. Polish Journal of Food and Nutrition Sciences. 2017;67(3):225-232
  116. 116. Gedikli S, Gelen V, Sengul E, Ozkanlar S, Gur C, Agırbas O, et al. Therapeutic effects of melatonin on liver and kidney damages In intensive exercise model of rats. Endocrine, Metabolic & Immune Disorders Drug Targets. 2015;15(4):308-314
  117. 117. Karbownik M, Reiter RJ. Antioxidative effects of melatonin in protection against cellular damage caused by ionizing radiation. Proceedings of the Society for Experimental Biology and Medicine. 2000;22:9-22
  118. 118. Goc Z, Szaroma W, Kapusta E, Dziubek K. Protective effects of melatonin on the activity of SOD, CAT, GSH-Px and GSH content in organs of mice after administration of SNP. The Chinese Journal of Physiology. 2017;60(1):1-10
  119. 119. Purushothaman A, Sheeja AA, Janardanan D. Hydroxyl radical scavenging activity of melatonin and its related indolamines, Free Radicals. Res. 2020;54(5):373-383
  120. 120. Loren P, Sanchez R, Arias ME, Felmer R, Risopatron J, Cheuqueman C. Melatonin scavenger properties against oxidative and nitrosative stress: Impact on gamete handling and in vitro embryo production in humans and other mammals. International Journal of Molecular Sciences. 2017;18(6):1119
  121. 121. Fan W, He Y, Guan X, Gu W, Wu Z, Zhu X. el al, Involvement of the nitric oxide in melatonin-mediated protection against injury. Life Sciences. 2018;200:142-147
  122. 122. Reina M, Martínez A. A new free radical scavenging cascade involving melatonin and three of its metabolites (3OHM, AFMK and AMK). Computational & Theoretical Chemistry. 2018;1123:111-118
  123. 123. Ghosh AK, Naaz S, Bhattacharjee B, Ghosal N, Chattopadhyay A, Roy S, et al. Mechanism of melatonin protection against copperascorbate-induced oxidative damage in vitro through isothermal titration calorimetry. Life Sciences. 2017;180:123-136
  124. 124. Dehghani S, Hosseini M, Haghgoo S, Changizi V, Akbari Javar H, Khoobi M, et al. Multifunctional MIL-Cur@ FC as a theranostic agent for magnetic resonance imaging and targeting drug delivery: in vitro and in vivo study. Journal of Drug Targeting. 2020;28(6):668-680
  125. 125. Okatani Y, Wakatsuki A, Shinohara K, Kaneda C, Fukaya T. Melatonin stimulates glutathione peroxidase activity in human chorion. Journal of Pineal Research. 2001;30(4):199-205
  126. 126. Zhang Y, Wei Z, Liu W, Wang J, He X, Huang H, et al. Melatonin protects against arsenic trioxide-induced liver injury by the upregulation of Nrf2 expression through the activation of PI3K/AKT pathway. Oncotarget. 2017;8(3):3773

Written By

Volkan Gelen, Emin Şengül and Abdulsamed Kükürt

Submitted: 20 June 2022 Reviewed: 15 September 2022 Published: 23 October 2022