Next Article in Journal
New Bacterial Phytase through Metagenomic Prospection
Next Article in Special Issue
Melatonin-Mediated Development of Ovine Cumulus Cells, Perhaps by Regulation of DNA Methylation
Previous Article in Journal
Efficiency of Osmotic Dehydration of Apples in Polyols Solutions
Previous Article in Special Issue
Exogenous Melatonin Mitigates Acid Rain Stress to Tomato Plants through Modulation of Leaf Ultrastructure, Photosynthesis and Antioxidant Potential
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Melatonin Regulates the Synthesis of Steroid Hormones on Male Reproduction: A Review

1
State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
2
National Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Molecules 2018, 23(2), 447; https://doi.org/10.3390/molecules23020447
Submission received: 19 January 2018 / Revised: 10 February 2018 / Accepted: 14 February 2018 / Published: 17 February 2018

Abstract

:
Melatonin is a ubiquitous molecule and exhibits different effects in long-day and short-day breeding animals. Testosterone, the main resource of androgens in the testis, is produced by Leydig cells but regulated mainly by cytokine secreted by Sertoli cells. Melatonin acts as a local modulator of the endocrine activity in Leydig cells. In Sertoli cells, melatonin influences cellular proliferation and energy metabolism and, consequently, can regulate steroidogenesis. These suggest melatonin as a key player in the regulation of steroidogenesis. However, the melatonin-induced regulation of steroid hormones may differ among species, and the literature data indicate that melatonin has important effects on steroidogenesis and male reproduction.

1. Introduction

Spermatogenesis, the process of male gamete differentiation and maturation, is regulated by various hormones, such as luteinizing hormone (LH), follicle-stimulating hormone (FSH) from the pituitary gland, and testosterone produced by Leydig cells in the testis. FSH binds to its receptor restrictively expressed by Sertoli cells, which increases cAMP concentration and stimulates androgen-binding protein (ABP) synthesis and LH release, and this regulates testosterone production in Leydig cells. Testosterone concentration in the seminiferous tubules is associated with ABP [1]. Spermatogenesis is a mostly testosterone-dependent cellular event in the testis. In the absence of testosterone or androgen receptor, spermatogenesis fails to proceed beyond meiosis stage [2]. As rate-limiting steps, steroidogenic acute regulatory protein (StAR) and GATA binding factor 4 (GATA-4) co-regulate steroid hormone synthesis. Cyclic AMP response element binding protein (CREB) positively regulates the expression of steroidogenesis-linked genes [3]. Melatonin is a major secretory product of the pineal gland with both lipophilic and hydrophilic properties, and can pass through the blood–testis barrier and enter testicular cells. It acts through several specific receptors, including membrane melatonin receptors 1 (MT1) and 2 (MT2), and retinoid acid receptor-related orphan receptor Α (RORα), which has been identified in a large variety of mammalian cell types [4]. MT1 and MT2 couple with G-protein to regulate testosterone synthesis by regulating cAMP signal transduction cascades [5]. At present, there is still controversy about whether melatonin is directly combined with RORα to play its biological role [6]. However, melatonin can regulate animal reproduction at the transcriptional level through the nuclear receptor. Research suggests that aromatase could be activated by RORα and, as a result, conversion of androgen into estrogen was promoted [7].
The effects of melatonin on the levels of reproductive hormones are variable and depend on physiological conditions and species of animals [8]. During the long day period of seasonal breeders, such as rodents, melatonin reduces the expression of the androgen receptor and ABP [9]. Injecting melatonin into Syrian hamster testes during the breeding period significantly decreases the content of testosterone, decreases testicular volume, and diminishes androgen synthesis [10]. However, a constant supplementation of melatonin to the short-day breeding animals promoted the gonad function [11,12]. Long-term treatment with melatonin induces early testicular development in sika deer [13], and melatonin implants advance sperm production in silver fox [14]. Subcutaneous injections of melatonin increase testosterone concentrations in goats [15]. These processes eventually lead to spermatozoa maturation [16]. Melatonin is positively correlated with androgen concentration in the short-day period of the seasonal breeding animals. Our study revealed that, after treatment with melatonin, the concentration of testosterone in the somatic cells of sheep testes was 3.54 ± 0.17 nmol/L which was twofold higher than that in the control [17].
Reproduction is synchronized by day length via the pineal hormone melatonin. Melatonin affects stimulators of gonadotropin release hormone (GnRH) by regulating the secretion of hypothalamus–pituitary–gonadal related hormones, and then regulates the secretion of LH and FSH. Pinealectomy and exogenous melatonin for a long time on sheep could lead to the release of GnRH and LH, thus activating the reproductive activity [18]. For long-day mammals, melatonin is an inhibitory factor for reproduction, and there is evidence that the effect of this resistant gland is achieved by inhibiting GnRH [19]. At present, the exact mechanism of melatonin regulation of GnRH is still unclear [20]. Kisspeptins (Kp), a family of potent hypothalamic stimulators of GnRH neurons, is essential to convey melatonin’s message [21,22]. In short winter days, the Syrian hamster displays a complete gonadal atrophy with a marked reduction in expression of Kp. Acute peritoneal injection of Kp-induced c-Fos expression in a large number of GnRH neurons and pituitary gonadotrophs together with a strong increase in circulating testosterone [23,24]. In long days, Kp is highly expressed in the anteroventral periventricular nucleus (AVPV), with low expression in the arcuate nucleus in Phodopus sungorus. The situation is exactly the opposite in short days [25]. Photoperiod, via melatonin, modulates kiss1 signaling to drive the reproductive axis [26]. It is found that melatonin can induce the expression of Kiss1 and kiss2 and GnRH3 genes in zebrafish brain, and the increase of LHβ in the pituitary gland, which indicates that melatonin can promote gonad maturation and significantly improve the reproductive capacity of zebrafish [27]. Exogenous melatonin treatment for male sea bass found the expression of kiss1 gene was significantly increased in the hypothalamus after 30 days, the expression of kiss2 was enhanced after 90 days, but the expression of Kp in the back side of the brain was significantly decreased after 150 days, and decreased the mRNA expression of GnRH-1, GnRH-3, and FSH gene in the pituitary [28]. Melatonin can cause changes in the expression of Kp, thus affecting the changes in the reproductive system. The precise mechanisms which melatonin affects kisspeptin remain unclear. The path between melatonin and Kp is also a hot spot of current exploration [29].

2. Melatonin Regulates Leydig Cell–Testosterone Secretions

Melatonin is involved in the function of the male reproductive system, particularly in the testes, since Leydig cells are sensitive to melatonin [30,31]. Melatonin regulates androgen secretion through a melatonin membrane receptor in Leydig cells [32]. The binding of phosphorylated CREB to the cAMP response element of the StAR promoter accelerates steroid synthesis. However, in steroidogenic cells, although not all cAMP-regulated genes, many of them have a regulatory sequence recognized by a GATA family transcription factor [33]. In the testes, GATA-4 predominantly regulates the transcription of StAR [34]. The inhibitory effects of melatonin on testosterone production are mediated by the downregulation of GATA-4 expression in a mouse Leydig cell line [10]. Our study shows that melatonin promoted testosterone production through RORα- enhanced GATA-4 expression in an in vitro goat spermatogonial stem cell differentiation culture system (Figure 1) [35]. Additionally, some studies have indicated that the increasing androgens may be primarily due to the stimulatory effect of melatonin on the steroidogenic enzyme 3β-hydroxysteroid dehydrogenase [36]. Other studies showed that activation of melatonin membrane receptors also increased the c-Jun-N-terminal kinase activity [37]. c-Fos and c-Jun were proposed to mediate the responses of Leydig cells to testosterone production in vivo [38]. These genes are involved in the StAR transcription [39].

3. Melatonin Regulates the Function of Sertoli Cells

The Leydig cell is a place where testosterone in the testis is synthesized and secreted, and regulated mainly by insulin-like growth factor secreted by Sertoli cells. Estrogen plays an important role in the function of testicular. Sertoli cells are the main source of estrogen production in immature males. Estrogen receptor-alpha (ERα) was expression in Leydig cells whereas ERβ was detected in Sertoli and germ cells, namely spermatocytes and spermatids [40]. The number of spermatogonial cells per testis was increased in ERβKO mice. The ERαKO mice had significant germ cell loss. The number of Leydig cells per testis was significantly increased in ERβKO mice but not in ERαKO mice [41]. The above results show that ERβ involved in regulation of Leydig cell proliferation and the production of testosterone in the adult mouse testis. The cytochrome P450 aromatase (P450arom) is a key enzyme responsible for the formation of estrogens from androgens and is exists in the endoplasmic reticulum of various tissues. P450arom has been immunolocalized in Leydig cells of numerous species as well as in germ cells of mouse, brown bear, and bank vole. Aromatase activity has been detected in vitro in immature and mature rat Leydig cells and Sertoli cells, while in pig, ram, and humans the enzyme activity is only present in Leydig cells. According to the stage of maturation of germ cell, the amount of aromatase transcripts decreases, being more elevated in younger than in mature rat germ cells [42]. Melatonin inhibits the activity and expression of aromatase, as well as decreases estrogen biosynthesis by regulating gene expression of aromatase via the promoter region [43,44,45]. We found that melatonin increased testosterone production in co-cultured Leydig and Sertoli cells from sheep. Melatonin increased the expression of stem cell factor and insulin-like growth factor-1 and decreased estrogen synthesis in Sertoli cells. It promoted insulin-like growth factor-1 and decreased estrogen content via the membrane melatonin receptor 1 [46]. Furthermore, melatonin regulates Sertoli cell metabolism and, thus, may affect spermatogenesis. Lactate produced by Sertoli cell provides nutritional support and has an anti-apoptotic effect in developing germ cells [47].

4. Conclusions

The melatonin receptor was expressed in testicular cells, and melatonin has effects on testicular development [48]. Serum and seminal plasma levels of melatonin are significantly lower in infertile patients [49]. In addition, environmental endocrine disruptors—such as estrogen analogues—significantly increased the rate of male infertility [50]. Male germ cells are extremely sensitive to reactive oxygen species (ROS), and excessive ROS can cause asthenozoospermia [51], however, melatonin can effectively reduce ROS and lipid peroxides. The simultaneous addition of melatonin during the transplantation of spermatogonial stem cells in azoospermia mouse testes increases the efficiency of transplantation and improves the structural properties of the testis tissue [52]. Further research on the mechanism of melatonin regulating the synthesis of steroid hormones and exploring the small molecules of melatonin receptors will help to cure the reproductive diseases caused by the disorder of steroid hormones.

Acknowledgments

This work was supported by grants from Major Research Plan “973” Project (2012CB944702), and Natural Science Foundation of China (31501953, 31471352, and 31171380).

Author Contributions

Shou-Long Deng, Kun Yu, Tie-Cheng Sun, Yuan-Yuan Li, and Yi-Xun Liu wrote the paper.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Stocco, D.M.; Wang, X.; Jo, Y.; Manna, P.R. Multiple signaling pathways regulating steroidogenesis and steroidogenic acute regulatory protein expression: more complicated than we thought. Mol. Endocrinol. 2005, 19, 2647–5269. [Google Scholar] [CrossRef] [PubMed]
  2. Chen, S.R.; Liu, Y.X. Regulation of spermatogonial stem cell self-renewal and spermatocyte meiosis by Sertoli cell signaling. J. Soc. Reprod. Fertil. 2015, 149, R159–R167. [Google Scholar] [CrossRef] [PubMed]
  3. Wen, Q.; Cheng, C.Y.; Liu, Y.X. Development, function and fate of fetal Leydig cells. Semin. Cell Dev. Biol. 2016, 59, 89–98. [Google Scholar] [CrossRef] [PubMed]
  4. Sanchez-Barcelo, E.J.; Mediavilla, M.D.; Vriend, J.; Reiter, R.J. Constitutive photomorphogenesis protein 1 (COP1) and COP9 signalosome, evolutionarily conserved photomorphogenic proteins as possible targets of melatonin. J. Pineal Res. 2016, 61, 41–51. [Google Scholar] [CrossRef] [PubMed]
  5. Cipolla-Neto, J.; Amaral, F.G.; Afeche, S.C.; Tan, D.X.; Reiter, R.J. Melatonin, energy metabolism, and obesity: A review. J. Pineal Res. 2014, 56, 371–381. [Google Scholar] [CrossRef] [PubMed]
  6. Slominski, A.T.; Zmijewski, M.A.; Jetten, A.M. RORα is not a receptor for melatonin (response to DOI 10.1002/bies.201600018). Bioessays 2016, 38, 1193–1194. [Google Scholar] [CrossRef] [PubMed]
  7. Odawara, H.; Iwasaki, T.; Horiguchi, J.; Rokutanda, N.; Hirooka, K.; Miyazaki, W.; Koibuchi, Y.; Shimokawa, N.; Iino, Y.; Takeyoshi, I.; et al. Activation of aromatase expression by retinoic acid receptor-related orphan receptor (ROR) alpha in breast cancer cells: Identification of a novel ROR response element. J Biol. Chem. 2009, 284, 17711–17719. [Google Scholar] [CrossRef] [PubMed]
  8. Reiter, R.J.; Tan, D.X.; Rosales-Corral, S.; Manchester, L.C. The universal nature, unequal distribution and antioxidant functions of melatonin and its derivatives. Mini Rev. Med. Chem. 2013, 13, 373–384. [Google Scholar] [PubMed]
  9. Ahmad, R.; Haldar, C. Effect of intra-testicular melatonin injection on testicular functions, local and general immunity of a tropical rodent Funambulus pennanti. Endocrine 2010, 37, 479–488. [Google Scholar] [CrossRef] [PubMed]
  10. Qin, F.; Zhang, J.; Zan, L.; Guo, W.; Wang, J.; Chen, L.; Cao, Y.; Shen, O.; Tong, J. Inhibitory effect of melatonin on testosterone synthesis is mediated via GATA-4/SF-1 transcription factors. Reprod. Biomed. Online 2015, 31, 638–646. [Google Scholar] [CrossRef] [PubMed]
  11. Casao, A.; Pérez-Pé, R.; Abecia, J.A.; Forcada, F.; Muiño-Blanco, T.; Cebrián-Pérez, J.Á. The effect of exogenous melatonin during the non-reproductive season on the seminal plasma hormonal profile and the antioxidant defence system of Rasa Aragonesa rams. Anim. Reprod. Sci. 2013, 138, 168–174. [Google Scholar] [CrossRef] [PubMed]
  12. Mura, M.C.; Luridiana, S.; Bodano, S.; Daga, C.; Cosso, G.; Diaz, M.L.; Bini, P.P.; Carcangiu, V. Influence of melatonin receptor 1A gene polymorphisms on seasonal reproduction in Sarda ewes with different body condition scores and ages. Anim. Reprod. Sci. 2014, 149, 173–177. [Google Scholar] [CrossRef] [PubMed]
  13. Wang, L.; Zhuo, Z.Y.; Shi, W.Q.; Tan, D.X.; Gao, C.; Tian, X.Z.; Zhang, L.; Zhou, G.B.; Zhu, S.E.; Yun, P.; et al. Melatonin promotes superovulation in sika deer (Cervus nippon). Int. J. Mol. Sci. 2014, 15, 12107–12118. [Google Scholar] [CrossRef] [PubMed]
  14. Forsberg, M.; Madej, A. Effects of melatonin implants on plasma concentrations of testosterone, thyroxine and prolactin in the male silver fox (Vulpes vulpes). J. Reprod. Fertil. 1990, 89, 351–358. [Google Scholar] [CrossRef] [PubMed]
  15. Rekik, M.; Taboubi, R.; Ben Salem, I.; Fehri, Y.; Sakly, C.; Lassoued, N.; Hilali, M.E. Melatonin administration enhances the reproductive capacity of young rams under a southern Mediterranean environment. Anim. Sci. J. 2015, 86, 66–672. [Google Scholar] [CrossRef] [PubMed]
  16. Gonzalez-A, M.; Luna, C.; Pérez-Pé, R.; Muiño-Blanco, T.; Cebrián-Pérez, J.A.; Casao, A. New evidence of melatonin receptor contribution to ram sperm functionality. Reprod. Fertil. Dev. 2016, 28, 924–935. [Google Scholar] [CrossRef] [PubMed]
  17. Deng, S.L.; Chen, S.R.; Wang, Z.P.; Zhang, Y.; Tang, J.X.; Li, J.; Wang, X.X.; Cheng, J.M.; Jin, C.; Li, X.Y.; et al. Melatonin promotes development of haploid germ cells from early developing spermatogenic cells of Suffolk sheep under in vitro condition. J. Pineal Res. 2016, 60, 435–447. [Google Scholar] [CrossRef] [PubMed]
  18. 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-D,L-aspartic acid-induced LHRH release during the stimulation of luteinizing hormone secretion by melatonin. Biol. Reprod. 1995, 52, 1156–1161. [Google Scholar] [CrossRef] [PubMed]
  19. Buchanan, K.L.; Yellon, S.M. Delayed puberty in the male Djungarian hamster: Effect of short photoperiod or melatonin treatment on the GnRH neuronal system. Neuroendocrinology 1991, 54, 96–102. [Google Scholar] [CrossRef] [PubMed]
  20. Roy, D.; Belsham, D.D. Melatonin receptor activation regulates GnRH gene expression and secretion in GT1-7 GnRH neurons. Signal transduction mechanisms. J. Biol. Chem. 2002, 277, 251–258. [Google Scholar] [CrossRef] [PubMed]
  21. Ancel, C.; Bentsen, A.H.; Sébert, M.E.; Tena-Sempere, M.; Mikkelsen, J.D.; 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. [Google Scholar] [CrossRef] [PubMed]
  22. Irwig, M.S.; Fraley, G.S.; Smith, J.T.; Acohido, B.V.; Popa, S.M.; Cunningham, M.J.; Gottsch, M.L.; Clifton, D.K.; Steiner, R.A. Kisspeptin activation of gonadotropin releasing hormone neurons and regulation of KiSS-1 mRNA in the male rat. Neuroendocrinology 2004, 80, 264–272. [Google Scholar] [CrossRef] [PubMed]
  23. Ansel, L.; Bentsen, A.H.; Ancel, C.; Bolborea, M.; Klosen, P.; Mikkelsen, J.D.; Simonneaux, V. Peripheral kisspeptin reverses short photoperiod-induced gonadal regression in Syrian hamsters by promoting GNRH release. Reproduction 2011, 142, 417–425. [Google Scholar] [CrossRef] [PubMed]
  24. Henningsen, J.B.; Poirel, V.J.; Mikkelsen, J.D.; 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. J. Comp. Neurol. 2016, 524, 1825–1838. [Google Scholar] [CrossRef] [PubMed]
  25. Mason, A.O.; Greives, T.J.; Scotti, M.A.; Levine, J.; Frommeyer, S.; Ketterson, E.D.; Demas, G.E.; Kriegsfeld, L.J. Suppression of kisspeptin expression and gonadotropic axis sensitivity following exposure to inhibitory day lengths in female Siberian hamsters. Horm. Behav. 2007, 52, 492–498. [Google Scholar] [CrossRef] [PubMed]
  26. Revel, F.G.; Saboureau, M.; Masson-Pévet, M.; Pévet, P.; Mikkelsen, J.D.; Simonneaux, V. Kisspeptin mediates the photoperiodic control of reproduction in hamsters. Curr. Biol. 2006, 16, 1730–1735. [Google Scholar] [CrossRef] [PubMed]
  27. Carnevali, O.; Gioacchini, G.; Maradonna, F.; Olivotto, I.; Migliarini, B. Melatonin induces follicle maturation in Danio rerio. PLoS One. 2011, 6, e19978. [Google Scholar] [CrossRef] [PubMed]
  28. Alvarado, M.V.; Carrillo, M.; Felip, A. Melatonin-induced changes in kiss/gnrh gene expression patterns in the brain of male sea bass during spermatogenesis. Comp. Biochem. Physiol. A Mol. Integr. Physiol. 2015, 185, 69–79. [Google Scholar] [CrossRef] [PubMed]
  29. Xu, J.; Li, P. Expression of EAP1 and CUX1 in the hypothalamus of female rats and relationship with KISS1 and GnRH. Endocr. J. 2016, 63, 681–690. [Google Scholar] [CrossRef] [PubMed]
  30. Li, C.; Zhou, X. Melatonin and male reproduction. Clin. Chim. Acta. 2015, 446, 175–180. [Google Scholar] [CrossRef] [PubMed]
  31. Baburski, A.Z.; Sokanovic, S.J.; Janjic, M.M.; Stojkov-Mimic, N.J.; Bjelic, M.M.; Andric, S.A.; Kostic, T.S. Melatonin replacement restores the circadian behavior in adult rat Leydig cells after pinealectomy. Mol. Cell Endocrinol. 2015, 413, 26–35. [Google Scholar] [CrossRef] [PubMed]
  32. 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+. J. Mol. Endocrinol. 1999, 23, 299–306. [Google Scholar] [CrossRef] [PubMed]
  33. Bouchard, M.F.; Taniguchi, H.; Viger, R.S. The effect of human GATA4 gene mutations on the activity of target gonadal promoters. J. Mol. Endocrinol. 2009, 42, 149–160. [Google Scholar] [CrossRef] [PubMed]
  34. Svechnikov, K.; Landreh, L.; Weisser, J.; Izzo, G.; Colón, E.; Svechnikova, I.; Söder, O. Origin, development and regulation of human Leydig cells. Horm. Res. Paediatr. 2010, 73, 93–101. [Google Scholar] [CrossRef] [PubMed]
  35. Deng, S.L.; Zhang, Y.; Yu, K.; Wang, X.X.; Chen, S.R.; Han, D.P.; Cheng, C.Y.; Lian, Z.X.; Liu, Y.X. Melatonin up-regulates the expression of the GATA-4 transcription factor and increases testosterone secretion from Leydig cells through RORα signaling in an in vitro goat spermatogonial stem cell differentiation culture system. Oncotarget 2017, 8, 110592–110605. [Google Scholar] [CrossRef] [PubMed]
  36. Srivastava, R.K.; Krishna, A. Melatonin affects steroidogenesis and delayed ovulation during winter in vespertilionid bat, Scotophilus heathi. J. Steroid. Biochem. Mol. Biol. 2010, 118, 107–116. [Google Scholar] [CrossRef] [PubMed]
  37. Chan, A.S.; Lai, F.P.; Lo, R.K.; Voyno-Yasenetskaya, T.A.; Stanbridge, E.J.; Wong, Y.H. Melatonin mt1 and MT2 receptors stimulate c-Jun N-terminal kinase via pertussis toxin-sensitive and -insensitive G proteins. Cell. Signal. 2002, 14, 249–257. [Google Scholar] [CrossRef]
  38. Rossi, S.P.; Matzkin, M.E.; Terradas, C.; Ponzio, R.; Puigdomenech, E.; Levalle, O.; Calandra, R.S.; Frungieri, M.B. New insights into melatonin/CRH signaling in hamster Leydig cells. Gen. Comp. Endocrinol. 2012, 178, 153–163. [Google Scholar] [CrossRef] [PubMed]
  39. Manna, P.R.; Stocco, D.M. Crosstalk of CREB and Fos/Jun on a single cis-element: transcriptional repression of the steroidogenic acute regulatory protein gene. J. Mol. Endocrinol. 2007, 39, 261–277. [Google Scholar] [CrossRef] [PubMed]
  40. Bilińska, 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. Mol. Cell. Endocrinol. 2001, 178, 189–198. [Google Scholar] [CrossRef]
  41. Gould, M.L.; Hurst, P.R.; Nichol, H.D. The effects of oestrogen receptors alpha and beta on testicular cell number and steroidogenesis in mice. Reproduction 2007, 134, 271–279. [Google Scholar] [CrossRef] [PubMed]
  42. Carreau, S. Germ cells: A new source of estrogens in the male gonad. Mol Cell Endocrinol 2001, 178, 65–72. [Google Scholar] [CrossRef]
  43. Letellier, K.; Azeddine, B.; Parent, S.; Labelle, H.; Rompré, P.H.; Moreau, A.; Moldovan, F. Estrogen cross-talk with the melatonin signaling pathway in human osteoblasts derived from adolescent idiopathic scoliosis patients. J. Pineal Res. 2008, 45, 383–393. [Google Scholar] [CrossRef] [PubMed]
  44. Yoshitane, H.; Honma, S.; Imamura, K.; Nakajima, H.; Nishide, S.Y.; Ono, D.; Kiyota, H.; Shinozaki, N.; Matsuki, H.; Wada, N.; et al. JNK regulates the photic response of the mammalian circadian clock. EMBO Rep. 2012, 13, 455–461. [Google Scholar] [CrossRef] [PubMed]
  45. Vriend, J.; Reiter, R.J. Breast cancer: Modulation by melatonin and the ubiquitin-proteasome system--A review. Mol. Cell. Endocrinol. 2015, 417, 1–9. [Google Scholar] [CrossRef] [PubMed]
  46. Deng, S.L.; Wang, Z.P.; Jin, C.; Kang, X.L.; Batool, A.; Zhang, Y.; Li, X.Y.; Wang, X.X.; Chen, S.R.; Chang, C.S.; et al. Melatonin promotes sheep Leydig cell testosterone secretion in a co-culture with Sertoli cells. Theriogenology 2018, 106, 170–177. [Google Scholar] [CrossRef] [PubMed]
  47. Rocha, C.S.; Martins, A.D.; Rato, L.; Silva, B.M.; Oliveira, P.F.; Alves, M.G. Melatonin alters the glycolytic profile of Sertoli cells: implications for male fertility. Mol. Hum. Reprod. 2014, 20, 1067–1076. [Google Scholar] [CrossRef] [PubMed]
  48. Kasahara, T.; Abe, K.; Mekada, K.; Yoshiki, A.; Kato, T. Genetic variation of melatonin productivity in laboratory mice under domestication. Proc. Natl. Acad. Sci. USA 2010, 107, 6412–6417. [Google Scholar] [CrossRef] [PubMed]
  49. 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, 6. [Google Scholar] [CrossRef] [PubMed]
  50. Deng, S.; Wang, X.; Wang, Z.; Chen, S.; Wang, Y.; Hao, X.; Sun, T.; Zhang, Y.; Lian, Z.; Liu, Y. In vitro production of functional haploid sperm cells from male germ cells of Saanen dairy goat. Theriogenology 2017, 90, 120–128. [Google Scholar] [CrossRef] [PubMed]
  51. Deng, S.L.; Sun, T.C.; Yu, K.; Wang, Z.P.; Zhang, B.L.; Zhang, Y.; Wang, X.X.; Lian, Z.X.; Liu, Y.X. Melatonin reduces oxidative damage and upregulates heat shock protein 90 expression in cryopreserved human semen. Free Radic. Biol. Med. 2017, 113, 347–354. [Google Scholar] [CrossRef] [PubMed]
  52. Gholami, M.; Saki, G.; Hemadi, M.; Khodadadi, A.; Mohammadi-Asl, J. Melatonin improves spermatogonial stem cells transplantation efficiency in azoospermic mice. Iran J. Basic Med. Sci. 2014, 17, 93–99. [Google Scholar] [PubMed]
Sample Availability: Samples of the compounds are not available from the authors.
Figure 1. A schematic illustration of the melatonin regulates the synthesis of steroid hormones in rams.
Figure 1. A schematic illustration of the melatonin regulates the synthesis of steroid hormones in rams.
Molecules 23 00447 g001

Share and Cite

MDPI and ACS Style

Yu, K.; Deng, S.-L.; Sun, T.-C.; Li, Y.-Y.; Liu, Y.-X. Melatonin Regulates the Synthesis of Steroid Hormones on Male Reproduction: A Review. Molecules 2018, 23, 447. https://doi.org/10.3390/molecules23020447

AMA Style

Yu K, Deng S-L, Sun T-C, Li Y-Y, Liu Y-X. Melatonin Regulates the Synthesis of Steroid Hormones on Male Reproduction: A Review. Molecules. 2018; 23(2):447. https://doi.org/10.3390/molecules23020447

Chicago/Turabian Style

Yu, Kun, Shou-Long Deng, Tie-Cheng Sun, Yuan-Yuan Li, and Yi-Xun Liu. 2018. "Melatonin Regulates the Synthesis of Steroid Hormones on Male Reproduction: A Review" Molecules 23, no. 2: 447. https://doi.org/10.3390/molecules23020447

Article Metrics

Back to TopTop