Melatonin: shedding light on infertility?--A review of the recent literature

Shavi Fernando, Luk Rombauts, Shavi Fernando, Luk Rombauts

Abstract

In recent years, the negative impact of oxidative stress on fertility has become widely recognised. Several studies have demonstrated its negative effect on the number and quality of retrieved oocytes and embryos following in-vitro fertilisation (IVF). Melatonin, a pineal hormone that regulates circadian rhythms, has also been shown to exhibit unique oxygen scavenging abilities. Some studies have suggested a role for melatonin in gamete biology. Clinical studies also suggest that melatonin supplementation in IVF may lead to better pregnancy rates. Here we present a critical review and summary of the current literature and provide suggestions for future well designed clinical trials.

Figures

Figure 1
Figure 1
Actions of melatonin and its metabolites. Extrapolated from Hardeland [46], Reiter et al. [13] and Watson [47]. GPx: glutathione peroxidase; SOD: superoxide dismutase; ROS: Reactive oxygen species; RNS: Reactive nitrogen species.
Figure 2
Figure 2
Relative concentrations of plasma melatonin, LH, estradiol and progesterone in hMG/hCG treated cycles. Adapted with permission from Tang et al. [59]. LH: Luteinising hormone.

References

    1. Lundsberg L, Pal L, Gariepy A, Xu X, Chu M, Illuzzi J. Knowledge, attitudes, and practices regarding conception and fertility: a population-based survey among reproductive-age United States women (in press) Fertil Steril. 2014;101:767. doi: 10.1016/j.fertnstert.2013.12.006.
    1. Chakravarty S, Rizvi SI. Physiological effects of melatonin: implications on human health. Biomedicine. 2008;28:252–255. doi: 10.7705/biomedica.v28i2.96.
    1. Madalinski M. Does a melatonin supplement alter the course of gastro-esophageal reflux disease? World J Gastrointest Pharmacol Ther. 2011;2:50–51. doi: 10.4292/wjgpt.v2.i6.50.
    1. Biran V, Phan Duy A, Decobert F, Bednarek N, Alberti C, Baud O. Is melatonin ready to be used in preterm infants as a neuroprotectant? Dev Med Child Neurol. 2014;56:717–723. doi: 10.1111/dmcn.12415.
    1. Bai J, Dong L, Song Z, Ge H, Cai X, Wang G, Liu P. The role of melatonin as an antioxidant in human lens epithelial cells. Free Radic Res. 2013;47:635–642. doi: 10.3109/10715762.2013.808743.
    1. de Matos CA, de Bruin P, de Bruin V, Nunes D, Pereira E, Cavalcante M, Andrade G. Melatonin reduces lung oxidative stress in patients with chronic obstructive pulmonary disease: a randomise, double-blind, placebo-controlled study. J Pineal Res. 2012;53:238–244. doi: 10.1111/j.1600-079X.2012.00992.x.
    1. Scheuer C, Pommergaard H, Rosenberg J, Gögenur I. Melatonin's protective effect against UV radiation: a systematic review of clinical and experimental studies [Epub ahead of print] Photodermatol Photoimmunol Photomed. 2013;30:180. doi: 10.1111/phpp.12080.
    1. Russcher M, Koch B, Nagtegaal E, van der Putten K, ter Wee P, Gaillard C. The role of melatonin treatment in chronic kidney disease. Front Biosci (Landmark Ed) 2012;1:2644–2656. doi: 10.2741/4075.
    1. Carbajo-Pescador S, Ordoñez R, Benet M, Jover R, García-Palomo A, Mauriz J, González-Gallego J. Inhibition of VEGF expression through blockade of Hif1α and STAT3 signalling mediates the anti-angiogenic effect of melatonin in HepG2 liver cancer cells. Br J Cancer. 2013;109:83–91. doi: 10.1038/bjc.2013.285.
    1. Nawrot-Porąbka K, Jaworek J, Leja-Szpak A, Szklarczyk J, Konturek S, Reiter R. Luminal melatonin stimulates pancreatic enzyme secretion via activation of serotonin-dependent nerves. Pharmacol Rep. 2013;65:494–504. doi: 10.1016/S1734-1140(13)71025-9.
    1. Csaba G. The pineal regulation of the immune system: 40 years since the discovery. Acta Microbiol Immunol Hung. 2013;60:77–91. doi: 10.1556/AMicr.60.2013.2.1.
    1. Acuña-Castroviejo D, Escames G, Venegas C, Díaz-Casado M, Lima-Cabello E, López L, Rosales-Corral S, DX T, Reiter R. Extrapineal melatonin: sources, regulation, and potential functions [Epub ahead of print] Cell Mol Life Sci. 2014;71:2997. doi: 10.1007/s00018-014-1579-2.
    1. Reiter R, Rosales-Corral S, Manchester L, Tan D. Peripheral reproductive organ health and melatonin: ready for prime time. Int J Mol Sci. 2013;14:7231–7272. doi: 10.3390/ijms14047231.
    1. Hsing A, Meyer T, Niwa S, Quraishi S, Chu L. Measuring serum melatonin in epidemiologic studies. Cancer Epidemiol Biomarkers Prev. 2010;19:932–937. doi: 10.1158/1055-9965.EPI-10-0004.
    1. Waldhauser F, Waldhauser M, Lieberman H, Deng M, Lynch H, Wurtmann R. Bioavailability of oral melatonin in humans. Neuroendocrinology. 1984;39:307–313. doi: 10.1159/000123997.
    1. Li C, Li G, Tan D, Li F, Ma X. A novel enzyme-dependent melatonin metabolite in humans. J Pineal Res. 2013;54:100–106.
    1. Shreeve N, Cagampang F, Sadek K, Tolhurst M, Houldey A, Hill C, Brook N, Macklon N, Cheong Y. Poor sleep in PCOS; is melatonin the culprit? Hum Reprod. 2013;28:1348–1353. doi: 10.1093/humrep/det013.
    1. de Almeida E, Di Mascio P, Harumi T, Spence D, Moscovitch A, Hardeland R, Cardinali D, Brown G, Pandi-Perumal S. Measurement of melatonin in body fluids: standards, protocols and procedures. Childs Nerv Syst. 2011;27:879–891. doi: 10.1007/s00381-010-1278-8.
    1. Reiter R, Tan D, Tamura H, Cruz M, Fuentes-Broto L. Clinical relevance of melatonin in ovarian and placental physiology: a review. Gynecol Endocrinol. 2014;30:83–89. doi: 10.3109/09513590.2013.849238.
    1. Zhu Y, Fu A, Hoffman A, Figueiro M, Carskadon M, Sharkey K, Rea M. Advanced sleep schedules affect circadian gene expression in young adults with delayed sleep schedules. Sleep Med. 2013;14:449–455. doi: 10.1016/j.sleep.2012.12.006.
    1. Wilhelmsen-Langeland A, Saxvig I, Pallesen S, Nordhus I, Vedaa O, Lundervold A, Bjorvatn B. A randomise controlled trial with bright light and melatonin for the treatment of delayed sleep phase disorder: effects on subjective and objective sleepiness and cognitive function. J Biol Rhythms. 2013;28:306–321. doi: 10.1177/0748730413500126.
    1. Cortesi F, Giannotti F, Sebastiani T, Panunzi S, Valente D. Controlled-release melatonin, singly and combined with cognitive behavioural therapy, for persistent insomnia in children with autism spectrum disorders: a randomise placebo-controlled trial. J Sleep Res. 2012;21:700–709. doi: 10.1111/j.1365-2869.2012.01021.x.
    1. Yadav S, Haldar C. Reciprocal interaction between melatonin receptors (Mel(1a), Mel(1b), and Mel(1c)) and androgen receptor (AR) expression in immunoregulation of a seasonally breeding bird, Perdicula asiatica: role of photoperiod. J Photochem Photobiol B. 2013;5:52–60. doi: 10.1016/j.jphotobiol.2013.03.005.
    1. Clarke I, Caraty A. Kisspeptin and seasonality of reproduction. Adv Exp Med Biol. 2013;784:411–430. doi: 10.1007/978-1-4614-6199-9_19.
    1. Abecia J, Forcada F, González-Bulnes A. Pharmaceutical control of reproduction in sheep and goats. Vet Clin North Am Food Anim Pract. 2011;27:67–79. doi: 10.1016/j.cvfa.2010.10.001.
    1. Zaidi J, Jurkovic D, Campbell S, Okokon E, Tan S. Circadian variation in uterine artery blood flow indices during the follicular phase of the menstrual cycle. Ultrasound Obstet Gynecol. 1995;5:406–410. doi: 10.1046/j.1469-0705.1995.05060406.x.
    1. Silman R. Melatonin: a contraceptive for the nineties. Eur J Obstet Gynecol Reprod Biol. 1993;49:3–9. doi: 10.1016/0028-2243(93)90099-X.
    1. Kojo S. Vitamin C: basic metabolism and its function as an index of oxidative stress. Curr Med Chem. 2004;11:1041–1064. doi: 10.2174/0929867043455567.
    1. Bouayed J, Bohn T. Exogenous antioxidants - double-edged swords in cellular redox state: health beneficial effects at physiologic doses versus deleterious effects at high doses. Oxid Med Cell Longev. 2010;3:228–237. doi: 10.4161/oxim.3.4.12858.
    1. Valko M, Leibfritz D, Moncol J, Cronin M, Mazur M, Telser J. Free radicals and antioxidants in normal physiological functions and human disease. Int J Biochem Cell Biol. 2007;39:44–84. doi: 10.1016/j.biocel.2006.07.001.
    1. Rahman K. Studies on free radicals, antioxidants, and co-factors. Clin Interv Aging. 2007;2:219–236.
    1. Wood S, Quinn A, Troupe S, Kingsland C, Lewis-Jones I. Seasonal variation in assisted conception cycles and the influence of photoperiodism on outcome in in-vitro fertilisation cycles. Hum Fertil. 2006;9:223–229. doi: 10.1080/14647270600806557.
    1. Agarwal A, Gupta S, Sharma RK: Role of oxidative stress in female reproduction.Reprod Biol Endocrinol 2005, 3. ., []
    1. Srinivasan V, Spence W, Pandi-Perumal SR, Zakharia R, Bhatnagar K, Brzezinski A. Melatonin and human reproduction: shedding light on the darkness hormone. Gynecol Endocrinol. 2009;25:779–785. doi: 10.3109/09513590903159649.
    1. Dubocovich M, Markowska M. Functional MT1 and MT2 melatonin receptors in mammals. Endocrine. 2005;27:101–110. doi: 10.1385/ENDO:27:2:101.
    1. Tamura H, Takasaki A, Taketani T, Tanabe M, Kizuka F, Lee L, Tamura I, Maekawa R, Aasada H, Yamagata Y, Sugino N: The role of melatonin as an antioxidant in the follicle.J Ovarian Res 2012, 5.. ., []
    1. Tan D, Manchester L, Reiter R, Qi W, Karbownik M, Calvo J. Significance of melatonin in antioxidative defense system: reactions and products. Biol Signals Recept. 2000;9:137–159. doi: 10.1159/000014635.
    1. Acuña-Castroviejo D, Reiter R, Menéndez-Peláez A, Pablos M, Burgos A. Characterization of high-affinity melatonin binding sites in purified cell nuclei of rat liver. J Pineal Res. 1994;16:100–112. doi: 10.1111/j.1600-079X.1994.tb00089.x.
    1. Benítez-King G, Huerto-Delgadillo L, Antón-Tay F. Binding of 3H-melatonin to calmodulin. Life Sci. 1993;53:201–207. doi: 10.1016/0024-3205(93)90670-X.
    1. Ressmeyer A, Mayo J, Zelosko V, Sáinz R, Tan D, Poeggeler B, Antolín I, Zsizsik B, Reiter R, Hardeland R. Antioxidant properties of the melatonin metabolite N1-acetyl-5-methoxykynuramine (AMK): scavenging of free radicals and prevention of protein destruction. Redox Rep. 2003;8:205–213. doi: 10.1179/135100003225002709.
    1. Galano A, Tan D, Reiter R. On the free radical scavenging activities of melatonin's metabolites, AFMK and AMK. J Pineal Res. 2013;54:245–257. doi: 10.1111/jpi.12010.
    1. Tan D, Reiter R, Manchester L, Yan M, El-Sawi M, Sainz R, Mayo J, Kohen R, Allegra M, Hardeland R. Chemical and physical properties and potential mechanisms: melatonin as a broad spectrum antioxidant and free radical scavenger. Curr Top Med Chem. 2002;2:181–197. doi: 10.2174/1568026023394443.
    1. Mayo J, Sainz R, Antoli I, Herrera F, Martin V, Rodriguez C. Melatonin regulation of antioxidant enzyme gene expression. Cell Moll Life Sci. 2002;59:1706–1713. doi: 10.1007/PL00012498.
    1. Tamura H, Nakamura Y, Korkmaz A, Manchester L, Tan D, Sugino N, Reiter R. Melatonin and the ovary: physiological and pathophysiological implications. Fertil Steril. 2009;92:328–343. doi: 10.1016/j.fertnstert.2008.05.016.
    1. Urata Y, Honma S, Goto S, Todoroki S, Iida T, Cho S, Honma K, Kondo T. Melatonin induces gamma-glutamylcysteine synthetase mediated by activator protein-1 in human vascular endothelial cells. Free Radic Biol Med. 1999;27:838–847. doi: 10.1016/S0891-5849(99)00131-8.
    1. Hardeland R. Melatonin metabolism in the central nervous system. Curr Neuropharmacol. 2010;8:168–181. doi: 10.2174/157015910792246164.
    1. Watson R. Melatonin in the Promotion of Health 2nd edition. United States: CRC Press; 2011.
    1. Sanchez-Barcelo EJ, Mediavilla MD, Tan DX, Reiter RJ. Clinical uses of melatonin: evaluation of human trials. Curr Med Chem. 2010;17:2070–2095. doi: 10.2174/092986710791233689.
    1. Kolli V, Kanakasabapathy I, Faith M, Ramamoorthy H, Isaac B, Natarajan K, Abraham P. A preclinical study on the protective effect of melatonin against methotrexate-induced small intestinal damage: effect mediated by attenuation of nitrosative stress, protein tyrosine nitration, and PARP activation. Cancer Chemother Pharmacol. 2013;71:1209–1218. doi: 10.1007/s00280-013-2115-z.
    1. Tomov B, Popov D, Tomova R, Vladov N, Den Otter W, Krastev Z. BCG and melatonin. 2013. Therapeutic response of untreatable hepatocellular carcinoma after application of the immune modulators IL-2; pp. 4531–4535.
    1. Tomov B, Popov D, Tomova R, Vladov N, Den Otter W, Krastev Z. Therapeutic response of untreatable hepatocellular carcinoma after application of the immune modulators IL-2, BCG and melatonin. Anticancer Res. 2013;33:4531–4535.
    1. Kim J, Lee Y, Kim B, Kim S, Kim D, Jo K, Lee S, Lee K, Baik H. Melatonin improves inflammatory cytokine profiles in lung inflammation associated with sleep deprivation. Mol Med Rep. 2012;5:1281–1284.
    1. Siah K, Wong R, Ho K. Melatonin for the treatment of irritable bowel syndrome. World J Gastroenterol. 2014;20:2492–2498. doi: 10.3748/wjg.v20.i10.2492.
    1. Jena G, Trivedi P. A review of the use of melatonin in ulcerative colitis: experimental evidence and new approaches. Inflamm Bowel Dis. 2014;20:553–563. doi: 10.1097/01.MIB.0000436962.32164.6e.
    1. Trivedi P, Jena G. Melatonin reduces ulcerative colitis-associated local and systemic damage in mice: investigation on possible mechanisms. Dig Dis Sci. 2013;58:3460. doi: 10.1007/s10620-013-2831-6.
    1. Chojnacki C, Wiśniewska-Jarosińska M, Kulig G, Majsterek I, Reiter R, Chojnacki J. Evaluation of enterochromaffin cells and melatonin secretion exponents in ulcerative colitis. World J Gastroenterol. 2013;19:3602–3607. doi: 10.3748/wjg.v19.i23.3602.
    1. Schwertner A, Conceição dos Santos C, Costa G, Deitos A, de Souza A, de Souza I, Torres I, da Cunha Filho J, Caumo W. Efficacy of melatonin in the treatment of endometriosis: a phase II, randomise, double-blind, placebo-controlled trial. Pain. 2013;154:874–881. doi: 10.1016/j.pain.2013.02.025.
    1. Turi A, Garzetti GG. The pattern of melatonin in amenorrheic women affected by sterility. Acta Eur Fertil. 1993;24:71–74.
    1. Tang P, Chan T, Tang G, Pang S. Plasma melatonin profile and hormonal interactions in the menstrual cycles of anovulatory infertile women treated with gonadotropins. Gynecol Obstet Invest. 1998;45:247–252. doi: 10.1159/000009977.
    1. Webley GE, Hearn JP. Local production of progesterone by the corpus luteum of the marmoset monkey in response to perfusion with chorionic gonadotrophin and melatonin in-vivo. J Endocrinol. 1987;112:449–457. doi: 10.1677/joe.0.1120449.
    1. Attarchi M, Darkhi H, Khodarahmian M, Dolati M, Kashanian M, Ghaffari M, Mirzamohammadi E, Mohammadi S. Characteristics of menstrual cycle in shift workers. Glob J Health Sci. 2013;5:163–172.
    1. Grajewski B, Nguyen MM, Whelan EA, Cole RJ, Hein MJ. Measuring and identifying large-study metrics for circadian rhythm disruption in female flight attendants. Scand J Work Environ Health. 2003;29:337–346. doi: 10.5271/sjweh.740.
    1. Lawson CC, Whelan EA, Lividoti Hibert EN, Spiegelman D, Schernhammer ES, Rich-Edwards JW. Rotating shift work and menstrual cycle characteristics. Epidemiology. 2011;22:305–312. doi: 10.1097/EDE.0b013e3182130016.
    1. Miyauchi F, Nanjo K, Otsuka K. Effects of night shift on plasma concentrations of melatonin, LH, FSH and prolactin, and menstrual irregularity. Sangyo Igaku. 1992;34:545–550. doi: 10.1539/joh1959.34.545.
    1. Boczek-Leszczyk E, Juszczak M. The influence of melatonin on human reproduction. Pol Merkur Lekarski. 2007;23:128–130.
    1. Voordouw B, Euser R, Verdonk R, Alberda B, de Jong F, Drogendijk A, Fauser B, Cohen M. Melatonin and melatonin-progestin combinations alter pituitary-ovarian function in women and can inhibit ovulation. J Clin Endocrinol Metab. 1992;74:108–117.
    1. Salhab M, Dhorne-Pollet S, Auclair S, Guyader-Joly C, Brisard D, Dalbies-Tran R, Dupont J, Ponsart C, Mermillod P, Uzbekova S. In-vitro maturation of oocytes alters gene expression and signaling pathways in bovine cumulus cells. Mol Reprod Dev. 2013;80:166–182. doi: 10.1002/mrd.22148.
    1. Itoh M, Ishizuka B, Kuribayashi Y, Amemiya A, Sumi Y. Melatonin, its precursors, and synthesizing enzyme activities in the human ovary. Mol Hum Reprod. 1999;5:402–408. doi: 10.1093/molehr/5.5.402.
    1. Wurtman R, Axelrod J, Potter L. The uptake of h3-melatonin in endocrine and nervous tissues and the effects of constant light exposure. J Pharmacol Exp Ther. 1964;143:314–318.
    1. Brzezinski A, Seibel M, Lynch H, Deng M, Wurtman R. Melatonin in human preovulatory follicular fluid. J Clin Endocrinol Metab. 1987;64:865–867. doi: 10.1210/jcem-64-4-865.
    1. Rönnberg L, Kauppila A, Leppäluoto J, Martikainen H, Vakkuri O. Circadian and seasonal variation in human preovulatory follicular fluid melatonin concentration. J Clin Endocrinol Metab. 1990;71:492–496. doi: 10.1210/jcem-71-2-493.
    1. Nakamura Y, Tamura H, Takayama H, Kato H. Increased endogenous level of melatonin in preovulatory human follicles does not directly influence progesterone production. Fertil Steril. 2003;80:1012–1016. doi: 10.1016/S0015-0282(03)01008-2.
    1. Carlomagno G, Nordio M, Chiu TT, Unfer V. Contribution of myo-inositol and melatonin to human reproduction. Eur J Obstet Gynecol Reprod Biol. 2011;159:267–272. doi: 10.1016/j.ejogrb.2011.07.038.
    1. Hobson S, Lim R, Gardiner E, Alers N, Wallace E. BMJ Open. 2013. Phase I pilot clinical trial of antenatal maternally administered melatonin to decrease the level of oxidative stress in human pregnancies affected by pre-eclampsia (PAMPR): study protocol.
    1. Tamura H, Nakamura Y, Pilar Terron M, Flores L, Manchester L, Tan D, Sugino N, Reiter R. Melatonin and pregnancy in the human. Reprod Toxicol. 2008;25:291–303. doi: 10.1016/j.reprotox.2008.03.005.
    1. Lemley C, Camacho L, Vonnahme K. Uterine infusion of melatonin or melatonin receptor antagonist alters ovine feto-placental hemodynamics during midgestation. 2013.
    1. Wong C, Jow G, Kaizaki A, Fan L, Tien L. Melatonin ameliorates brain injury induced by systemic lipopolysaccharide in neonatal rats. Neuroscience. 2014;267:147–156. doi: 10.1016/j.neuroscience.2014.02.032.
    1. Miller SL, Yawno T, Alers NO, Castillo-Melendez M, Supramaniam VG, VanZyl N, Sabaretnam T, Loose JM, Drummond GR, Walker DW, Jenkin G, Wallace EM. Antenatal antioxidant treatment with melatonin to decrease newborn neurodevelopmental deficits and brain injury caused by fetal growth restriction. J Pineal Res. 2014;56(3):283–94. doi: 10.1111/jpi.12121.
    1. Sack R, Lewy A, Erb D, Vollmer W, Singer C. Human melatonin production decreases with age. J Pineal Res. 1986;3:379–388. doi: 10.1111/j.1600-079X.1986.tb00760.x.
    1. Toffol E, Kalleinen N, Haukka J, Vakkuri O, Partonen T, Polo-Kantola P. Melatonin in perimenopausal and postmenopausal women: associations with mood, sleep, climacteric symptoms, and quality of life. Menopause. 2014;21:493–500. doi: 10.1097/GME.0b013e3182a6c8f3.
    1. Bellipanni G, DI Marzo F, Blasi F, Di Marzo A. Effects of melatonin in perimenopausal and menopausal women: our personal experience. Ann N Y Acad Sci. 2005;1057:393–402. doi: 10.1196/annals.1356.030.
    1. Maria S, Witt-Enderby PA: Melatonin effects on bone: potential use for the prevention and treatment for osteopenia, osteoporosis, and periodontal disease and for use in bone-grafting procedures.J Pineal Res 2013.
    1. Kotlarczyk MP, Lassila HC, O'Neil CK, D'Amico F, Enderby LT, Witt-Enderby PA, Balk JL. Melatonin osteoporosis prevention study (MOPS): a randomise, double-blind, placebo-controlled study examining the effects of melatonin on bone health and quality of life in perimenopausal women. J Pineal Res. 2012;52:414–426. doi: 10.1111/j.1600-079X.2011.00956.x.
    1. Meliska CJ, Martinez LF, Lopez AM, Sorenson DL, Nowakowski S, Parry BL. Relationship of morningness-eveningness questionnaire score to melatonin and sleep timing, body mass index and atypical depressive symptoms in peri- and post-menopausal women. Psychiatry Res. 2011;188:88–95. doi: 10.1016/j.psychres.2010.12.010.
    1. Barron ML. Light exposure, melatonin secretion, and menstrual cycle parameters: an integrative review. Biol Res Nurs. 2007;9:49–69. doi: 10.1177/1099800407303337.
    1. Hemadi M, Shokri S, Pourmatroud E, Moramezi F, Khodadai A. Follicular dynamic and immunoreactions of the vitrified ovarian graft after host treatment with variable regimens of melatonin. Am J Reprod Immunol. 2012;67:401–412. doi: 10.1111/j.1600-0897.2011.01087.x.
    1. Jahnke G, Marr M, Myers C, Wilson R, Travlos G, Price C. Maternal and developmental toxicity evaluation of melatonin administered orally to pregnant Sprague–Dawley rats. Toxicol Sci. 1999;50:271–279. doi: 10.1093/toxsci/50.2.271.
    1. Showell M, Brown J, Clarke J, Hart R: Antioxidants for female subfertility.Cochrane Database Syst Rev 2013.
    1. Kucuktulu E. Protective effect of melatonin against radiation induced nephrotoxicity in rats. Asian Pac J Cancer Prev. 2012;13:4101–4105. doi: 10.7314/APJCP.2012.13.8.4101.
    1. Staikou C, Kyrozis A, Moschovos C, Fassoulaki A. Effects of morning melatonin administration on electroencephalographic theta to alpha power ratio in reproductive versus postmenopausal healthy female volunteers. Neurosci Lett. 2012;507:90–93. doi: 10.1016/j.neulet.2011.11.061.
    1. Weaver D. Reproductive safety of melatonin: a “wonder drug” to wonder about. J Biol Rhythms. 1997;12:682. doi: 10.1177/074873049701200625.
    1. Gill A. Book Poisons Standard 2013, Standard for the Uniform Scheduling of Medicines and Poisons No. 4 (SUSMP 4) City: Australian Government Department of Health and Ageing; 2013. Poisons Standard 2013, Standard for the Uniform Scheduling of Medicines and Poisons No. 4 (SUSMP 4), Australian Government Department of Health and Ageing.
    1. Maestroni GJ, Cardinali DP, Esquifino AI, Pandi-Perumal SR. Does melatonin play a disease-promoting role in rheumatoid arthritis? J Neuroimmunol. 2005;158:106–111. doi: 10.1016/j.jneuroim.2004.08.015.
    1. Hong YG, Riegler JL. Is melatonin associated with the development of autoimmune hepatitis? J Clin Gastroenterol. 1997;25:376–378. doi: 10.1097/00004836-199707000-00020.
    1. Fourman LT, Robert Meyer B. Autoimmune hepatitis in association with ramelteon. J Clin Gastroenterol. 2013;47:651–654. doi: 10.1097/MCG.0b013e31829174f0.
    1. Constantinescu CS. Melanin, melatonin, melanocyte-stimulating hormone, and the susceptibility to autoimmune demyelination: a rationale for light therapy in multiple sclerosis. Med Hypotheses. 1995;45:455–458. doi: 10.1016/0306-9877(95)90220-1.
    1. Sulli A, Maestroni GJ, Villaggio B, Hertens E, Craviotto C, Pizzorni C, Briata M, Seriolo B, Cutolo M. Melatonin serum levels in rheumatoid arthritis. Ann N Y Acad Sci. 2002;966:276–283. doi: 10.1111/j.1749-6632.2002.tb04227.x.
    1. Guerin P, El Mouatassim S, Menezo Y. Oxidative stress and protection against reactive oxygen species in the pre-implantation embryo and its surroundings. Hum Reprod Update. 2001;7:175–189. doi: 10.1093/humupd/7.2.175.
    1. Palini S, Benedetti S, Tagliamonte MC, De Stefani S, Primiterra M, Polli V, Rocchi P, Catalani S, Battistelli S, Canestrari F, Bulletti C. Influence of ovarian stimulation for IVF/ICSI on the antioxidant defence system and relationship to outcome. Reprod Biomed Online. 2014;29:65. doi: 10.1016/j.rbmo.2014.03.010.
    1. Yoshida M, Ishigati K, Nagai T, Chikyu M, Pursel V. Glutathione concentration during maturation and after fertilisation in pig ooctes: relevance to the ability of oocytes to form male pronucleus. Biol Reprod. 1993;49:89–94. doi: 10.1095/biolreprod49.1.89.
    1. Fatehi A, Roelen B, Colenbrander B, Schoevers E, Gadella B, Beverst M, van den Hurk R. Presence of cumulus cells during in-vitro fertilisation protects the bovine oocyte against oxidative stress and improves first cleavage but does not affect further development. Zygote. 2005;13:177–185. doi: 10.1017/S0967199405003126.
    1. Huang B, Li Z, Ai J, Zhu L, Li Y, Jin L, Zhang H. Antioxidant capacity of follicular fluid from patients undergoing in-vitro fertilisation. Int J Clin Exp Pathol. 2014;7:2273–2282.
    1. Papis K, Poleszczuk O, Wenta-Muchalska E, Modlinski J. Melatonin effect on bovine embryo development in-vitro in relation to oxygen concentration. J Pineal Res. 2007;43:321–326. doi: 10.1111/j.1600-079X.2007.00479.x.
    1. Takahashi T, Takahashi E, Igarashi H, Tezuka N, Kurachi H. Impact of oxidative stress in aged mouse oocytes on calcium oscillations at fertilisation. Mol Reprod Dev. 2003;66:143–152. doi: 10.1002/mrd.10341.
    1. du Plessis S, Makker K, Desai N, Agarwal A. Impact of oxidative stress on IVF. Expert Rev Obstet Gynecol. 2008;3:539–554. doi: 10.1586/17474108.3.4.539.
    1. Combelles C, Gupta S, Agarwal A. Could oxidative stress influence the in-vitro maturation of oocytes? Reprod Biomed Online. 2009;18:864–880. doi: 10.1016/S1472-6483(10)60038-7.
    1. Bedaiwy M, Elnashar S, Goldberg J, Sharma R, Mascha E, Arrigain S, Agarwal A, Falcone T. Effect of follicular fluid oxidative stress parameters on intracytoplasmic sperm injection outcome. Gynecol Endocrinol. 2012;28:51–55. doi: 10.3109/09513590.2011.579652.
    1. Ghiselli A, Serafini M, Natella F, Scaccini C. Total antioxidant capacity as a tool to assess redox status: critical view and experimental data. Free Radic Biol Med. 2000;29:1106–1114. doi: 10.1016/S0891-5849(00)00394-4.
    1. O'Flaherty C, de Lamirande E, Gagnon C. Positive role of reactive oxygen species in mammalian sperm capacitation: triggering and modulation of phosphorylation events. Free Radic Biol Med. 2006;41:528–540. doi: 10.1016/j.freeradbiomed.2006.04.027.
    1. Donà G, Fiore C, Tibaldi E, Frezzato F, Andrisani A, Ambrosini G, Fiorentin D, Armanini D, Bordin L, Clari G. Endogenous reactive oxygen species content and modulation of tyrosine phosphorylation during sperm capacitation. Int J Androl. 2011;34:411. doi: 10.1111/j.1365-2605.2010.01097.x.
    1. Donà G, Fiore C, Andrisani A, Ambrosini G, Brunati A, Ragazzi E, Armanini D, Bordin L, Clari G. Evaluation of correct endogenous reactive oxygen species content for human sperm capacitation and involvement of the NADPH oxidase system. Hum Reprod. 2011;26:3264–3273. doi: 10.1093/humrep/der321.
    1. Aitken R, Clarkson J. Cellular basis of defective sperm function and its association with the genesis of reactive oxygen species by human spermatozoa. J Reprod Fertil. 1987;81:459–469. doi: 10.1530/jrf.0.0810459.
    1. Alvarez J, Touchstone J, Blasco L, Storey B. Spontaneous lipid peroxidation and production of hydrogen peroxide and superoxide in human spermatozoa. superoxide dismutase as major enzyme protectant against oxygen toxicity. J Androl. 1987;8:338–348. doi: 10.1002/j.1939-4640.1987.tb00973.x.
    1. Guz J, Gackowski D, Foksinski M, Rozalski R, Zarakowska E, Siomek A, Szpila A, Kotzbach M, Kotzbach R, Olinski R: Comparison of oxidative stress/DNA damage in semen and blood of fertile and infertile men.PLoS One 2013, 8. e68490.
    1. Jones R, Mann T, Sherins R. Peroxidative breakdown of phospholipids in human spermatozoa, spermicidal properties of fatty acid peroxides, and protective action of seminal plasma. Fertil Steril. 1979;31:531–537.
    1. Koppers A, Mitchell L, Wang P, Lin M, Aitken R. Phosphoinositide 3-kinase signalling pathway involvement in a trunctated apoptotic cascade associated with motility loss and oxidative DNA damage in human spermatozoa. Biochem J. 2011;2011:687–698. doi: 10.1042/BJ20110114.
    1. Aitken R, De Iuliis G. On the possible origins of DNA damage in human spermatozoa. Mol Hum Reprod. 2010;16:3–13. doi: 10.1093/molehr/gap059.
    1. Wright C, Milne S, Leeson H. Sperm DNA damage caused by oxidative stress: modifiable clinical, lifestyle and nutritional factors in male infertility. Reprod Biomed Online. 2014;28:684–703. doi: 10.1016/j.rbmo.2014.02.004.
    1. Seli E, Gardner D, Schoolcraft W, Moffatt O, Sakkas D. Extent of nuclear DNA damage in ejaculated spermatozoa impacts on blastocyst development after in-vitro fertilisation. Fertil Steril. 2004;82:378–383. doi: 10.1016/j.fertnstert.2003.12.039.
    1. Lewis S, Aitken R. DNA damage to spermatozoa has impacts on fertilisation and pregnancy. Cell Tissue Res. 2005;322:33–41. doi: 10.1007/s00441-005-1097-5.
    1. El-Raey M, Geshi M, Somfai T, Kaneda M, Hirako M, Abdel-Ghaffar A, Sosa G, El-Roos M, 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. doi: 10.1002/mrd.21295.
    1. Hardeland R. Antioxidative protection by melatonin: multiplicity of mechanisms from radical detoxification to radical avoidance. Endocrine. 2005;27:119–130. doi: 10.1385/ENDO:27:2:119.
    1. Lowes D, Webster N, Murphy M, Galley H. Antioxidants that protect mitochondria reduce interleukin-6 and oxidative stress, improve mitochondrial function, and reduce biochemical markers of organ dysfunction in a rat model of acute sepsis. Br J Anaesth. 2013;110:472–480. doi: 10.1093/bja/aes577.
    1. Espey L. Current status of the hypothesis that mammalian ovulation is comparable to an inflammatory reaction. Biol Reprod. 1994;50:233–238. doi: 10.1095/biolreprod50.2.233.
    1. Tamura H, Takasaki A, Miwa I, Taniguchi K, Maekawa R, Asada H, Taketani T, Matsuoka A, Yamagata Y, Shimamura K, Morioka H, Ishikawa H, Reiter RJ, Sugino N. Oxidative stress impairs oocyte quality and melatonin protects oocytes from free radical damage and improves fertilisation rate. J Pineal Res. 2008;44(3):280–87. doi: 10.1111/j.1600-079X.2007.00524.x.
    1. Tamura H, Taketani T, Takasaki A, Sugino N. Book Influence of oxidative stress on oocyte quality and protective role of melatonin as an antioxidant. 7. Munich, Germany: Journal fur Reproduktionsmedizin und Endokrinologie; 2010. Influence of oxidative stress on oocyte quality and protective role of melatonin as an antioxidant; p. 366.
    1. Lord T. Melatonin prevents postovulatory oocyte aging in the mouse and extends the window for optimal fertilisation in-vitro. Biol Reprod. 2013;88:67. doi: 10.1095/biolreprod.112.106450.
    1. Knapen M, Zusterzeel P, Peters W, Steegers E. Glutathione and glutathione-related enzymes in reproduction: a review. Eur J Obstet Gynecol Reprod Biol. 1999;82:171–184. doi: 10.1016/S0301-2115(98)00242-5.
    1. Kang J, Koo O, Kwon D, Park H, Jang G, Kang S, Lee B. Effects of melatonin on in-vitro maturation of porcine oocyte and expression of melatonin receptor RNA in cumulus and granulosa cells. J Pineal Res. 2009;46:22–28. doi: 10.1111/j.1600-079X.2008.00602.x.
    1. Salimi M, Salehi M, Masteri Farahani R, Dehghani M, Abadi M, Novin M, Nourozian M, Hosseini A. The effect of melatonin on maturation, glutathione level and expression of HMGB1 gene in Brilliant Cresyl Blue (BCB) stained immature oocyte. Cell J. 2014;15:294–301.
    1. Bahadori MH, Ghasemian F, Ramezani M, Asgari Z. Melatonin effect during different maturation stages of oocyte and subsequent embryo development in mice. Iran J Reprod Med. 2013;11:11–18.
    1. Wei D, Zhang C, Xie J, Song X, Yin B, Liu Q, Hu L, Hao H, Geng J, Wang P. Supplementation with low concentrations of melatonin improves nuclear maturation of human oocytes in-vitro. J Assist Reprod Genet. 2013;30:933–938. doi: 10.1007/s10815-013-0021-2.
    1. Kim M, Park E, Kim H, Choi W, Cho J, Lee W, Cha K, Kim Y, Lee D, Yoon T. Does supplementation of in-vitro culture medium with melatonin improve IVF outcome in PCOS? Reprod Biomed Online. 2013;26:22–29. doi: 10.1016/j.rbmo.2012.10.007.
    1. Nishihara T, Hashimoto S, Ito K, Nakaoka Y, Matsumoto K, Hosoi Y, Morimoto Y. Oral melatonin supplementation improves oocyte and embryo quality in women undergoing in-vitro fertilisation-embryo transfer. Gynecol Endocrinol. 2014;30:359–362. doi: 10.3109/09513590.2013.879856.
    1. Barnett AG, van der Pols JC, Dobson AJ. Regression to the mean: what it is and how to deal with it. Int J Epidemiol. 2005;34:215–220. doi: 10.1093/ije/dyh299.
    1. Rombauts L. Is there a recommended maximum starting dose of FSH in IVF? J Assist Reprod Genet. 2007;24:343–349. doi: 10.1007/s10815-007-9134-9.
    1. Eryilmaz O, Devran A, Sarikaya E, Aksakal F, Mollamahmutoğlu L, Cicek N. Melatonin improves the oocyte and the embryo in IVF patients with sleep disturbances, but does not improve the sleeping problems. J Assist Reprod Genet. 2011;28:815–820. doi: 10.1007/s10815-011-9604-y.
    1. Batıoğlu A, Şahin U, Gürlek B, Öztürk N, Ünsal E. The efficacy of melatonin administration on oocyte quality. Gynecol Endocrinol. 2012;28:91–93. doi: 10.3109/09513590.2011.589925.
    1. Rizzo P, Raffone E, Benedetto V. Effect of the treatment with myo-inositol plus folic acid plus melatonin in comparison with a treatment with myo-inositol plus folic acid on oocyte quality and pregnancy outcome in IVF cycles. a prospective, clinical trial. Embase European Rev Med Pharmacol Sci. 2010;14:555–561.
    1. Unfer V, Carlomagno G, Rizzo P, Raffone E, Roseff S. Myo-inositol rather than D-chiro-inositol is able to improve oocyte quality in intracytoplasmic sperm injection cycles. a prospective, controlled, randomise trial. Eur Rev Med Pharmacol Sci. 2011;15:452–457.
    1. Konakchieva R: Physiological role of melatonin in human reproductive system - Signaling, targets and receptor localization. In Book Physiological role of melatonin in human reproductive system - Signaling, targets and receptor localization. Bulgaria: 2012.
    1. Ortiz A, Espino J, Bejarano I, Lozano G, Monllor F, Gracía J, Pariente J, Rodríguez A. High endogenous melatonin concentrations enhance sperm quality and short-term in-vitro exposure to melatonin improves aspects of sperm motility. J Pineal Res. 2011;50:132–139.
    1. Espino J, Ortiz A, Bejarano I, Lozano G, Monllor F, Garcia J, Rodriguez A, Pariente J. Melatonin protects human spermatozoa from apoptosis via melatonin receptor- and extracellular signal-regulated kinase-mediated pathways. Fertil Steril. 2011;95:2290–2296. doi: 10.1016/j.fertnstert.2011.03.063.
    1. Espino J, Bejarano I, Ortiz A, Lozano G, Gracía J, Pariente J, Rodríguez A. Melatonin as a potential tool against oxidative damage and apoptosis in ejaculated human spermatozoa. Fertil Steril. 2010;94:1915–1917. doi: 10.1016/j.fertnstert.2009.12.082.
    1. Ashrafi I, Kohram H, Ardabili F. Antioxidative effects of melatonin on kinetics, microscopic and oxidative parameters of cryopreserved bull spermatozoa. Anim Reprod Sci. 2013;139:25–30. doi: 10.1016/j.anireprosci.2013.03.016.
    1. Nasiraei-Moghadam S, Parivar K, Ahmadiani A, Movahhedin M, Vaez Mahdavi M. Protective effect of melatonin against inequality-induced damages on testicular tissue and sperm parameters. Int J Fertil Steril. 2014;7:313–322.
    1. Kim S, Lee I, Baek H, Shin I, Moon C, Kim S, Yun W, Nam K, Kim H, Kim J. Melatonin prevents gentamicin-induced testicular toxicity and oxidative stress in rats [EPub ahead of print] Andrologia. 2013;46:1032. doi: 10.1111/and.12191.
    1. Liu C, Gao P, Xu S, Wang Y, Chen C, He M, Yu Z, Zhang L, Zhou Z. Mobile phone radiation induces mode-dependent DNA damage in a mouse spermatocyte-derived cell line: a protective role of melatonin. Int J Radiat Biol. 2013;89:993–1001. doi: 10.3109/09553002.2013.811309.
    1. du Plessis 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. doi: 10.1111/j.1439-0272.2009.00964.x.
    1. Rodriguez-Osorio N, Kim I, Wang H, Kaya A, Memili E. Melatonin increases cleavage rate of porcine preimplantation embryos in-vitro. J Pineal Res. 2007;43:283–288. doi: 10.1111/j.1600-079X.2007.00475.x.
    1. Ishizuka B, Kuribayashi Y, Murai K, Amemiya A, Itoh M. The effect of melatonin on in-vitro fertilisation and embryo development in mice. J Pineal Res. 2000;28:48–51. doi: 10.1034/j.1600-079x.2000.280107.x.
    1. Choi J, Park SM, Lee E, Kim JH, Jeong YI, Lee JY, Park SW, Kim HS, Hossein MS, Jeong YW, Kim S, Hyun SH, Hwang WS. Anti-apoptotic effect of melatonin on preimplantation development of porcine parthenogenetic embryos. Mol Reprod Dev. 2008;75(7):1127–35. doi: 10.1002/mrd.20861.
    1. Asgari Z, Ghasemian F, Ramezani M, Bahadori MH. The effect of melatonin on the developmental potential and implantation rate of mouse embryos. Cell J. 2012;14:203–208.
    1. Mehaisen GM, Saeed AM. In-vitro development rate of preimplantation rabbit embryos cultured with different levels of melatonin. Zygote. 2013;28:1–5.
    1. Wang F, Tian X, Zhang L, Gao C, He C, Fu Y, Ji P, Li Y, Li N, Liu G. Beneficial effects of melatonin on in-vitro bovine embryonic development are mediated by melatonin receptor 1. J Pineal Res. 2014;56:333–342. doi: 10.1111/jpi.12126.
    1. Wang F, Tian X, Zhou Y, Tan D, Zhu S, Dai Y, Liu G. Melatonin improves the quality of in-vitro produced (IVP) bovine embryos: implications for blastocyst development, cryotolerance, and modifications of relevant gene expression. PLoS One. 2014;9:e93641. doi: 10.1371/journal.pone.0093641.
    1. Succu S, Pasciu V, Manca ME, Chelucci S, Torres-Rovira L, Leoni GG, Zinellu A, Carru C, Naitana S, Berlinguer F. Dose-dependent effect of melatonin on postwarming development of vitrified ovine embryos. Theriogenology. 2014;81:1058–1066. doi: 10.1016/j.theriogenology.2014.01.032.
    1. Sugino N, Takiguchi S, Kashida S, Takayama H, Yamagata Y, Nakamura Y, Kato H. Suppression of intracellular superoxide dismutase activity by antisense oligonucleotides causes inhibition of progesterone production by rat luteal cells. Biol Reprod. 1999;61:1133–1138. doi: 10.1095/biolreprod61.4.1133.
    1. Scarpellini F, Mastrone M, Sbracia M, Scarpellini L. Serum lipoperoxide level variations in normal and luteal phase defect cycles. Gynecol Obstet Invest. 1996;42:28–30. doi: 10.1159/000291884.
    1. Taketani T, Tamura H, Takasaki A, Lee L, Kizuka F, Tamura I, Taniguchi K, Maekawa R, Asada H, Shimamura K, Reiter RJ, Sugino N. Protective role of melatonin in progesterone production by human luteal cells. J Pineal Res. 2011;51(2):207–13. doi: 10.1111/j.1600-079X.2011.00878.x.
    1. Takasaki A, Tamura H, Taniguchi K, Asada H, Taketani T, Matsuoka A, Yamagata Y, Shimamura K, Morioka H, Sugino N: Luteal blood flow and luteal function.J Ovarian Res 2009, 2.., []
    1. Ciotta L, Stracquadanio M, Pagano I, Carbonaro A, Palumbo M, Gulino F. Effects of myo-inositol supplementation on oocyte's quality in PCOS patients: a double blind trial. Eur Rev Med Pharmacol Sci. 2011;15:509–514.
    1. Nazzaro A, Salerno A, Marino S, Granato C, Pastore E. The addiction of melatonin to myo-inositol plus folic acid improve oocyte quality and pregnancy outcome in IVF cycle. a prospective clinical trial. Hum Reprod. 2011;26:i227. doi: 10.1093/humrep/deq308.
    1. Pacchiarotti A, Carlomagno G, Unfer V, Frati P, Pacchiarotti A, Prapas N: Role of Myo-inositol and melatonin supplementation in follicular fluid of IVF patients with polycystic ovarian syndrome: a randomise controlled trial.ClinicalTrialsgov 2013. ., []
    1. Unfer V, Raffone E, Rizzo P, Buffo S. Effect of a supplementation with myo-inositol plus melatonin on oocyte quality in women who failed to conceive in previous in-vitro fertilisation cycles for poor oocyte quality: a prospective, longitudinal, cohort study. Gynaecol Endocrinol. 2011;27:857–861. doi: 10.3109/09513590.2011.564687.
    1. Seko L, Moroni R, Leitao V, Teixeira D, Nastri C, Martins W. Melatonin supplementation during controlled ovarian stimulation for women undergoing assisted reproductive technology: systematic review and meta-analysis of randomise controlled trials. Fertil Steril. 2014;101:154–161. doi: 10.1016/j.fertnstert.2013.09.036.
    1. Vidor LP, Torres IL, de Souza IC C, Fregni F, Caumo W. Analgesic and sedative effects of melatonin in temporomandibular disorders: a double-blind, randomise, parallel-group, placebo-controlled study. J Pain Symptom Manage. 2013;46:422–432. doi: 10.1016/j.jpainsymman.2012.08.019.

Source: PubMed

3
Abonnieren