Next Article in Journal
Negative Effects of Chronic High Intake of Fructose on Lung Diseases
Next Article in Special Issue
Lower Fiber Consumption in Women with Polycystic Ovary Syndrome: A Meta-Analysis of Observational Studies
Previous Article in Journal
Reduced Consumption of Sugar-Sweetened Beverages Is Associated with Lower Body Mass Index Z-Score Gain among Chinese Schoolchildren
Previous Article in Special Issue
Relevance of Vitamin D and Its Deficiency for the Ovarian Follicle and the Oocyte: An Update
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Melatonin in Endometriosis: Mechanistic Understanding and Clinical Insight

1
Department of Obstetrics & Gynaecology, The Chinese University of Hong Kong, Hong Kong 999077, China
2
Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
3
Laboratory of Reproduction and Development, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
4
School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
5
Chinese University of Hong Kong-Sichuan University Joint Laboratory in Reproductive Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Nutrients 2022, 14(19), 4087; https://doi.org/10.3390/nu14194087
Submission received: 15 August 2022 / Revised: 27 September 2022 / Accepted: 27 September 2022 / Published: 1 October 2022
(This article belongs to the Special Issue Impact of Nutrition on Female Reproductive Disorders)

Abstract

:
Endometriosis is defined as the development of endometrial glands and stroma outside the uterine cavity. Pathophysiology of this disease includes abnormal hormone profiles, cell survival, migration, invasion, angiogenesis, oxidative stress, immunology, and inflammation. Melatonin is a neuroendocrine hormone that is synthesized and released primarily at night from the mammalian pineal gland. Increasing evidence has revealed that melatonin can be synthesized and secreted from multiple extra-pineal tissues where it regulates immune response, inflammation, and angiogenesis locally. Melatonin receptors are expressed in the uterus, and the therapeutic effects of melatonin on endometriosis and other reproductive disorders have been reported. In this review, key information related to the metabolism of melatonin and its biological effects is summarized. Furthermore, the latest in vitro and in vivo findings are highlighted to evaluate the pleiotropic functions of melatonin, as well as to summarize its physiological and pathological effects and treatment potential in endometriosis. Moreover, the pharmacological and therapeutic benefits derived from the administration of exogenous melatonin on reproductive system-related disease are discussed to support the potential of melatonin supplements toward the development of endometriosis. More clinical trials are needed to confirm its therapeutic effects and safety.

1. Introduction

Melatonin (N-acetyl-5-methoxytryptamine) is a neurohormone synthesized and released at night from the pineal gland. This hormone has been shown to regulate the circadian rhythm of the body, pubertal development, and seasonal adaptation [1,2]. It has also been reported to act directly on the hypothalamic–pituitary–gonad (HPG) axis to regulate reproduction [3,4]. For many years, melatonin has only been considered as a hormone to be secreted exclusively by the pineal gland. In recent years, it has been shown that this hormone can be produced by many extra-pineal tissues to exert its biological functions locally [5]. As a pleiotropic molecule, melatonin can interact with female reproductive organs, including the ovaries, uterus, mammary glands, and placenta, to regulate many physiological and pathological events [6,7]. Several studies have shown the capabilities of melatonin to improve fertilization in sub-fertile patients [8]. The beneficial effects of melatonin in reproduction can be attributed to its well-characterized antioxidative effect and other biological functions, such as the regulation of steroid hormone production, anti-cell proliferation, pro-apoptosis, anti-cell adhesion, anti-invasion, anti-epithelial-mesenchymal transition (EMT), anti-angiogenic activity, and regulation of immune modulation as well as neurotrophic effects [9].
Endometriosis is a chronic disease that can be defined as the development of endometrial glands and stroma on the uterine cavity [10]. It is estimated that endometriosis affects 5 to 10% of women between 25 and 35 years old [10]. Endometriosis is an estrogen-dependent inflammatory disease that affects women at reproductive stage and leads to sterility. Typical symptoms of endometriosis include chronic pelvic pain, dysmenorrhea, and dyspareunia.
The pathogenesis of endometriosis is not yet fully understood. Despite several proposed theories on the etiology of endometriosis, none of these models can fully explain the pathogenesis of this disease. Regardless of this, it is commonly theorized to arise from the endometrial fragments escaping into the peritoneal cavity through the process of retrograde menstruation. Meanwhile, abnormal endocrine, immunology, pro-inflammatory, and proangiogenic pathways may contribute to the development of endometriosis [11,12]. Additionally, many abnormal cellular functions, including cell proliferation and migration/invasion, have been shown to induce the process [13]. Misinterpretation of endometriosis-induced pain, such as menstruation-associated abdominal cramps, can often delay the diagnosis of endometriosis by up to 8 to 10 years [14]. Treatment of endometriosis is mainly used to alleviate the symptoms and increase fertility rates [15]. Hormonal compounds and symptomatic treatments are the major medications to treat endometriosis, including non-steroidal anti-inflammatory drugs, gonadotropin-releasing hormone agonists, progestins, and oral contraceptive pills [16]. As these medications bring side effects by creating a hypoestrogenic state to suppress ovarian functions, women of childbearing age should instead only use pain relief [15]. As a consequence, it is highly crucial to develop potential new treatments that do not interfere with the reproductive functions in women suffering from endometriosis. As the occurrence of endometriosis cannot be explained by any single pathophysiological event, therapeutic drugs should be multi-targeted to resolve the disease complexity [17]. Regarding this, melatonin could be a potential candidate as an ideal medication for the treatment of endometriosis. With melatonin demonstrating a potential therapeutic effect against endometriosis in an early clinical study, its therapeutic targets and molecular mechanism are worth exploring further [18].
Considering that several melatonin-involved pathways might be relevant to the pathophysiology of endometriosis, there is currently no review dedicated to melatonin as a potential treatment of endometriosis. Hence, a more comprehensive evaluation of the effects of melatonin in treating endometriosis is worth deliberating. This review aims to explain the biology and molecular mechanism of melatonin and emphasizes its potential clinical applications for treating endometriosis.

2. The Biology of Melatonin

2.1. Synthesis

The classic melatonin biosynthetic pathway includes five enzymatic steps in animals. In the first step, tryptophan is hydroxylated to 5-hydroxytryptophan by tryptophan-5-hydroxylase (TPH); consequently, it is decarboxylated to serotonin (5-hydroxytryptamine) by the aromatic amino acid decarboxylase (AADC). In the two final steps, it is proposed that serotonin is acetylated by aralkylamine N-acetyltransferase (AANAT) to N-acetylserotonin. Ultimately, this works as an intermediate to form N-acetyl-5-methoxytryptamine, also known as melatonin, exclusively in the pineal gland in animals [19]. After synthesis from the pineal gland, melatonin passes to the hypothalamic suprachiasmatic nuclei (SCN) via the polysynaptic pathway. Additionally, in recent year, multiple studies have also shown that melatonin could be produced in several extra-pineal organs, including the retina, skin, gastrointestinal tract, and bone marrow, and act locally through autocrine and/or paracrine manners [20], while for the female reproductive system, melatonin was found to be secreted by the uterus, ovaries, and placenta [21].

2.2. Metabolism

The circadian melatonin cycle is at maximum at night and minima during the day. The plasma half-life of melatonin is relatively short, around 20 to 50 minutes [22]. Melatonin is highly soluble in both water and lipids. Therefore, it easily diffuses through the cell membrane and the blood–brain barrier [23]. In humans, melatonin is mainly metabolized to 6-hydroxymelatonin (6-HMEL) that serves as a surrogate of circulatory melatonin levels by the liver enzyme cytochrome P450 1A2 (CYP1A2), which is then subsequently conjugated with sulfate and is excreted in the urine [24]. For exogenous melatonin, the peak plasma/serum concentration (Tmax) is achieved at approximately 50 min after oral administration of melatonin in an immediate-release dosage form. In accordance, the elimination half-life (T1/2) of melatonin for both oral and intravenous administration was 45 min [25].

2.3. Receptors

In humans, there have been two main isoforms of the membrane-bound melatonin receptors (MTs), namely MT1 and MT2, identified in the plasma and peripheral tissues [22]. Both of them belong to the G protein-coupled receptors (GPCRs) family. The key biological effects of melatonin are exerted by activating through MT1 and MT2 in humans [22]. After the two receptors couple to Gαi/o proteins, intracellular levels of the second messenger cyclic adenosine monophosphate (cAMP) are consequently decreased, which is the most common signaling pathway activated by melatonin [26], such that MT1 and MT2 receptors have a high density of expressions in the neuroendocrine system, such as the hypothalamic SCN, pituitary pars tuberalis (PT), and gonadotroph cells [27,28]. These receptors are also expressed in several peripheral organs, including skin, vasculature, gastrointestinal tract, and kidney [29,30,31,32]. In the reproductive system, melatonin receptors can be detected in the placenta, uterus, and endometrium [33,34,35]. These biological effects are mediated by the interaction of melatonin with MT1/MT2 for activating other molecular pathways. For instance, melatonin may act as a ligand for the family of orphan retinoid-linked nuclear receptors (RZR/ROR), including RZRα, RORα, RORα2, and RZRβ [36].

2.4. Biological Effects

Besides being known to regulate the circadian rhythm, new evidence indicates that melatonin can act as a regulator in the endocrine, cardiovascular, immune, and neutral systems [37,38,39,40]. Melatonin exerts these effects by working as a potent antioxidant that can directly scavenge free radicals, oxygen-centered radicals, and other toxic hydroxyl radicals [41,42]. Owing to its potent antioxidant properties, melatonin can promote apoptosis with anti-inflammatory and anti-angiogenesis capabilities toward various carcinomas [43,44,45]. Likewise, melatonin has been shown to play a key role in reproduction and women’s health. For instance, studies have shown a significant and positive impact on the maturation of the oocyte and subsequent embryonic development [46]. In addition, patients who were given melatonin during in vitro fertilization and embryo transfer (IVF-ET) are likely to increase the success rate of the IVF-ET treatment [47]. Regarding endometriosis, women who worked during night shifts were reported to have decreased pineal gland melatonin production and an increased risk of endometriosis, hinting at a possible correlation between melatonin and endometriosis [48,49].

3. Potential Therapeutic Mechanisms of Melatonin in Endometriosis

In studies using a model of endometriosis, melatonin was shown to prevent and treat endometriosis by reducing the size and weight of the endometriotic lesions [50,51,52], while in women with endometriosis, MT2 protein expression was significantly decreased in peritoneal lesions compared to eutopic endometrium [53]. A randomized phase II double-blinded, placebo-controlled clinical trial demonstrated that melatonin could alleviate pain in endometriosis [18]. Details are summarized below.
Current evidence suggests that melatonin supplements are safe for short-term use [54]. Even when taken at extreme doses, only mild adverse effects such as dizziness, sleepiness, nausea, and headache have been reported [55,56]. No studies have indicated that exogenous melatonin would induce any serious adverse effects. In addition, clinical studies have proven the safety profile of exogenous melatonin to be used as a supplement, particularly in obstetrics and reproductive-related conditions, such as for fertility [57]. These studies showed the potential of melatonin as a therapeutic drug for endometriosis without any negative effect on the reproductive system. Similarly, randomized clinical studies indicate that long-term melatonin treatment causes only mild adverse effects comparable to a placebo. Long-term safety of melatonin in children and adolescents, however, requires further investigation. The schematic diagram of the potential therapeutic effects of melatonin on endometriosis is shown in Figure 1. The studies related to the determined effects of melatonin on endometriosis are summarized in Table 1. In addition, the effects of exogenous melatonin in the rat model of endometriosis are compared in Figure 2.
Table 1. Experimental studies of melatonin for endometriosis.
Table 1. Experimental studies of melatonin for endometriosis.
StudiesStudy TypeStudy
Sample
Specie
Models/Methods Dose Route Duration ResultsMechanism
Guney
2008 [58]
Animal
in vivo
ratsEndometrium fragments sutured to the abdominal wall10 mg/kg dailyip4 weeksDecreased lesion sizes and weightIncreased SOD, CAT
Decreased MDA, COX-2
Paul
2008 [59]
Animal
in vivo
miceIntraperitoneal transplantation48 mg/kg dailyip10 or 20 days/Increased TIMP-1
Decreased MMP-9
Paul
2010 [60]
Animal
in vivo
miceIntraperitoneal transplantation48 mg/kg dailyip10 or 20 days/Increased TIMP-3
Decreased MMP3
Yildirim
2010 [61]
Animal
in vivo
ratsEndometrium fragments sutured to the abdominal wall10 mg/kg dailyip 2 weeksDecreased lesion sizes
Decreased histopathologic scores
Increased SOD, CAT
Koc
2010 [62]
Animal
in vivo
ratsEndometrium fragments sutured to the abdominal wall10 mg/kg dailyip4 weeksDecreased histopathologic score Increased SOD, CAT
Decreased MDA
Kocadal
2013 [63]
Animal
in vivo
ratsIntraperitoneal transplantation20 mg/kg dailyim and ip2 weeksDecreased lesion sizes
Decreased histopathologic score
/
Yilmaz
2015 [51]
Animal
in vivo
ratsEndometrium fragments sutured to the abdominal wall10 mg/kg dailyip4 weeksDecreased lesion weight
Decreased histopathologic scores
Increased SOD, MDA, TIMP-2
Decreased VEGF, MMP-9
Cetinkaya 2015 [50] Animal
in vivo
ratsEndometrium fragments sutured to the abdominal wall10 or 20 mg/kg dailyim and ip2 weeksDecreased lesion sizes in both treatment groups without significant difference/
Shasha
2018 [64]
Cell
in vitro
human Endometriotic eutopic epithelial cell culture 1 mMculture medium72 h/Increased Numb, E-cadherin
Decreased migration, invasion and Notch1, Vimentin, Slug, Snail
ip, Intraperitoneal injection; im, Intramuscular injection; SOD, Superoxide dismutase; CAT, Catalase; MDA, Malondialdehyde; COX-2, Cyclooxygenase 2; TIMP, Tissue inhibitors of metalloprotease; MMP, Matrix metalloproteinase; VEGF, Vascular endothelial growth factor.
Figure 2. Forest plots of effectiveness of melatonin treatment compared with control in endometriosis. Forest plot of included studies comparing the effect of exogenous melatonin (10 mg/kg) in a rat model of endometriosis. (A) Lesion size at 2 weeks [50,51,61]. (B) Lesion size at 4 weeks [50,58,61,62]. (C) Histopathological score after 2 weeks [50,51,61]. (D) Histopathological score after 4 weeks [50,58,61,62]. (E) Level of superoxide dismutase (SOD) after 2 weeks [50,58,62].
Figure 2. Forest plots of effectiveness of melatonin treatment compared with control in endometriosis. Forest plot of included studies comparing the effect of exogenous melatonin (10 mg/kg) in a rat model of endometriosis. (A) Lesion size at 2 weeks [50,51,61]. (B) Lesion size at 4 weeks [50,58,61,62]. (C) Histopathological score after 2 weeks [50,51,61]. (D) Histopathological score after 4 weeks [50,58,61,62]. (E) Level of superoxide dismutase (SOD) after 2 weeks [50,58,62].
Nutrients 14 04087 g002

3.1. Anti-Oxidation

As endometriosis patients are prominently known to have an upregulated level of reactive oxygen species (ROS), the insufficient antioxidants would prompt a deleterious effect from the imbalance between ROS and antioxidants [65]. The elevated ROS in endometriosis can lead to cellular damage and other undesirable side effects, including a systemic inflammatory reaction and endometriosis-related infertility [66]. With melatonin being a free-radical scavenger and a potent stimulator of antioxidant enzymes, it has been proposed as a potential therapeutic agent to prevent the onset of endometriosis by enhancing the antioxidant levels. Based on an animal study, melatonin was shown to downregulate cyclooxygenase 2 (COX-2) and malondialdehyde (MDA) protein levels and upregulate SOD and catalase (CAT) in the endometriotic lesions [58], while in another in vivo study, melatonin also demonstrated upregulating levels of antioxidants, such as SOD and CAT [52]. Likewise, activities of lipid peroxidation (LP) were induced, and antioxidants, including SOD and CAT, were reduced in an endometriosis rat model after undergoing pinealectomy [62]. Interestingly, these effects were effectively reversed by the introduction of exogenous melatonin [62]. Similarly, the MDA levels and histopathological scores of endometriosis were significantly reduced, with no alteration in SOD levels, in the human xenograft model of endometriosis when treated with melatonin for 4 weeks [67]. Another study also revealed similar results in demonstrating the capability of melatonin to upregulate SOD levels and decreased histopathological scores, but with a discrepancy in the increased MAD level [51].
In general, melatonin has been shown to elicit a tendency to prevent oxidative stress in organs requiring high oxygen consumption (e.g., the brain) and those colonized with certain microbes to create a hostile environment, such as in the gut and skin [68,69]. Besides these, the beneficial role of melatonin as a potent antioxidant has also been demonstrated in the human reproductive system. As melatonin can reduce oxidative stress in follicles, it functioned by preventing the free radicals generated from causing harm on the oocytes [47]. In addition, melatonin can improve the uterine microenvironment by promoting the expression of antioxidant enzymes, such as SOD and CAT [70]. Moreover, these findings clearly illustrate that melatonin can potentially act as an antioxidant to treat endometriosis through the upregulation of antioxidant enzymes and downregulation of pro-oxidant enzymes. The studies related to the determined effects of melatonin on endometriosis are summarized in Table 1.

3.2. Regulation of Steroid Hormone Production and Function

Endometriosis is a disease of hormone imbalance, particularly referred to as an estrogen-dependent and progesterone (P4)-resistance disorder [10]. In contrast to normal endometrium, endometriotic tissues have a higher capability to synthesize E2 locally, due to the high expression of two of the key enzymes for estrogen biosynthesis: aromatase (CYP19A1) and steroidogenic acute regulatory protein (StAR) [71]. As the accumulated E2 can induce cell proliferation and inflammation, this would further stimulate the advancement and progression of endometriosis by promoting lesion growth and undermining the endometrial sensitivity [12,72]. In normal endometrium, the mediators of estrogenic action can be through the two receptors estrogen receptor alpha (ERα) (ESR1) and estrogen receptor beta (ERβ) (ESR2) [73]. Their regulation in endometriosis remains uncertain, such that one study suggested that patients with endometriosis would have downregulation of ERα and upregulation of Erβ [74]. On the contrary, another study showed that ERα and ERβ expression levels were both increased in ectopic tissue in comparison with the normal and eutopic endometrium [74]. Although there were mixed results, both studies supported that the ESR2/ESR1 ratio was increased in the endometriotic lesions. The upregulated estrogen activities would therefore cause progesterone resistance [75]. In addition, patients with endometriosis were reported to be unable to manifest P4 to activate its receptor (PGR) or the transcription of its target gene [76]. As a consequence, PGR-B in mRNA and protein concentrations were significantly downregulated in both endometriotic lesions and endometrium [77,78,79]. In this regard, current studies have unveiled the regulatory effects of melatonin on P4 and E2 that would take place in tissue-specific manners and through a series of ongoing mechanisms [80].

3.2.1. Estrogen

Significant research has confirmed the tendency of melatonin in suppressing E2 production. A study conducted on hamster ovary showed that melatonin inhibited E2 synthesis from the preovulatory follicle via the MAPK pathway following treatment with human chorionic gonadotropin (hCG), a hormone to induce the corpus luteum to secrete E2 [81]. Long-term melatonin treatment in rats has been shown to significantly reduce 17 beta-estradiol levels in plasma, ERα expression levels in the ovary, and ERβ levels in the uterine tissue [82]. In a clinical trial, daily administration of 300 mg of melatonin for 4 months resulted in a significant reduction in estradiol level [83]. In preclinical studies on hormone-dependent breast cancer, melatonin was found to lower serum E2 levels, ESR expressions, and tumor growth in mice models, as well as reduce cell viability through the estrogen response pathway in a human breast cancer cell line [84,85,86]. On the other hand, melatonin was noted to stimulate the production of E2 through the activation of the PKA-CREB signaling pathway in human granulosa cells [87,88]. Aromatase, which is encoded by CYP19A1, is the core enzyme to catalyze the androgen biosynthesis to form E2 [89]. In human MCF-7 breast cancer cells, aromatase expression was suppressed by the bounding of melatonin to MT1 for inhibiting downstream functions, such as cell growth, via the cAMP pathway [90,91]. Likewise, melatonin also can reduce E2 biosynthesis and blocked the growth of human umbilical vein endothelial cells (HUVECs) and rat glioma cells via the suppression of the aromatase level [92,93]. Interestingly, melatonin had a stimulating effect on the aromatase expression level in human granulosa cells [88]. The underlying cause of differential effects of melatonin could be explained by CYP19A1 being driven by different promoters at the upstream of the first exons in various tissues, which ultimately leads to the respective outcomes in different tissues [94,95]. Pinealectomy in rats has been reported to increase E2 synthesis and interfere with LH and FSH releases to result in anovulation [96]. Treatment with melatonin inhibited the proliferation of endometrial epithelial cells in the presence of E2 at 48 h after exposure [53]. In a rat endometriosis model, treatment with melatonin resulted in greater regression of endometriosis foci and reduced recurrence rate when compared to those treated with an aromatase inhibitor, letrozole [52]. Moreover, these studies illustrated that melatonin would not only regulate the E2 response pathway but would contribute to suppressing the E2 biosynthesis via aromatase activity.

3.2.2. Progesterone

To date, there is a great deal of related evidence suggesting that melatonin may play a role in regulating P4 resistance. In human granulosa cells, multiple findings showed that melatonin increased the production of P4 [97,98]. On the contrary, a study conducted on patients with uterine cancer showed that melatonin inhibited the production of P4 in human granulosa cells [99]. Meanwhile, some studies showed that melatonin had no direct effect on the production of P4 in human granulosa cells [100,101]. P4 resistance was a consequence of PGR dysregulation, as well as due to the reduction of StAR, a key enzyme in regulating the synthesis of P4. Melatonin enhanced PGR expression in mouse cumulus cells and induced the expression of StAR in human granulosa and bovine theca cells via the activation of the PI3K/AKT signaling pathway [98,102]. A similar upregulation effect was also observed in mammalian Leydig cells [103]. Other contradictory findings suggested that melatonin reduced PGR expression in the MCF-7 human breast cancer cell line [104,105]. P4 is essential for the development of endometrial pinopodes and microvilli. With the levels of P4 being reduced after pinealectomy, it further contributes to the outcome of a reduced number of implantations, indicating the potential role of melatonin [106]. Nevertheless, further studies on the function of melatonin on P4 regulation would still be warranted on hormonal balance.

3.3. Anti-Proliferation and Pro-Apoptosis

The endometrium maintains complex controls on proliferation and apoptosis as part of repetitive menstrual cycles that prepare the endometrium for the window of implantation and pregnancy. However, the occurrence of endometriosis would disrupt the balance between cell proliferation and apoptosis [10]. Melatonin was shown to regulate several pathways associated with proliferation and apoptosis in normal and cancer cells [107]. These pathways mainly mediate through nuclear factor-kappaB (NF-κB), PI3K/Akt, and cyclin-dependent kinases (CDKs) to promote cell proliferation [108], such that the regulatory activity of melatonin can be demonstrated by its capability to inhibit NF-κB activation in human lung cancer cell lines and liver cancer cell lines [109,110]. Likewise, melatonin (or in combination with vitamin D3) was found to inhibit proliferation via the PI3K/Akt pathway in a human breast cancer cell line [111,112]. Additionally, melatonin was shown to inhibit the RNA expressions of cyclins and CDKs to suppress the progression of the G1-phase in dopaminergic neuroblastoma cells in rats [113]. In addition, melatonin can inhibit the RNA expression level of cyclin D1 in human breast cancer cells and the RNA and protein expression levels of cyclin D1, cyclin B1, CDK1, and CDK4 in osteosarcoma cells [114,115]. Although there is currently no literature documenting the capability to downregulate proliferation via the above-mentioned pathways in endometriosis, several studies have shown that melatonin significantly suppressed eutopic endometrial lesions in endometriosis mouse models [52,58,63]. Additionally, melatonin could also inhibit the cell proliferation induced by estrogen in human endometrial epithelial cells [53]. However, melatonin can facilitate apoptosis through the modulation of various apoptotic mediators including mitochondria, Bax, Bcl-2, endogenous ROS, and apoptosis receptors [116]. In a study using a xenograft of endometriosis, it showed that the treatment of melatonin can enhance apoptotic cells while reducing the expression of Bcl-2 via the upregulation of Bax expression and promoting caspase 9 activation [60]. Taken together, these studies can indicate the potential of melatonin to possibly restore the balance of the endometrium on proliferation and apoptosis disrupted by the occurrence of endometriosis.

3.4. Anti-Adhesion and Anti-Invasion

Ectopic endometriosis cells have an advanced potential to invade immunosurveillance and attach to the lymph nodes and the abdominal cavity [117]. Viable glandular and stromal endometrial cells attach to the abdominal cavity through interacting with cell surface receptors, including integrins and membrane adhesion molecules in the extracellular matrix (ECM), such as fibronectin and laminin [117,118]. The mRNA and protein levels of collagen I and fibronectin were found to be inhibited by melatonin in a hindlimb ischemia mouse model [119]. Matrix metalloproteinases (MMPs) are a large family that can remodel ECM and regulate adhesion and invasion of cells [120]. MMP proteins were highly expressed in endometriotic tissues when compared to normal endometrium [121,122,123]. As the tissue inhibitors of metalloproteases (TIMPs) are endogenous inhibitors of MMPs, a significant reduction in the protein expressions of TIMP-1 and TIMP-2 in endometriotic diseased tissue was reported [124,125,126]. In another study, it was shown that eutopic endometrium and ectopic lesions of women with endometriosis had lower levels of TIMP-3 mRNA expression, while the MMP-9 mRNA level was solely enhanced in ectopic lesions [121]. Other studies also showed that the expression levels of MMP2, MMP3, and MMP9 in ectopic endometrium and endometriosis cells were amplified [127,128]. In accordance, melatonin was found to regulate MMP gene expression and activity in many diseases, such that melatonin could protect against endometriosis by regulating TIMP-1/MMP9 and TIMP3/MMP3 in the peritoneal endometriosis mice model on degrading ECM and regressing the growth of the endometriotic lesion [59,60]. Similarly, melatonin was found to increase TIMP-2 expression levels in the endometriosis rat model [51]. On the other hand, melatonin was shown to downregulate MMP2 and MMP9 expression levels in the gastric mucosa of rats, human gastric cancer cell lines, and human breast cancer cell lines [129,130,131]. It was also found to reduce the concentration of MMP9 in rat retina, human ovarian cells, and breast cancer cells [132,133,134]. The downregulation in mRNA and protein levels of MMP2 and MMP9 can induce high glucose and IL-1β levels in human retinal endothelial cells and retinal pigment epithelial cells [135]. In addition, mice that were kept under different light conditions showed a dose-dependence effect of melatonin to inhibit the adhesion of the integrin-mediated granulocyte to intercellular adhesion molecule-coated plates via melatonin receptors [136]. Taken together, the pleiotropic role of melatonin would contribute to the effect of downregulation toward adhesion and invasion.

3.5. Anti-Epithelial–Mesenchymal Transition (Anti-EMT)

As EMT is defined as the process whereby epithelial cells are transformed into mesenchymal cells, it is an essential process to regulate endometriosis progression and metastasis [137]. To undergo the process of EMT, cells are required in the cycle of migration, invasion, and proliferation. In addition, signaling pathways, such as NF-κB, Wnt, Notch, and other crucial factors (e.g., Snail, Slug, and Twist), commonly regulate the EMT process via modulating the expression levels of E-cadherin, N-cadherin, and vimentin [138,139,140,141], such that the Notch signaling pathway has been shown to be critical for the induction of EMT and the development of various diseases, including endometriosis [142,143]. Melatonin has been shown to inhibit Estradiol-17β-induced EMT in normal epithelial cells and endometriotic cells by increasing the expression of Numb and decreasing activity of the Notch signaling pathway [64]. Besides its anti-EMT effect on endometriosis, melatonin inhibited the EMT process and tumor progression in human gastric cancer cell lines by upregulating E-cadherin expression via the NF-κB signaling pathway [144,145]. A reduction in vimentin expression was equally observed when the rates of migration and invasion of mammary spheres formed by canine and human breast cancer cell lines were suppressed by NF-κB signaling after melatonin treatment [146]. Likewise, the Wnt/β-catenin pathway is another important regulator of EMT for β-catenin to interact with E-cadherin, which acts as a key factor for adhesive binding [139]. In a similar matter, melatonin can activate GSK3β via upregulating the Akt phosphorylation level to stimulate β-catenin metabolism, which ultimately inhibits EMT [147]. In addition, melatonin was found to suppress RSK2 via the HER2/MAPK/Erk pathway to inhibit EMT in breast cancer cells [148]. Besides these pathways, melatonin has also been shown to inhibit transforming growth factor beta-induced EMT in MG-63 osteosarcoma cells via the HIF-1/Snail/MMP-9 pathway [149].

3.6. Anti-Angiogenesis

Angiogenesis serves as a critical function during the development of endometriosis for facilitating the formation of new blood vessels to circulate nutrients and oxygen and to stimulate the progression of endometriotic lesions [150]. Activities of angiogenic factors, in particular vascular endothelial growth factor (VEGF), were higher in both peritoneal fluid and endometrium in women with endometriosis than controls [151,152]. The stimulation by VEGF can promote proliferation, migration, tubular organization, and increased permeability of endothelial cells, ultimately favoring angiogenesis [153]. Based a meta-analysis evaluating the treatment with inhibitors of VEGF/VEGFR agents in animal models of endometriosis, the capability to reduce the endometriotic lesion sizes without influencing the number of follicles was shown [154]. Similarly, a surgically induced endometriosis animal model demonstrated that suppression of the isoform of VEGF, notably VEGFC, can also suppress angiogenesis and the growth of endometriotic lesions [155,156]. In another study, melatonin was found to decrease the VEGF expression level for anti-angiogenic activity for reducing implant volumes in an endometriosis model [51]. Additionally, melatonin was also shown to regulate VEGF and VEGFR expression in physiological and pathological situations. In a breast cancer xenograft model, melatonin suppressed the expression of VEGFR-2, the most critical receptor of VEGF, to significantly reduce vascular density and angiogenesis [157], while in HUVECs, melatonin showed a dose-dependent inhibition of VEGFR-2 induced by VEGF [158]. In addition, oral administration of melatonin decreased serum VEGF levels in patients with metastatic cancer [159]. Additionally, in MCF-7 cells, melatonin treatment reduced VEGF-C and VEGF receptors (VEGFR2 and VEGFR3) during hypoxia [134]. Several in vitro cancer studies demonstrated that melatonin interfered with hypoxia-inducible factor-1a (HIF-1α), which is the transcription factor and an upstream of VEGF. In a mouse model of renal cancer, the effect of melatonin on the expression of HIF-1α and VEGF was determined to be dependent on its antioxidant activity [159]. Likewise, melatonin reduced the growth of ectopic uterine tissue in an endometriosis rat model through the modulation of VEGF [51]. Furthermore, two vascular growth factors, angiopoietin-1 (Ang-1) and Ang-2, were noted to play crucial roles in angiogenesis. Their expressions in the eutopic endometrium of women with endometriosis were found to be increased when compared to normal uterine endometrium [160]. Melatonin lowered levels of ANG-1, ANG-2, and VEGF in the co-culture medium and their respective mRNA expressions in HUVEC and MCF-7 cell lines, respectively [45]. Unlike other angiogenic inhibitors solely targeting specific molecules, melatonin would act to alter several important angiogenic factors and processes to suppress vascularization in various diseases.

3.7. Immune Modulation

The immune system plays an important role in the pathophysiology and symptomatology of endometriosis [161,162]. Both the peripheral immune system and the eutopic endometrial immune situation were altered in women with endometriosis [163,164]. Unlike healthy individuals, macrophages (Mø), immature dendritic cells (DC), and regulatory T cells displayed different behaviors in women with endometriosis [165]. Besides immune cells, cytokines would also be altered in women suffering from endometriosis. Such cytokines as interleukin (IL) 6, IL-8, and cancer antigen (CA) 125 were shown to be upregulated, while tumor necrosis factor (TNF) α was found to be downregulated in patients with endometriosis. These alternations in the immune system would evidently reduce the immunological surveillance in endometriosis. Hence, the determination of these cytokines can often act as a diagnostic index of endometriosis [166,167]. In a mice study, the numbers of monocytes and natural killer (NK) cells were increased in those treated with either 7 or 14 days of melatonin [168]. Melatonin also enhanced antigen presentation via interaction between macrophages and T-lymphocytes, which can lead to the activation and proliferation of cytotoxic T lymphocytes [169]. In tumor-bearing rats, melatonin also showed its stimulatory potential on lymphocytes, monocytes/macrophages, and NK cells [170]. Recently, a meta-analysis analyzed 31 randomized and non-randomized placebo-controlled clinical trials with a total sample size of 1517 participants. It showed that melatonin supplementation significantly decreased TNF-α and IL-6 levels [171]. Taken together, melatonin has a direct immune regulatory effect in both animals and human studies.

3.8. Neurotrophic Effect

Pain is the most common symptom resulting from endometriosis. Endometriotic lesions cause chronic pelvic pain via compressing or infiltrating the nerves located close to the lesions [172]. Although the underlying mechanisms are elusive, emerging evidence has shown that endometriotic lesions and peritoneal fluid in women with endometriosis exhibit outstanding neurotrophic properties, including the increased expression of new nerve fibers, and the upregulation of various neurotrophies [173]. Brain-derived neurotrophic factor (BDNF), or abrineurin, is a member of the neurotrophic class of growth factors. This is a key regulator of neuronal survival and neurogenesis [174]. As reported, BDNF has the capacity to modulate pain pathways at different levels, from the peripheral nociceptors to spinal cord neurons and the brain [175]. A phase II clinical trial demonstrated that melatonin downregulated the secretion of BDNF to reduce pain scores as indexed by the visual analog scale (VAS) that includes daily pain, dysmenorrhea, dyspareunia, dysuria, and dyschezia. In addition, this study showed that melatonin treatment could be associated with an improved sleeping quality in patients with endometriosis [18]. Typically, melatonin has been suggested to be a well-tolerated alternative treatment for the pain-associated symptoms of endometriosis. In addition, melatonin possesses neuroprotective functions and is often considered as a potential neurological drug in clinics, such that treatment with melatonin in newborn piglets increased hypothermic neuroprotection by improving brain energy metabolism and reducing brain damage [176]. It reduced the expression of astrogliosis, whose accumulation would trigger the production of pro-inflammatory cytokines leading to secondary damage [177]. In rat experimental models of neuropathic conditions, which include Alzheimer’s disease, Parkinson’s disease, and ischemic stroke, melatonin showed its effectiveness in arresting neurodegenerative phenomena [178]. Furthermore, during the dark phase, it appeared that mice with a higher melatonin level in plasma were less susceptible to a nociceptive stimulus [179]. The analgesic effects of melatonin were undeniable and evident, which had been documented in various animal models and clinical trials of patients with different pain syndromes. Animals treated with melatonin demonstrated its antinociceptive effect, thus alleviating the hypersensitivity induced in the hot plate test, electrical stimulation, and nerve ligation [180,181,182]. In clinical studies, melatonin reduced the frequency and intensity of pain in migraines, cluster headaches, tension headaches, chronic back pain, and fibromyalgia [183,184,185,186,187]. Considering the therapeutic application of melatonin from both its neuroprotective and antinociceptive functions, melatonin should be considered as a potential drug for treating endometriosis-associated pain.

4. Clinical Insights of Melatonin in Endometriosis and Other Reproductive Diseases

Currently, there are many ongoing clinical studies on using melatonin as a therapeutic strategy, including insomnia, diabetes, fibromyalgia, hypertension, anticancer, and aging-related diseases, such as Parkinson’s and Alzheimer’s disease [188,189,190,191]. In the obstetrics and gynecology discipline, there was only one randomized controlled trial in melatonin for endometriosis in 2013. For other reproductive disorders, melatonin has shown its clinical therapeutic values in various aspects (Table 2). Based on a clinical trial, it was shown that melatonin supplements could relieve pelvic pain and improve the sleeping quality of women with endometriosis [18]. During application for IVF-ET with infertile patients, the administration of melatonin can improve the oocyte quality [192]. The number of degenerated oocytes was significantly reduced in the 3 mg melatonin group when compared to those in the control group [193]. Additionally, the number of fertilized embryos was also shown to have an increased tendency in the 3 mg melatonin group [194]. Likewise, intrafollicular melatonin concentrations were slightly increased in the 1 mg melatonin group, and concentrations were further increased in the 3 mg melatonin group. After treatment of melatonin, the concentration of intrafollicular lipid peroxide, a free-radical reaction that causes injury to oocytes, showed a tendency towards a lower level, the level of significance being dependent on melatonin dosage [195]. Melatonin increased follicular concentrations of it, thereby inhibiting oxidative stress and improving the quality of oocytes [195]. A further study also examined the possibility of whether melatonin therapy inhibited oxidative stress to improve the quality of oocytes and the clinical outcome of IVF-ET [196]. The result in this study reported improved concurrent fertilization and pregnancy rates with melatonin treatment [196]. These initial studies inspired other researchers to examine whether the use of melatonin can help to increase the number of mature oocytes for improving the fertilization rate and the clinical outcome of IVF-ET. In general, melatonin supplementation was shown to increase the number and quality of both oocytes and embryos. Moreover, it could increase the clinical pregnancy rates of patients with infertility disorders or low fertility rates [47,192,195,197]. As poor oocyte quality is a major cause of infertility in endometriosis, whether melatonin can increase the fertility rate through improving oocyte qualities requires further studies. In addition, treatment with melatonin was able to restore menstrual cyclicity in women with polycystic ovarian syndrome (PCOS) [198]. Daily supplementation of melatonin in PCOS women showed a significant improvement in pregnancy rates [199]. Importantly, the overall effects of melatonin supplementation in fetal growth restriction, pre-eclampsia, and climacteric symptoms appear to be promising and clinically relevant [200,201,202]. Likewise, women with endometriosis also have menstrual and poor obstetrics outcomes as summarized in Table 2.
Therapeutic strategies that are safe and with minimal reproductive side effects are essential to women with endometriosis. Melatonin is believed to be a potential candidate as a safe supplementation toward treating endometriosis and endometriosis-associated symptoms [210]. Owing to the limited clinical trials of melatonin in endometriosis, the other clinical trials utilizing melatonin as a potential therapeutic solution on other reproductive diseases may help to support its application toward endometriosis.

5. Conclusions

Importantly, this review has helped to indicate the potential therapeutic capabilities of melatonin for the treatment of endometriosis and its associated symptoms. This is attributed to its many properties, including antioxidation and anti-inflammation, its ability to modulate the endocrine functions via hormonal signaling pathways, and the absence of toxic side effects. Additionally, there is accumulating evidence which illustrates the application of exogenous melatonin to suppress ectopic endometriotic lesions, relieve endometriosis-associated pelvic pain, and enhance the sleeping quality of women with endometriosis. Nevertheless, further analysis on determining the melatonin levels in the eutopic and ectopic endometrium of women with endometriosis for therapeutic efficacy is still warranted. Moreover, corroborating evidence from mechanistic studies and randomized controlled trials would be necessary for better understanding the pharmakinetics and the clinical application of melatonin for endometriosis treatment.

Author Contributions

Conceptualization, Y.L., T.Z. and C.-C.W.; data curation, Y.L. and R.Z.; writing—original draft preparation, Y.L. and S.-W.H.; writing—review and editing, J.P.-W.C., G.C.-W.M., L.F. and C.-C.W. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Cavallo, A. Melatonin and human puberty: Current perspectives. J. Pineal Res. 1993, 15, 115–121. [Google Scholar] [CrossRef]
  2. Cagnacci, A. Melatonin in relation to physiology in adult humans. J. Pineal Res. 1996, 21, 200–213. [Google Scholar] [CrossRef] [PubMed]
  3. Malpaux, B.; Daveau, A.; Maurice-Mandon, F.; Duarte, G.; Chemineau, P. Evidence That Melatonin Acts in the Premammillary Hypothalamic Area to Control Reproduction in the Ewe: Presence of Binding Sites and Stimulation of Luteinizing Hormone Secretion by in Situ Microimplant Delivery. Endocrinology 1998, 139, 1508–1516. [Google Scholar] [CrossRef]
  4. Gingerich, S.; Wang, X.; Lee, P.; Dhillon, S.; Chalmers, J.; Koletar, M.; Belsham, D. The generation of an array of clonal, immortalized cell models from the rat hypothalamus: Analysis of melatonin effects on kisspeptin and gonadotropin-inhibitory hormone neurons. Neuroscience 2009, 162, 1134–1140. [Google Scholar] [CrossRef]
  5. Acuña-Castroviejo, D.; Escames, G.; Venegas, C.; Díaz-Casado, M.E.; Lima-Cabello, E.; Lopez, L.C.; Rosales-Corral, S.; Tan, D.-X.; Reiter, R.J. Extrapineal melatonin: Sources, regulation, and potential functions. Cell. Mol. Life Sci. 2014, 71, 2997–3025. [Google Scholar] [CrossRef] [PubMed]
  6. Pang, S.; Li, L.; Ayre, E.; Pang, C.; Lee, P.; Xu, R.; Chow, P.; Yu, Z.; Shiu, S. Neuroendocrinology of melatonin in reproduction: Recent developments. J. Chem. Neuroanat. 1998, 14, 157–166. [Google Scholar] [CrossRef]
  7. Yellon, S.M.; Longo, L.D. Melatonin rhythms in fetal and maternal circulation during pregnancy in sheep. Am. J. Physiol. Metab. 1987, 252, E799–E802. [Google Scholar] [CrossRef] [PubMed]
  8. Showell, M.G.; Mackenzie-Proctor, R.; Jordan, V.; Hart, R.J. Antioxidants for female subfertility. Cochrane Database Syst. Rev. 2020, 2020, 8. [Google Scholar] [CrossRef]
  9. Hardeland, R.; Cardinali, D.P.; Srinivasan, V.; Spence, D.W.; Brown, G.M.; Pandi-Perumal, S.R. Melatonin—A pleiotropic, orchestrating regulator molecule. Prog. Neurobiol. 2011, 93, 350–384. [Google Scholar] [CrossRef] [Green Version]
  10. Bulun, S.; Yilmaz, B.D.; Sison, C.; Miyazaki, K.; Bernardi, L.; Liu, S.; Kohlmeier, A.; Yin, P.; Milad, M.; Wei, J. Endometriosis. Endocr. Rev. 2019, 40, 1048–1079. [Google Scholar] [CrossRef]
  11. Horne, A.W.; Saunders, P. SnapShot: Endometriosis. Cell 2019, 179, 1677–1677.e1. [Google Scholar] [CrossRef] [PubMed]
  12. Zondervan, K.T.; Becker, C.M.; Missmer, S.A. Endometriosis. N. Engl. J. Med. 2020, 382, 1244–1256. [Google Scholar] [CrossRef]
  13. Vercellini, P.; Viganò, P.; Somigliana, E.; Fedele, L. Endometriosis: Pathogenesis and treatment. Nat. Rev. Endocrinol. 2014, 10, 261–275. [Google Scholar] [CrossRef] [PubMed]
  14. Greene, R.; Stratton, P.; Cleary, S.D.; Ballweg, M.L.; Sinaii, N. Diagnostic experience among 4334 women reporting surgically diagnosed endometriosis. Fertil. Steril. 2009, 91, 32–39. [Google Scholar] [CrossRef] [PubMed]
  15. Ferrero, S.; Evangelisti, G.; Barra, F. Current and emerging treatment options for endometriosis. Expert Opin. Pharmacother. 2018, 19, 1109–1125. [Google Scholar] [CrossRef] [PubMed]
  16. Dunselman, G.A.J.; Vermeulen, N.; Becker, C.; Calhaz-Jorge, C.; D’Hooghe, T.; De Bie, B.; Heikinheimo, O.; Horne, A.W.; Kiesel, L.; Nap, A.; et al. ESHRE guideline: Management of women with endometriosis. Hum. Reprod. 2014, 29, 400–412. [Google Scholar] [CrossRef] [PubMed]
  17. Hung, S.W.; Zhang, R.; Tan, Z.; Chung, J.P.W.; Zhang, T.; Wang, C.C. Pharmaceuticals targeting signaling pathways of endometriosis as potential new medical treatment: A review. Med. Res. Rev. 2021, 41, 2489–2564. [Google Scholar] [CrossRef]
  18. Schwertner, A.; Dos Santos, C.C.C.; Costa, G.D.; Deitos, A.; Souza, A.; Custodio-De-Souza, I.-C.; Torres, I.L.; Cunha-Filho, J.; Caumo, W. Efficacy of melatonin in the treatment of endometriosis: A phase II, randomized, double-blind, placebo-controlled trial. Pain 2013, 154, 874–881. [Google Scholar] [CrossRef]
  19. Mannino, G.; Pernici, C.; Serio, G.; Gentile, C.; Bertea, C.M. Melatonin and Phytomelatonin: Chemistry, Biosynthesis, Metabolism, Distribution and Bioactivity in Plants and Animals—An Overview. Int. J. Mol. Sci. 2021, 22, 9996. [Google Scholar] [CrossRef]
  20. Pandi-Perumal, S.R.; Srinivasan, V.; Maestroni, G.J.M.; Cardinali, D.P.; Poeggeler, B.; Hardeland, R. Melatonin: Nature’s most versatile biological signal? FEBS J. 2006, 273, 2813–2838. [Google Scholar] [CrossRef]
  21. Kvetnoy, I.M. Extrapineal Melatonin: Location and Role within Diffuse Neuroendocrine System. Histochem. J. 1999, 31, 1–12. [Google Scholar] [CrossRef]
  22. Ekmekcioglu, C. Melatonin receptors in humans: Biological role and clinical relevance. Biomed. Pharmacother. 2006, 60, 97–108. [Google Scholar] [CrossRef]
  23. Yu, H.; Dickson, E.J.; Jung, S.-R.; Koh, D.-S.; Hille, B. High membrane permeability for melatonin. J. Gen. Physiol. 2015, 147, 63–76. [Google Scholar] [CrossRef] [Green Version]
  24. Ma, X.; Idle, J.R.; Krausz, K.W.; Gonzalez, F.J. Metabolism of melatonin by human cytochromes p450. Drug Metab. Dispos. 2005, 33, 489–494. [Google Scholar] [CrossRef]
  25. Harpsøe, N.G.; Andersen, L.P.H.; Gögenur, I.; Rosenberg, J. Clinical pharmacokinetics of melatonin: A systematic review. Eur. J. Clin. Pharmacol. 2015, 71, 901–909. [Google Scholar] [CrossRef]
  26. Dubocovich, M.L.; Delagrange, P.; Krause, D.N.; Sugden, D.; Cardinali, D.P.; Olcese, J. International Union of Basic and Clinical Pharmacology. LXXV. Nomenclature, Classification, and Pharmacology of G Protein-Coupled Melatonin Receptors. Pharmacol. Rev. 2010, 62, 343–380. [Google Scholar] [CrossRef] [Green Version]
  27. Mazzucchelli, C.; Pannacci, M.; Nonno, R.; Lucini, V.; Fraschini, F.; Stankov, B.M. The melatonin receptor in the human brain: Cloning experiments and distribution studies. Mol. Brain Res. 1996, 39, 117–126. [Google Scholar] [CrossRef]
  28. Weaver, D.R.; Reppert, S.M. The Mel1a melatonin receptor gene is expressed in human suprachiasmatic nuclei. NeuroReport 1996, 8, 109–112. [Google Scholar] [CrossRef]
  29. Viswanathan, M.; Laitinen, J.T.; Saavedra, J.M. Expression of melatonin receptors in arteries involved in thermoregulation. Proc. Natl. Acad. Sci. USA 1990, 87, 6200–6203. [Google Scholar] [CrossRef] [Green Version]
  30. Konturek, S.J.; Konturek, P.C.; Brzozowska, I.; Pawlik, M.; Sliwowski, Z.; Cześnikiewicz-Guzik, M.; Kwiecień, S.; Brzozowski, T.; Bubenik, G.; Pawlik, W.W. Localization and biological activities of melatonin in intact and diseased gastrointestinal tract (GIT). J. Physiol. Pharmacol. Off. J. Pol. Physiol. Soc. 2007, 58, 381–405. [Google Scholar]
  31. Drew, J.; Williams, L.; Hannah, L.; Barrett, P. Abramovich Melatonin receptors in the human fetal kidney: 2-[125I]iodomelatonin binding sites correlated with expression of Mel1a and Mel1b receptor genes. J. Endocrinol. 1998, 156, 261–267. [Google Scholar] [CrossRef] [Green Version]
  32. Slominski, A.; Tobin, D.; Zmijewski, M.; Wortsman, J.; Paus, R. Melatonin in the skin: Synthesis, metabolism and functions. Trends Endocrinol. Metab. 2008, 19, 17–24. [Google Scholar] [CrossRef]
  33. Iwasaki, S.; Nakazawa, K.; Sakai, J.; Kometani, K.; Iwashita, M.; Yoshimura, Y.; Maruyama, T. Melatonin as a local regulator of human placental function. J. Pineal Res. 2005, 39, 261–265. [Google Scholar] [CrossRef]
  34. Schlabritz-Loutsevitch, N.; Hellner, N.; Middendorf, R.; Muller, D.; Olcese, J. The Human Myometrium as a Target for Melatonin. J. Clin. Endocrinol. Metab. 2003, 88, 908–913. [Google Scholar] [CrossRef] [Green Version]
  35. Kobayashi, Y.; Itoh, M.T.; Kondo, H.; Okuma, Y.; Sato, S.; Kanishi, Y.; Hamada, N.; Kiguchi, K.; Ishizuka, B. Melatonin binding sites in estrogen receptor-positive cells derived from human endometrial cancer. J. Pineal Res. 2003, 35, 71–74. [Google Scholar] [CrossRef]
  36. Becker-André, M.; Wiesenberg, I.; Schaeren-Wiemers, N.; André, E.; Missbach, M.; Saurat, J.-H.; Carlberg, C. Pineal gland hormone melatonin binds and activates an orphan of the nuclear receptor superfamily. J. Biol. Chem. 1997, 272, 28531–28534. [Google Scholar] [CrossRef] [Green Version]
  37. Zisapel, N. New perspectives on the role of melatonin in human sleep, circadian rhythms and their regulation. J. Cereb. Blood Flow Metab. 2018, 175, 3190–3199. [Google Scholar] [CrossRef] [Green Version]
  38. Takayama, H.; Nakamura, Y.; Tamura, H.; Yamagata, Y.; Harada, A.; Nakata, M.; Sugino, N.; Kato, H. Pineal Gland(Melatonin) Affects the Parturition Time, but not Luteal Function and Fetal Growth, in Pregnant Rats. Endocr. J. 2003, 50, 37–43. [Google Scholar] [CrossRef] [Green Version]
  39. Sagrillo-Fagundes, L.; Salustiano, E.M.A.; Yen, P.W.; Soliman, A.; Vaillancourt, C. Melatonin in Pregnancy: Effects on Brain Development and CNS Programming Disorders. Curr. Pharm. Des. 2016, 22, 978–986. [Google Scholar] [CrossRef]
  40. Pandi-Perumal, S.R.; Bahammam, A.S.; Ojike, N.I.; Akinseye, O.A.; Kendzerska, T.; Buttoo, K.; Dhandapany, P.S.; Brown, G.M.; Cardinali, D.P. Melatonin and Human Cardiovascular Disease. J. Cardiovasc. Pharmacol. Ther. 2016, 22, 122–132. [Google Scholar] [CrossRef]
  41. Hardeland, R. Melatonin and the electron transport chain. Cell. Mol. Life Sci. 2017, 74, 3883–3896. [Google Scholar] [CrossRef]
  42. Mehrzadi, S.; Safa, M.; Kamrava, S.K.; Darabi, R.; Hayat, P.; Motevalian, M. Protective mechanisms of melatonin against hydrogen-peroxide-induced toxicity in human bone-marrow-derived mesenchymal stem cells. Can. J. Physiol. Pharmacol. 2017, 95, 773–786. [Google Scholar] [CrossRef] [PubMed]
  43. Chuffa, L.G.; Alves, M.; Martinez, M.; Camargo, I.C.C.; Pinheiro, P.F.F.; Domeniconi, R.F.; Júnior, L.A.L.; Martinez, F.E. Apoptosis is triggered by melatonin in an in vivo model of ovarian carcinoma. Endocr. Relat. Cancer 2015, 23, 65–76. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Cuesta, S.; Kireev, R.; Forman, K.; García, C.; Escames, G.; Ariznavarreta, C.; Vara, E.; Tresguerres, J.A. Melatonin improves inflammation processes in liver of senescence-accelerated prone male mice (SAMP8). Exp. Gerontol. 2010, 45, 950–956. [Google Scholar] [CrossRef] [PubMed]
  45. González-González, A.; González, A.; Alonso-González, C.; Menéndez-Menéndez, J.; Martínez-Campa, C.; Cos, S. Complementary actions of melatonin on angiogenic factors, the angiopoietin/Tie2 axis and VEGF, in co-cultures of human endothelial and breast cancer cells. Oncol. Rep. 2017, 39, 433–441. [Google Scholar] [CrossRef]
  46. Tamura, H.; Nakamura, Y.; Terron, M.P.; Flores-Alvarado, L.J.; Manchester, L.C.; Tan, D.-X.; Sugino, N.; Reiter, R.J. Melatonin and pregnancy in the human. Reprod. Toxicol. 2008, 25, 291–303. [Google Scholar] [CrossRef]
  47. Tamura, H.; Takasaki, A.; Miwa, I.; Taniguchi, K.; Maekawa, R.; Asada, H.; Taketani, T.; Matsuoka, A.; Yamagata, Y.; Shimamura, K.; et al. Oxidative stress impairs oocyte quality and melatonin protects oocytes from free radical damage and improves fertilization rate. J. Pineal Res. 2007, 44, 280–287. [Google Scholar] [CrossRef]
  48. Song, G.; Yoon, K.-A.; Chi, H.; Roh, J.; Kim, J.-H. Decreased concentration of serum melatonin in nighttime compared with daytime female medical technologists in South Korea. Chronobiol. Int. 2016, 33, 1305–1310. [Google Scholar] [CrossRef]
  49. Marino, J.L.; Holt, V.L.; Chen, C.; Davis, S. Shift Work, hCLOCK T3111C Polymorphism, and Endometriosis Risk. Epidemiology 2008, 19, 477–484. [Google Scholar] [CrossRef] [Green Version]
  50. Cetinkaya, N.; Attar, R.; Yildirim, G.; Ficicioglu, C.; Ozkan, F.; Yilmaz, B.; Yesildaglar, N. The effects of different doses of melatonin treatment on endometrial implants in an oophorectomized rat endometriosis model. Arch. Gynecol. Obstet. 2014, 291, 591–598. [Google Scholar] [CrossRef]
  51. Yilmaz, B.; Kilic, S.; Aksakal, O.; Ertas, I.E.; Tanrisever, G.G.; Aksoy, Y.; Lortlar, N.; Kelekci, S.; Gungor, T. Melatonin causes regression of endometriotic implants in rats by modulating angiogenesis, tissue levels of antioxidants and matrix metalloproteinases. Arch. Gynecol. Obstet. 2014, 292, 209–216. [Google Scholar] [CrossRef] [PubMed]
  52. Yildirim, G.; Attar, R.; Ozkan, F.; Kumbak, B.; Ficicioglu, C.; Yesildaglar, N. The effects of letrozole and melatonin on surgically induced endometriosis in a rat model: A preliminary study. Fertil. Steril. 2010, 93, 1787–1792. [Google Scholar] [CrossRef] [PubMed]
  53. Mosher, A.A.; Tsoulis, M.W.; Lim, J.; Tan, C.; Agarwal, S.; Leyland, N.A.; Foster, W.G. Melatonin activity and receptor expression in endometrial tissue and endometriosis. Hum. Reprod. 2019, 34, 1215–1224. [Google Scholar] [CrossRef] [PubMed]
  54. Barchas, J.; Dacosta, F.; Spector, S. Acute Pharmacology of Melatonin. Nature 1967, 214, 919–920. [Google Scholar] [CrossRef]
  55. Cavalcante, A.G.D.M.; De Bruin, P.F.C.; De Bruin, V.M.S.; Nunes, D.M.; Pereira, E.D.B.; Cavalcante, M.M.; Andrade, G.M. Melatonin reduces lung oxidative stress in patients with chronic obstructive pulmonary disease: A randomized, double-blind, placebo-controlled study. J. Pineal Res. 2012, 53, 238–244. [Google Scholar] [CrossRef]
  56. Nickkholgh, A.; Schneider, H.; Sobirey, M.; Venetz, W.P.; Hinz, U.; Pelzl, L.H.; Gotthardt, D.N.; Cekauskas, A.; Manikas, M.; Mikalauskas, S.; et al. The use of high-dose melatonin in liver resection is safe: First clinical experience. J. Pineal Res. 2011, 50, 381–388. [Google Scholar] [CrossRef]
  57. Genario, R.; Morello, E.; Bueno, A.A.; Santos, H.O. The usefulness of melatonin in the field of obstetrics and gynecology. Pharmacol. Res. 2019, 147, 104337. [Google Scholar] [CrossRef]
  58. Güney, M.; Oral, B.; Karahan, N.; Mungan, T. Regression of endometrial explants in a rat model of endometriosis treated with melatonin. Fertil. Steril. 2008, 89, 934–942. [Google Scholar] [CrossRef]
  59. Paul, S.; Sharma, A.V.; Das Mahapatra, P.; Bhattacharya, P.; Reiter, R.J.; Swarnakar, S. Role of melatonin in regulating matrix metalloproteinase-9 via tissue inhibitors of metalloproteinase-1 during protection against endometriosis. J. Pineal Res. 2008, 44, 439–449. [Google Scholar] [CrossRef]
  60. Paul, S.; Bhattacharya, P.; Das Mahapatra, P.; Swarnakar, S. Melatonin protects against endometriosis via regulation of matrix metalloproteinase-3 and an apoptotic pathway. J. Pineal Res. 2010, 49, 156–168. [Google Scholar] [CrossRef]
  61. Yıldırım, G.; Attar, R.; Fıçıcıoğlu, C.; Karateke, A.; Ozkan, F.; Kılıç, E.; Yılmaz, B.; Yeşildağlar, N. The combination of letrozole and melatonin causes regression in size not histopathological scores on endometriosis in an experimental rat model. J. Turk. Gynecol. Assoc. 2009, 10, 199–204. [Google Scholar]
  62. Koc, O.; Gunduz, B.; Topcuoglu, A.; Bugdayci, G.; Yilmaz, F.; Duran, B. Effects of pinealectomy and melatonin supplementation on endometrial explants in a rat model. Eur. J. Obstet. Gynecol. Reprod. Biol. 2010, 153, 72–76. [Google Scholar] [CrossRef]
  63. Kocadal, N.C.; Attar, R.; Yıldırım, G.; Fıçıcıoğlu, C.; Ozkan, F.; Yılmaz, B.; Yesildaglar, N. Melatonin treatment results in regression of endometriotic lesions in an ooferectomized rat endometriosis model. J. Turk. Gynecol. Assoc. 2013, 14, 81–86. [Google Scholar] [CrossRef] [PubMed]
  64. Qi, S.; Yan, L.; Liu, Z.; Mu, Y.-L.; Li, M.; Zhao, X.; Chen, Z.-J.; Zhang, H. Melatonin inhibits 17β-estradiol-induced migration, invasion and epithelial-mesenchymal transition in normal and endometriotic endometrial epithelial cells. Reprod. Biol. Endocrinol. 2018, 16, 62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Scutiero, G.; Iannone, P.; Bernardi, G.; Bonaccorsi, G.; Spadaro, S.; Volta, C.A.; Greco, P.; Nappi, L. Oxidative Stress and Endometriosis: A Systematic Review of the Literature. Oxid. Med. Cell. Longev. 2017, 2017, 7265238. [Google Scholar] [CrossRef]
  66. Donnez, J.; Binda, M.M.; Donnez, O.; Dolmans, M.-M. Oxidative stress in the pelvic cavity and its role in the pathogenesis of endometriosis. Fertil. Steril. 2016, 106, 1011–1017. [Google Scholar] [CrossRef] [Green Version]
  67. Yesildaglar, N.; Yıldırım, G.Y.; Attar, R.; Ozkan, F.; Akkaya, H.; Yilmaz, B. The effects of melatonin on endometriotic lesions induced by implanting human endometriotic cells in the first SCID-mouse endometriosis-model developed in Turkey. Clin. Exp. Obstet. Gynecol. 2016, 43, 25–30. [Google Scholar] [CrossRef]
  68. Hardeland, R. Antioxidative Protection by Melatonin: Multiplicity of Mechanisms from Radical Detoxification to Radical Avoidance. Endocrine 2005, 27, 119–130. [Google Scholar] [CrossRef]
  69. Tan, D.-X.; Manchester, L.C.; Terron, M.P.; Flores-Alvarado, L.J.; Reiter, R.J. One molecule, many derivatives: A never-ending interaction of melatonin with reactive oxygen and nitrogen species? J. Pineal Res. 2006, 42, 28–42. [Google Scholar] [CrossRef]
  70. Richter, H.G.; Hansell, J.A.; Raut, S.; Giussani, D.A. Melatonin improves placental efficiency and birth weight and increases the placental expression of antioxidant enzymes in undernourished pregnancy. J. Pineal Res. 2009, 46, 357–364. [Google Scholar] [CrossRef]
  71. Mori, T.; Ito, F.; Koshiba, A.; Kataoka, H.; Takaoka, O.; Okimura, H.; Khan, K.N.; Kitawaki, J. Local estrogen formation and its regulation in endometriosis. Reprod. Med. Biol. 2019, 18, 305–311. [Google Scholar] [CrossRef] [PubMed]
  72. Han, S.J.; O’Malley, B.W. The dynamics of nuclear receptors and nuclear receptor coregulators in the pathogenesis of endometriosis. Hum. Reprod. Updat. 2014, 20, 467–484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Tang, Z.-R.; Zhang, R.; Lian, Z.-X.; Deng, S.-L.; Yu, K. Estrogen-Receptor Expression and Function in Female Reproductive Disease. Cells 2019, 8, 1123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Pellegrini, C.; Gori, I.; Achtari, C.; Hornung, D.; Chardonnens, E.; Wunder, D.; Fiche, M.; Canny, G.O. The expression of estrogen receptors as well as GREB1, c-MYC, and cyclin D1, estrogen-regulated genes implicated in proliferation, is increased in peritoneal endometriosis. Fertil. Steril. 2012, 98, 1200–1208. [Google Scholar] [CrossRef] [PubMed]
  75. Chantalat, E.; Valera, M.-C.; Vaysse, C.; Noirrit, E.; Rusidze, M.; Weyl, A.; Vergriete, K.; Buscail, E.; Lluel, P.; Fontaine, C.; et al. Estrogen Receptors and Endometriosis. Int. J. Mol. Sci. 2020, 21, 2815. [Google Scholar] [CrossRef] [Green Version]
  76. Patel, B.G.; Rudnicki, M.; Yu, J.; Shu, Y.; Taylor, R.N. Progesterone resistance in endometriosis: Origins, consequences and interventions. Acta Obstet. Gynecol. Scand. 2017, 96, 623–632. [Google Scholar] [CrossRef] [Green Version]
  77. Attia, G.R.; Zeitoun, K.; Edwards, D.; Johns, A.; Carr, B.R.; Bulun, S.E. Progesterone Receptor Isoform A But Not B Is Expressed in Endometriosis. J. Clin. Endocrinol. Metab. 2000, 85, 2897–2902. [Google Scholar] [CrossRef] [Green Version]
  78. Flores, V.; Vanhie, A.; Dang, T.; Taylor, H.S. Progesterone Receptor Status Predicts Response to Progestin Therapy in Endometriosis. J. Clin. Endocrinol. Metab. 2018, 103, 4561–4568. [Google Scholar] [CrossRef] [Green Version]
  79. Liang, B.; Wu, L.; Xu, H.; Cheung, C.W.; Fung, W.Y.; Wong, S.W.; Wang, C.C. Efficacy, safety and recurrence of new progestins and selective progesterone receptor modulator for the treatment of endometriosis: A comparison study in mice. Reprod. Biol. Endocrinol. 2018, 16, 32. [Google Scholar] [CrossRef]
  80. Cipolla-Neto, J.; Amaral, F.G.; Soares, J.M., Jr.; Gallo, C.C.; Furtado, A.; Cavaco, J.E.; Gonçalves, I.; Santos, C.R.A.; Quintela, T. The crosstalk between melatonin and sex steroid hormones. Neuroendocrinology 2021, 112, 115–129. [Google Scholar] [CrossRef]
  81. Tamura, H.; Nakamura, Y.; Takiguchi, S.; Kashida, S.; Yamagata, Y.; Sugino, N.; Kato, H. Melatonin directly suppresses steroid production by preovulatory follicles in the cyclic hamster. J. Pineal Res. 1998, 25, 135–141. [Google Scholar] [CrossRef] [PubMed]
  82. Chuffa, L.G.; Seiva, F.R.; Fávaro, W.J.; Teixeira, G.R.; Amorim, J.P.; Mendes, L.O.; A Fioruci, B.; Pinheiro, P.F.F.; Fernandes, A.A.H.; Franci, J.A.; et al. Melatonin reduces LH, 17 beta-estradiol and induces differential regulation of sex steroid receptors in reproductive tissues during rat ovulation. Reprod. Biol. Endocrinol. 2011, 9, 108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Voordouw, B.C.; Euser, R.; Verdonk, R.E.; Alberda, B.T.; De Jong, F.H.; Drogendijk, A.C.; Fauser, B.C.; 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. [Google Scholar] [CrossRef] [PubMed]
  84. Cos, S.; Sánchez-Barceló, E.J. Melatonin, experimental basis for a possible application in breast cancer prevention and treatment. Histol. Histopathol. 2000, 15, 637–647. [Google Scholar] [CrossRef] [PubMed]
  85. González, A. Melatonin: An Endogenous Antiestrogen with Oncostatic Properties. In Melatonin From Molecules to Therapy; Nova Science Publishers Inc.: New York, NY, USA, 2007; pp. 261–272. [Google Scholar]
  86. Hill, S.M.; Spriggs, L.L.; Simon, M.A.; Muraoka, H.; Blask, D.E. The growth inhibitory action of melatonin on human breast cancer cells is linked to the estrogen response system. Cancer Lett. 1992, 64, 249–256. [Google Scholar] [CrossRef]
  87. Bódis, J.; Koppán, M.; Kornya, L.; Tinneberg, H.; Török, A. Influence of Melatonin on Basal and Gonadotropin-Stimulated Progesterone and Estradiol Secretion of Cultured Human Granulosa Cells and in the Superfused Granulosa Cell System. Gynecol. Obstet. Investig. 2001, 52, 198–202. [Google Scholar] [CrossRef]
  88. Cheng, J.-C.; Fang, L.; Li, Y.; Wang, S.; Li, Y.; Yan, Y.; Jia, Q.; Wu, Z.; Wang, Z.; Han, X.; et al. Melatonin stimulates aromatase expression and estradiol production in human granulosa-lutein cells: Relevance for high serum estradiol levels in patients with ovarian hyperstimulation syndrome. Exp. Mol. Med. 2020, 52, 1341–1350. [Google Scholar] [CrossRef]
  89. Chen, S.; Besman, M.J.; Sparkes, R.S.; Zollman, S.; Klisak, I.; Mohandas, T.; Hall, P.F.; Shively, J.E. Human Aromatase: cDNA Cloning, Southern Blot Analysis, and Assignment of the Gene to Chromosome 15. DNA 1988, 7, 27–38. [Google Scholar] [CrossRef]
  90. González, A.; Martínez-Campa, C.; Mediavilla, M.D.; Alonso-González, C.; Sánchez-Mateos, S.; Hill, S.M.; Sánchez-Barceló, E.J.; Cos, S. Effects of MT1 melatonin receptor overexpression on the aromatase-suppressive effect of melatonin in MCF-7 human breast cancer cells. Oncol. Rep. 2007, 17, 947–953. [Google Scholar] [CrossRef]
  91. Martínez-Campa, C.M.; González, A.; Mediavilla, M.D.; Alonso-Gonzalez, C.; Alvarezgarcia, V.; Sanchez-Barcelo, E.J.; Cos, S. Melatonin inhibits aromatase promoter expression by regulating cyclooxygenases expression and activity in breast cancer cells. Br. J. Cancer 2009, 101, 1613–1619. [Google Scholar] [CrossRef] [Green Version]
  92. Alvarez-García, V.; González, A.; Martínez-Campa, C.; Alonso-González, C.; Cos, S. Melatonin modulates aromatase activity and expression in endothelial cells. Oncol. Rep. 2013, 29, 2058–2064. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Gonzalez, A.; Martínez-Campa, C.M.; Mediavilla, M.D.; Alonso-Gonzalez, C.; Sanchez-Barcelo, E.J.; Cos, S. Inhibitory effects of pharmacological doses of melatonin on aromatase activity and expression in rat glioma cells. Br. J. Cancer 2007, 97, 755–760. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Simpson, E.R.; Misso, M.; Hewitt, K.N.; Hill, R.; Boon, W.C.; Jones, M.; Kovacic, A.; Zhou, J.; Clyne, C.D. Estrogen—The Good, the Bad, and the Unexpected. Endocr. Rev. 2005, 26, 322–330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Chen, D.; Reierstad, S.; Lu, M.; Lin, Z.; Ishikawa, H.; Bulun, S.E. Regulation of breast cancer-associated aromatase promoters. Cancer Lett. 2009, 273, 15–27. [Google Scholar] [CrossRef]
  96. Dardes, R.C.; Baracat, E.C.; Simões, M.J. Modulation of estrous cycle and LH, FSH and melatonin levels by pinealectomy and sham-pinealectomy in female rats. Prog. Neuro Psychopharmacol. Biol. Psychiatry 2000, 24, 441–453. [Google Scholar] [CrossRef]
  97. Webley, G.E.; Luck, M.R. Melatonin directly stimulates the secretion of progesterone by human and bovine granulosa cells in vitro. Reproduction 1986, 78, 711–717. [Google Scholar] [CrossRef] [Green Version]
  98. Fang, L.; Li, Y.; Wang, S.; Yu, Y.; Li, Y.; Guo, Y.; Yan, Y.; Sun, Y.-P. Melatonin induces progesterone production in human granulosa-lutein cells through upregulation of StAR expression. Aging 2019, 11, 9013–9024. [Google Scholar] [CrossRef]
  99. Schaeffer, H.-J.; Sirotkin, A.V. Melatonin and serotonin regulate the release of insulin-like growth factor-I, oxytocin and progesterone by cultured human granulosa cells. Exp. Clin. Endocrinol. Diabetes 1997, 105, 109–112. [Google Scholar] [CrossRef]
  100. Woo, M.M.M.; Tai, C.-J.; Kang, S.K.; Nathwani, P.S.; Pang, S.F.; Leung, P. Direct Action of Melatonin in Human Granulosa-Luteal Cells. J. Clin. Endocrinol. Metab. 2001, 86, 4789–4797. [Google Scholar] [CrossRef]
  101. Taketani, T.; Tamura, H.; Takasaki, A.; Lee, L.; Kizuka, F.; Tamura, I.; Taniguchi, K.; Maekawa, R.; Asada, H.; Shimamura, K.; et al. Protective role of melatonin in progesterone production by human luteal cells. J. Pineal Res. 2011, 51, 207–213. [Google Scholar] [CrossRef]
  102. Wang, X.; Meng, K.; He, Y.; Wang, H.; Zhang, Y.; Quan, F. Melatonin Stimulates STAR Expression and Progesterone Production via Activation of the PI3K/AKT Pathway in Bovine Theca Cells. Int. J. Biol. Sci. 2019, 15, 404–415. [Google Scholar] [CrossRef] [PubMed]
  103. Yang, M.; Guan, S.; Tao, J.; Zhu, K.; Lv, D.; Wang, J.; Li, G.; Gao, Y.; Wu, H.; Liu, J.; et al. Melatonin promotes male reproductive performance and increases testosterone synthesis in mammalian Leydig cells. Biol. Reprod. 2021, 104, 1322–1336. [Google Scholar] [CrossRef] [PubMed]
  104. Ezzati, M.; Roshangar, L.; Soleimani, J.; Karimian, N. Evaluating the Effect of Melatonin on HAS2, PGR, Cumulus Expansion, and Fertility Potential in Mouse. Cell J. 2018, 20, 108–112. [Google Scholar] [CrossRef]
  105. Ram, P.; Yuan, L.; Dai, J.; Kiefer, T.; Klotz, D.M.; Spriggs, L.L.; Hill, S.M. Differential responsiveness of MCF-7 human breast cancer cell line stocks to the pineal hormone, melatonin. J. Pineal Res. 2000, 28, 210–218. [Google Scholar] [CrossRef] [PubMed]
  106. Chuffa, L.G.D.A.; Lupi, L.A.; Cucielo, M.S.; Silveira, H.S.; Reiter, R.J.; Seiva, F.R.F. Melatonin Promotes Uterine and Placental Health: Potential Molecular Mechanisms. Int. J. Mol. Sci. 2019, 21, 300. [Google Scholar] [CrossRef] [Green Version]
  107. Sainz, R.M.; Mayo, J.C.; Rodriguez, C.; Tan, D.X.; Lopez-Burillo, S.; Reiter, R.J. Melatonin and cell death: Differential actions on apoptosis in normal and cancer cells. Cell Mol. Life Sci. 2003, 60, 1407–1426. [Google Scholar] [CrossRef]
  108. Vermeulen, K.; Berneman, Z.N.; Van Bockstaele, D.R. Cell cycle and apoptosis. Cell Prolif. 2003, 36, 165–175. [Google Scholar] [CrossRef]
  109. Lu, J.-J.; Fu, L.; Tang, Z.; Zhang, C.; Qin, L.; Wang, J.; Yu, Z.; Shi, D.; Xiao, X.; Xie, F.; et al. Melatonin inhibits AP-2β/hTERT, NF-κB/COX-2 and Akt/ERK and activates caspase/Cyto C signaling to enhance the antitumor activity of berberine in lung cancer cells. Oncotarget 2015, 7, 2985–3001. [Google Scholar] [CrossRef] [Green Version]
  110. Ordoñez, R.; Carbajo-Pescador, S.; Prieto-Domínguez, N.; Palomo, A.G.; González-Gallego, J.; Mauriz, J.L. Inhibition of matrix metalloproteinase-9 and nuclear factor kappa B contribute to melatonin prevention of motility and invasiveness in HepG2 liver cancer cells. J. Pineal Res. 2013, 56, 20–30. [Google Scholar] [CrossRef]
  111. Wang, J.; Xiao, X.; Zhang, Y.; Shi, D.; Chen, W.; Fu, L.; Liu, L.; Xie, F.; Kang, T.; Huang, W.; et al. Simultaneous modulation of COX-2, p300, Akt, and Apaf-1 signaling by melatonin to inhibit proliferation and induce apoptosis in breast cancer cells. J. Pineal Res. 2012, 53, 77–90. [Google Scholar] [CrossRef]
  112. Proietti, S.; Cucina, A.; D’Anselmi, F.; DiNicola, S.; Pasqualato, A.; Lisi, E.; Bizzarri, M. Melatonin and vitamin D3 synergistically down-regulate Akt and MDM2 leading to TGFβ-1-dependent growth inhibition of breast cancer cells. J. Pineal Res. 2010, 50, 150–158. [Google Scholar] [CrossRef] [PubMed]
  113. Pizarro, J.G.; Yeste-Velasco, M.; Esparza, J.L.; Verdaguer, E.; Palls, M.; Camins, A.; Folch, J.; Pallàs, M.; Esparza, J.L. The antiproliferative activity of melatonin in B65 rat dopaminergic neuroblastoma cells is related to the downregulation of cell cycle-related genes. J. Pineal Res. 2008, 45, 8–16. [Google Scholar] [CrossRef] [PubMed]
  114. Cini, G.; Neri, B.; Pacini, A.; Cesati, V.; Sassoli, C.; Quattrone, S.; D’Apolito, M.; Fazio, A.; Scapagnini, G.; Provenzani, A.; et al. Antiproliferative activity of melatonin by transcriptional inhibition of cyclin D1 expression: A molecular basis for melatonin-induced oncostatic effects. J. Pineal Res. 2005, 39, 12–20. [Google Scholar] [CrossRef] [PubMed]
  115. Liu, L.; Xu, Y.; Reiter, R.J. Melatonin inhibits the proliferation of human osteosarcoma cell line MG-63. Bone 2013, 55, 432–438. [Google Scholar] [CrossRef] [PubMed]
  116. Mortezaee, K.; Najafi, M.; Farhood, B.; Ahmadi, A.; Potes, Y.; Shabeeb, D.; Musa, A.E. Modulation of apoptosis by melatonin for improving cancer treatment efficiency: An updated review. Life Sci. 2019, 228, 228–241. [Google Scholar] [CrossRef] [PubMed]
  117. Klemmt, P.A.; Carver, J.G.; Koninckx, P.; McVeigh, E.J.; Mardon, H.J. Endometrial cells from women with endometriosis have increased adhesion and proliferative capacity in response to extracellular matrix components: Towards a mechanistic model for endometriosis progression. Hum. Reprod. 2007, 22, 3139–3147. [Google Scholar] [CrossRef] [Green Version]
  118. Rai, V.; Hopkisson, J.; Kennedy, S.; Bergqvist, A.; Barlow, D.H.; Mardon, H.J. Integrins alpha 3 and alpha 6 are differentially expressed in endometrium and endometriosis. J. Pathol. 1996, 180, 181–187. [Google Scholar] [CrossRef]
  119. Han, Y.-S.; Lee, J.H.; Lee, S.H. Melatonin suppresses ischemia-induced fibrosis by regulating miR-149. Biochem. Biophys. Res. Commun. 2020, 525, 354–359. [Google Scholar] [CrossRef]
  120. Egeblad, M.; Werb, Z. New functions for the matrix metalloproteinases in cancer progression. Nat. Cancer 2002, 2, 161–174. [Google Scholar] [CrossRef]
  121. Chung, H.-W.; Wen, Y.; Chun, S.-H.; Nezhat, C.; Woo, B.-H.; Polan, M.L. Matrix metalloproteinase-9 and tissue inhibitor of metalloproteinase-3 mRNA expression in ectopic and eutopic endometrium in women with endometriosis: A rationale for endometriotic invasiveness. Fertil. Steril. 2001, 75, 152–159. [Google Scholar] [CrossRef]
  122. Shaco-Levy, R.; Sharabi, S.; Benharroch, D.; Piura, B.; Sion-Vardy, N. Matrix metalloproteinases 2 and 9, E-cadherin, and β-catenin expression in endometriosis, low-grade endometrial carcinoma and non-neoplastic eutopic endometrium. Eur. J. Obstet. Gynecol. Reprod. Biol. 2008, 139, 226–232. [Google Scholar] [CrossRef] [PubMed]
  123. Di Carlo, C.; Bonifacio, M.; Tommaselli, G.A.; Bifulco, G.; Guerra, G.; Nappi, C. Metalloproteinases, vascular endothelial growth factor, and angiopoietin 1 and 2 in eutopic and ectopic endometrium. Fertil. Steril. 2009, 91, 2315–2323. [Google Scholar] [CrossRef] [PubMed]
  124. Collette, T.; Maheux, R.; Mailloux, J.; Akoum, A. Increased expression of matrix metalloproteinase-9 in the eutopic endometrial tissue of women with endometriosis. Hum. Reprod. 2006, 21, 3059–3067. [Google Scholar] [CrossRef] [PubMed]
  125. Chung, H.W.; Lee, J.Y.; Moon, H.-S.; Hur, S.E.; Park, M.H.; Wen, Y.; Polan, M.L. Matrix metalloproteinase-2, membranous type 1 matrix metalloproteinase, and tissue inhibitor of metalloproteinase-2 expression in ectopic and eutopic endometrium. Fertil. Steril. 2002, 78, 787–795. [Google Scholar] [CrossRef]
  126. Collette, T.; Bellehumeur, C.; Kats, R.; Maheux, R.; Mailloux, J.; Villeneuve, M.; Akoum, A. Evidence for an increased release of proteolytic activity by the eutopic endometrial tissue in women with endometriosis and for involvement of matrix metalloproteinase-9. Hum. Reprod. 2004, 19, 1257–1264. [Google Scholar] [CrossRef] [Green Version]
  127. Matsuzaki, S.; Botchorishvili, R.; Pouly, J.L.; Canis, M. Targeting the Wnt/β-catenin pathway in endometriosis: A potentially effective approach for treatment and prevention. Mol. Cell. Ther. 2014, 2, 36. [Google Scholar] [CrossRef] [Green Version]
  128. Cox, K.E.; Piva, M.; Sharpe-Timms, K.L. Differential regulation of matrix metalloproteinase-3 gene expression in endometriotic lesions compared with endometrium. Biol. Reprod. 2001, 65, 1297–1303. [Google Scholar] [CrossRef] [Green Version]
  129. Ganguly, K.; Maity, P.; Reiter, R.J.; Swarnakar, S. Effect of melatonin on secreted and induced matrix metalloproteinase-9 and -2 activity during prevention of indomethacin-induced gastric ulcer. J. Pineal Res. 2005, 39, 307–315. [Google Scholar] [CrossRef]
  130. Liu, D.; Shi, K.; Fu, M.; Chen, F. Melatonin indirectly decreases gastric cancer cell proliferation and invasion via effects on cancer-associated fibroblasts. Life Sci. 2021, 277, 119497. [Google Scholar] [CrossRef]
  131. Mao, L.; Yuan, L.; Slakey, L.M.; Jones, F.E.; E Burow, M.; Hill, S.M. Inhibition of breast cancer cell invasion by melatonin is mediated through regulation of the p38 mitogen-activated protein kinase signaling pathway. Breast Cancer Res. 2010, 12, R107. [Google Scholar] [CrossRef] [Green Version]
  132. Djordjevic, B.; Cvetkovic, T.; Stoimenov, T.J.; Despotovic, M.; Zivanovic, S.; Basic, J.; Veljkovic, A.; Velickov, A.; Kocic, G.; Pavlovic, D.; et al. Oral supplementation with melatonin reduces oxidative damage and concentrations of inducible nitric oxide synthase, VEGF and matrix metalloproteinase 9 in the retina of rats with streptozotocin/nicotinamide induced pre-diabetes. Eur. J. Pharmacol. 2018, 833, 290–297. [Google Scholar] [CrossRef] [PubMed]
  133. Akbar, M.A.; Movassaghpour, A.A.; Ghanbari, H.; Kheirandish, M.; Maroufi, N.F.; Rahbarghazi, R.; Nouri, M.; Samadi, N. The potential therapeutic effect of melatonin on human ovarian cancer by inhibition of invasion and migration of cancer stem cells. Sci. Rep. 2017, 7, 17062. [Google Scholar] [CrossRef] [Green Version]
  134. Jardim-Perassi, B.V.; Lourenço, M.R.; Doho, G.M.; Grígolo, I.H.; Gelaleti, G.B.; Ferreira, L.C.; Borin, T.F.; Moschetta, M.G.; Zuccari, D.A.P.D.C. Melatonin Regulates Angiogenic Factors under Hypoxia in Breast Cancer Cell Lines. Anti-Cancer Agents Med. Chem. 2016, 16, 347–358. [Google Scholar] [CrossRef] [PubMed]
  135. Xie, M.; Hu, A.; Luo, Y.; Sun, W.; Hu, X.; Tang, S. Interleukin-4 and melatonin ameliorate high glucose and interleukin-1β stimulated inflammatory reaction in human retinal endothelial cells and retinal pigment epithelial cells. Mol. Vis. 2014, 20, 921–928. [Google Scholar] [PubMed]
  136. Černyšiov, V.; Mauricas, M.; Girkontaite, I. Melatonin inhibits granulocyte adhesion to ICAM via MT3/QR2 and MT2 receptors. Int. Immunol. 2015, 27, 599–608. [Google Scholar] [CrossRef] [Green Version]
  137. Matsuzaki, S.; Darcha, C. Epithelial to mesenchymal transition-like and mesenchymal to epithelial transition-like processes might be involved in the pathogenesis of pelvic endometriosis. Hum. Reprod. 2012, 27, 712–721. [Google Scholar] [CrossRef]
  138. Thiery, J.P.; Acloque, H.; Huang, R.Y.J.; Nieto, M.A. Epithelial-Mesenchymal Transitions in Development and Disease. Cell 2009, 139, 871–890. [Google Scholar] [CrossRef]
  139. Heerboth, S.; Housman, G.; Leary, M.; Longacre, M.; Byler, S.; Lapinska, K.; Willbanks, A.; Sarkar, S. EMT and tumor metastasis. Clin. Transl. Med. 2015, 4, 6. Available online: http://www.clintransmed.com/%5Cnhttp://ovidsp.ovid.com/ovidweb.cgi?T=JS&PAGE=reference&D=emed13&NEWS=N&AN=2015793182 (accessed on 26 September 2022). [CrossRef]
  140. Min, C.; Eddy, S.F.; Sherr, D.H.; Sonenshein, G.E. NF-κB and epithelial to mesenchymal transition of cancer. J. Cell. Biochem. 2008, 104, 733–744. [Google Scholar] [CrossRef]
  141. Lamouille, S.; Xu, J.; Derynck, R. Molecular mechanisms of epithelial–mesenchymal transition. Nat. Rev. Mol. Cell Biol. 2014, 15, 178–196. [Google Scholar] [CrossRef] [Green Version]
  142. Li, Y.; Ma, J.; Qian, X.; Wu, Q.; Xia, J.; Miele, L.H.; Sarkar, F.H.; Wang, Z. Regulation of EMT by Notch signaling pathway in tumor progression. Curr. Cancer Drug Targets 2013, 13, 957–962. [Google Scholar] [CrossRef] [PubMed]
  143. De Francesco, E.M.; Maggiolini, M.; Musti, A.M. Crosstalk between Notch, HIF-1α and GPER in Breast Cancer EMT. Int. J. Mol. Sci. 2018, 19, 2011. [Google Scholar] [CrossRef] [Green Version]
  144. Wu, S.-M.; Lin, W.-Y.; Shen, C.-C.; Pan, H.-C.; Bin, W.K.; Chen, Y.-C.; Jan, Y.-J.; Lai, D.-W.; Tang, S.-C.; Tien, H.-R.; et al. Melatonin set out to ER stress signaling thwarts epithelial mesenchymal transition and peritoneal dissemination via calpain-mediated C/EBPβ and NFκ B cleavage. J. Pineal Res. 2015, 60, 142–154. [Google Scholar] [CrossRef] [PubMed]
  145. Wang, X.; Wang, B.; Xie, J.; Hou, D.; Zhang, H.; Huang, H. Melatonin inhibits epithelial-to-mesenchymal transition in gastric cancer cells via attenuation of IL-1β/NF-κB/MMP2/MMP9 signaling. Int. J. Mol. Med. 2018, 42, 2221–2228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Ndo, N.G.; Colombo, J.; Lopes, J.R.; Gelaleti, G.B.; Moschetta, M.G.; Sonehara, N.M.; Hellmén, E.; Cde, F.Z.; Oliani, S.M.; Zuccari, D.A. Effect of Melatonin in Epithelial Mesenchymal Transition Markers and Invasive Properties of Breast Cancer Stem Cells of Canine and Human Cell Lines. PLoS ONE 2016, 11, e0150407. [Google Scholar] [CrossRef] [Green Version]
  147. Mao, L.; Dauchy, R.T.; Blask, D.E.; Slakey, L.M.; Xiang, S.; Yuan, L.; Dauchy, E.M.; Shan, B.; Brainard, G.C.; Hanifin, J.P.; et al. Circadian Gating of Epithelial-to-Mesenchymal Transition in Breast Cancer Cells Via Melatonin-Regulation of GSK3β. Mol. Endocrinol. 2012, 26, 1808–1820. [Google Scholar] [CrossRef]
  148. Mao, L.; Summers, W.; Xiang, S.; Yuan, L.; Dauchy, R.T.; Reynolds, A.; Wren-Dail, M.A.; Pointer, D.; Frasch, T.; Blask, D.E.; et al. Melatonin Represses Metastasis in Her2-Postive Human Breast Cancer Cells by Suppressing RSK2 Expression. Mol. Cancer Res. 2016, 14, 1159–1169. [Google Scholar] [CrossRef] [Green Version]
  149. Chen, Y.; Zhang, T.; Liu, X.; Li, Z.; Zhou, D.; Xu, W. Melatonin suppresses epithelial-to-mesenchymal transition in the MG-63 cell line. Mol. Med. Rep. 2019, 21, 1356–1364. [Google Scholar] [CrossRef]
  150. Samimi, M.; Pourhanifeh, M.H.; Mehdizadehkashi, A.; Eftekhar, T.; Asemi, Z. The role of inflammation, oxidative stress, angiogenesis, and apoptosis in the pathophysiology of endometriosis: Basic science and new insights based on gene expression. J. Cell. Physiol. 2019, 234, 19384–19392. [Google Scholar] [CrossRef]
  151. McLaren, J.; Prentice, A.; Charnock-Jones, D.S.; Smith, S.K. Vascular Endothelial Growth Factor (VEGF) Concentrations Are Elevated in Peritoneal Fluid of Women With Endometriosis. Obstet. Gynecol. Surv. 1996, 51, 488–490. [Google Scholar] [CrossRef]
  152. McLaren, J. Vascular endothelial growth factor and endometriotic angiogenesis. Hum. Reprod. Updat. 2000, 6, 45–55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Melincovici, C.S.; Boşca, A.B.; Şuşman, S.; Mărginean, M.; Mihu, C.; Istrate, M.; Moldovan, I.M.; Roman, A.L.; Mihu, C.M. Vascular endothelial growth factor (VEGF)—Key factor in normal and pathological angiogenesis. Rom. J. Morphol. Embryol. 2018, 59, 455–467. [Google Scholar]
  154. Liu, S.; Xin, X.; Hua, T.; Shi, R.; Chi, S.; Jin, Z.; Wang, H. Efficacy of Anti-VEGF/VEGFR Agents on Animal Models of Endometriosis: A Systematic Review and Meta-Analysis. PLoS ONE 2016, 11, e0166658. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Xu, H.; Zhang, T.; Man, G.C.W.; May, K.E.; Becker, C.M.; Davis, T.N.; Kung, A.L.; Birsner, A.E.; D’Amato, R.J.; Wong, A.W.Y.; et al. Vascular endothelial growth factor C is increased in endometrium and promotes endothelial functions, vascular permeability and angiogenesis and growth of endometriosis. Angiogenesis 2013, 16, 541–551. [Google Scholar] [CrossRef]
  156. Xu, H.; Becker, C.M.; Lui, W.T.; Chu, C.Y.; Davis, T.N.; Kung, A.; Birsner, A.E.; D’Amato, R.J.; Man, C.W.; Wang, R. Green tea epigallocatechin-3-gallate inhibits angiogenesis and suppresses vascular endothelial growth factor C/vascular endothelial growth factor receptor 2 expression and signaling in experimental endometriosis in vivo. Fertil. Steril. 2011, 96, 1021–1028.e1. [Google Scholar] [CrossRef]
  157. Jardim-Perassi, B.V.; Arbab, A.S.; Ferreira, L.C.; Borin, T.F.; Varma, N.R.S.; Iskander, A.S.M.; Shankar, A.; Ali, M.M.; Zuccari, D.A.P.D.C. Effect of Melatonin on Tumor Growth and Angiogenesis in Xenograft Model of Breast Cancer. PLoS ONE 2014, 9, e85311. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Cerezo, A.B.; Hornedo-Ortega, R.; Álvarez-Fernández, M.A.; Troncoso, A.M.; García-Parrilla, M.C. Inhibition of VEGF-Induced VEGFR-2 Activation and HUVEC Migration by Melatonin and Other Bioactive Indolic Compounds. Nutrients 2017, 9, 249. [Google Scholar] [CrossRef] [Green Version]
  159. Lissoni, P.; Rovelli, F.; Malugani, F.; Bucovec, R.; Conti, A.; Maestroni, G.J. Anti-angiogenic activity of melatonin in advanced cancer patients. Neuro Endocrinol. Lett. 2001, 22, 45–48. [Google Scholar]
  160. Hur, S.E.; Lee, J.Y.; Moon, H.-S.; Chung, H.W. Angiopoietin-1, angiopoietin-2 and Tie-2 expression in eutopic endometrium in advanced endometriosis. Mol. Hum. Reprod. 2006, 12, 421–426. [Google Scholar] [CrossRef]
  161. Zondervan, K.T.; Becker, C.M.; Koga, K.; Missmer, S.A.; Taylor, R.N.; Viganò, P. Endometriosis. Nat. Rev. Dis. Prim. 2018, 4, 9. [Google Scholar] [CrossRef]
  162. Burney, R.O.; Giudice, L.C. Pathogenesis and pathophysiology of endometriosis. Fertil. Steril. 2012, 98, 511–519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Lessey, B.A.; Lebovic, D.I.; Taylor, R.N. Eutopic Endometrium in Women with Endometriosis: Ground Zero for the Study of Implantation Defects. Semin. Reprod. Med. 2013, 31, 109–124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Agic, A.; Xu, H.; Finas, D.; Banz, C.; Diedrich, K.; Hornung, D. Is Endometriosis Associated with Systemic Subclinical Inflammation? Gynecol. Obstet. Investig. 2006, 62, 139–147. [Google Scholar] [CrossRef] [PubMed]
  165. Vallvé-Juanico, J.; Houshdaran, S.; Giudice, L.C. The endometrial immune environment of women with endometriosis. Hum. Reprod. Updat. 2019, 25, 565–592. [Google Scholar] [CrossRef] [PubMed]
  166. Mihalyi, A.; Gevaert, O.; Kyama, C.M.; Simsa, P.; Pochet, N.; De Smet, F.; De Moor, B.; Meuleman, C.; Billen, J.; Blanckaert, N.; et al. Non-invasive diagnosis of endometriosis based on a combined analysis of six plasma biomarkers. Hum. Reprod. 2009, 25, 654–664. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Vodolazkaia, A.; El-Aalamat, Y.; Popovic, D.; Mihalyi, A.; Bossuyt, X.; Kyama, C.M.; Fassbender, A.; Bokor, A.; Schols, D.; Huskens, D.; et al. Evaluation of a panel of 28 biomarkers for the non-invasive diagnosis of endometriosis. Hum. Reprod. 2012, 27, 2698–2711. Available online: http://linker2.worldcat.org/?rft.institution_id=129954&spage=2698&pkgName=UKPMC&issn=0268-1161&linkclass=to_article&jKey=Hum%2BReprod&issue=9&provider=NLM&date=2012-09&aulast=Vodolazkaia+A.%3B+Mihalyi+A.%3B+Kyama+C.M.%3B+Fassbender+A.%3B+Bokor+A.%3B+Meule (accessed on 26 September 2022). [CrossRef] [Green Version]
  168. Currier, N.L.; Sun, L.Z.-Y.; Miller, S.C. Exogenous melatonin: Quantitative enhancement in vivo of cells mediating non-specific immunity. J. Neuroimmunol. 2000, 104, 101–108. [Google Scholar] [CrossRef]
  169. Miller, S.C.; Pandi, P.S.R.; Esquifino, A.I.; Cardinali, D.P.; Maestroni, G.J.M. The role of melatonin in immuno-enhancement: Potential application in cancer. Int. J. Exp. Pathol. 2006, 87, 81–87. [Google Scholar] [CrossRef]
  170. Martins, E., Jr.; Fernandes, L.; Bartol, I.; Cipolla-Neto, J.; Rosa, L.C. The effect of melatonin chronic treatment upon macrophage and lymphocyte metabolism and function in Walker-256 tumour-bearing rats. J. Neuroimmunol. 1998, 82, 81–89. [Google Scholar] [CrossRef]
  171. Cho, J.H.; Bhutani, S.; Kim, C.H.; Irwin, M.R. Anti-inflammatory effects of melatonin: A systematic review and meta-analysis of clinical trials. Brain Behav. Immun. 2021, 93, 245–253. [Google Scholar] [CrossRef]
  172. Stratton, P.; Berkley, K.J. Chronic pelvic pain and endometriosis: Translational evidence of the relationship and implications. Hum. Reprod. Update 2010, 17, 327–346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Morotti, M.; Vincent, K.; Brawn, J.; Zondervan, K.T.; Becker, C.M. Peripheral changes in endometriosis-associated pain. Hum. Reprod. Update 2014, 20, 717–736. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Nockher, W.A.; Renz, H. Neurotrophins in allergic diseases: From neuronal growth factors to intercellular signaling molecules. J. Allergy Clin. Immunol. 2006, 117, 583–589. [Google Scholar] [CrossRef] [PubMed]
  175. Nijs, J.; Meeus, M.; Versijpt, J.; Moens, M.; Bos, I.; Knaepen, K.; Meeusen, R. Brain-derived neurotrophic factor as a driving force behind neuroplasticity in neuropathic and central sensitization pain: A new therapeutic target? Expert Opin. Ther. Targets 2014, 19, 565–576. [Google Scholar] [CrossRef] [PubMed]
  176. Robertson, N.J.; Faulkner, S.; Fleiss, B.; Bainbridge, A.; Andorka, C.; Price, D.; Powell, E.; Lecky-Thompson, L.; Thei, L.; Chandrasekaran, M.; et al. Melatonin augments hypothermic neuroprotection in a perinatal asphyxia model. Brain 2012, 136, 90–105. [Google Scholar] [CrossRef] [PubMed]
  177. Babaee, A.; Eftekhar-Vaghefi, S.H.; Asadi-Shekaari, M.; Shahrokhi, N.; Soltani, S.D.; Malekpour-Afshar, R.; Basiri, M. Melatonin treatment reduces astrogliosis and apoptosis in rats with traumatic brain injury. Iran. J. Basic Med. Sci. 2015, 18, 867–872. [Google Scholar] [CrossRef]
  178. Srinivasan, V.; Pandi-Perumal, S.R.; Maestroni, G.J.M.; Esquifino, A.I.; Hardeland, R.; Cardinali, D.P. Role of melatonin in neurodegenerative diseases. Neurotox. Res. 2005, 7, 293–318. [Google Scholar] [CrossRef]
  179. Morris, R.W.; Lutsch, E.F. Daily Susceptibility Rhythm to Morphine Analgesia. J. Pharm. Sci. 1969, 58, 374–376. [Google Scholar] [CrossRef]
  180. Yu, C.-X.; Zhu, C.-B.; Xu, S.-F.; Cao, X.-D.; Wu, G.-C. The analgesic effects of peripheral and central administration of melatonin in rats. Eur. J. Pharmacol. 2000, 403, 49–53. [Google Scholar] [CrossRef]
  181. El-Shenawy, S.M.; Abdel-Salam, O.M.; Baiuomy, A.R.; El-Batran, S.; Arbid, M.S. Studies on the anti-inflammatory and anti-nociceptive effects of melatonin in the rat. Pharmacol. Res. 2002, 46, 235–243. [Google Scholar] [CrossRef]
  182. Lopez-Canul, M.; Palazzo, E.; Dominguez-Lopez, S.; Luongo, L.; Lacoste, B.; Comai, S.; Angeloni, D.; Fraschini, F.; Boccella, S.; Spadoni, G.; et al. Selective melatonin MT2 receptor ligands relieve neuropathic pain through modulation of brainstem descending antinociceptive pathways. Pain 2015, 156, 305–317. [Google Scholar] [CrossRef] [PubMed]
  183. Masruha, M.R.; Vieira, D.S.D.S.; Minett, T.S.C.; Cipolla-Neto, J.; Zukerman, E.; Vilanova, L.C.P.; Peres, M.F.P. Low urinary 6-sulphatoxymelatonin concentrations in acute migraine. J. Headache Pain 2008, 9, 221–224. [Google Scholar] [CrossRef] [Green Version]
  184. Leone, M.; D’Amico, D.; Moschiano, F.; Fraschini, F.; Bussone, G. Melatonin Versus Placebo in the Prophylaxis of Cluster Headache: A double-blind pilot study with parallel groups. Cephalalgia 1996, 16, 494–496. [Google Scholar] [CrossRef] [PubMed]
  185. Miano, S.; Parisi, P.; Pelliccia, A.; Luchetti, A.; Paolino, M.C.; Villa, M.P. Melatonin to prevent migraine or tension-type headache in children. Neurol. Sci. 2008, 29, 285–287. [Google Scholar] [CrossRef]
  186. Kurganova, Y.M.; Danilov, A. A role of melatonin in the treatment of low back pain. Zhurnal Nevrol. Psikhiatrii Im. SS Korsakova 2015, 115, 30–35. [Google Scholar] [CrossRef]
  187. Hussain, S.A.-R.; Alkhalifa, I.; Jasim, N.A.; Gorial, F.I. Adjuvant use of melatonin for treatment of fibromyalgia. J. Pineal Res. 2010, 50, 267–271. [Google Scholar] [CrossRef] [PubMed]
  188. Alghamdi, B.S. The neuroprotective role of melatonin in neurological disorders. J. Neurosci. Res. 2018, 96, 1136–1149. [Google Scholar] [CrossRef] [Green Version]
  189. Karamitri, A.; Jockers, R. Melatonin in type 2 diabetes mellitus and obesity. Nat. Rev. Endocrinol. 2018, 15, 105–125. [Google Scholar] [CrossRef]
  190. Wu, H.; Liu, J.; Yin, Y.; Zhang, D.; Xia, P.; Zhu, G. Therapeutic Opportunities in Colorectal Cancer: Focus on Melatonin Antioncogenic Action. BioMed Res. Int. 2019, 2019, 9740568. [Google Scholar] [CrossRef]
  191. Alston, M.; Cain, S.W.; Rajaratnam, S.M.W. Advances of Melatonin-Based Therapies in the Treatment of Disturbed Sleep and Mood. Sleep-Wake Neurobiol. Pharmacol. 2018, 253, 305–319. [Google Scholar] [CrossRef]
  192. Eryilmaz, O.G.; Devran, A.; Sarikaya, E.; Aksakal, F.N.; 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Batıoğlu, A.S.; Şahin, U.; Gürlek, B.; Öztürk, N.; Unsal, E. The efficacy of melatonin administration on oocyte quality. Gynecol. Endocrinol. 2011, 28, 91–93. [Google Scholar] [CrossRef]
  194. Fernando, S.; Wallace, E.M.; Vollenhoven, B.; Lolatgis, N.; Hope, N.; Wong, M.; Lawrence, M.; Lawrence, A.; Russell, C.; Leong, K.; et al. Melatonin in Assisted Reproductive Technology: A Pilot Double-Blind Randomized Placebo-Controlled Clinical Trial. Front. Endocrinol. 2018, 9, 545. [Google Scholar] [CrossRef] [Green Version]
  195. Takasaki, A.; Nakamura, Y.; Tamura, H.; Shimamura, K.; Morioka, H. Melatonin as a new drug for improving oocyte quality. Reprod. Med. Biol. 2003, 2, 139–144. [Google Scholar] [CrossRef] [PubMed]
  196. Espino, J.; Macedo, M.; Lozano, G.; Ortiz, A.; Rodríguez, C.; Rodríguez, A.B.; Bejarano, I. Impact of Melatonin Supplementation in Women with Unexplained Infertility Undergoing Fertility Treatment. Antioxidants 2019, 8, 338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Fernando, S.; Osianlis, T.; Vollenhoven, B.; Wallace, E.; Rombauts, L. A pilot double-blind randomised placebo-controlled dose-response trial assessing the effects of melatonin on infertility treatment (MIART): Study protocol. BMJ Open 2014, 4, e005986. [Google Scholar] [CrossRef] [Green Version]
  198. Tagliaferri, V.; Romualdi, D.; Scarinci, E.; De Cicco, S.; Di Florio, C.; Immediata, V.; Tropea, A.; Santarsiero, C.M.; Lanzone, A.; Apa, R. Melatonin Treatment May Be Able to Restore Menstrual Cyclicity in Women With PCOS: A Pilot Study. Reprod. Sci. 2017, 25, 269–275. [Google Scholar] [CrossRef]
  199. Mokhtari, F.; Asbagh, F.A.; Azmoodeh, O.; Bakhtiyari, M.; Almasi-Hashiani, A. Effects of Melatonin Administration on Chemical Pregnancy Rates of Polycystic Ovary Syndrome Patients Undergoing Intrauterine Insemination: A Randomized Clinical Trial. Int. J. Fertil. Steril. 2019, 13, 225–229. [Google Scholar] [CrossRef]
  200. Miller, S.L.; Yawno, T.; Alers, N.O.; Castillo-Melendez, M.; Supramaniam, V.G.; Vanzyl, N.; Sabaretnam, T.; Loose, J.M.; Drummond, G.R.; Walker, D.W.; et al. Antenatal antioxidant treatment with melatonin to decrease newborn neurodevelopmental deficits and brain injury caused by fetal growth restriction. J. Pineal Res. 2014, 56, 283–294. [Google Scholar] [CrossRef]
  201. Hobson, S.; Gurusinghe, S.; Lim, R.; Alers, N.O.; Miller, S.L.; Kingdom, J.C.; Wallace, E.M. Melatonin improves endothelial function in vitro and prolongs pregnancy in women with early-onset preeclampsia. J. Pineal Res. 2018, 65, e12508. [Google Scholar] [CrossRef]
  202. Parandavar, N.; Abdali, K.; Keshtgar, S.; Emamghoreishi, M.; Amooee, S. The Effect of Melatonin on Climacteric Symptoms in Menopausal Women; A Double-Blind, Randomized Controlled, Clinical Trial. Iran. J. Public Health 2014, 43, 1405–1416. [Google Scholar] [PubMed]
  203. 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. Eur. Rev. Med. Pharmacol. Sci. 2010, 14, 555–561. [Google Scholar] [PubMed]
  204. 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 fertilization-embryo transfer. Gynecol. Endocrinol. 2014, 30, 359–362. [Google Scholar] [CrossRef]
  205. Fernando, S.; Biggs, S.N.; Horne, R.S.C.; Vollenhoven, B.; Lolatgis, N.; Hope, N.; Wong, M.; Lawrence, M.; Lawrence, A.; Russell, C.; et al. The impact of melatonin on the sleep patterns of women undergoing IVF: A double blind RCT. Hum. Reprod. Open 2018, 2017, hox027. [Google Scholar] [CrossRef] [Green Version]
  206. Wdowiak, A.; Filip, M. The effect of myo-inositol, vitamin D3 and melatonin on the oocyte quality and pregnancy in in vitro fertilization: A randomized prospective controlled trial. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 8529–8536. [Google Scholar] [CrossRef] [PubMed]
  207. Jahromi, B.N.; Sadeghi, S.; Alipour, S.; Parsanezhad, M.E.; Alamdarloo, S.M. Effect of melatonin on the outcome of assisted repro-ductive technique cycles in women with diminished ovarian reserve: A double-blinded randomized clinical trial. Iran J. Med. Sci. 2017, 42, 73–78. [Google Scholar]
  208. Parandavar, N.; Hojat, M.; Abdali, K.; Keshtgar, S.; Emamghoreishi, M.; Yeganeh, B.S. The effect of melatonin on the lipid levels in menopausal women: A double-blind, controlled, clinical trial. J. Educ. Health Promot. 2018, 7, 144. [Google Scholar] [CrossRef]
  209. Chojnacki, C.; Kaczka, A.; Gasiorowska, A.; Fichna, J.; Chojnacki, J.; Brzozowski, T. The effect of long-term melatonin supplementation on psychosomatic disorders in postmenopausal women. J. Physiol. Pharmacol. 2018, 69, 297–304. [Google Scholar] [CrossRef]
  210. Andersen, L.P.H.; Gögenur, I.; Rosenberg, J.; Reiter, R.J. The Safety of Melatonin in Humans. Clin. Drug Investig. 2015, 36, 169–175. [Google Scholar] [CrossRef]
Figure 1. Therapeutic targets of melatonin toward inhibiting the development and progression of endometriosis. The effect of melatonin with respect to various molecular targets involved in the processes of endometriosis, including adhesion, invasion, EMT, oxidative stress, angiogenesis, and chronic pelvic pain. Abbreviations: MMP, Matrix metalloproteinase; TIMP, Tissue inhibitors of metalloprotease; EMT, Epithelial–mesenchymal transition; MDA, Malondialdehyde; COX-2, Cyclooxygenase 2; SOD, Superoxide dismutase; CAT, Catalase; VEGF, Vascular endothelial growth factor; BDNF, Brain-derived neurotrophic factor.
Figure 1. Therapeutic targets of melatonin toward inhibiting the development and progression of endometriosis. The effect of melatonin with respect to various molecular targets involved in the processes of endometriosis, including adhesion, invasion, EMT, oxidative stress, angiogenesis, and chronic pelvic pain. Abbreviations: MMP, Matrix metalloproteinase; TIMP, Tissue inhibitors of metalloprotease; EMT, Epithelial–mesenchymal transition; MDA, Malondialdehyde; COX-2, Cyclooxygenase 2; SOD, Superoxide dismutase; CAT, Catalase; VEGF, Vascular endothelial growth factor; BDNF, Brain-derived neurotrophic factor.
Nutrients 14 04087 g001
Table 2. Clinical trials of melatonin for endometriosis and other female reproduction.
Table 2. Clinical trials of melatonin for endometriosis and other female reproduction.
Studies Disorder Sample Size
(Melatonin/
Control)
Age
(Means ± SD/
SEM or Range)
Daily DoseDuration or PeriodMain OutcomesAdverse EventsTrial Registration Number
Schwertner 2013 [18]Endometriosis20/2018–4510 mg8 weeksReduced pain scores and BDNF levels
Increased sleep quality
//
Takasaki
2003 [195]
Infertility 27/NA34 ± 4.41 or
3 mg
From 2nd to 5th day of the menstrual cycle to hCG dayDecreased number of degenerate oocytes (only in 3 mg group)//
Tamura
2008 [47]
Infertility 56/5934.8 ± 4.83 mgFrom 2nd to 5th day of the menstrual cycle to OPU dayIncreased oocytes quality and fertility rate//
Rizzo
2010 [203]
Infertility 32/3335–423 mgFrom GnRH administration day to pregnancy test result was confirmedIncreased tendency on number of mature oocytes, embryo quality, clinical pregnancy rate, and implantation rate//
Batioglu
2012 [193]
Infertility40/4520–403 mgDuring the IVF-ET procedureIncreased number and percentage of mature oocytes
Increased number of embryos of top quality
Increased tendency on clinical pregnancy rate
//
Nishihara 2014 [204]Infertility97/NA≤423 mgAt least 2 weeksIncreased fertilization and embryo quality //
Fernando 2018 [205]Infertility32/29
29/29
26/29
18–452, 4, or 8 mgFrom 2nd to 5th day of the menstrual cycle to OPU dayNo significant differences in daytime Karolinska sleepiness score between groups
No differences in objective measures of sleep quantity or quality
Improved the subjective sleep quality scores except 8 mg group
/ACTRN12613001317785
Fernando 2018 [194]Infertility41/41
39/41
40/41
18–452, 4, or 8 mgFrom 2nd to 5th day of the menstrual cycle to OPU dayThere were no differences between all the groups in total oocyte number, number of MII oocytes, number of fertilized oocytes, the number or quality of embryos, clinical pregnancy rate, or live birth rateHeadache, fatigue (no significant difference)ACTRN12613001317785
Espino
2019 [196]
Infertility20/20 3 or 6 mgFrom 2nd to 5th day of the menstrual cycle to OPU day Ameliorated intrafollicular oxidative balance and oocyte quality
Increased the rate of pregnancies/live births
No significant difference between 3 mg group and 6 mg group
//
Wdowiak 2020 [206]Infertility50/5020–351 mg6 monthsIncreased blastocyst and oocyte quality
Increased rate of clinical pregnancy
//
Jahromi
2017 [207]
Diminished ovarian reserve40/4022–423 mgFrom 2nd to 5th day of the menstrual cycle to OPU dayIncreased serum estradiol level on the triggering day
Increased number of MII oocytes, top quality embryos with G1 and G2
/IRCT2014041417264N1
Tagliaferri 2017 [198]PCOS40/NA23.25 ± 4.072 mg6 months Improved the menstrual irregularities
Decreased biochemical hyperandrogenism
//
Mokhtari
2019 [199]
PCOS98/10028.9 ± 5.53 mgFrom 2nd to 5th day of the menstrual cycle to hCG dayIncreased chemical pregnancies and endometrial thickness//
Parandavar 2018 [208] Menopausal women 98/14253.22 ± 4.213 mg3 months Increased amount of triglyceride //
Chojnacki 2018 [209] Postmenopausal women 30/3051–648 mg12 months Decreased Kupperman Index and body mass index No adverse influence on patients’ psychosomatic activity/
Miller
2014 [200]
Fetal growth restriction 6/6/8 mgFor the duration of pregnancy Decreased placental oxidative stress No adverse maternal or fetal effects NCT01695070
Parandavar
2014 [202]
Climacteric symptoms 99/10140–603 mg3 months Decreased climacteric symptoms score Sleepiness, nausea, vomiting, and headache (no significant difference)/
Hobson
2018 [201]
Preeclampsia 20/48Control: 30.6 ± 0.8
Melatonin: 32.7 ± 1.1
30 mgFrom recruitment until delivery No adverse events and adverse drug reactions
Prolonged gestation
Reduced the pharmacological need for antihypertensives
No adverse events/
ACTRN, Australian clinical trials registration number; BDNF, Brain-derived neurotrophic factor; hCG, Human chorionic gonadotropin; GnRH, Gonadotropin-releasing hormone; IRCT, Iranian Registry of Clinical Trials; IVF-ET, In vitro fertilization and embryo transfer; MII, Metaphase II; NA, Not applicable; NCT, National clinical trial; OPU, Oocyte pick up; PCOS, Polycystic ovary syndrome; SD, Standard deviation; SEM, Standard error of mean; SOD, Superoxide dismutase.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Li, Y.; Hung, S.-W.; Zhang, R.; Man, G.C.-W.; Zhang, T.; Chung, J.P.-W.; Fang, L.; Wang, C.-C. Melatonin in Endometriosis: Mechanistic Understanding and Clinical Insight. Nutrients 2022, 14, 4087. https://doi.org/10.3390/nu14194087

AMA Style

Li Y, Hung S-W, Zhang R, Man GC-W, Zhang T, Chung JP-W, Fang L, Wang C-C. Melatonin in Endometriosis: Mechanistic Understanding and Clinical Insight. Nutrients. 2022; 14(19):4087. https://doi.org/10.3390/nu14194087

Chicago/Turabian Style

Li, Yiran, Sze-Wan Hung, Ruizhe Zhang, Gene Chi-Wai Man, Tao Zhang, Jacqueline Pui-Wah Chung, Lanlan Fang, and Chi-Chiu Wang. 2022. "Melatonin in Endometriosis: Mechanistic Understanding and Clinical Insight" Nutrients 14, no. 19: 4087. https://doi.org/10.3390/nu14194087

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop