Open Access

Insight into the roles of melatonin in bone tissue and bone‑related diseases (Review)

  • Authors:
    • Xiaofeng Lu
    • Shaoling Yu
    • Guangjin Chen
    • Wenhao Zheng
    • Jinfeng Peng
    • Xiaofei Huang
    • Lili Chen
  • View Affiliations

  • Published online on: March 17, 2021     https://doi.org/10.3892/ijmm.2021.4915
  • Article Number: 82
  • Copyright: © Lu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Bone‑related diseases comprise a large group of common diseases, including fractures, osteoporosis and osteoarthritis (OA), which affect a large number of individuals, particularly the elderly. The progressive destruction and loss of alveolar bone caused by periodontitis is a specific type of bone loss, which has a high incidence and markedly reduces the quality of life of patients. With the existing methods of prevention and treatment, the incidence and mortality of bone‑related diseases are still gradually increasing, creating a significant financial burden to societies worldwide. To prevent the occurrence of bone‑related diseases, delay their progression or reverse the injuries they cause, new alternative or complementary treatments need to be developed. Melatonin exerts numerous physiological effects, including inducing anti‑inflammatory and antioxidative functions, resetting circadian rhythms and promoting wound healing and tissue regeneration. Melatonin also participates in the health management of bone and cartilage. In the present review, the potential roles of melatonin in the pathogenesis and progression of bone injury, osteoporosis, OA and periodontitis are summarized. Furthermore, the high efficiency and diversity of the physiological regulatory effects of melatonin are highlighted and the potential benefits of the use of melatonin for the clinical prevention and treatment of bone‑related diseases are discussed.

1. Introduction

Bone-related diseases, such as fractures, osteoporosis and osteoarthritis (OA), severely affect the quality of life of patients due to their high incidence, slow recovery, associated pain and negative effects on patient behavior. Due to the aging of the population in several countries, the prevalence of bone-related diseases is projected to further increase in the following few decades. Data from 2013 indicated that ~57 million Americans aged >50 years suffered from bone diseases, among which 48 million had osteopenia and 9 million had osteoporosis. The diseases were accompanied by a risk of fracture in these individuals. Without intervention, the prevalence of osteopenia is projected to increase to 64.3 million American individuals and that of osteoporosis to 11.9 million by the year 2030 (1). Bone disease is the focus of a large number of clinical studies, due to the accompanying high combined lifetime risk of forearm, hip and vertebral fractures (40%), which is comparable to the rate of cardiovascular diseases (2). In addition to the high morbidity and mortality rates, fractures related to osteoporosis have also created a significant financial burden to societies worldwide. For instance, osteoporosis-related costs in the European Union amounted to ~€37 billion in 2010; the majority of that amount was spent on fracture therapy and long-term fracture care, accounting for 66 and 29%, respectively, of the budget (3).

Melatonin, a common molecule with a simple structure known as N-acetyl-5-methoxytryptamine, exists in almost all living organisms (4). Melatonin is secreted by pinealocytes in the pineal gland, while certain tissues can also produce a small amount of melatonin locally (5). There is a synchronization between melatonin production and the light/dark (L/D) cycle. Melatonin is synthesized and secreted in dark environments. When retinal photoreceptive ganglion cells are stimulated by light (mainly in the blue range), the synthesis and secretion of melatonin reduces until no more is secreted (6,7). Generally, levels of melatonin begin to increase early in the evening and peak at 12-2 a.m., followed by a progressive decrease thereafter (8). As the organism ages, melatonin production gradually declines. Melatonin levels continuously decrease from the age of 40-45 years (6).

Currently, melatonin is considered a potent cytoprotective agent, rather than a hormone in the classical sense (9). Melatonin has excellent lipophilic properties and can easily enter the cell membrane and subcellular compartment (10). Melatonin can synchronize the circadian clock in peripheral tissues, maintain the synchronization of bone metabolism with L/D cycles and participate in numerous important physiological processes, such as anti-inflammatory, antitumor and antioxidation processes, as well as regulating circadian and endocrine rhythms, regulating immunity, and promoting wound healing and tissue regeneration (7,11). Melatonin plays a positive role in bone-related diseases by exerting multiple effects. Although there are several physical and drug treatments for bone-related diseases, melatonin has the advantage over other drugs of being inexpensive, and having a wide safety margin, a wide impact on tissues and almost no side effect, suggesting its potential as a main or complementary treatment strategy for a large range of bone diseases.

2. Bone injury

Basic study of bone injury

Bone injury is very common in clinical practice. A variety of pathologies, such as tumors, trauma and surgery, as well as other factors, are likely to cause varying degrees of bone injury. This is a major issue for clinical treatment at present, and an important challenge that will threaten human health in the ensuing 50 years. The bone defect size largely determines the amount of bone repair. The larger the defect, the more difficult to obtain a satisfactory repair (12). Bone repair is often affected by a variety of negative effects, such as possible infection and ischemia of the bone injury site or adjacent tissues, and systemic diseases. Therefore, clinical intervention is required for bone repair.

Possible effects of melatonin on bone injury repair

As shown in Fig. 1, considerable evidence has demonstrated that melatonin contributes to bone repair. There are different claims about the effects of melatonin on osteogenic and osteoclastic activities. Melatonin has been shown to enhance the vertical bone augmentation of rat calvaria by increasing new bone regeneration, neovascularization and the number of osteoblast-like cells (13). It has also been shown to increase the cartilage and callus at the fracture site (14). At pharmacological doses, melatonin has been shown to stimulate osteoblast proliferation and alkaline phosphatase (ALP) activity in a dose-dependent manner. In vitro, melatonin increases the expression of collagen type I (Col1), bone sialoprotein, osteopontin and osteocalcin (OCN), and promotes the production of mineralized extracellular matrix (ECM). At the same time, femoral neocortical bone is enhanced in mice administered an intraperitoneal (i.p.) injection of melatonin (15). However, Histing et al (14) reported that melatonin delayed bone healing in a mouse model of femoral fracture. They explained that melatonin exerted a positive effect on bone repair by inhibiting bone resorption instead of promoting bone regeneration. The levels of receptor activator of NF-κB ligand (RANKL) were reduced in the mice with fractures that received a daily dose of 50 mg/kg melatonin, which may inhibit bone resorption by damaging the balance between osteoprotegerin (OPG) and RANKL.

In addition to exhibiting osteogenic and osteoclast activity, the role of osteoblast and osteoclast differentiation during the process of bone repair is also of interest. Sethi et al (16) noted that osteoblast differentiation requires chronic and uninterrupted melatonin exposure. Melatonin may promote mesenchymal stem cell (MSC) proliferation and migration, at least partly by upregulating neuropeptide Y (NPY) and NPY receptor Y1 (NPY1R), accelerating osteogenic differentiation and promoting fracture healing in the rat femur. Concurrently, NPY/NPY1R expression has been shown to be increased in the fracture zone and serum (17). Osteogenic and chondrogenic differentiation can be promoted, and adipogenesis can be inhibited through the enhancement of runt-related transcription factor 2 expression and the Wnt/β-catenin signaling pathway, as well as the inhibition of peroxisome proliferator-activated receptor (PPAR)-γ (18). The upregulation of the platelet-derived growth factor/protein kinase B pathway partly contributes to the enhanced osteogenic potential and weakened osteoclastic differentiation of MC3T3-E1 cells, and promotes fracture healing in mice with femoral fracture following melatonin treatment (19). In addition, the BMP, extracellular regulated kinase (ERK) and Wnt signaling pathways also participate in the process, while melatonin can improve wound healing and trigger osteogenesis markers in a dose-dependent manner (20). In addition, endochondral bone formation is an important form of osteogenesis. Melatonin increases the expression of chondrogenic differentiation genes in MSCs, which could be partly blocked by luzindole (21).

Inflammation and oxidative stress are inevitable during bone injury. Melatonin is known for its potent antioxidant and anti-inflammatory properties. Oxidative stress produces reactive oxygen species (ROS) and is usually promoted by aging (22), which can lead to excessive bone resorption (23,24). Melatonin is considered a potent natural antioxidant, not only due to the direct inhibition of ROS, but also due to the mobilization of the intracellular antioxidative enzyme system. Melatonin can protect MSCs against oxidation-induced apoptosis by reducing ROS production, enhancing cell viability and promoting continued differentiation (25). Melatonin can increase solute carrier family 39 member 1 expression, activate the mitogen-activated protein kinase (MAPK)/ERK pathway, increase phosphorylated-ERK1/2/5 levels and significantly inhibit the production of ROS; moreover, zinc uptake in cells is increased (26). All the above-mentioned processes can inhibit cell apoptosis (27). In addition, melatonin is beneficial for the inhibition of oxygen free radical activity during fracture healing and the regulation of antioxidant enzyme activity, which promotes fracture healing. In a previous study, it was observed that in contrast to the fracture group, more bone binding was observed in the melatonin treatment fracture group at the same healing time (28 days after the fracture) (28).

Vascular injury often exists simultaneously in bone injuries, such as fractures, which inevitably leads to ischemia and/or hypoxia at the injury site and is extremely unfavorable for the repair of defects. Ischemia/reperfusion can cause excessive ROS production in tissues and lead to cell damage (29,30). Melatonin can eliminate these adverse effects and can be used in fractures with vascular injury and compartment syndrome (31). In hypoxic environments, MC3T3-E1 cells stimulated by melatonin prefer to differentiate towards osteoblasts and promote mineralization through the p38 mitogen-activated protein kinase and protein kinase D1 pathways (32). The effect of melatonin on angiogenesis mediators also plays an important role in bone regeneration (33). Growth factors are considered potential modulators of angiogenesis. For instance, vascular endothelial growth factor (VEGF) contributes to angiogenesis (34). Melatonin treatment has been found to elevate the level of VEGF during granulation tissue formation and accelerate the angiogenic process (35), indicating that melatonin can provide beneficial effects to bone defect repair with vascular injury.

Moreover, diabetes mellitus (DM) induces high levels of ROS production. In DM model rats with fractures, the process of fiber formation and trabecular mineralization was more rapid in the melatonin treatment group compared with the control group (36).

As previously demonstrated, the use of melatonin pre-operatively helped improve the quality sleep of patients and reduced the use of opioid drugs during surgery. The normal circadian rhythm of melatonin secretion can be altered by anesthesia (37). The pre-operative use of melatonin may also have the potential to reduce the incidence of delirium; however, certain existing studies have reached varying conclusions. Al-Aama et al (38) thought exogenous melatonin administered nightly may decrease delirium occurrence in elderly medical in-patients. Sultan (39) pointed out that melatonin was successful in decreasing post-operative delirium. However, the study by de Jonghe et al (40) came to the conclusion that treatment with melatonin did not reduce the incidence of delirium in older-aged patients with hip fracture surgery. Melatonin can be used in combination with various bone graft materials to stimulate bone regeneration in large or comminuted bone injuries (41). In addition, melatonin can significantly protect bones from radiation injury and prevent epiphyseal growth plate damage (42).

The time and dose of melatonin treatment is worthy of consideration, since high doses, such as 50 mg/kg, can cause decreased bone remodeling in mice, thus delaying fracture healing (14). Melatonin supplementation should also be carried out at night as far as possible, to adhere to its natural secretion law, to avoid breaking the normal secretory circadian rhythm, which can have adverse consequences. Evidence from the relevant animal studies is presented in Table I.

Table I

Evidence of the effects of melatonin on bone injury in animal studies.

Table I

Evidence of the effects of melatonin on bone injury in animal studies.

Refs.ObjectivesModelRoute of administrationTime of administrationFrequencyDosesDurationOutcomes
13Fischer ratsCalvarium holesPowder local implantation-Only once10 mg-After 12 weeks: Increased new bone regeneration, neovascularization and osteoblast-like cells
14CD-1 miceFemur fracturei.p.-Daily50 mg/kg2 and 5 weeks2 weeks: Delayed healing 5 weeks: Increased cartilage and callus at the fracture site
15Mice-i.p.-Daily100 mg/kg21 daysMore new femoral neocortical bone formation
17Sprague-Dawley ratsFemoral fracturei.p.MorningDaily30 mg/kg8 weeksPromotive fracture healing and ALP activity, inhibited osteoclasts differentiation
19C57BL/6J miceFemoral fracturei.p.-Daily50 mg/kg5 weeksPromotive fracture healing and ALP activity, inhibited osteoclasts differentiation and obviously bridging callus formation
28Sprague-Dawley ratsFemoral fracturei.p.-Daily30 mg/kg28 daysMore bony union
31Wistar-albino ratsTibia fracturei.p.-Daily25 mg/kg14 daysMelatonin eradicated adverse effects of ischemia on fracture healing
33Albino New Zealand rabbitsTibial defectPowder local implantation-Only once1.2 mg-After 1 week and 2 weeks: Longer cortical bone and more blood vessels formation
36Sprague-Dawley ratsTibial defect and DMi.p.19:00-20:00Daily250 µg10 and 30 days10 days: Higher number of osteoblasts and blood vessels as well as larger new mineralized surface 30 days: Lower level of advanced oxidation protein products and malondialdehyde
42Sprague-Dawley ratsRadiationi.p.30 min prior to radiationDaily15 mg/kg3 days6 weeks later: A superior radioprotective function of melatonin over amifostine in preventing radiation-induced epiphyseal growth plate injury

[i] i.p., intraperitoneal; DM, diabetes mellitus.

3. Osteoporosis

Study basics of osteoporosis

Osteoporosis is considered one of the most common diseases, and is becoming increasingly prevalent with the aging of the global population. Millions of individuals worldwide suffer from osteoporosis, particularly postmenopausal women (43-45) and the elderly (46,47). The reduced bone density and damaged bone architecture caused by osteoporosis can increase the risk of fragility fractures, and lead to a higher morbidity and mortality. A variety of drugs with varying degrees of efficacy and side-effects have been used for the treatment of osteoporosis (48). Even with the existing osteoporosis treatments, the prevalence of osteoporosis is steadily increasing (49), which has created a huge economic burden to societies worldwide (50-52). Thus, novel strategies need to be developed to prevent or combat bone loss for the treatment of osteoporosis and its complications.

Potential effects of melatonin on osteoporosis

Melatonin is involved in the regulation of bone mass accumulation and loss (Fig. 2). Egermann et al (53) confirmed that the bone mass significantly decreases following pinealectomy. The decrease of melatonin secretion is associated with menopause and is one of the most important causes of osteoporosis (54). The production of melatonin decreases with age (55), which may lead to a higher bone loss among the elderly. In addition, the expression of melatonin receptor 1A (MTNR1A) on the surface of human osteoblasts decreases with age, which is more pronounced in women (15).

It has been demonstrated that melatonin supplementation can improve perimenopausal- and age-related osteoporosis. Melatonin supplementation is well-tolerated and can attenuate perimenopausal symptoms, as well as restore the balance of bone remodeling to avoid bone loss and osteoporosis (56). The daily oral melatonin administration (100 mg/kg body weight) has been reported to increase bone formation to prevent ovariectomy-induced bone degeneration in mice (57). Although its efficacy requires further confirmation, melatonin is considered a safe nutritional supplement for peri- and postmenopausal women to improve bone density (58). Moreover, a dietary melatonin supplement has been shown to improve the microstructure and biomechanical properties of the bones of aged rats (59).

Estrogen deficiency, the major characteristic of menopause, contributes to osteoporosis. Increased levels of nucleotide-binding domain and the leucine-rich repeat pyrin 3 domain (NLRP3) inflammasome have been observed in the hippocampus of female mice with estrogen deficiency (60). Melatonin can attenuate osteoporosis induced by estrogen deficiency and can improve the osteoblastic differentiation potential by inhibiting NOD-, LRR- and pyrin domain-containing protein 3 inflammasome activation. The modulation of the Wnt/β-catenin pathway is involved in this process (61).

Type 2 DM and osteoporosis are both negatively affected by aging and lifestyle changes and quite often coexist. A high risk of fracture has been identified in patients with type 2 DM, particularly those with long periods of DM, poor glycemic control and diabetic complications (62,63). Reduced bone remodeling is one of the characteristics of DM, and autophagy is considered to be a potential target for the management of diabetic osteoporosis (64,65). The level of autophagy in osteoblasts may be reduced, and the process of DM-induced osteoporosis may be delayed by melatonin by inhibiting the ERK signaling pathway (66). Patients with multiple sclerosis (MS) have decreased serum melatonin levels and are also at risk of osteoporosis, while melatonin therapy can reduce the risk and normalize bone metabolites in MS (67). In addition to the modulation of bone formation and resorption, there are other effects of melatonin on bone metabolism, such as modulating calcium metabolism to prevent osteoporosis and hypocalcemia (68). Related evidence from relevant human and animal studies is presented in Table II.

Table II

Evidence of the effects of melatonin on osteoporosis in animal and clinical studies.

Table II

Evidence of the effects of melatonin on osteoporosis in animal and clinical studies.

Refs.ObjectivesModelRoute of administrationTime of administrationFrequencyDosesDurationOutcomes
56Perimenopausal women-OralNightDaily3 mg6 monthsThe ratio of type-I collagen cross-linked N-telopeptide (NTX): OCN trended downward to 1:1. Improved physical domain scores
57C57BL/6 mice (female)OvariectomyOral gavage6 weeks after surgeryDaily100 mg/kg6 weeksIncreased bone formation
59Wistar rats-Diluted in drinking water--10 mg/kg/day10 weeksHigher bone volume, bone trabecular number, trabecular thickness, cortical thickness, bone stiffness, flexural modulus and ultimate load
61C57BL/6J mice (female)Ovariectomyi.p.-Daily10, 50 mg/kg8 weeksMelatonin alleviated bone loss in a dose- dependent manner
66Sprague-Dawley ratsType 2 DMi.p.-Daily50, 100 mg/kg4, 8, 12 weeksMelatonin improved the bone microstructure and reduced the level of autophagy (50 mg/kg was better than 100 mg/kg)
67C57BL/6 mice (female)Experimental autoimmune encephalomyelitisi.p.-Daily10 mg/kg13 daysIncreased 25-hydroxyvitamin D, calcium and OCN

[i] DM, diabetes mellitus; OCN, osteocalcin.

4. Osteoarthritis

Basic study of OA

OA accompanied by chronic joint degeneration is one of the most common joint diseases, affecting ~3.8% of the world population (69). Aging, obesity, sex, genetics, diet-related factors, specific bone/joint shapes and numerous other factors may cause the degenerative injury of articular cartilage, and reactive hyperplasia of the articular margin and subchondral bone, which may lead to the occurrence of OA (70-72).

The functions of articular cartilage depend on cartilage ECM, which primarily comprises proteoglycan and Col2α1 (73). OA is characterized by the imbalance between cartilage ECM anabolism and catabolism (74). The development of OA occurs due to the presence of oxidative stress and inflammation (75). For instance, interleukin (IL)-1β is considered a primary inflammatory mediator in joints with local OA, and is involved in the early inflammatory process of OA, inducing chondrocyte metabolic disorders and cartilage dysfunction, ultimately resulting in joint dysfunction (76,77).

The current therapies for OA mainly focus on reducing joint load and large motion, with the aim of relieving symptoms, delaying the pathological process and improving the quality of life of patients with OA (78). Appropriate physical therapy, proper exercise, drug therapy and joint replacement surgery, among others, can be considered as treatment methods. Anti-inflammatory and analgesic drugs can be used to alleviate the symptoms of patients. However, drugs currently used in the treatment of OA, such as glucocorticoids and analgesics, have certain side-effects (79). Therefore, it is necessary to evaluate the risks of drugs and to identify novel types of low-risk drugs.

Potential effects of melatonin on OA

Inflammation plays a crucial role in the pathogenesis of OA, since mild and chronic inflammation have been shown to contribute to the symptoms and progression of OA (80,81). The self-repair ability of cartilage is limited, with the cell-based articular cartilage repair ability in inflamed joints being even lower. Melatonin intervention can partly restore the chondrogenic differentiation ability of MSCs affected by IL-1β-induced inflammation (21,77). In addition, the accumulation of ECM increases due the enhancement of ECM synthesis and the reduction of the degeneration enzyme expression induced by IL-1β (82). The effect of long-term intervention (21 days) is significant. Melatonin can also reduce the phosphorylation of p65 and IκBα, thereby inhibiting downstream NF-κB signaling pathway activation, which plays a key role in metabolism, inflammation and apoptosis (83).

ROS can be detected in the joints of OA model rats, which can cause hyaluronic acid depolymerization and molecular configuration changes, resulting in a decrease in the viscosity of synovial fluid (84). The age-related imbalance in ROS production is responsible for cartilage degradation and chondrocyte death (85). Pro-inflammatory cytokines mediate intracellular ROS production during inflammation, impairing the viability of cells and leading to apoptosis and senescence in various cell types (86). As part of the anti-inflammatory properties of melatonin, the dynamic action of the sirtuin 1 (SIRT1) pathway is notable. Oxidative stress upregulates SIRT1 in chondrocytes, while melatonin can reduce the production of nitric oxide, cyclooxygenase-2, inducible nitric oxide synthase and prostaglandin E2 by decreasing the expression and activity of SIRT1 (87). The expression of SIRT1-dependent nuclear factor of activated T cells 5 and nicotinamide phosphoribosyltransferase in IL-1β-stimulated chondrocytes can be suppressed by melatonin to alleviate OA (88). At the same time, melatonin also functions by activating antioxidant enzymes. Excessive ROS production reduces antioxidant enzyme expression in the progression of OA (89), while melatonin can induce the production of antioxidant enzymes, such as superoxide dismutase (90), while inhibiting ROS production (25), further suppressing oxidative stress.

The improper production of circadian clock-regulated hormones may also be involved in the occurrence of OA (91). The expression of circadian clock genes in chondrocytes is altered during the inflammatory process of OA (92), as the expression peak of brain and muscle ARNT-like 1 (Bmal1) is decreased, while that of period circadian regulator 2 (Per2) is increased (93). Per2 knockdown can reduce the expression of major cartilage degenerative enzymes, suggesting that the high expression of Per2 is one of the reasons for the progression of OA. The decline in Bmal1 expression is also associated with the mechanisms of OA (94), and can be restored by melatonin (95). Another study demonstrated that clock-related gene expression decreased in abnormal cartilage samples, and a nano-molar dose of melatonin restored clock-related gene expression and corrected the abnormal chondrocyte phenotype (95).

Multiple microRNAs (miRNAs/miRs) are involved in OA (96); among these, miR-140-5p has been shown to be expressed in cartilage and plays an important role in the differentiation of chondrocytes and the degeneration of cartilage (97). OA-associated cartilage changes occur in mice that lack miR-140 (98), while the overexpression of miR-140 has been shown to inhibit the synthesis of matrix catabolic enzyme (99). Elevated levels of pro-inflammatory cytokines in cartilage may reduce miR-140 expression (100). The protective roles of melatonin in OA-induced cartilage degradation are partly associated with the upregulation of miR-140 and the activation of the SMAD signaling pathways (82), which can inhibit NF-κB pathways in articular cartilage (101). In addition, other miRNAs participating in the protection of the cartilage, such as miR-526b-3p and miR-590-5p, can be upregulated by melatonin, improving the chondrogenic differentiation of MSCs (102).

An intra-articular glucocorticoid (GC) injection is a method used to alleviate inflammation and chronic pain in patients with OA. However, evidence has suggested that treatment with GCs may only be effective in the short-term, while the long-term use of GCs may not be effective, or may even aggravate cartilage degradation (103,104). It has been reported that dexamethasone-induced ECM degradation in chondrocytes in a dose-dependent manner and reduced the intracellular proportion of nicotinamide adenine dinucleotide (NAD)+/NAD + hydrogen (NADH) and the supernatant concentration of NADP/NADPH. Melatonin pre-treatment can reverse these negative effects, possibly via the NAD+-dependent activation of SIRT1 (105), which can promote chondrocyte survival and ECM synthesis to prevent dexamethasone-induced damage to chondrocytes. On the other hand, NADPH oxidase has been shown to mediate the proliferation of multiple cell types, including stem cells; however, it is a major source of ROS (106). Melatonin can effectively regulate the ROS production levels of NADPH oxidase. In general, NADPH oxidase produces ROS at non-cytotoxic levels; when ROS production increases to a harmful level, excess ROS are removed by melatonin (25).

Transforming growth factor (TGF)-β is considered to be one of the synthetic factors engaged in cartilage maintenance (107). A large amount of TGF-β is present in healthy cartilage; however, the level of TGF-β is decreased in OA-affected cartilage (108). Although TGF-β exerts protective effects on cartilage, it has been proven that it can also be a destructive factor (109). Following the exogenous melatonin (1-10 ng/ml) treatment of porcine articular chondrocytes, the level of intracellular TGF-β1 has been shown to increase. This suggests that melatonin can promote porcine chondrocyte ECM synthesis, possibly through the TGF-β signaling pathway, and the use of melatonin instead of TGF-β in the treatment of OA may provide a suitable amount of TGF-β and reduce adverse reactions (110).

Melatonin can be detected in synovial fluid (111), indicating that melatonin secreted in its natural state can reach the articular cavity. The nutrition of chondrocytes in articular cartilage mainly originates from the synovial fluid, and therefore an intra-articular melatonin injection may be a potentially effective method of melatonin therapy for OA. However, melatonin treatment may only be effective in the short-term and at low concentrations. When melatonin stimulation persists, the proteolytic cleavage of RANKL proteins in the synovium is promoted, causing severe subchondral bone erosion (95). Low concentrations of melatonin, such as 1 nM, can restore Col2α1 expression by inhibiting matrix metalloproteinase (MMP)-13; on the contrary, high concentrations, such as 1 mM, cannot rescue the reduced expression of Col2α1 caused by tumor necrosis factor (TNF)-α exposure (112). ROS generation is promoted if the concentration of melatonin is too high or the incubation time is too long (113). It has been reported that pharmacological concentrations (µm-mM) of melatonin promote the production of ROS and pro-inflammatory cytokines (114). The results of the study by Liu et al (25) demonstrated that short-term melatonin treatment (5 days) promoted MSC proliferation, while long-term culture had no significant effect. These results suggested extra attention should be paid to the dose and duration of melatonin therapy, in order to avoid severe side-effects. Mild to moderate exercise can inhibit the inflammatory processes through joint compression-mediated biomechanical stimulation, helping to reduce the negative effects of high-dose and/or long-term melatonin use. There may be a synergistic effect on cartilage protection between melatonin therapy and treadmill exercise, which appears to be more effective in the early stage of the disease (112). Moreover, melatonin-combined exercise can not only restore disordered molecular clocks and correct cartilage abnormalities, but also reduce periarticular bone defects and maintain articular bone homeostasis in late-stage OA (95). However, only mild to moderate exercise is beneficial, as strenuous daily exercise aggravates OA (Fig. 3) (115). Evidence on this matter from relevant animal studies is presented in Table III.

Figure 3

Pathogenesis of OA and potential roles of melatonin. The direct cause of OA is the decrease of cartilage ECM. Inflammation and strenuous exercise lead to an increased ROS production, activating oxidative stress, inhibiting chondrocytes viability, and decreasing the production of ECM components. Pro-inflammatory cytokines promote chondrocytes hypertrophy and then reduce ECM synthesis. Additionally, pro-inflammatory cytokines decrease miR-140 production in chondrocytes, inhibit the viability and chondrogenic differentiation of MSCs and increase the production of ECM degradation enzymes. All these mechanisms promote the loss of ECM. Oxidative stress also leads to decreased viscosity of synovial fluid. Pro-inflammatory cytokines are also associated with pain and hyperalgesia. The abnormal expression of clock-related genes, such as the decrease in Bmal1 expression and the increase in Per2 expression, is also considered to be related to the occurrence of OA. Melatonin application can effectively antagonize the above-mentioned processes, reducing the level of oxidative stress and inflammation and restoring the normal expression of clock genes. Moreover, melatonin can upregulate TGF-β expression to increase ECM synthesis. Intra-articular injections of glucocorticoids can relieve inflammation and pain, but may also be associated with the risk of aggravating ECM loss. Melatonin can alleviate this adverse reaction. In addition, melatonin treatment combined with mild to moderate exercise may do better in OA therapeutic process. OA, osteoarthritis; ECM, extracellular matrix; ROS, reactive oxygen species; Bmal1, brain and muscle ARNT-like 1; Per2, period circadian regulator 2; TGF-β, transforming growth factor β.

Table III

Evidence of the effects of melatonin on osteoarthritis in animal studies.

Table III

Evidence of the effects of melatonin on osteoarthritis in animal studies.

Refs.ObjectivesModelRoute of administrationTime of administrationFrequencyDosesDurationOutcomes
82C57BL/6J miceSurgically-induced osteoarthritisIntra-articular injectionAfter surgeryTwice a week10 mg/ml (10 µl)4 weeksAttenuated OA progression
87New Zealand white rabbits (female)Surgically-induced osteoarthritisIntra-articular injectionBeginning on the day of surgeryWeekly20 mg/kg4 weeksReduced cartilage degradation
88Lewis ratsSurgically-induced osteoarthritisIntra-articular injectionOn day 3 following the surgeryOnce10 mg/ml (20 µl)-Repressed expression of relevant genes in rat OA pathogenesis after 3 weeks
95Sprague-Dawley ratsIntra-articular collagenase injection- induced osteoarthritisSubcutaneous injection-Twice daily10 mg/kg4 weeksMelatonin prevented periarticular muscle damage and cartilage degeneration. But prolonged melatonin administration leaded to subchondral bone erosion
112Sprague Dawley ratsIntra-articular collagenase injection- induced osteoarthritisSubcutaneous injection07:00 and 19:00Twice daily10 mg/kg1, 4 weeksMelatonin with treadmill exercise may have both preventive and synergistic effects on rescue from cartilage degeneration and is more effective in the initial phase (1 week)

5. Periodontitis

Basic study of periodontitis

Periodontitis, an inflammatory and destructive disease of the periodontal tissues, is characterized by the loss of periodontal attachment. Periodontitis is also considered as inflammation of the alveolar bone, since marginal alveolar bone loss is a key secondary feature of periodontitis, with teeth loosening and complete loss occurring at the terminal stage. The common characteristics of periodontitis usually include redness in the periodontal tissue, pain, pus overflow, halitosis and dental stone formation, which progresses to alveolar bone resorption. Dental plaque is the primary initiating factor of periodontitis, creating and maintaining an inflammatory environment in the periodontal area. Gingival inflammation (gingivitis) induced by dental plaque is the most common and mildest type of periodontal disease, with the potential to develop into periodontitis in the case of no intervention. ROS generation is another important feature of periodontitis. Oral bacteria, as well as the inflammatory and immune reaction, lead to the generation of ROS, contributing to the progression of the disease (116,117).

Scaling and root planning (SRP) is considered an extremely effective basic treatment method for periodontitis. Generally, adherence to basic periodontal therapy can improve periodontal status in most patients with chronic periodontitis. However, in certain patients, progressive attachment loss cannot be terminated by SRP alone (118). For this reason, adjuvant treatment with SRP should be considered.

Potential effects of melatonin on periodontitis

Saliva and plasma melatonin levels are significantly lower in individuals with periodontal disease compared to clinically healthy subjects (119); however, the ratio of saliva and plasma melatonin levels is similar to that in healthy subjects (120). In a previous study, serum melatonin was introduced into the oral cavity through the salivary glands at a stable proportion (~33%, the ratio of salivary and serum melatonin) (121). Moreover, the gingiva is one of the external sites of melatonin synthesis, and melatonin receptor 1 has been found in human gingiva, indicating that melatonin may play a receptor-mediating role in the oral cavity (122). Certain studies have suggested that melatonin may serve as a potential supplementary therapy and a biomarker detecting the dynamics of periodontal disease (123,124).

Melatonin levels in saliva and gingival crevicular fluid vary inversely with the severity of periodontitis (125,126), suggesting that melatonin may provide protective effects against the destruction of periodontal tissues. Srinath et al (119) suggested that melatonin had antibacterial properties, since Prevotella intermedia, Streptococcus mutans and Porphyromonas gingivalis, the primary bacteria in the occurrence and progression of periodontitis, were sensitive to melatonin.

In a previous study, patients with severe periodontitis received nonsurgical periodontal therapy (NSPT) following the oral administration of melatonin at 1 mg/day (a dietary supplement dosage advised by the Italian Ministry of Health) for 1 month (127). All patients were able to tolerate melatonin well, demonstrating a significant reduction in probing depth (PD) within 6 months, suggesting that melatonin supplementation is likely to promote the healing process of periodontal pockets following long-term treatment (127). A small number of mild adverse reactions were observed at the initial stage of oral administration, which disappeared within a few days without affecting the compliance of the patients (127). Another study reported that, when combined with NSPT, the oral administration of 2 mg melatonin daily for 30 days improved the clinical attachment level (CAL) and PD significantly following long-term treatment (3 and 6 months) (128). Bazyar et al (129) obtained similar results with 6 mg melatonin daily administration daily with NPST. In a rat model of periapical periodontitis, following an i.p. injection of melatonin (10 mg/kg) for 21 days, radiological periapical bone loss and osteoclasts were decreased, the OPG level was increased, and the IL-1β, RANK and RANKL levels were decreased, as compared with the positive control. In addition, the bacteria localization score has been shown to be significantly lower following melatonin treatment (130). Renn et al (131) reported that preventive melatonin supplementation suppressed the Toll-like receptor 4/myeloid differentiation factor 88 pathway to inhibit the activation of pro-inflammatory cytokines and normalize the balance between RANKL and OPG, thus suppressing the progression of periodontitis.

There may be a bidirectional association between DM and periodontitis, both of which are common chronic diseases. According to previous studies, DM is considered to be a risk factor of periodontitis development, increasing the prevalence and severity and promoting the progression of periodontitis (132,133). On the other hand, periodontitis may also increase the complications of DM (134). Patients with DM or periodontal disease have reduced melatonin levels in serum and saliva, and when the two diseases coexist, these levels are further reduced (135). In a previous study, in patients with mild to moderate periodontitis with DM, melatonin supplementation at 6 mg once a day for 8 weeks clearly increased the serum melatonin levels; moreover, PD, CAL loss and the high sensitivity-C reactive protein and IL-6 levels were reduced during NSPT (129). Balci Yuce et al (136) reported that melatonin decreased osteoclasts and inhibited alveolar bone resorption in rats suffering from both DM and periodontitis; however, no decrease in bone loss was observed in rats with periodontitis alone. Oral local melatonin application was found to delay bone loss during periodontitis in patients with DM by downregulating pro-inflammatory factors (137,138).

There is a clear association between periodontitis and obesity. The prevalence of periodontitis and degree of inflammation in obese or overweight patients seem to be higher compared to individuals with normal weight (139,140). An experimental study reported that, when the two diseases coexisted, a significant elevation was observed in periodontal destruction, lipid dysbolism, glucose levels and hepatic damage parameters, thus revealing comorbidity effects (141). These comorbidity effects may be associated with the circadian clock (142). Considered as an important modulator of the circadian clock, melatonin may be a key mechanism in this comorbidity effect (11,143).

Reduced levels of melatonin have been found to be associated with obesity (144). On the other hand, patients with periodontitis, and aggressive periodontitis in particular, are likely to have significantly lower levels of melatonin in saliva and gingival crevicular fluid (120,123). Melatonin supplementation helps restore lipid and glucose metabolism, reduce pro-inflammatory factor expression, improve body weight control and avoid obesity-related complications in obese patients (144-146). Rats with obesity or periodontitis have been shown to exhibited significantly lower circulating melatonin levels, although these levels are further reduced in rats with both obesity and periodontitis (147). In addition, a markedly increased destruction of periodontal tissue was observed in rats suffering from both obesity and periodontitis, with evident inflammatory infiltration and osteoclastic activity (147). There were almost significant negative associations between circulating melatonin levels and periodontal pocket depth, dental plaque index and modified gingival index (147). Combined therapy with SRP and melatonin supplementation significantly reduced alveolar bone destruction and pro-inflammatory cytokines in rats with comorbidities of obesity and periodontitis, providing a protective effect (148). When periodontitis, pinealectomy, or a combination of both are present, the TNF and insulin concentration, as well as the homeostasis model assessment of insulin resistance index, are increased, indicating insulin resistance (149). Pineal excision can also lead to lipid profile dysregulation, which may be improved to a similar degree to the control group by melatonin alternative therapy, indicating that alternative melatonin therapy provides a therapeutic effect against dyslipidemia (149).

Periodontal ligament cells can secrete various cytokines to modulate and maintain the homeostasis of periodontal tissues, thereby playing an arrestive role in alveolar bone metabolism (150). Periodontal tissue regeneration can be enhanced by conditioned medium from periodontal ligament stem cells in a concentration-dependent manner by suppressing TNF-α production (151). There is an important balance between cementum formation and bone loss during the maintenance of periodontal health. Melatonin inhibited ethanol-induced ROS production and senescence-like phenotypes in human periodontal ligament stem cells and cementoblasts. In addition, it restored the decreased osteoblastic/cementoblastic differentiation, and increased osteoclastic differentiation through the protein never in mitosis gene A interacting-1 pathway. Furthermore, the downregulation of certain pathways, such as the MAPK, AMP-activated protein kinase, mammalian target of rapamycin (mTOR) and nuclear factor of activated T-cells c-1 pathways, has been suggested to exert protective effects against ethanol-induced senescence (152).

El-Sharkawy et al (153) reported that a daily dietary supplement of 10 mg melatonin may be an effective complementary treatment for patients with insomnia with generalized chronic periodontitis, resulting in an improved CAL and sleep quality, as well as lower PD and salivary TNF-α levels. During the entire study period, the improvement in insomnia was maintained for up to 6 months without any rebound, even though the daily dietary melatonin supplement was administered for only 2 months. As shown by previous data, the level of systemic inflammatory markers in sleep disorders increased significantly (154). There may be a certain degree of bidirectional association between sleep disorders and periodontal disease. Therefore, improving sleep quality may itself improve the response to periodontal therapy (Fig. 4). Evidence from relevant human and animal studies on this matter are presented in Table IV.

Table IV

Evidence of the effects of melatonin on periodontitis in animal and clinical studies.

Table IV

Evidence of the effects of melatonin on periodontitis in animal and clinical studies.

Refs.ObjectivesModelRoute of administrationTime of administrationFrequencyDosesDurationOutcomes
127Patients with untreated severe periodontitisWith NSPTOralAfter NSPTDaily1 mg1 monthPD reduced even at 6 months
128Patients with chronic periodontitisWith NSPTOralAfter NSPTDaily2 mg4 weeksImproved CAL and lower PD in a long time as 3 and even 6 months
129Patients with chronic periodontitis and type 2 DMWith NSPTOral1 h before bed timeDaily6 mg8 weeksImproved CAL and lower PD
130Sprague-Dawley ratsPeriapical lesionsi.p.-Daily10 mg/kg21 daysReduced radiological periapical bone loss
131Wistar ratsExperimental periodontitisi.p.10:00-10:30Daily10, 50, 100 mg/kg14 and 28 daysReduced pro-inflammatory cytokine level and suppressed progression of periodontitis
136Wistar ratsExperimental DM and periodontitisi.p.-Daily10 mg/kg4 weeksInhibited resorption of alveolar bone
137Patients with periodontal disease and DM-Topical application-Daily1% orabase cream20 daysSignificant decrease of the gingival index, PD and salivary levels of RANKL, and significant rise of salivary OPG
138Patients with periodontal disease and DM-Topical application-Daily1% orabase cream21 daysStatistically significant decrease of the gingival index, PD, and IL-1β, IL-6 and prostaglandin E2 in gingival crevicular fluid
148Wistar ratsObesity and periodontitisDissolved in drinking water-Daily25 µg/ml4 weeksCombined therapy of SRP and melatonin supplement significantly reduced alveolar bone destruction and proinflammatory cytokines
149Wistar albino ratsPinealectomy and periodontitisDissolved in drinking water7:00 p.m. - 7:00 a.m.Daily5 mg/kg28 daysMelatonin efficiently prevented insulinresistance, improved lipid profile, and increased plasma levels of insulin and TNF
153Patients with generalized chronic periodontitis and primary insomniaWith SRPOral1 h before bed timeDaily10 mg2 monthsGreater CAL and sleep quality, lower PD and salivary TNF-α levels

[i] NSPT, nonsurgical periodontal therapy; PD, probing depth; CAL, clinical attachment level; DM, diabetes mellitus; OPG, osteoprotegerin; IL, interleukin; SRP, scaling and root planning; TNF, tumor necrosis factor.

6. Differential effects of melatonin administered at various concentrations and times

In the majority of previous studies, melatonin has been found to play a positive role in bone tissue and bone-related diseases. Melatonin supplementation in humans has a generally favorable safety profile. Clinical studies have demonstrated that the use of melatonin in the short-(days) and medium-term (weeks to months) is safe, with only minor, transient adverse reactions reported (155). In addition to the most commonly reported adverse reactions, which are related to fatigue, mood and psychomotor or neurocognitive performance, a few studies have reported adverse events associated with endocrine and cardiovascular function, as summarized in a critical systematic review of clinical evidence (156). The safety of melatonin application in pregnant and lactating women is unknown, due to the lack of relevant research.

Although there is considerable evidence to suggest that melatonin exerts a positive effect on bone health, certain studies have reached different conclusions. Frisher et al (157) reported an association between melatonin and a significantly increased risk of fracture. In addition, in that study, it was shown that the concentration and administration time may impact the effects of melatonin treatment. As shown in Tables I-IV, the usual strategy for animal experiments is i.p. injection at doses between 10-50 mg/kg. A small number of studies have used higher doses, such as 100 mg/kg (15,66,131). The intra-articular injection is a method commonly used in studies on OA, where the doses are lower (0.1-0.2 µg) (82,88). In addition, the normal dose used for subcutaneous injection is 10 mg/kg (95,112). A few studies have used other methods, such as oral gavage (57), the addition of melatonin to drinking water (59,148,149) and melatonin powder implantation (13,33). Apart from one-off administration (13,33), the shortest duration of melatonin treatment was 3 days (42), and the longest being up to 12 weeks (66). The majority of experiments used melatonin for 2-4 weeks. For clinical research, the most common route is by oral administration, with doses fluctuating between 1-10 mg/day (56,127-129,153). In addition, melatonin can be used locally in the form of 1% orabase cream for the treatment of periodontitis (137,138). The drug treatment durations were ~1-2 months, and 6 months in one study (56).

To date, there is no consensus on the optimal route, dosage and time of melatonin administration. Further research is therefore required to explore the optimal route, dosage and administration time of melatonin, and determine whether long-term melatonin supplementation has any adverse effects, as well as whether melatonin can be used as a daily adjuvant in elderly, perimenopausal and postmenopausal women, and in patients with periodontitis.

7. Conclusion and future prospects

Melatonin is a common molecule mainly produced by the pineal gland. As an important regulatory factor of circadian rhythm, melatonin has the ability to synchronize and maintain the circadian clock in peripheral tissues with L/D cycles. In addition, melatonin is also considered cytoprotective, due to its anti-inflammatory, antitumor and antioxidant effects, and its ability to regulate hormones, the immune system and tissue regeneration.

Bone-related diseases have a high incidence, and are associated with severe and persistent symptoms, a slow recovery and a high impact on the lives of patients, as well as a heavy economic burden. The most common treatment usually exerts a curative effect and some side-effects. Since melatonin is inexpensive, with a wide safety margin, has a wide impact on tissues and almost no side-effects, the use of melatonin as a supplementary treatment may be a potential therapeutic option for bone disease (Fig. 5). As a widely available and versatile molecule in vivo, melatonin has potent antioxidant and anti-inflammatory properties in a variety of bone diseases. In addition, melatonin also plays a vital role in promoting osteogenesis and inhibiting osteoclastogenesis (13,14). The promotion of vascularization will also provide a good boost for bone repair (31). A close association has been identified between cartilage and bone. Subchondral osteogenesis is an important type of osteogenesis (21), and cartilage and bone metabolic disorder is a common challenge in OA (72). Melatonin also protects and promotes the differentiation of cartilage (77), which is important for bone regeneration and bone development. Melatonin always drives MSCs to differentiate toward osteoblasts (17-19) and chondroblasts (18,21,77) and protects MSCs from disease-induced apoptosis (25,27,83). Patients with bone disease may also have other chronic metabolic diseases, such as diabetes (62,63,132-134), which have adverse effects on bone health and lead to comorbidity effects. Despite these comorbidity effects, melatonin can still have beneficial effects (66,129,136), which provides a novel insight for future multi-disease combination therapy.

Stomatologists pay great attention to the protection of periodontal tissue. The protective effects of melatonin on periodontal tissue are significant, helping patients with periodontitis retain more alveolar bone, providing more possibilities for natural tooth retention and subsequent tooth defect repair. In addition, the protective effects of melatonin on alveolar bone can also accelerate the process of osteointegration during implant restoration, to obtain a better implantation effect (158). Melatonin can be used to inhibit the enhancement of oxidative stress in oral tissues in the period immediately following tooth extraction to avoid excessive alveolar bone loss (159).

Human tissues are regulated by a circadian rhythm. In particular, bone metabolism is closely associated with the circadian rhythm, including the development, repair and remodeling of bone tissue; cartilage metabolism is also regulated in a similar manner (160-162). The disruption of the circadian rhythm leads to a series of adverse effects. As an important hormone regulating the circadian rhythm, melatonin helps restore the circadian rhythm (143). Sleep disorders and depression are also common symptoms among the elderly (163) and perimenopausal women (164). Melatonin can improve the sleep quality and mental health of patients, thus improving patient compliance; thus, the efficacy of melatonin treatment can be further consolidated.

In conclusion, melatonin functions as a protector in bone injury, osteoporosis, OA and periodontitis by exerting multiple effects. Melatonin supplementation in humans has a generally favorable safety profile. Due to the protective effects of melatonin, as well as its low price and high safety, exploring melatonin as a supplement to bone tissue and bone-related disease therapy is worthwhile. However, since a few studies have reported adverse effects, and there is no consensus on the optimal program of melatonin administration, further research is required to explore the optimal administration conditions and safety of long-term melatonin supplementation.

Availability of data and materials

Not applicable.

Authors' contributions

XL was involved in data curation, investigation and visualization, and in the writing of the original draft. SY was involved in the conceptualization of the study, as well as in the preparation of the figures, writing of the original draft, and in the writing, reviewing and editing of the manuscript. GC, WZ, JP and XH were involved in in the writing, reviewing and editing of the present review article. LC was involved in the conceptualization of the study, as well as in funding acquisition, project administration, study supervision, and in the writing, reviewing and editing of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China for Distinguished Young Scholars (grant no. 31725011 to LC).

Abbreviations:

ALP

alkaline phosphatase

CAL

clinical attachment level

DM

diabetes mellitus

ECM

extracellular matrix

GC

glucocorticoid

i.p. injection

intraperitoneal injection

L/D

light/dark

MSCs

mesenchymal stem cells

MTNR1A

melatonin receptor 1A

NSPT

nonsurgical periodontal therapy

OA

osteoarthritis

OCN

osteocalcin

OPG

osteoprotegerin

PD

probing depth

RANKL

receptor activator of NF-κB ligand

ROS

reactive oxygen species, SRP, scaling and root planning

TGF

transforming growth factor

TNF

tumor necrosis factor

VEGF

vascular endothelial growth factor

References

1 

Maria S and Witt-Enderby PA: Melatonin effects on bone: Potential use for the prevention and treatment for osteopenia, osteoporosis, and periodontal disease and for use in bone-grafting procedures. J Pineal Res. 56:115–125. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Johnell O and Kanis JA: An estimate of the worldwide prevalence and disability associated with osteoporotic fractures. Osteoporos Int. 17:1726–1733. 2006. View Article : Google Scholar : PubMed/NCBI

3 

Kanis JA: Diagnosis of osteoporosis and assessment of fracture risk. Lancet. 359:1929–1936. 2002. View Article : Google Scholar : PubMed/NCBI

4 

Meng X, Li Y, Li S, Zhou Y, Gan RY, Xu DP and Li HB: Dietary sources and bioactivities of melatonin. Nutrients. 9:3672017. View Article : Google Scholar :

5 

Cipolla-Neto J and Amaral FGD: Melatonin as a hormone: New physiological and clinical insights. Endocr Rev. 39:990–1028. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Reiter RJ: Pineal melatonin: Cell biology of its synthesis and of its physiological interactions. Endocr Rev. 12:151–180. 1991. View Article : Google Scholar : PubMed/NCBI

7 

Amaral FGD and Cipolla-Neto J: A brief review about melatonin, a pineal hormone. Arch Endocrinol Metab. 62:472–479. 2018. View Article : Google Scholar : PubMed/NCBI

8 

Simonneaux V and Ribelayga C: Generation of the melatonin endocrine message in mammals: A review of the complex regulation of melatonin synthesis by norepinephrine, peptides, and other pineal transmitters. Pharmacol Rev. 55:325–395. 2003. View Article : Google Scholar : PubMed/NCBI

9 

Tan DX, Manchester LC, Hardeland R, Lopez-Burillo S, Mayo JC, Sainz RM and Reiter RJ: Melatonin: A hormone, a tissue factor, an autocoid, a paracoid, and an antioxidant vitamin. J Pineal Res. 34:75–78. 2003. View Article : Google Scholar

10 

Permuy M, López-Peña M, González-Cantalapiedra A and Muñoz F: Melatonin: A review of its potential functions and effects on dental diseases. Int J Mol Sci. 18:8652017. View Article : Google Scholar :

11 

Tordjman S, Chokron S, Delorme R, Charrier A, Bellissant E, Jaafari N and Fougerou C: Melatonin: Pharmacology, functions and therapeutic benefits. Curr Neuropharmacol. 15:434–443. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Nauth A, Schemitsch E, Norris B, Nollin Z and Watson JT: Critical-size bone defects: Is there a consensus for diagnosis and treatment? J Orthop Trauma. 32(Suppl 1): S7–S11. 2018. View Article : Google Scholar : PubMed/NCBI

13 

Shino H, Hasuike A, Arai Y, Honda M, Isokawa K and Sato S: Melatonin enhances vertical bone augmentation in rat calvaria secluded spaces. Med Oral Patol Oral Cir Bucal. 21:e122–e126. 2016. View Article : Google Scholar :

14 

Histing T, Anton C, Scheuer C, Garcia P, Holstein JH, Klein M, Matthys R, Pohlemann T and Menger MD: Melatonin impairs fracture healing by suppressing RANKL-mediated bone remodeling. J Surg Res. 173:83–90. 2012. View Article : Google Scholar

15 

Satomura K, Tobiume S, Tokuyama R, Yamasaki Y, Kudoh K, Maeda E and Nagayama M: Melatonin at pharmacological doses enhances human osteoblastic differentiation in vitro and promotes mouse cortical bone formation in vivo. J Pineal Res. 42:231–239. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Sethi S, Radio NM, Kotlarczyk MP, Chen CT, Wei YH, Jockers R and Witt-Enderby PA: Determination of the minimal melatonin exposure required to induce osteoblast differentiation from human mesenchymal stem cells and these effects on downstream signaling pathways. J Pineal Res. 49:222–238. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Dong P, Gu X, Zhu G, Li M, Ma B and Zi Y: Melatonin induces osteoblastic differentiation of mesenchymal stem cells and promotes fracture healing in a rat model of femoral fracture via neuropeptide Y/neuropeptide Y receptor Y1 signaling. Pharmacology. 102:272–280. 2018. View Article : Google Scholar : PubMed/NCBI

18 

Luchetti F, Canonico B, Bartolini D, Arcangeletti M, Ciffolilli S, Murdolo G, Piroddi M, Papa S, Reiter RJ and Galli F: Melatonin regulates mesenchymal stem cell differentiation: A review. J Pineal Res. 56:382–397. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Zhu G, Ma B, Dong P, Shang J, Gu X and Zi Y: Melatonin promotes osteoblastic differentiation and regulates PDGF/AKT signaling pathway. Cell Biol Int. 44:402–411. 2020. View Article : Google Scholar

20 

Park KH, Kang JW, Lee EM, Kim JS, Rhee YH, Kim M, Jeong SJ, Park YG and Kim SH: Melatonin promotes osteoblastic differentiation through the BMP/ERK/Wnt signaling pathways. J Pineal Res. 51:187–194. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Gao W, Lin M, Liang A, Zhang L, Chen C, Liang G, Xu C, Peng Y, Chen C, Huang D and Su P: Melatonin enhances chondrogenic differentiation of human mesenchymal stem cells. J Pineal Res. 56:62–70. 2014. View Article : Google Scholar

22 

Zhang B, Bailey WM, McVicar AL and Gensel JC: Age increases reactive oxygen species production in macrophages and potentiates oxidative damage after spinal cord injury. Neurobiol Aging. 47:157–167. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Fraser JH, Helfrich MH, Wallace HM and Ralston SH: Hydrogen peroxide, but not superoxide, stimulates bone resorption in mouse calvariae. Bone. 19:223–226. 1996. View Article : Google Scholar : PubMed/NCBI

24 

Lee NK, Choi YG, Baik JY, Han SY, Jeong DW, Bae YS, Kim N and Lee SY: A crucial role for reactive oxygen species in RANKL-induced osteoclast differentiation. Blood. 106:852–859. 2005. View Article : Google Scholar : PubMed/NCBI

25 

Liu X, Gong Y, Xiong K, Ye Y, Xiong Y, Zhuang Z, Luo Y, Jiang Q and He F: Melatonin mediates protective effects on inflammatory response induced by interleukin-1 beta in human mesenchymal stem cells. J Pineal Res. 55:14–25. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Liu XW, Zi Y, Liu YE, Zhang YB, Xiang LB and Hou MX: Melatonin exerts protective effect on N2a cells under hypoxia conditions through Zip1/ERK pathway. Neurosci Lett. 595:74–80. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Xu S, Yang Y, Han S and Wu Z: ZIP1 and zinc inhibits fluoride-induced apoptosis in MC3T3-E1 cells. Biol Trace Elem Res. 159:399–409. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Halıcı M, Öner M, Güney A, Canöz Ö, Narin F and Halıcı C: Melatonin promotes fracture healing in the rat model. Eklem Hastalik Cerrahisi. 21:172–177. 2010.

29 

Quesnelle KM, Bystrom PV and Toledo-Pereyra LH: Molecular responses to ischemia and reperfusion in the liver. Arch Toxicol. 89:651–657. 2015. View Article : Google Scholar : PubMed/NCBI

30 

Bagheri F, Khori V, Alizadeh AM, Khalighfard S, Khodayari S and Khodayari H: Reactive oxygen species-mediated cardiac-reperfusion injury: Mechanisms and therapies. Life Sci. 165:43–55. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Erdem M, Gulabi D, Asci M, Bostan B, Gunes T and Koseoglu RD: The effects of melatonin and caffeic acid phenethyl ester (CAPE) on fracture healing under ischemic conditions. Acta Orthop Traumatol Turc. 48:339–345. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Son JH, Cho YC, Sung IY, Kim IR, Park BS and Kim YD: Melatonin promotes osteoblast differentiation and mineralization of MC3T3-E1 cells under hypoxic conditions through activation of PKD/p38 pathways. J Pineal Res. 57:385–392. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Ramírez-Fernández MP, Calvo-Guirado JL, de-Val JE, Delgado-Ruiz RA, Negri B, Pardo-Zamora G, Peñarrocha D, Barona C, Granero JM and Alcaraz-Baños M: Melatonin promotes angiogenesis during repair of bone defects: A radiological and histomorphometric study in rabbit tibiae. Clin Oral Investig. 17:147–158. 2013. View Article : Google Scholar

34 

Melincovici CS, Boşca AB, Şuşman S, Mărginean M, Mihu C, Istrate M, Moldovan IM, Roman AL and Mihu CM: Vascular endothelial growth factor (VEGF)-key factor in normal and pathological angiogenesis. Rom J Morphol Embryol. 59:455–467. 2018.

35 

Pugazhenthi K, Kapoor M, Clarkson AN, Hall I and Appleton I: Melatonin accelerates the process of wound repair in full-thickness incisional wounds. J Pineal Res. 44:387–396. 2008. View Article : Google Scholar : PubMed/NCBI

36 

Yildirimturk S, Batu S, Alatli C, Olgac V, Firat D and Sirin Y: The effects of supplemental melatonin administration on the healing of bone defects in streptozotocin-induced diabetic rats. J Appl Oral Sci. 24:239–249. 2016. View Article : Google Scholar : PubMed/NCBI

37 

de Carvalho Nogueira EF, de Oliveira Vasconcelos R, Teixeira Correia SS, Souza Catunda I, Amorim JA and do Egito Cavalcanti Vasconcelos B: Is there a benefit to the use of melatonin in preoperative zygomatic fractures? J Oral Maxillofac Surg. 77:2017.e1–2017.e7. 2019. View Article : Google Scholar

38 

Al-Aama T, Brymer C, Gutmanis I, Woolmore-Goodwin SM, Esbaugh J and Dasgupta M: Melatonin decreases delirium in elderly patients: A randomized, placebo-controlled trial. Int J Geriatr Psychiatry. 26:687–694. 2011. View Article : Google Scholar

39 

Sultan SS: Assessment of role of perioperative melatonin in prevention and treatment of postoperative delirium after hip arthroplasty under spinal anesthesia in the elderly. Saudi J Anaesth. 4:169–173. 2010. View Article : Google Scholar : PubMed/NCBI

40 

de Jonghe A, van Munster BC, Goslings JC, Kloen P, van Rees C, Wolvius R, van Velde R, Levi M, de Haan RJ and de Rooij SE; Amsterdam Delirium Study Group: Effect of melatonin on incidence of delirium among patients with hip fracture: A multicentre, double-blind randomized controlled trial. CMAJ. 186:E547–E556. 2014. View Article : Google Scholar : PubMed/NCBI

41 

Majidinia M, Reiter RJ, Shakouri SK, Mohebbi I, Rastegar M, Kaviani M, Darband SG, Jahanban-Esfahlan R, Nabavi SM and Yousefi B: The multiple functions of melatonin in regenerative medicine. Ageing Res Rev. 45:33–52. 2018. View Article : Google Scholar : PubMed/NCBI

42 

Topkan E, Tufan H, Yavuz AA, Bacanli D, Onal C, Kosdak S and Yavuz MN: Comparison of the protective effects of melatonin and amifostine on radiation-induced epiphyseal injury. Int J Radiat Biol. 84:796–802. 2008. View Article : Google Scholar : PubMed/NCBI

43 

Rachner TD, Khosla S and Hofbauer LC: Osteoporosis: Now and the future. Lancet. 377:1276–1287. 2011. View Article : Google Scholar : PubMed/NCBI

44 

Cipriani C, Pepe J, Bertoldo F, Bianchi G, Cantatore FP, Corrado A, Di Stefano M, Frediani B, Gatti D, Giustina A, et al: The epidemiology of osteoporosis in Italian postmenopausal women according to the National Bone Health Alliance (NBHA) diagnostic criteria: A multicenter cohort study. J Endocrinol Invest. 41:431–438. 2018. View Article : Google Scholar

45 

Parizad N, Baghi V, Karimi EB and Ghanei Gheshlagh R: The prevalence of osteoporosis among Iranian postmenopausal women with type 2 diabetes: A systematic review and meta-analysis. Diabetes Metab Syndr. 13:2607–2612. 2019. View Article : Google Scholar : PubMed/NCBI

46 

Melton LJ III: The prevalence of osteoporosis: Gender and racial comparison. Calcif Tissue Int. 69:179–181. 2001. View Article : Google Scholar : PubMed/NCBI

47 

Coughlan T and Dockery F: Osteoporosis and fracture risk in older people. Clin Med (Lond). 14:187–191. 2014. View Article : Google Scholar

48 

Liu GF, Wang ZQ, Liu L, Zhang BT, Miao YY and Yu SN: A network meta-analysis on the short-term efficacy and adverse events of different anti-osteoporosis drugs for the treatment of postmenopausal osteoporosis. J Cell Biochem. 119:4469–4481. 2018. View Article : Google Scholar

49 

Cui Z, Meng X, Feng H, Zhuang S, Liu Z, Zhu T, Ye K, Xing Y, Sun C, Zhou F and Tian Y: Estimation and projection about the standardized prevalence of osteoporosis in mainland China. Arch Osteoporos. 15:22019. View Article : Google Scholar : PubMed/NCBI

50 

Lane NE: Epidemiology, etiology, and diagnosis of osteoporosis. Am J Obstet Gynecol. 194(2 Suppl): S3–S11. 2006. View Article : Google Scholar : PubMed/NCBI

51 

Mohd-Tahir NA and Li SC: Economic burden of osteoporosis-related hip fracture in Asia: A systematic review. Osteoporos Int. 28:2035–2044. 2017. View Article : Google Scholar : PubMed/NCBI

52 

Hopkins RB, Burke N, Von Keyserlingk C, Leslie WD, Morin SN, Adachi JD, Papaioannou A, Bessette L, Brown JP, Pericleous L and Tarride J: The current economic burden of illness of osteoporosis in Canada. Osteoporos Int. 27:3023–3032. 2016. View Article : Google Scholar : PubMed/NCBI

53 

Egermann M, Gerhardt C, Barth A, Maestroni GJ, Schneider E and Alini M: Pinealectomy affects bone mineral density and structure-an experimental study in sheep. BMC Musculoskelet Disord. 12:2712011. View Article : Google Scholar

54 

Pines A: Circadian rhythm and menopause. Climacteric. 19:551–552. 2016. View Article : Google Scholar : PubMed/NCBI

55 

Sack RL, Lewy AJ, Erb DL, Vollmer WM and Singer CM: Human melatonin production decreases with age. J Pineal Res. 3:379–388. 1986. View Article : Google Scholar : PubMed/NCBI

56 

Kotlarczyk MP, Lassila HC, O'Neil CK, D'Amico F, Enderby LT, Witt-Enderby PA and Balk JL: Melatonin osteoporosis prevention study (MOPS): A randomized, double-blind, placebo-controlled study examining the effects of melatonin on bone health and quality of life in perimenopausal women. J Pineal Res. 52:414–426. 2012. View Article : Google Scholar : PubMed/NCBI

57 

Sharan K, Lewis K, Furukawa T and Yadav VK: Regulation of bone mass through pineal-derived melatonin-MT2 receptor pathway. J Pineal Res. 63. pp. e124232017, View Article : Google Scholar

58 

Bao T, Zeng L, Yang K, Li Y, Ren F, Zhang Y and Gao Z: Can melatonin improve the osteopenia of perimenopausal and postmenopausal women? A meta-analysis. Int J Endocrinol. 2019:51516782019. View Article : Google Scholar : PubMed/NCBI

59 

Tresguerres IF, Tamimi F, Eimar H, Barralet JE, Prieto S, Torres J, Calvo-Guirado JL and Tresguerres JA: Melatonin dietary supplement as an anti-aging therapy for age-related bone loss. Rejuvenation Res. 17:341–346. 2014. View Article : Google Scholar : PubMed/NCBI

60 

Xu Y, Sheng H, Bao Q, Wang Y, Lu J and Ni X: NLRP3 inflammasome activation mediates estrogen deficiency-induced depression- and anxiety-like behavior and hippocampal inflammation in mice. Brain Behav Immun. 56:175–186. 2016. View Article : Google Scholar : PubMed/NCBI

61 

Xu L, Zhang L, Wang Z, Li C, Li S, Li L, Fan Q and Zheng L: Melatonin suppresses estrogen deficiency-induced osteoporosis and promotes osteoblastogenesis by inactivating the NLRP3 inflammasome. Calcif Tissue Int. 103:400–410. 2018. View Article : Google Scholar : PubMed/NCBI

62 

Si Y, Wang C, Guo Y, Xu G and Ma Y: Prevalence of osteoporosis in patients with type 2 diabetes mellitus in the Chinese mainland: A systematic review and meta-analysis. Iran J Public Health. 48:1203–1214. 2019.PubMed/NCBI

63 

Paschou SA, Dede AD, Anagnostis PG, Vryonidou A, Morganstein D and Goulis DG: Type 2 diabetes and osteoporosis: A guide to optimal management. J Clin Endocrinol Metab. 102:3621–3634. 2017. View Article : Google Scholar : PubMed/NCBI

64 

Lecka-Czernik B: Diabetes, bone and glucose-lowering agents: Basic biology. Diabetologia. 60:1163–1169. 2017. View Article : Google Scholar : PubMed/NCBI

65 

Meng HZ, Zhang WL, Liu F and Yang MW: Advanced glycation end products affect osteoblast proliferation and function by modulating autophagy via the receptor of advanced glycation end products/raf protein/mitogen-activated protein kinase/extracellular signal-regulated kinase kinase/extracellular signal-regulated kinase (RAGE/Raf/MEK/ERK) pathway. J Biol Chem. 290:28189–28199. 2015. View Article : Google Scholar : PubMed/NCBI

66 

Zhang WL, Meng HZ, Yang RF, Yang MW, Sun GH, Liu JH, Shi PX, Liu F and Yang B: Melatonin suppresses autophagy in type 2 diabetic osteoporosis. Oncotarget. 7:52179–52194. 2016. View Article : Google Scholar : PubMed/NCBI

67 

Ghareghani M, Scavo L, Arnoult D, Zibara K and Farhadi N: Melatonin therapy reduces the risk of osteoporosis and normalizes bone formation in multiple sclerosis. Fundam Clin Pharmacol. 32:181–187. 2018. View Article : Google Scholar

68 

Witt-Enderby PA, Radio NM, Doctor JS and Davis VL: Therapeutic treatments potentially mediated by melatonin receptors: Potential clinical uses in the prevention of osteoporosis, cancer and as an adjuvant therapy. J Pineal Res. 41:297–305. 2006. View Article : Google Scholar : PubMed/NCBI

69 

March L, Smith EU, Hoy DG, Cross MJ, Sanchez-Riera L, Blyth F, Buchbinder R, Vos T and Woolf AD: Burden of disability due to musculoskeletal (MSK) disorders. Best Pract Res Clin Rheumatol. 28:353–366. 2014. View Article : Google Scholar : PubMed/NCBI

70 

Neogi T and Zhang Y: Epidemiology of osteoarthritis. Rheum Dis Clin North Am. 39:1–19. 2013. View Article : Google Scholar : PubMed/NCBI

71 

Thomas AC, Hubbard-Turner T, Wikstrom EA and Palmieri-Smith RM: Epidemiology of Posttraumatic Osteoarthritis. J Athl Train. 52:491–496. 2017. View Article : Google Scholar :

72 

Vina ER and Kwoh CK: Epidemiology of osteoarthritis: Literature update. Curr Opin Rheumatol. 30:160–167. 2018. View Article : Google Scholar :

73 

Gentili C and Cancedda R: Cartilage and bone extracellular matrix. Curr Pharm Des. 15:1334–1348. 2009. View Article : Google Scholar : PubMed/NCBI

74 

Mobasheri A, Rayman MP, Gualillo O, Sellam J, van der Kraan P and Fearon U: The role of metabolism in the pathogenesis of osteoarthritis. Nat Rev Rheumatol. 13:302–311. 2017. View Article : Google Scholar : PubMed/NCBI

75 

Buckwalter JA, Anderson DD, Brown TD, Tochigi Y and Martin JA: The roles of mechanical stresses in the pathogenesis of osteoarthritis: Implications for treatment of joint injuries. Cartilage. 4:286–294. 2013. View Article : Google Scholar

76 

Kapoor M, Martel-Pelletier J, Lajeunesse D, Pelletier JP and Fahmi H: Role of proinflammatory cytokines in the pathophysiology of osteoarthritis. Nat Rev Rheumatol. 7:33–42. 2011. View Article : Google Scholar

77 

Gao B, Gao W, Wu Z, Zhou T, Qiu X, Wang X, Lian C, Peng Y, Liang A, Qiu J, et al: Melatonin rescued interleukin 1β-impaired chondrogenesis of human mesenchymal stem cells. Stem Cell Res Ther. 9:1622018. View Article : Google Scholar

78 

Taruc-Uy RL and Lynch SA: Diagnosis and treatment of osteoarthritis. Prim Care. 40:821–836. vii2013. View Article : Google Scholar : PubMed/NCBI

79 

Hainque B, Dominice J, Jaffray P, Ronot X and Adolphe M: Effects of dexamethasone on the growth of cultured rabbit articular chondrocytes: Relation with the nuclear glucocorticoid-receptor complex. Ann Rheum Dis. 46:146–152. 1987. View Article : Google Scholar : PubMed/NCBI

80 

Robinson WH, Lepus CM, Wang Q, Raghu H, Mao R, Lindstrom TM and Sokolove J: Low-grade inflammation as a key mediator of the pathogenesis of osteoarthritis. Nat Rev Rheumatol. 12:580–592. 2016. View Article : Google Scholar : PubMed/NCBI

81 

Liu-Bryan R and Terkeltaub R: Emerging regulators of the inflammatory process in osteoarthritis. Nat Rev Rheumatol. 11:35–44. 2015. View Article : Google Scholar :

82 

Zhang Y, Lin J, Zhou X, Chen X, Chen AC, Pi B, Pan G, Pei M, Yang H, Liu T and He F: Melatonin prevents osteoarthritis-induced cartilage degradation via targeting MicroRNA-140. Oxid Med Cell Longev. 2019:97059292019. View Article : Google Scholar

83 

Hosseinzadeh A, Kamrava SK, Joghataei MT, Darabi R, Shakeri-Zadeh A, Shahriari M, Reiter RJ, Ghaznavi H and Mehrzadi S: Apoptosis signaling pathways in osteoarthritis and possible protective role of melatonin. J Pineal Res. 61:411–425. 2016. View Article : Google Scholar : PubMed/NCBI

84 

Milam SB, Zardeneta G and Schmitz JP: Oxidative stress and degenerative temporomandibular joint disease: A proposed hypothesis. J Oral Maxillofac Surg. 56:214–223. 1998. View Article : Google Scholar : PubMed/NCBI

85 

Bolduc JA, Collins JA and Loeser RF: Reactive oxygen species, aging and articular cartilage homeostasis. Free Radic Biol Med. 132:73–82. 2019. View Article : Google Scholar :

86 

Bakker AD, Silva VC, Krishnan R, Bacabac RG, Blaauboer ME, Lin YC, Marcantonio RA, Cirelli JA and Klein-Nulend J: Tumor necrosis factor alpha and interleukin-1beta modulate calcium and nitric oxide signaling in mechanically stimulated osteocytes. Arthritis Rheum. 60:3336–3345. 2009. View Article : Google Scholar : PubMed/NCBI

87 

Lim HD, Kim YS, Ko SH, Yoon IJ, Cho SG, Chun YH, Choi BJ and Kim EC: Cytoprotective and anti-inflammatory effects of melatonin in hydrogen peroxide-stimulated CHON-001 human chondrocyte cell line and rabbit model of osteoarthritis via the SIRT1 pathway. J Pineal Res. 53:225–237. 2012. View Article : Google Scholar : PubMed/NCBI

88 

Guo JY, Li F, Wen YB, Cui HX, Guo ML, Zhang L, Zhang YF, Guo YJ and Guo YX: Melatonin inhibits Sirt1-dependent NAMPT and NFAT5 signaling in chondrocytes to attenuate osteoarthritis. Oncotarget. 8:55967–55983. 2017. View Article : Google Scholar : PubMed/NCBI

89 

Cuzzocrea S, Zingarelli B, Gilad E, Hake P, Salzman AL and Szabó C: Protective effect of melatonin in carrageenan-induced models of local inflammation: Relationship to its inhibitory effect on nitric oxide production and its peroxynitrite scavenging activity. J Pineal Res. 23:106–116. 1997. View Article : Google Scholar : PubMed/NCBI

90 

Ozturk G, Coşkun S, Erbaş D and Hasanoglu E: The effect of melatonin on liver superoxide dismutase activity, serum nitrate and thyroid hormone levels. Jpn J Physiol. 50:149–153. 2000. View Article : Google Scholar : PubMed/NCBI

91 

Hossain FM and Hong Y, Jin Y, Choi J and Hong Y: Physiological and pathological role of circadian hormones in osteoarthritis: Dose-dependent or time-dependent? J Clin Med. 8:14152019. View Article : Google Scholar :

92 

Jahanban-Esfahlan R, Mehrzadi S, Reiter RJ, Seidi K, Majidinia M, Baghi HB, Khatami N, Yousefi B and Sadeghpour A: Melatonin in regulation of inflammatory pathways in rheumatoid arthritis and osteoarthritis: Involvement of circadian clock genes. Br J Pharmacol. 175:3230–3238. 2018. View Article : Google Scholar :

93 

Rong J, Zhu M, Munro J, Cornish J, McCarthy GM, Dalbeth N and Poulsen RC: Altered expression of the core circadian clock component PERIOD2 contributes to osteoarthritis-like changes in chondrocyte activity. Chronobiol Int. 36:319–331. 2019. View Article : Google Scholar

94 

Yang W, Kang X, Liu J, Li H, Ma Z, Jin X, Qian Z, Xie T, Qin N, Feng D, et al: Clock gene Bmal1 modulates human cartilage gene expression by crosstalk with Sirt1. Endocrinology. 157:3096–3107. 2016. View Article : Google Scholar : PubMed/NCBI

95 

Hong Y, Kim H, Lee S, Jin Y, Choi J, Lee SR, Chang KT and Hong Y: Role of melatonin combined with exercise as a switch-like regulator for circadian behavior in advanced osteoarthritic knee. Oncotarget. 8:97633–97647. 2017. View Article : Google Scholar : PubMed/NCBI

96 

Nugent M: MicroRNAs: Exploring new horizons in osteoarthritis. Osteoarthritis Cartilage. 24:573–580. 2016. View Article : Google Scholar

97 

Miyaki S and Asahara H: Macro view of microRNA function in osteoarthritis. Nat Rev Rheumatol. 8:543–552. 2012. View Article : Google Scholar : PubMed/NCBI

98 

Miyaki S, Sato T, Inoue A, Otsuki S, Ito Y, Yokoyama S, Kato Y, Takemoto F, Nakasa T, Yamashita S, et al: MicroRNA-140 plays dual roles in both cartilage development and homeostasis. Genes Dev. 24:1173–1185. 2010. View Article : Google Scholar : PubMed/NCBI

99 

Si HB, Zeng Y, Liu SY, Zhou ZK, Chen YN, Cheng JQ, Lu YR and Shen B: Intra-articular injection of microRNA-140 (miRNA-140) alleviates osteoarthritis (OA) progression by modulating extracellular matrix (ECM) homeostasis in rats. Osteoarthritis Cartilage. 25:1698–1707. 2017. View Article : Google Scholar : PubMed/NCBI

100 

Miyaki S, Nakasa T, Otsuki S, Grogan SP, Higashiyama R, Inoue A, Kato Y, Sato T, Lotz MK and Asahara H: MicroRNA-140 is expressed in differentiated human articular chondrocytes and modulates interleukin-1 responses. Arthritis Rheum. 60:2723–2730. 2009. View Article : Google Scholar : PubMed/NCBI

101 

Karlsen TA, de Souza GA, Ødegaard B, Engebretsen L and Brinchmann JE: microRNA-140 inhibits inflammation and stimulates chondrogenesis in a model of interleukin 1β-induced osteoarthritis. Mol Ther Nucleic Acids. 5:e3732016. View Article : Google Scholar

102 

Wu Z, Qiu X, Gao B, Lian C, Peng Y, Liang A, Xu C, Gao W, Zhang L, Su P, et al: Melatonin-mediated miR-526b-3p and miR-590-5p upregulation promotes chondrogenic differentiation of human mesenchymal stem cells. J Pineal Res. 65:e124832018. View Article : Google Scholar : PubMed/NCBI

103 

Jüni P, Hari R, Rutjes AW, Fischer R, Silletta MG, Reichenbach S and da Costa BR: Intra-articular corticosteroid for knee osteoarthritis. Cochrane Database Syst Rev. pp. CD0053282015

104 

McAlindon TE, LaValley MP, Harvey WF, Price LL, Driban JB, Zhang M and Ward RJ: Effect of intra-articular triamcinolone vs saline on knee cartilage volume and pain in patients with knee osteoarthritis: A randomized clinical trial. JAMA. 317:1967–1975. 2017. View Article : Google Scholar : PubMed/NCBI

105 

Yang W, Kang X, Qin N, Li F, Jin X, Ma Z, Qian Z and Wu S: Melatonin protects chondrocytes from impairment induced by glucocorticoids via NAD+-dependent SIRT1. Steroids. 126:24–29. 2017. View Article : Google Scholar : PubMed/NCBI

106 

Garrido-Urbani S, Jaquet V and Imhof BA: ROS and NADPH oxidase: Key regulators of tumor vascularisation. Med Sci (Paris). 30:415–421. 2014.In French. View Article : Google Scholar

107 

Ahn J, Kim SA, Kim KW, Oh JH and Kim SJ: Optimization of TGF-β1-transduced chondrocytes for cartilage regeneration in a 3D printed knee joint model. PLoS One. 14:e02176012019. View Article : Google Scholar

108 

Verdier MP, Seité S, Guntzer K, Pujol JP and Boumédiène K: Immunohistochemical analysis of transforming growth factor beta isoforms and their receptors in human cartilage from normal and osteoarthritic femoral heads. Rheumatol Int. 25:118–124. 2005. View Article : Google Scholar

109 

Fang J, Xu L, Li Y and Zhao Z: Roles of TGF-beta 1 signaling in the development of osteoarthritis. Histol Histopathol. 31:1161–1167. 2016.PubMed/NCBI

110 

Pei M, He F, Wei L and Rawson A: Melatonin enhances cartilage matrix synthesis by porcine articular chondrocytes. J Pineal Res. 46:181–187. 2009. View Article : Google Scholar

111 

Maestroni GJ, Sulli A, Pizzorni C, Villaggio B and Cutolo M: Melatonin in rheumatoid arthritis: Synovial macrophages show melatonin receptors. Ann N Y Acad Sci. 966:271–275. 2002. View Article : Google Scholar : PubMed/NCBI

112 

Hong Y, Kim H, Lee Y, Lee S, Kim K, Jin Y, Lee SR, Chang KT and Hong Y: Salutary effects of melatonin combined with treadmill exercise on cartilage damage. J Pineal Res. 57:53–66. 2014. View Article : Google Scholar : PubMed/NCBI

113 

Osseni RA, Rat P, Bogdan A, Warnet JM and Touitou Y: Evidence of prooxidant and antioxidant action of melatonin on human liver cell line HepG2. Life Sci. 68:387–399. 2000. View Article : Google Scholar

114 

Zhang HM and Zhang Y: Melatonin: A well-documented anti-oxidant with conditional pro-oxidant actions. J Pineal Res. 57:131–146. 2014. View Article : Google Scholar : PubMed/NCBI

115 

Saito R, Muneta T, Ozeki N, Nakagawa Y, Udo M, Yanagisawa K, Tsuji K, Tomita M, Koga H and Sekiya I: Strenuous running exacerbates knee cartilage erosion induced by low amount of mono-iodoacetate in rats. BMC Musculoskelet Disord. 18:362017. View Article : Google Scholar : PubMed/NCBI

116 

Gustafsson A and Asman B: Increased release of free oxygen radicals from peripheral neutrophils in adult periodontitis after Fc delta-receptor stimulation. J Clin Periodontol. 23:38–44. 1996. View Article : Google Scholar : PubMed/NCBI

117 

Battino M, Bullon P, Wilson M and Newman H: Oxidative injury and inflammatory periodontal diseases: The challenge of anti-oxidants to free radicals and reactive oxygen species. Crit Rev Oral Biol Med. 10:458–476. 1999. View Article : Google Scholar

118 

Walters J and Lai PC: Should antibiotics be prescribed to treat chronic periodontitis? Dent Clin North Am. 59:919–933. 2015. View Article : Google Scholar : PubMed/NCBI

119 

Srinath R, Acharya AB and Thakur SL: Salivary and gingival crevicular fluid melatonin in periodontal health and disease. J Periodontol. 81:277–283. 2010. View Article : Google Scholar : PubMed/NCBI

120 

Gómez-Moreno G, Cutando-Soriano A, Arana C, Galindo P, Bolaños J, Acuña-Castroviejo D and Wang HL: Melatonin expression in periodontal disease. J Periodontal Res. 42:536–540. 2007. View Article : Google Scholar : PubMed/NCBI

121 

Laakso ML, Porkka-Heiskanen T, Alila A, Stenberg D and Johansson G: Correlation between salivary and serum melatonin: Dependence on serum melatonin levels. J Pineal Res. 9:39–50. 1990. View Article : Google Scholar : PubMed/NCBI

122 

Madapusi BT and Rao SR: Preliminary evaluation of human gingiva as an extrapineal site of melatonin biosynthesis in states of periodontal health and disease. J Clin Diagnostic Res. 12:ZF1–ZF7. 2018.

123 

Ghallab NA, Hamdy E and Shaker OG: Malondialdehyde, superoxide dismutase and melatonin levels in gingival crevicular fluid of aggressive and chronic periodontitis patients. Aust Dent J. 61:53–61. 2016. View Article : Google Scholar

124 

Lodhi K, Saimbi CS, Khan MA, Nath C and Shukla R: Evaluation of melatonin levels in saliva in gingivitis and periodontitis cases: A pilot study. Contemp Clin Dent. 7:519–523. 2016. View Article : Google Scholar : PubMed/NCBI

125 

Cutando A, Galindo P, Gómez-Moreno G, Arana C, Bolaños J, Acuña-Castroviejo D and Wang HL: Relationship between salivary melatonin and severity of periodontal disease. J Periodontol. 77:1533–1538. 2006. View Article : Google Scholar : PubMed/NCBI

126 

Almughrabi OM, Marzouk KM, Hasanato RM and Shafik SS: Melatonin levels in periodontal health and disease. J Periodont Res. 48:315–321. 2013. View Article : Google Scholar

127 

Tinto M, Sartori M, Pizzi I, Verga A and Longoni S: Melatonin as host modulating agent supporting nonsurgical periodontal therapy in patients affected by untreated severe periodontitis: A preliminary randomized, triple-blind, placebo-controlled study. J Periodont Res. 55:61–67. 2020. View Article : Google Scholar

128 

Chitsazi M, Faramarzie M, Sadighi M, Shirmohammadi A and Hashemzadeh A: Effects of adjective use of melatonin and vitamin C in the treatment of chronic periodontitis: A randomized clinical trial. J Dent Res Dent Clin Dent Prospects. 11:236–240. 2017.

129 

Bazyar H, Gholinezhad H, Moradi L, Salehi P, Abadi F, Ravanbakhsh M and Zare Javid A: The effects of melatonin supplementation in adjunct with non-surgical periodontal therapy on periodontal status, serum melatonin and inflammatory markers in type 2 diabetes mellitus patients with chronic periodontitis: A double-blind, placebo-controlled trial. Inflammopharmacology. 27:67–76. 2019. View Article : Google Scholar

130 

Sarıtekin E, Üreyen Kaya B, Aşcı H and Özmen Ö: Anti-inflammatory and antiresorptive functions of melatonin on experimentally induced periapical lesions. Int Endod J. 52:1466–1478. 2019. View Article : Google Scholar

131 

Renn TY, Huang YK, Feng SW, Wang HW, Lee WF, Lin CT, Burnouf T, Chen LY, Kao PF and Chang HM: Prophylactic supplement with melatonin successfully suppresses the pathogenesis of periodontitis through normalizing RANKL/OPG ratio and depressing the TLR4/MyD88 signaling pathway. J Pineal Res. 64:2018. View Article : Google Scholar

132 

Botero JE, Yepes FL, Roldán N, Castrillón CA, Hincapie JP, Ochoa SP, Ospina CA, Becerra MA, Jaramillo A, Gutierrez SJ and Contreras A: Tooth and periodontal clinical attachment loss are associated with hyperglycemia in patients with diabetes. J Periodontol. 83:1245–1250. 2012. View Article : Google Scholar : PubMed/NCBI

133 

Daniel R, Gokulanathan S, Shanmugasundaram N, Lakshmigandhan M and Kavin T: Diabetes and periodontal disease. J Pharm Bioallied Sci. 4(Suppl 2): S280–S282. 2012. View Article : Google Scholar : PubMed/NCBI

134 

Grover HS and Luthra S: Molecular mechanisms involved in the bidirectional relationship between diabetes mellitus and periodontal disease. J Indian Soc Periodontol. 17:292–301. 2013. View Article : Google Scholar : PubMed/NCBI

135 

Abdolsamadi H, Goodarzi MT, Ahmadi Motemayel F, Jazaeri M, Feradmal J, Zarabadi M, Hoseyni M and Torkzaban P: Reduction of melatonin level in patients with type II diabetes and periodontal diseases. J Dent Res Dent Clin Dent Prospects. 8:160–165. 2014.PubMed/NCBI

136 

Balci Yuce H, Karatas O, Aydemir Turkal H, Pirim Gorgun E, Ocakli S, Benli I and Cayli S: The effect of melatonin on bone loss, diabetic control, and apoptosis in rats with diabetes with ligature-induced periodontitis. J Periodontol. 87:e35–e43. 2016. View Article : Google Scholar

137 

Cutando A, López-Valverde A, de Diego RG, de Vicente J, Reiter R, Fernández MH and Ferrera MJ: Effect of topical application of melatonin to the gingiva on salivary osteoprotegerin, RANKL and melatonin levels in patients with diabetes and periodontal disease. Odontology. 102:290–296. 2014.

138 

Montero J, López-Valverde N, Ferrera MJ and López-Valverde A: Changes in crevicular cytokines after application of melatonin in patients with periodontal disease. J Clin Exp Dent. 9:e1081–e1087. 2017.PubMed/NCBI

139 

Martens L, De Smet S, Yusof MY and Rajasekharan S: Association between overweight/obesity and periodontal disease in children and adolescents: A systematic review and meta-analysis. Eur Arch Paediatr Dent. 18:69–82. 2017. View Article : Google Scholar : PubMed/NCBI

140 

Keller A, Rohde JF, Raymond K and Heitmann BL: Association between periodontal disease and overweight and obesity: A systematic review. J Periodontol. 86:766–776. 2015. View Article : Google Scholar : PubMed/NCBI

141 

Virto L, Cano P, Jiménez-Ortega V, Fernández-Mateos P, González J, Esquifino AI and Sanz M: Obesity and periodontitis: An experimental study to evaluate periodontal and systemic effects of comorbidity. J Periodontol. 89:176–185. 2018.

142 

Shimizu I, Yoshida Y and Minamino T: A role for circadian clock in metabolic disease. Hypertens Res. 39:483–491. 2016. View Article : Google Scholar : PubMed/NCBI

143 

Zisapel N: New perspectives on the role of melatonin in human sleep, circadian rhythms and their regulation. Br J Pharmacol. 175:3190–3199. 2018. View Article : Google Scholar : PubMed/NCBI

144 

Szewczyk-Golec K, Woźniak A and Reiter RJ: Inter-relationships of the chronobiotic, melatonin, with leptin and adiponectin: Implications for obesity. J Pineal Res. 59:277–291. 2015. View Article : Google Scholar : PubMed/NCBI

145 

Ríos-Lugo MJ, Cano P, Jiménez-Ortega V, Fernández-Mateos MP, Scacchi PA, Cardinali DP and Esquifino AI: Melatonin effect on plasma adiponectin, leptin, insulin, glucose, triglycerides and cholesterol in normal and high fat-fed rats. J Pineal Res. 49:342–348. 2010. View Article : Google Scholar : PubMed/NCBI

146 

Favero G, Stacchiotti A, Castrezzati S, Bonomini F, Albanese M, Rezzani R and Rodella LF: Melatonin reduces obesity and restores adipokine patterns and metabolism in obese (ob/ob) mice. Nutr Res. 35:891–900. 2015. View Article : Google Scholar : PubMed/NCBI

147 

Virto L, Haugen HJ, Fernández-Mateos P, Cano P, González J, Jiménez-Ortega V, Esquifino AI and Sanz M: Melatonin expression in periodontitis and obesity: An experimental in-vivo investigation. J Periodont Res. 53:825–831. 2018. View Article : Google Scholar

148 

Virto L, Cano P, Jiménez-Ortega V, Fernández-Mateos P, González J, Haugen HJ, Esquifino AI and Sanz M: Melatonin as adjunctive therapy in the treatment of periodontitis associated with obesity. J Clin Periodontol. 45:1336–1346. 2018. View Article : Google Scholar : PubMed/NCBI

149 

Santos RMD, Marani F, Chiba FY, Mattera MSLC, Tsosura TVS, Tessarin GWL, Pereira RF, Belardi BE, Pinheiro BCES and Sumida DH: Melatonin promotes reduction in TNF levels and improves the lipid profile and insulin sensitivity in pinealectomized rats with periodontal disease. Life Sci. 213:32–39. 2018. View Article : Google Scholar : PubMed/NCBI

150 

Diomede F, Zingariello M, Cavalcanti MFXB, Merciaro I, Pizzicannella J, De Isla N, Caputi S, Ballerini P and Trubiani O: MyD88/ERK/NFkB pathways and pro-inflammatory cytokines release in periodontal ligament stem cells stimulated by Porphyromonas gingivalis. Eur J Histochem. 61:27912017.

151 

Nagata M, Iwasaki K, Akazawa K, Komaki M, Yokoyama N, Izumi Y and Morita I: Conditioned medium from periodontal ligament stem cells enhances periodontal regeneration. Tissue Eng Part A. 23:367–377. 2017. View Article : Google Scholar :

152 

Bae WJ, Park JS, Kang SK, Kwon IK and Kim EC: Effects of melatonin and its underlying mechanism on ethanol-stimulated senescence and osteoclastic differentiation in human periodontal ligament cells and cementoblasts. Int J Mol Sci. 19:17422018. View Article : Google Scholar :

153 

El-Sharkawy H, Elmeadawy S, Elshinnawi U and Anees M: Is dietary melatonin supplementation a viable adjunctive therapy for chronic periodontitis?-A randomized controlled clinical trial. J Periodont Res. 54:190–197. 2019. View Article : Google Scholar

154 

Irwin MR, Olmstead R and Carroll JE: Sleep disturbance, sleep duration, and inflammation: A Systematic review and meta-analysis of cohort studies and experimental sleep deprivation. Biol Psychiatry. 80:40–52. 2016. View Article : Google Scholar

155 

Andersen LP, Gögenur I, Rosenberg J and Reiter RJ: The safety of melatonin in humans. Clin Drug Investig. 36:169–175. 2016. View Article : Google Scholar

156 

Foley HM and Steel AE: Adverse events associated with oral administration of melatonin: A critical systematic review of clinical evidence. Complement Ther Med. 42:65–81. 2019. View Article : Google Scholar : PubMed/NCBI

157 

Frisher M, Gibbons N, Bashford J, Chapman S and Weich S: Melatonin, hypnotics and their association with fracture: A matched cohort study. Age Ageing. 45:801–806. 2016. View Article : Google Scholar : PubMed/NCBI

158 

Sun T, Li J, Xing HL, Tao ZS and Yang M: Melatonin improves the osseointegration of hydroxyapatite-coated titanium implants in senile female rats. Z Gerontol Geriatr. 53:770–777. 2020. View Article : Google Scholar

159 

Cutando A, Arana C, Gómez-Moreno G, Escames G, López A, Ferrera MJ, Reiter RJ and Acuña-Castroviejo D: Local application of melatonin into alveolar sockets of beagle dogs reduces tooth removal-induced oxidative stress. J Periodontol. 78:576–583. 2007. View Article : Google Scholar : PubMed/NCBI

160 

Swanson CM, Kohrt WM, Buxton OM, Everson CA, Wright KP Jr, Orwoll ES and Shea SA: The importance of the circadian system & sleep for bone health. Metabolism. 84:28–43. 2018. View Article : Google Scholar :

161 

Wu QY, Wang J, Tong X, Chen J, Wang B, Miao ZN, Li X, Ye JX and Yuan FL: Emerging role of circadian rhythm in bone remodeling. J Mol Med (Berl). 97:19–24. 2019. View Article : Google Scholar

162 

Yang N and Meng QJ: Circadian clocks in articular cartilage and bone: A compass in the sea of matrices. J Biol Rhythms. 31:415–427. 2016. View Article : Google Scholar : PubMed/NCBI

163 

Tian Y and Li LM: Epidemiological study of sleep disorder in the elderly. Zhonghua Liu Xing Bing Xue Za Zhi. 38:988–992. 2017.In Chinese. PubMed/NCBI

164 

Santoro N: Perimenopause: From research to practice. J Womens Health (Larchmt). 25:332–339. 2016. View Article : Google Scholar

Related Articles

Journal Cover

May-2021
Volume 47 Issue 5

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lu X, Yu S, Chen G, Zheng W, Peng J, Huang X and Chen L: Insight into the roles of melatonin in bone tissue and bone‑related diseases (Review). Int J Mol Med 47: 82, 2021
APA
Lu, X., Yu, S., Chen, G., Zheng, W., Peng, J., Huang, X., & Chen, L. (2021). Insight into the roles of melatonin in bone tissue and bone‑related diseases (Review). International Journal of Molecular Medicine, 47, 82. https://doi.org/10.3892/ijmm.2021.4915
MLA
Lu, X., Yu, S., Chen, G., Zheng, W., Peng, J., Huang, X., Chen, L."Insight into the roles of melatonin in bone tissue and bone‑related diseases (Review)". International Journal of Molecular Medicine 47.5 (2021): 82.
Chicago
Lu, X., Yu, S., Chen, G., Zheng, W., Peng, J., Huang, X., Chen, L."Insight into the roles of melatonin in bone tissue and bone‑related diseases (Review)". International Journal of Molecular Medicine 47, no. 5 (2021): 82. https://doi.org/10.3892/ijmm.2021.4915