Skip to main content

Melatonin: new insights on its therapeutic properties in diabetic complications

Abstract

Diabetes and diabetic complications are considered as leading causes of both morbidity and mortality in the world. Unfortunately, routine medical treatments used for affected patients possess undesirable side effects, including kidney and liver damages as well as gastrointestinal adverse reactions. Therefore, exploring the novel therapeutic strategies for diabetic patients is a crucial issue. It has been recently shown that melatonin, as main product of the pineal gland, despite its various pharmacological features including anticancer, anti-aging, antioxidant and anti-inflammatory effects, exerts anti-diabetic properties through regulating various cellular mechanisms. The aim of the present review is to describe potential roles of melatonin in the treatment of diabetes and its complications.

Background

Diabetes mellitus (DM), a frequent life-threatening metabolic disorder worldwide, is characterized by hyperglycemia caused by either insulin resistance or declined insulin secretion [1]. In the last decades, the incidence of diabetes mellitus has been enhancing [2, 3]. This disease has been one of the leading cause of death worldwide, and the International Diabetes Mellitus Federation estimated that 592 million people will suffer from diabetes mellitus by the year 2035 [4]. Several investigations have indicated that diabetes mellitus, could mediate various complications, including diabetic cardiovascular complications, diabetic neuropathy, retinopathy, nephropathy and liver complications, which have been the main causes of its mortality and morbidity [4, 5]. Thus, it is essential to search for efficient methods for the management and prevention of diabetes mellitus and its complications [6].

The impact of pineal hormone on insulin secretion, carbohydrate metabolism and blood glucose has been recently demonstrated. Melatonin (N-acetyl-5-methoxytryptamine), a tryptophan-derived endocrine agent, is mainly synthetized by the pineal gland and locally by numerous other tissues [7]. Despite the higher insulin levels in diabetic subjects, diabetes mellitus is accompanied by lower serum concentrations of melatonin in diabetic GotoKakizaki rats [8]. Furthermore, melatonin functions as an anti-aging [9], anti-inflammatory, antioxidant [10] as well as antihypertensive agent [11]. This review summarizes the therapeutic potentials of melatonin in the treatment of diabetes mellitus and its complications, based on findings of recent studies.

Melatonin: potentials, safety and bioavailability

Melatonin, a broad spectrum antioxidant, can be used in various pathologic conditions mainly due to its regulatory effects on autophagy, endoplasmic reticulum (ER) stress, and oxidative stress. This molecule has potential therapeutic effects for treating neurodegenerative diseases, malignancies, cardiovascular diseases and many other clinical problems [12]. Of note, it also has a regulatory role in sleep disorders and reproduction [13, 14]. Furthermore, melatonin has immunomodulatory effect [15], which depends on its capability to increase the cytokines production and its anti-apoptotic and antioxidant properties. By affecting cytokine release from immunocompetent cells, melatonin has been suggested to modulate the immune system [16]. Melatonin modulates reproduction by regulating the production of sex hormones in seasonally breeding animals. In long-breeders, melatonin decreases estrogen production. However in short-breeders, melatonin increases the level of estrogen synthesis in winter [17, 18]. In humans, melatonin role on reproduction is not totally clear. During the dark season, the high level of melatonin is associated with the low estrogen production rate [19]. In addition, melatonin directly increases the steroidogenesis in human granulosa-luteal cells [20]. Furthermore, melatonin increases progesterone production via its M2 receptors on corpus luteum [21]. However, there are reports that indicate no role for melatonin in up-regulating estrogen synthesis by granulosa cells [22]. Taken together, melatonin affects the activity of ovaries; however this control and the exact mechanisms remain to be determined.

As mentioned earlier, melatonin is a very potent anti-oxidant partially due to its molecule structure which is both lipophilic and hydrophilic, so it can cross all bio-barriers easily and accumulate in high amounts within subcellular organelles including mitochondria which are the main sites for reactive oxygen species (ROS) production [23,24,25,26]. The anti-oxidative effects of melatonin is attributed to its direct free radical scavenging properties as well as inducing the production of other anti-oxidant enzymes including glutathione peroxidase (GPx), glutathione reductase (GR), catalase (CAT) and superoxide dismutase (SOD) [27,28,29,30]. Melatonin also inhibits the expression of pro-oxidant enzymes such as nitric oxide (NO) synthase (NOS), cyclooxygenase-2 (COX-2), myeloperoxidase and eosinophil peroxidase [31, 32]. Free radicals such as ROS are waste products of cellular metabolism. The accumulation of these agents within cells can cause damage to cell DNA, proteins and lipids and may give rise to cancerous cells. Melatonin can avoid these damages by neutralizing free radicals [30, 33, 34].

The safety of exogenous melatonin is still under investigation; however, with the limiting data available, melatonin can be considered as a safe drug. Melatonin administration for adolescents, children and preterm infants with different diseases has shown no significant side effects except at high doses or multiple time administration [35,36,37]. Of note, melatonin could affect sexual maturation in the mentioned groups [38]. In adults, oral administration of melatonin for improving of dyspnea (3 mg fast release for 3 months) in chronic obstructive pulmonary disease (COPD) patients and protecting the patients from depression and anxiety after breast cancer surgery (6 mg for 3 months) has shown no major side effects. However, minor adverse effects such as transient dizziness, headache, numbness, paresthesia of mouth, arms or legs, and deterioration of dyspnea has been considerable in oral administration [39, 40]. Melatonin use (9 mg sustained release per day for 4 weeks) in order to improve the sleep quality of the children with epilepsy had some adverse effects including: morning drowsiness, gastrointestinal symptoms, increased enuresis, headache, dizziness, diarrhea, rash, and hypothermia [41].

Since melatonin is mainly metabolized by cytochrome P450 (CYP) 1A2 and CYP2C19, inhibitors of these enzymes lead to higher concentration of melatonin in the body [42]. Melatonin lowers blood pressure and glucose level, therefore, patients receiving antihypertensive or blood glucose lowering drugs should use melatonin with cautious [43]. Since the administration of melatonin during pregnancy is not studied, its use is not recommended for pregnant women [38]. The use of melatonin by breastfeeding women causes daytime drowsiness for their infants [44].

Pharmacokinetics and optimal dosage of this therapeutic agent are not thoroughly clear yet and more studies are required on these subjects; however, there are some inconsistent studies on oral and intravenous administration of melatonin. The absorption of this hormone is site dependent making the rectum as the site with the highest absorption rate. Orally administered melatonin at doses up to 80 mg is absorbed fast and follows first-order kinetic [45]. Melatonin is expected to reach maximal plasma concentration after 30–45 min and 30–60 min (t max) in intravenous (IV) and oral administration, respectively [46, 47]. The elimination rate of melatonin from the body depends on the dose and route of administration. Because of structural characteristics, melatonin is both water and lipid soluble. So, it can run through tissues, cells and cellular compartments easily [48]. The elimination half-life (t1/2) for 100 mg melatonin in IV administration is 45 min [46], while it is 28–61 min for 0.005–2 mg [49]. The elimination half-life for oral administration of melatonin is a little higher and is about 46–65 min for 0.5–6 mg [47]. Melatonin is considered a drug with a high hepatic first pass effect. In oral administration, just 10–15% of melatonin reaches the systemic circulation and the rest of it is metabolized by CYP1A2 to 6-hydroxymelatonin and excreted in the urine after conjugation with sulfate or glucuronic acid [48, 50,51,52,53]. Of note, melatonin elimination occurs faster in children in comparison to adults [54]. Low absorption from the gastrointestinal tract, high first-pass effect and high rate of metabolizing make melatonin a drug with low bioavailability [53]. Because of poor bioavailability, other routes of administration are suggested; for example: subcutaneous injection, transdermal, oral transmucosal and intranasal [43, 55].

Anti-diabetic effects of Melatonin

Melatonin has been shown to have a beneficial role in controlling blood glucose in both animal and human studies. The expression level of glucose transporter type 4 (GLUT4) gene is reduced in pinealectomized animals, which consequently results in glucose intolerance and insulin resistance. These conditions are alleviated by melatonin treatment [56, 57]. Furthermore, the level of melatonin decreases in human dental pulp tissue in type 2 diabetic participants. Melatonin in pharmacological concentration could improve iNOS and SOD activity in hyperglyceamic human dental pulp cells (hDPCs), suggesting the protective effects of melatonin in human dental pulp tissue under hyperglycaemia [58].  In rats with streptozotocin (STZ)-induced diabetes, eight-week treatment with insulin (NPH, 1.5 U/100gr/day) and melatonin (0.2 mg/kg/day in drinking water) improves glucose homeostasis and insulin sensitivity of white adipose tissue comparing with using one of these drugs individually [59]. The production of melatonin has been reported to reduce in animal models of diabetes and melatonin treatment (100 mg/kg/day in drinking water for 8 weeks) in high-fat diet-fed mouse with insulin resistance results in better glucose tolerance [60]. Diabetes also results in decreased testosterone production. It has been shown that melatonin (10 mcg/kg/day in drinking water) is able ameliorate the deleterious effects of diabetes on testosterone production in rats by improving glucose metabolism in the Leydig cells and inducing acetate production which is a precursor for cholesterol synthesis [61].

Patients with type 2 diabetes mellitus (T2DM) have a lower nocturnal melatonin production compared to ones without diabetes [62]. Several studies indicated that single dose of melatonin in healthy post-menopausal and pre-menopausal women (1 mg and 5 mg respectively) worsens the glucose tolerance test in the morning and the evening [63, 64]. On the other hand, chronic treatment with prolonged-release melatonin (2 mg) over a 5-month period decreases the level of HbA1c and betters glycemic control [65]. Furthermore, administration of melatonin (6 mg) for 3 months resulted in better glycemic control in patients with T2DM [66]. In patients with poorly controlled T2DM, addition of melatonin (10 mg) and zinc acetate (50 mg) to metformin resulted in the better tissue response to metformin alone [67]. In obese patients with Acanthosis Nigricans, melatonin (3 mg/day) supplementation for 12 weeks improved insulin sensitivity as well as inflammatory status [57].

Oxidative stress, which has an important role in the induction of various complications of diseases such as diabetes, is effectively attenuated by the anti-oxidative activity of melatonin. Moreover, this hormone protects the beta cells of pancreas, with low antioxidant content, by neutralizing reactive oxygen species [68, 69]. Based on a recent systematic review, melatonin may have a potential role in better glycemic control through increasing insulin sensitivity and lowering fasting glucose [70].

Melatonin and diabetic complications

As mentioned above, diabetes mellitus causes numerous micro- and macrovascular complications in affected patients. Melatonin is considered as an appropriate candidate for the prevention and treatment of diabetes mellitus complications. Here, we discuss about its therapeutic potentials for cardiomyopathy, retinopathy, central nervous system (CNS)-related complications of diabetes, neuropathy, and nephropathy.

Effects of melatonin on diabetic cardiomyopathy

Diabetic cardiomyopathy is a common complication of diabetes mellitus, which is defined as adverse changes in myocardial structure and function in the absence of other cardiac risk factors including coronary artery disease or hypertension [71]. Diabetic cardiomyopathy is characterized by increased myocardial fibrosis and stiffness, with this being associated with the impaired diastolic function, late systolic dysfunction resulting in the inability of the heart to pump enough blood through the body, a state called heart failure [72]. The main pathogenetic factors involved in the development and progression of diabetic cardiomyopathy include hyperglycemia, systemic insulin resistance, and impaired cardiac insulin metabolic signaling; these factors induce several pathways including vascular endothelial dysfunction, adrenergic activity, impairment of mitochondria calcium (Ca2+) handling, activation of renin–angiotensin system, myocardial ischemia/functional hypoxia, oxidative stress, mitochondria dysfunction, inflammation, endoplasmic reticulum stress and cardiomyocyte death [72, 73]. Excessive level of reactive oxygen species (ROS) induced by high glucose contributes to the peroxidation of lipids, induction of apoptosis and inhibition of autophagy in muscle fibers [74]; diabetes impairs myocardial mitochondrial biogenesis and suppresses the activity of myocardial mitochondrial Complexes I, III and IV resulting in the loss of mitochondrial number, impairment of mitochondrial function, increased generation of ROS and escape of death‐inducing factors [75]. Although hyperglycemia increases the risk of cardiomyopathy in diabetic patients and good glycemic control lowers the progression of diabetic microvascular complications, intensive glucose control in patients with established diabetes mellitus cannot reduce the mortality induced by diabetic cardiomyopathy [76]. This highlights the need for effective therapeutic agents to prevent or treat diabetic cardiomyopathy.

Melatonin as an effective antioxidant has been demonstrated to prevent diabetes-induced harmful effects on the heart (Fig. 1a). Melatonin reduces oxidative damage in myocardial cells and inhibits extrinsic and intrinsic pathways of apoptosis; in STZ‐induced diabetic rats, melatonin improves antioxidative status and reduces lipid peroxidation level and apoptotic markers in heart tissue to near control values [77, 78]. Melatonin preserves mitochondrial function in cardiac of diabetic rats through increasing the mitochondrial biogenesis and deacetylation of mitochondrial anti-oxidative enzymes, which this effect results from activation of cGMP-PKGIα, sirtuin-1 (SIRT1)‐peroxisome proliferator-activated receptor gamma coactivator 1α (PGC1α) and 5′ AMP-activated protein kinase (AMPK)-PGC1α-SIRT3 signaling pathways [79,80,81]. Activation of PGC1α-SIRT3 signaling plays a key role in cardioprotective action of melatonin; this signaling pathway leads to the enhancement of mitochondrial SOD activity, oxidative phosphorylation of Complexes I, III and IV, and reduction of mitochondrial lipid peroxidation, ROS generation and myocardial apoptosis [79]. In addition, it has been emphasized that melatonin treatment ameliorates myocardial apoptosis through suppressing ER-stress and spleen tyrosine kinase (Syk)/mitochondrial complex I/sarcoendoplasmic reticulum calcium transport ATPase (SERCA) axis [82, 83]. Chronic high-glucose activates Syk leading to the repression of the expression and activity of mitochondrial complex I causing increased generation of ROS and subsequent peroxidation of SERCA; this results in the inhibition of cytoplasmic calcium re-uptake by ER leading to cellular calcium overload and cardiomyocytes death via activation of mitochondria‐ and ER stress‐mediated apoptosis [82, 83].

Fig. 1
figure 1

a Melatonin improves diabetic cardiomyopathy through inhibiting mitochondrial fission by activation of SIRT1‐PGC1α pathway, increasing mitochondrial biogenesis by activation of cGMP-PKGIα, SIRT1 and AMPK-PGC1α-SIRT3 signaling pathways, inhibiting cardiac hypertrophy by reduction of the expression of VEGF-A, inhibiting apoptotic pathway by decrease the expression of caspase-3, -9, -8, Bax, PERK, Syk/MC1/SERCA, IRS-1/Akt and MAPK signaling pathways and inhibiting oxidative stress by increase the activity of SOD, CAT, GPx, cGMP-PKGIα and Nrf-2-HO-1 signaling pathways and the level of GSH and reduction of ROS, MDA and NO levels. b Melatonin reduces diabetic retinopathy through inhibiting oxidative stress by reduction of ROS, MDA and NO levels and activation of CAT and PI3K/Akt-Nrf2 pathway, improving blood-retinal barrier by reduction of the expression of HIF-1α, VEGF-A and PEDF, inhibiting apoptotic pathway by decrease the expression of caspase-3, Bax and MAPKs pathways, and inhibiting inflammation by inhibition of the expression of TNF-α, NOS and the activity of NFκB. c Melatonin ameliorates diabetic neuropathy through inhibiting oxidative stress by increasing CAT, SOD and GPx activity and GSH level and activating Nrf-2-HO-1 pathway, inhibiting inflammation by reduction of TNF-α, iNOS, IL-6 and COX-2 expressions and the activity of NFκB, inhibiting apoptotic pathway by alleviation of the expression of caspase-3 and -9, Bax, PARP, and p38 MAPKs and elevation of PINK-1 level, and increasing the level of GABA and decreasing astrogliosis, which this effect leads to the improvement of memory and cognitive ability. d Melatonin improves diabetic nephropathy through inhibiting fibrotic process by reduction of the expression of TGF-β, inhibiting oxidative stress by enhancement of mitochondrial complex III, CAT, SOD, GPx and GST activities and reduction of NOX activity and MDA and NO generation, inhibiting inflammation by decreasing the level of IL-β, IL-6 and IL-33, inhibiting apoptosis by reduction of caspase-3 and Bax expression and JAK/STAT activity, inducing autophagy pathway by enhancement of the expression of Beclin-1, and inhibiting EMT by elevation the level of miR-49, which results in the alleviation of the level of ROCK. SIRT sirtuin, PGC1α peroxisome proliferator-activated receptor gamma coactivator 1α, AMPK 5′ AMP-activated protein kinase, cGMP Cyclic guanosine monophosphate, PKGIα Protein kinase G Iα, VEGF-A Vascular endothelial growth factor-A, Syk Spleen tyrosine kinase, SERCA sarcoendoplasmic reticulum calcium transport ATPase, Nrf2 erythroid 2‐related factor 2, HO-1 heme oxygenase-1, IRS‐1 insulin receptor substrate, Akt Protein kinase B, GPx glutathione peroxidase, CAT catalase, SOD superoxide dismutase, NOS NO synthase, iNOS inducible NOS, MDA malondialdehyde, COX-2 cyclooxygenase-2, ROS reactive oxygen species, NF‐κB nuclear factor-κB, TNF-α tumor necrosis factor α, IL interleukin, mTOR mammalian target of rapamycin, GSH glutathione, NO nitric oxide, JAK janus kinase, STAT signal transducer and activator of transcription, GABA gamma-aminobutyric acid, PARP poly(ADP-ribose) polymerase, MAPK mitogen-activated protein kinase, TGF-β transforming growth factor-β, EMT endothelial-to-mesenchymal transition, ROCK RhoA/Rho kinase, GST glutathione S-transferases, NOX NADPH oxidase

Melatonin modulates nuclear factor erythroid 2-related factor 2 (NRF2)-hemeoxygenase 1 (HO-1) and mitogen-activated protein kinase (MAPK) signaling through activating membrane receptors (especially MT2 receptor)-dependent cyclic guanosine monophosphate (cGMP)-protein kinase G Iα (PKGIα) signaling pathway, which contributes to the improvement of diabetic cardiac function by inhibiting myocardial apoptosis and oxidative stress [80]. Activation of SIRT1‐PGC1α pathway by melatonin contributes to the inhibition of dynamin‐related protein 1 (Drp1)‐mediated mitochondrial fission, suppression of oxidative stress, reduction of cardiomyocyte apoptosis, and improvement of mitochondrial and cardiac function in STZ‐induced diabetic mice [81]. Furthermore, maternal melatonin treatment improves the tolerance to myocardial ischemia/reperfusion injury in diabetic mother–offspring mice. This beneficial effect of melatonin results from activation of cardiac insulin receptor substrate (IRS)‐1/Protein kinase B (Akt) signaling leading to the suppression of mitochondria‐ and ER stress-mediated apoptosis and oxidative stress [84]. Co-administration of PPAR-g agonist, thiazolidinediones, with melatonin has been reported to have beneficial effects on diabetes-induced cardiovascular complications. Combination of pioglitazone or rosiglitazone with melatonin is effective to normalize levels of SOD, glutathione (GSH), CAT, and lipid peroxidation as well as inhibit myonecrosis, vacuolar changes and infiltration of inflammatory cells in the heart tissue of alloxan-induced diabetic rats; this indicates that melatonin may be a potential drug to increase beneficial effect of thiazolidinediones on diabetic cardiomyopathy [73]. Melatonin treatment may also prevent the development of diabetic cardiomyopathy through increasing the phosphorylation of vascular endothelial growth factor-A (VEGF-A) resulting in the inhibition of cardiac enlargement and hypertrophy in STZ-induced diabetic rats [85].

The protective effect of melatonin against diabetic cardiomyopathy may also result from its modulatory effect on autophagy pathway. Melatonin has been reported to alleviate cardiac remodeling through inhibiting the expression of mammalian target of rapamycin (mTOR); mTOR is an important molecule inhibiting autophagy pathway, which remarkably elevates in the myocardium of diabetic rats leading to the occurrence of both cardiomyopathy and cardiac hypertrophy [74]. Melatonin also regulates the activity of mammalian Ste20-like kinase 1 (Mst1)/Sirt3 signaling pathway. Suppression of Mst1 by melatonin contributes to the up-regulation of autophagy/Parkin-mediated mitophagy, inhibition of apoptosis and modulation of mitochondrial integrity and biogenesis leading to the alleviation of cardiac remodeling and dysfunction in diabetic mice [86, 87]. Table 1 summarizes studies indicating the protective effects of melatonin on diabetic cardiomyopathy.

Table 1 Investigations on melatonin and diabetic cardiomyopathy

The therapeutic effects of melatonin on diabetic retinopathy

Diabetic retinopathy (DR), a prevalent asymptomatic microvascular complication of diabetes mellitus, is a leading cause of vision loss worldwide [88, 89]. According to recent reports, one-third of diabetic patients possess some degree of diabetic retinopathy [90]. Due to the increasing prevalence of diabetes worldwide, the number of diabetic retinopathy patients has been estimated to elevate from 424.9 million in 2017 to 628 million by 2045 [91].

Nowadays, as mentioned earlier, inflammation and oxidative stress have crucial roles in diabetic retinopathy pathogenesis [92, 93]. Therefore, treatments such as melatonin restricting inflammatory and oxidative impacts of diabetes mellitus could be greatly potential to affect individuals through preventing or decreasing the retinal complications (Fig. 1b).

Various and heterogeneous factors, such as advanced glycation end products (AGEs), growth factors, hyperglycaemia as well as great levels of vitreous or circulating chemokines, cytokines and ROS can trigger the inflammatory responses in the retinal vasculature [94, 95]. Thus, mounting evidence suggests chronic inflammation as an essential process in the development of diabetic retinopathy, primarily in early stages [96].

Several investigations have shown enhanced levels of different pro-inflammatory cytokines, including COX2, inducible NO synthase (iNOS), nuclear factor kappa B (NF-κB), intercellular adhesion molecule-1 (ICAM), and vascular endothelial growth factor (VEGF) in the retinas and vitreous humours of diabetic animals and humans [92, 97, 98]. Although various stimuli are able to promote the production of VEGF leading to pathological angiogenesis during retinal hypoxia [99], oxidative stress has a key role via enhancing the level of growth factor and mediating the evolution of diabetic retinopathy to pathological advanced stages. Utilizing retinal pigment epithelial cells, Simão and colleagues showed that high levels of glucose regulate the expression of different VEGF isoforms through ROS generation [100].

Melatonin synthesis has been reported to reduce in diabetic rats, which this is characterized by the arylalkylamine N‐acetyl transferase (AANAT) activity reduction, as a significant enzyme regulating melatonin daily production; insulin therapy has been reported to ameliorate decreased melatonin levels [101, 102]. In agreement with these findings, Hikichi et al. also reported that nighttime levels of melatonin were considerably lower in diabetic patients compared to non-diabetic subjects, and these levels were lower in patients with proliferative diabetic retinopathy than non-proliferative diabetic retinopathy individuals [103]. In diabetic rats, melatonin reduces histopathological alterations of the retina through decreasing oxidative stress [104]. Dehdashtian et al. [30] reviewed the involvement of inflammation, oxidative stress and autophagy in the pathogenesis of diabetic retinopathy. They indicated that melatonin is able to modulate autophagy cascade and diminishes oxidative stress and inflammation. Melatonin enhances cellular antioxidant defenses and reduces the production of VEGF in Müller cells through activating the PI3K/Akt-Nrf2 signaling pathway; these anti-angiogenic and anti-oxidative effects of melatonin in Müller cells have shown to be receptor-dependent [105]. Melatonin treatment triggers Akt-Nrf2-induced antioxidant system and suppresses NF-κB pathway; it is suggested that regulating retinal melatonin and its receptors could be a beneficial method for treatment of diabetic retinopathy [106]. Melatonin reduces the mean scores of fluorescein leakage and the level of ROS and malondialdehyde (MDA) as well as suppresses the apoptosis and inflammation in rats with diabetic retinopathy through repressing the MAPK pathway [107, 108].

Djordjevic et al. [109] investigated the protective effects of melatonin on retina in pre-diabetic conditions. They showed that oral administration of melatonin (85 μg/animal/day) increases HDL cholesterol concentrations in treated rats and decreases fructosamine and fasting serum glucose levels. In addition, it affected quantitative insulin sensitivity check index and serum insulin in treated groups but had no considerable effects on non-fasting glucose. Totally, melatonin supplementation significantly diminished matrix metallopeptidase 9 (MMP9), VEGF, iNOS, advanced oxidation protein products (AOPP), and MDA levels, hence showing overall positive antioxidant impacts as well as pro-angiogenic signaling in the pre-diabetic retina. Ma and his colleagues investigated the therapeutic effects of melatonin on diabetic retinopathy rats. They showed that the expression of Bax, a pro-apoptotic gene, significantly increases in the retina of diabetic retinopathy rats, while the expression of Bcl-2, an anti-apoptosis gene, decreases. After melatonin therapy, the serum levels of interleukin (IL)-1β and IL-18 also reduces. These results showed that melatonin has anti-inflammatory and anti-apoptosis effects in rats with diabetic retinopathy [107]. Ozdemir et al. [110] showed that daily intraperitoneal administration of melatonin for 12 weeks declines retinal levels of MDA and nitrotyrosine, indicating an antioxidant support. Melatonin also normalizes retinal vascular alterations through decreasing the vasculoregulator cytokines. Studies reporting melatonin effects on diabetic retinopathy have been illustrated in Table 2.

Table 2 Effects of melatonin on diabetic retinopathy reported by various investigations

The therapeutic effects of melatonin on diabetic neuronal complications

Melatonin and CNS-related complications of diabetes

Cognitive functions

Acute and chronic vascular and metabolic perturbations impair the structural and functional brain integrity in diabetic patients. In other words, DM contributes to various structural and functional disorders related to peripheral and central nervous systems [111].

Cerebrovascular alterations, such as neurotropic changes like decreased IGF (insulin growth factor) levels, disrupted vascular reactivity, and attenuated blood flow to the brain demonstrate that mentioned aberrations precede apoptotic neuronal loss as well as functional cognitive disturbances in hippocampus [112, 113]. DM is correlated with moderate impairment in cognitive functions, Alzheimer’s disease and dementia [114,115,116,117]. Cognitive deficit is probably linked to hyperglycemia neurotoxic impacts and alterations in neuronal and neurotransmission functionalities. Enhanced oxidative stress promotes oxidative damages in multiple brain regions, such as the hippocampus [118]. As mentioned above, oxidative stress plays key contributory roles in the development of diabetic complications, while antioxidant treatment may restore or protect physiological functions [118]. Prior researchers have reported that antioxidants decrease the hippocampal neuronal cell damages induced by diabetes-mediated excitotoxicity [119,120,121]. It has been revealed that free radical scavengers protect neurons from diverse neurodegenerative situations. Melatonin, by its antioxidant ability, has been indicated to exert neuroprotective effects under different circumstances [122, 123].

In a study, Tuzuc and colleagues examined and compared the vitamin E and melatonin impacts on cognitive function of diabetic animals. Memory and learning behaviors were studies utilizing a spatial version of the Morris water maze test. Glutathione and lipid peroxidation levels were measured in frontal cortex and hippocampus. The diabetic animals developed considerable impairments in memory and learning behaviors in comparison with control rats. Moreover, the concentration of glutathione is reduced and the level of lipid peroxidation is enhanced in diabetic animals. Vitamin E and melatonin remarkably improved memory and learning performance. In addition, glutathione levels and lipid peroxidation were reversed by these nutraceuticals toward the control values. It can be concluded that, in diabetic patients, oxidative stress contributes to memory and learning deficits, suggesting that antioxidants including vitamin E and melatonin are able to ameliorate cognitive impairments in diabetes [124].

Administration of melatonin in STZ-induced diabetic rats with elevated level of glial fibrillary acidic protein (GFAP) and S100B, as an indicator of astrogliosis, prevents lipid peroxidation, GFAP and and S100B synthesis and the loss of neural cell adhesion molecule, with these effects being associated with the improvement of cognitive ability [125, 126]. Melatonin may show these effects through its antioxidant activity contributing to the increased level of brain antioxidant, decreased brain NOSs activities and plasma cytokines levels, and inhibition of astrocytes apoptosis playing an important role in the CNS homeostasis [104, 127, 128]. Inhibition of oxidative stress-induced activation of Poly(ADP-ribose) polymerase (PARP) cascade is another mechanism by which melatonin can improve high glucose-induced changes in neurotransmitter (glutamate and gamma-Aminobutyric acid (GABA)) levels and neurobehavioral parameters in diabetic rats with neuropathy; over-activation of PARP in response to ROS-induced DNA damage initiates an energy-consuming cycle leading to the depletion of intracellular NAD+ and ATP culminating into energy failure and cell death [129, 130]. A 2-week combination treatment with nicotinamide (300, 1000 mg/kg/day) and melatonin (3, 10 mg/kg/day), or their application as single therapeutic agents ameliorated the neurobehavioral parameters, and GABA and glutamate levels. In general, they showed that concurrent suppression of oxidative stress-PARP overactivation pathway can be valuable in the therapy of DM-related CNS disorders [130].

Depression

DM patients are approximately three times more probable to develop depression [131, 132]. Even pregnant females with gestational diabetes have greater incidence of pregnancy-related depression [133, 134]. Similar to other complications, hyperglycemia is correlated with depression [135], and glycemic therapy reduces depression risk even postpartum depression [136, 137]. Antioxidant defense breakdown may occur in chronic depression, due to a promoted ROS production and an attenuated antioxidant system activity [138].

Regarding antioxidant properties of melatonin, Ergenc and co-workers examined melatonin effects on S100B, AGE receptor (AGER), and AGE levels, oxidative stress, and anxiety and depressive like-behaviors in streptozotocin-mediated diabetic rats. They showed that, in addition to the amelioration of anxiety and depressive like-behaviors, melatonin reversed DM-mediated reduction of S100B and GSH, and enhancement of AGE and MDA levels, indicating anxiolytic and antidepressant potentials in diabetic rats via normalizing oxidative stress, S100B, and AGE/AGER in the hippocampus and prefrontal cortex [69].

Furthermore, Rebai et al. also elucidated that melatonin and fluoxetine decreased anxiety and depression signs, and Hemoglobin A1C (HbA1c) levels in diabetic rats. Melatonin and fluoxetine diminished thiobarbituric acid reactive substances in both cortices. The activity of CAT was restored by melatonin and fluoxetine, while only melatonin ameliorated GPx and glutathione S-transferases (GST) activity in the prefrontal cortex. Moreover, melatonin restored GPx activity in the hippocampus, while fluoxetine had no impact. Overall, they illustrated that antioxidants and antidepressants can counter oxidative disorders and mood related to DM [139]. Altogether, melatonin is a promising therapeutic agent for the treatment of CNS-related complications of DM and further studies are needed to be carried out to prove its efficacy and find possible actions mechanisms as well. Table 3 summarizes investigations evaluating the effect of melatonin on CNS-related complications of diabetes.

Table 3 Studies of melatonin effects on CNS-related complications of diabetes

Therapeutic potentials of melatonin for diabetic neuropathy

Neuropathy is a common complication of diabetes mellitus which approximately affects life quality of 50% of diabetic patients [140]. Oxidative stress and inflammation induced by hyperglycemia plays an important role in the pathogenesis of diabetic neuropathy [141]. The high incidence of diabetic neuropathy is due to the high oxygen requirement of the brain, high content of polyunsaturated fatty acids (PUFAs) and inefficient antioxidant capacity of the brain tissue, which results in the increased vulnerability of brain to diabetes-induced oxidative stress [128]. Prolonged hyperglycaemia promotes hippocampal and cortical neuron apoptosis in diabetic patients leading to the moderate impairments in cognitive function and neurodegenerative diseases such as Alzheimer’s disease [142]. Diabetes mellitus also damages somatic or autonomic nerves causing neuropathic pain, which contributes to the disruption of patients’ daily functions and reduction of life quality [143]. Due to the role of oxidative stress in the induction of diabetic neuropathy, administration of antioxidants such as melatonin may be a therapeutic approach to prevention of diabetic neuropathy (Fig. 1c).

Melatonin secretory patterns is disturbed in diabetic patients with autonomic neuropathy indicating a complex interaction between melatonin dynamics and autonomous neuropathy [144, 145]. Administration of melatonin has been reported to prevent hyperglycemia-induced neurotic damage and occurrence of diabetic neuropathy characterized by increasing motor nerve conduction velocity (MNCV) and sciatic nerve diameter as well as reduction of thermal hyperalgesia [146, 147]. The antihyperalgesic effect of melatonin is suggested to be mediated by the inhibition of l-arginine–NO pathway and induction of opioidergic system [148]. Furthermore, the analgesic properties of melatonin have been reported to results from it activity on melatonin MT2 receptor through modulation of brainstem descending antinociceptive pathways [149, 150]. In dorsal root ganglia neurons of mice, the analgesic effects of melatonin cause from MT2-dependent and independent pathways. The MT2-dependent pathway mediates by the activation of RAR-related orphan receptor alpha (RORα) leading to the inhibition of MAPK1 and subsequent calcium signaling pathways; this effect results in the c-fos, calcitonin gene-related peptide (CGRP), and neuro-inflammatory cytokines such as tumor necrosis factor (TNF)-α and IL-1β. The MT2-independent pathway mediates by inhibition of the expression of nitric oxide synthase 1 (NOS1) [151].

As mentioned previously, high glucose induces oxidative stress and increases Ca2+ ion entry playing important roles in the pathogenesis of diabetic neuropathy [152]. Treatment with melatonin improves hippocampal injury and peripheral neuropathic pain in STZ-induced diabetic rats; these protective activities of melatonin are characterized by the improvement in the spatial navigation memory and results from electrophysiological studies including tibial motor nerve conduction and cortical tibial nerve somatosensory evoked potentials [153,154,155]. Melatonin inhibits oxidative stress-induced Ca2+ mobilization through transient receptor potential (TRP) melastatin 2 (TRPM2) and TRP vanilloid type 1 (TRPV1) channels resulting in the prevention of mitochondrial depolarization, intracellular ROS production and apoptosis in neurons [153]. Histopathological, immunohistochemical and ultrastructural studies also have confirmed the neuroprotective effect of melatonin in STZ-induced diabetic rats with peripheral and central neuropathy. In the sciatic nerves, melatonin has been reported to reduce vacuolization of the myelin sheath, axon–myelin separation and the number of degenerated fibers. In cerebral cortices, melatonin-treated diabetic rats showed the reduction in the neurodegenerative and necrotic changes; spongiosis and reactive astrogliosis were absent and scattered pyknotic neurons with perineural vacuolations and swollen neurons with karolysis were remarkably reduced in animals with cerebral diabetic neuropathy [156, 157].

The modulatory effect of melatonin on inflammatory and autophagy pathways is considered as an important mechanism against diabetic neuropathy. Melatonin elevates the expression of nuclear factor erythroid 2‐related factor 2 (Nrf2) resulting in the up-regulation of the expression of phase II antioxidant/detoxifying enzymes such as heme oxygenase-1 (HO-1) decreasing oxidative stress and activation of nuclear factor-κB (NF‐κB) cascade [158].The expression of PINK1 and microtubule-associated proteins 1A/1B light chain 3 B (LC-3 B), mitophagy markers, increased in neuronal cells following treatment with melatonin. Since silencing of PINK1 expression impairs melatonin-mediated reduction of mitochondrial ROS production and apoptotic marker activation, it is suggested that melatonin prevents neuronal cell apoptosis under high glucose conditions through stimulating PINK1 expression; this effect of melatonin has been demonstrated to be MT2 receptor dependent [159].

Erectile dysfunction is an intractable health problem impressing more than half of male diabetic patients. Evaluation of the effect of melatonin on erectile dysfunction in diabetic rats revealed that melatonin treatment improves the erectile function through ameliorating neuropathy in dorsal penile nerve and major pelvic ganglion. This effect of melatonin is accompanied by alleviation of fibrosis through reduction of collagen deposition, oxidative stress, and p38 MAPK signaling pathway in penis [160]. Retina neuronal cell apoptosis is one of the main pathological symptoms of diabetic retinal neuropathy, which induces by the high glucose-induced excessive production of ROS resulting in the structure and function damages of mitochondria triggering the release of apoptosis-inducing factors and subsequent activation of neuronal apoptosis process [161]. Table 4 summarizes in vivo and in vitro studies evaluating the effect of melatonin on diabetic neuropathy.

Table 4 Studies of melatonin effects on diabetic neuropathy

Melatonin is an appropriate agent for the treatment of diabetic nephropathy

Diabetic nephropathy, as one of the main microvascular complications of both type 1 and type 2 diabetes mellitus, is the most frequent cause of end-stage renal diseases. Diabetic nephropathy is characterized by nephron enlargement, glomerular hyperfiltration as well as the hypertrophy of mesangial cells, which eventually develops into glomerulosclerosis [162]. Several mechanisms have been proposed for diabetic nephropathy progression and development, including lipid disorders, oxidative stress, pro-fibrotic and fibrotic cytokines generation (such as fibronectin (FN)-1, plasminogen activator inhibitor (PAI)-1, and connective tissue growth factor (CTGF) [163,164,165,166].

As mentioned above, ROS have a key role in diabetic complications [167]. In hyperglycemia state, the persistent oxidative stress contributes to damage to the nuclear DNA and the mitochondrial genetic material [168].

Hyperglycemia leads to apoptosis in different types of cells in diabetic nephropathy, such as the proximal tubule epithelial cells [169]. It has been previously explained that the apoptosis occurs through activating numerous intracellular signaling pathway [170]. Now, it is acknowledged that hyperglycemia mediates apoptosis and promotes the gradual loss of kidney function in diabetic nephropathy [171].

Structural melatonin’s features, such as electron-richness, hydrophilicity, and lipophilicity yield potential impacts of this agent in terms of antioxidant capacity and insulin secretion impact by the MT1 receptor [172]. Melatonin elevates numerous antioxidant enzymes, such as SOD, peroxidase, GSH and lipid peroxidase [29]. Melatonin easily crosses cell membranes and leads to protection against free radical-induced damage to biomolecules [173].

Diabetic kidney is more sensitive to ischemia/reperfusion (I/R) injury correlated with enhanced oxidative stress. In a 4-week in vivo study, Shi et al. evaluated the therapeutic effects of melatonin on renal I/R of diabetic rats. Animals were treated with or without melatonin, and renal I/R injury was induced by clamping right and left renal pedicles for 30 min followed by reperfusion for 48 h. Diabetic rats treated with melatonin were protected against renal injury, in aspects of oxidative stress and cell apoptosis in kidney [174].

In accordance with the findings of Parving et al. [175], Motawi and colleagues indicating that the combination of melatonin with Losartan Potassium considerably ameliorates the urine creatinine and serum uric acid levels as well as kidney injury molecule (KIM-1) level, as glomerular damage index [176]. In addition, these agents decrease diabetes mellitus effects on the level of oxidative stress biomarkers, where melatonin and LSP combination significantly ameliorate the levels of GSH, SOD, MDA, and NO in rats with diabetic nephropathy [176]. Aberrant angiogenesis has a pathological role in diabetic nephropathy; however, endothelial-to-mesenchymal transition of glomerular endothelial cells results in enhanced permeability, leading to proteinuria [177, 178]. Melatonin inhibits endothelial-to-mesenchymal transition (EMT) in glomerular endothelial cells exposed to transforming growth factor-β2 (TGF-β2) and in the glomeruli of diabetic rats; this effect is mediated by the up-regulation of miR-497 expression leading to inhibited RhoA/Rho kinase (ROCK) activity [179].

Rashed and his colleagues have shown that incubation of mesenchymal stem cells with melatonin before administration resulted in the improvement of autophagy pathway, elevation of SOD1 level and further decline in TGF-β level in the kidney tissue of diabetic rats compared to those treated with mesenchymal stem cells alone [180].

Onk et al. [173] found enhanced IL-33 levels in the kidney tissue following contrast-induced nephropathy (CIN) in diabetic rats; administration of melatonin in these animals considerably attenuated kidney tissue concentrations of inflammatory cytokines, oxidative stress biomarkers, and IL-33, indicating potential roles of melatonin in the treatment of diabetic nephropathy. Melatonin also reduced serum creatinine in rats with diabetic nephropathy.

In an in vitro study, Ji et al. evaluated protective roles of melatonin against angiotensin II–mediated diabetic nephropathy. They revealed that melatonin markedly enhanced the proliferative rate of cells and decreased angiotensin II-mediated apoptosis, as shown by declined concentrations of apoptotic proteins expression, such as Bax and caspase-3. Also, the recovery of mitochondrial function and reduced oxidative stress were implicated in the protective effect of melatonin. Additionally, the inhibition of Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway demonstrated that cytokine-induced inflammation is also targeted by melatonin [181].

Gumustekin et al. assessed the efficacy of melatonin in the treatment of rats with diabetic nephropathy. After 14 weeks of diabetes induction, 10 mg/kg melatonin was given to diabetic rats for 5 days. Melatonin could not improve hemorheological abnormalities, such as erythrocyte deformability and aggregation, but ameliorated renal injuries by suppressing apoptosis process [182].

It is obvious that melatonin affects various signaling pathways resulting in the reduction of diabetes-induced renal damages (Fig. 1d); however, further studies should be performed to find the exact mechanism of action of melatonin for diabetic nephropathy. Notably, it seems that melatonin deserves to be used in the human trials as a complementary treatment to assess its effectiveness in the treatment of patients with diabetic nephropathy. Herein, Table 5 summarizes in vivo and in vitro studies on the effect of melatonin on diabetic nephropathy.

Table 5 The application of melatonin for the treatment of diabetic nephropathy

Melatonin and diabetic wound healing

Diabetic wounds are leading cause of diabetes-associated amputations, resulting in poor quality of patients’ life and high medical cost [183]. It is characterized by impaired or diminished production of growth factor, granulation tissue quantity, epidermal barrier function, collagen accumulation, macrophage function, and angiogenic response [184, 185]. Disruption in healing process may result from ischemic conditions caused by intrinsic factors such as signaling abnormalities, neuropathy, and vascular problems as well as extrinsic factors including trauma and wound infection [186, 187]. Denervation with sensory abnormalities of the nerves further exacerbates the slowed microcirculation [188].

Melatonin, which is also produced in the skin [189], accelerates wound healing in full-thickness incisional wounds [190]. Ji e al. carried out an investigation to evaluate melatonin potential in improving diabetic wound healing and the possible mechanism. Treatment of mice bone marrow-derived endothelial progenitor cells (EPCs) with melatonin inhibits advanced AGE-mediated cellular dysfunction and apoptosis. Melatonin is reported to enhance autophagy flux characterized by the elevation of LC3 level, and reduction of p62 accumulation. Moreover, AMPK/mTOR signaling pathway is implicated in stimulating autophagy via melatonin. In a diabetic wound healing model, melatonin improved disrupted wound healing. Therefore, melatonin is suggested to improve disrupted wound healing through inhibiting apoptotic events by stimulation of autophagy pathway [191].

Melatonin also promotes umbilical cord blood (UCB)-mesenchymal stem cells (MSCs) motility through enhancing cytoskeletal reorganization which this effect is mediated by melatonin MT2 receptor triggering Gq protein alpha (Gαq)-dependent protein kinase (PK)Cζ phosphorylation regulating Focal adhesion kinase (FAK)/paxillin-mediated cell division cycle 42 (Cdc42)/actin-related proteins (Arp2/3) activation [192].

In keratinocytes, melatonin reduces high glucose-induced the release of inflammatory cytokines, such as interleukin (IL)-1β, IL-6, IL-8 and tumor necrosis factor-α associated with the reduction of ROS formation and the elevation of SOD activity. In diabetic wound, melatonin causes essential events in the healing process including suppression of apoptosis, promotion of migration and proliferation, reduction of oxidative stress, and suppression of inflammatory process in keratinocytes [193]. Hence, melatonin is suggested to be useful in diabetic foot ulcer through regulating keratinocyte activity. Table 6 summarizes investigations have evaluated the effect of melatonin on diabetic wound.

Table 6 Investigations on melatonin and diabetic wound healing

Melatonin application for diabetic patients in clinical practice

Raygan et al. carried out a randomized, double-blind clinical trial on diabetic patients with coronary heart disease. Sixty patients were divided into two groups to receive either placebo or 10 mg melatonin for 12 weeks. Melatonin had appropriate impacts on glycemic control, HDL-cholesterol, serum levels of high-sensitivity C-reactive protein (hs-CRP), blood pressure, plasma MDA, NO, and GSH as well as mental health parameters [194].

Kadhim et al. performed a clinical trial on patients with type 2 diabetes who were poorly controlled with metformin. Forty-six patients with type 2 diabetes were divided into three groups and were treated for 90 days with daily oral administration of melatonin (10 mg), zinc acetate (50 mg), or both supplements in addition to the routine used metformin or placebo. Daily intake of zinc and melatonin reduced the level of microalbuminuria and ameliorated the lipid profile. In conclusion, the addition of these supplements to metformin ameliorated the tissue responses to this oral hypoglycemic agent [67].

Another investigation was conducted to assess therapeutic efficacy of melatonin and zinc in glycemic control of type 2 diabetic patients poorly controlled with metformin. The supplements were administered at the same doses in 90 days. They revealed that the combination of zinc acetate and melatonin, when utilizes alone or in combination with metformin, ameliorates fasting and post-prandial glucose levels in diabetic subjects [195]. Regarding what discussed melatonin supplementations could be used for diabetic complications in human studies. To prove the effectiveness and safety of melatonin as a complementary treatment for diabetic patients, it is important to design large clinical trials.

Conclusions

Due to the rapid increase of the global DM prevalence, exploring efficient therapies for diabetic complications is an important issue. Routine treatments for DM are generally based on metabolic control, and existing drugs cannot totally control the development of complications. Researches have recently indicated the capability of melatonin to modulate various signaling pathways and demonstrated its therapeutic and preventive effects in several diseases, such as DM. Preclinical and clinical studies have shown different mechanisms of action of melatonin including reduction of glucose production, and improvement of pro-inflammatory pathways as well as oxidative stress state in DM patients. Previously, it has been revealed that melatonin treatment ameliorates DM through regulating the lipid and glucose metabolism, reducing the insulin resistance and increasing the sensitivity to insulin in experimental models of DM. It will be crucial to perform human clinical trials utilizing combinations of melatonin with current therapeutic agents for treatment and prevention of cardiomyopathy, neuropathy, retinopathy, nephropathy, and other diabetic complications.

Availability of data and materials

Not applicable

References

  1. American Diabetes Association. Diagnosis and classification of diabetes mellitus. Diabetes Care. 2014;37(Suppl 1):S81–90.

    Article  Google Scholar 

  2. Shaw JE, Sicree RA, Zimmet PZ. Global estimates of the prevalence of diabetes for 2010 and 2030. Diabetes Res Clin Pract. 2010;87(1):4–14.

    Article  CAS  PubMed  Google Scholar 

  3. Whiting DR, Guariguata L, Weil C, Shaw J. IDF diabetes atlas: global estimates of the prevalence of diabetes for 2011 and 2030. Diabetes Res Clin Pract. 2011;94(3):311–21.

    Article  PubMed  Google Scholar 

  4. Guariguata L, Whiting DR, Hambleton I, Beagley J, Linnenkamp U, Shaw JE. Global estimates of diabetes prevalence for 2013 and projections for 2035. Diabetes Res Clin Pract. 2014;103(2):137–49.

    Article  CAS  PubMed  Google Scholar 

  5. Du WH, Peng SM, Liu ZH, Shi L, Tan LF, Zou XQ. Hypoglycemic effect of the water extract of Pu-erh tea. J Agric Food Chem. 2012;60(40):10126–32.

    Article  CAS  PubMed  Google Scholar 

  6. Li Y, Wang C, Huai Q, Guo F, Liu L, Feng R, et al. Effects of tea or tea extract on metabolic profiles in patients with type 2 diabetes mellitus: a meta-analysis of ten randomized controlled trials. Diabetes Metabol Res Rev. 2016;32(1):2–10.

    Article  Google Scholar 

  7. Szewczyk PB, Dziuba AM, Poniewierka E. Melatonin–metabolism and the role of pineal hormone. Pielęgniarstwo i Zdrowie Publiczne Nursing and Public Health. 2018;8(2):135–9.

    Google Scholar 

  8. Frese T, Bach AG, Muhlbauer E, Ponicke K, Bromme HJ, Welp A, et al. Pineal melatonin synthesis is decreased in type 2 diabetic Goto-Kakizaki rats. Life Sci. 2009;85(13–14):526–33.

    Article  CAS  PubMed  Google Scholar 

  9. Hardeland R. Melatonin in healthy aging and longevity. Hormones in ageing and longevity. Berlin: Springer; 2017. p. 209–42.

    Book  Google Scholar 

  10. Esposito E, Cuzzocrea S. Antiinflammatory activity of melatonin in central nervous system. Curr Neuropharmacol. 2010;8(3):228–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Yousaf F, Seet E, Venkatraghavan L, Abrishami A, Chung F. Efficacy and safety of melatonin as an anxiolytic and analgesic in the perioperative period: a qualitative systematic review of randomized trials. Anesthesiology. 2010;113(4):968–76.

    Article  CAS  PubMed  Google Scholar 

  12. Boga JA, Caballero B, Potes Y, Perez-Martinez Z, Reiter RJ, Vega-Naredo I, et al. Therapeutic potential of melatonin related to its role as an autophagy regulator: a review. J Pineal Res. 2019;66(1):e12534.

    Article  PubMed  CAS  Google Scholar 

  13. She Q, Han Z, Liang S, Xu W, Li X, Zhao Y, et al. Impacts of circadian rhythm and melatonin on the specific activities of immune and antioxidant enzymes of the Chinese mitten crab (Eriocheir sinensis). Fish Shellfish Immunol. 2019;89:345–53.

    Article  CAS  PubMed  Google Scholar 

  14. Lv D, Tan T, Zhu T, Wang J, Zhang S, Zhang L, et al. Leptin mediates the effects of melatonin on female reproduction in mammals. J Pineal Res. 2019;66(3):e12559.

    Article  PubMed  CAS  Google Scholar 

  15. Zhang C, Zhang Q, Pang Y, Song X, Zhou N, Wang J, et al. The protective effects of melatonin on oxidative damage and the immune system of the Chinese mitten crab (Eriocheir sinensis) exposed to deltamethrin. Sci Total Environ. 2019;653:1426–34.

    Article  CAS  PubMed  Google Scholar 

  16. Garcia-Maurino S, Pozo D, Carrillo-Vico A, Calvo JR, Guerrero JM. Melatonin activates Th1 lymphocytes by increasing IL-12 production. Life Sci. 1999;65(20):2143–50.

    Article  CAS  PubMed  Google Scholar 

  17. Reiter RJ. The pineal and its hormones in the control of reproduction in mammals. Endocr Rev. 1980;1(2):109–31.

    Article  CAS  PubMed  Google Scholar 

  18. Yellon SM, Foster DL. Melatonin rhythms time photoperiod-induced puberty in the female lamb. Endocrinology. 1986;119(1):44–9.

    Article  CAS  PubMed  Google Scholar 

  19. Kauppila A, Kivelä A, Pakarinen A, Vakkuri O. Inverse seasonal relationship between melatonin and ovarian activity in humans in a region with a strong seasonal contrast in luminosity. J Clin Endocrinol Metabol. 1987;65(5):823–8.

    Article  CAS  Google Scholar 

  20. Woo MM, Tai C-J, Kang SK, Nathwani PS, Pang SF, Leung PC. Direct action of melatonin in human granulosa-luteal cells. J Clin Endocrinol Metabol. 2001;86(10):4789–97.

    Article  CAS  Google Scholar 

  21. Scarinci E, Tropea A, Notaristefano G, Arena V, Alesiani O, Fabozzi SM, et al. “Hormone of darkness” and human reproductive process: direct regulatory role of melatonin in human corpus luteum. J Endocrinol Invest. 2019;42(10):1191–7.

    Article  CAS  PubMed  Google Scholar 

  22. Webley G, Luck M. Melatonin directly stimulates the secretion of progesterone by human and bovine granulosa cells in vitro. Reproduction. 1986;78(2):711–7.

    Article  CAS  Google Scholar 

  23. Reiter RJ, Tan D-X, Qi W, Manchester LC, Karbownik M, Calvo JR. Pharmacology and physiology of melatonin in the reduction of oxidative stress in vivo. Neurosignals. 2000;9(3–4):160–71.

    Article  CAS  Google Scholar 

  24. Coto-Montes A, Boga JA, Rosales-Corral S, Fuentes-Broto L, Tan D-X, Reiter RJ. Role of melatonin in the regulation of autophagy and mitophagy: a review. Mol Cell Endocrinol. 2012;361(1):12–23.

    Article  CAS  PubMed  Google Scholar 

  25. Reiter RJ. Functional pleiotropy of the neurohormone melatonin: antioxidant protection and neuroendocrine regulation. Front Neuroendocrinol. 1995;16(4):383–415.

    Article  CAS  PubMed  Google Scholar 

  26. Venegas C, García JA, Escames G, Ortiz F, López A, Doerrier C, et al. Extrapineal melatonin: analysis of its subcellular distribution and daily fluctuations. J Pineal Res. 2012;52(2):217–27.

    Article  CAS  PubMed  Google Scholar 

  27. Barlow-Walden L, Reiter RJ, Abe M, Pablos M, Menendez-Pelaez A, Chen L-D, et al. Melatonin stimulates brain glutathione peroxidase activity. Neurochem Int. 1995;26(5):497–502.

    Article  CAS  PubMed  Google Scholar 

  28. Pablos MI, Agapito MT, Gutierrez R, Recio JM, Reiter RJ, Barlow-Walden L, et al. Melatonin stimulates the activity of the detoxifying enzyme glutathione peroxidase in several tissues of chicks. J Pineal Res. 1995;19(3):111–5.

    Article  CAS  PubMed  Google Scholar 

  29. Rodriguez C, Mayo JC, Sainz RM, Antolin I, Herrera F, Martin V, et al. Regulation of antioxidant enzymes: a significant role for melatonin. J Pineal Res. 2004;36(1):1–9.

    Article  CAS  PubMed  Google Scholar 

  30. Dehdashtian E, Mehrzadi S, Yousefi B, Hosseinzadeh A, Reiter RJ, Safa M, et al. Diabetic retinopathy pathogenesis and the ameliorating effects of melatonin; involvement of autophagy, inflammation and oxidative stress. Life Sci. 2018;193:20–33.

    Article  CAS  PubMed  Google Scholar 

  31. Reiter RJ, Mayo JC, Tan DX, Sainz RM, Alatorre-Jimenez M, Qin L. Melatonin as an antioxidant: under promises but over delivers. J Pineal Res. 2016;61(3):253–78.

    Article  CAS  PubMed  Google Scholar 

  32. Maiocchi SL, Morris JC, Rees MD, Thomas SR. Regulation of the nitric oxide oxidase activity of myeloperoxidase by pharmacological agents. Biochem Pharmacol. 2017;135:90–115.

    Article  CAS  PubMed  Google Scholar 

  33. Marshall K-A, Reiter RJ, Poeggeler B, Aruoma OI, Halliwell B. Evaluation of the antioxidant activity of melatonin in vitro. Free Radical Biol Med. 1996;21(3):307–15.

    Article  CAS  Google Scholar 

  34. Mirza-Aghazadeh-Attari M, Mohammadzadeh A, Mostavafi S, Mihanfar A, Ghazizadeh S, Sadighparvar S, et al. Melatonin: an important anticancer agent in colorectal cancer. J Cell Physiol. 2020;235(2):804–17.

    Article  CAS  PubMed  Google Scholar 

  35. Gitto E, Aversa S, Salpietro CD, Barberi I, Arrigo T, Trimarchi G, et al. Pain in neonatal intensive care: role of melatonin as an analgesic antioxidant. J Pineal Res. 2012;52(3):291–5.

    Article  CAS  PubMed  Google Scholar 

  36. Doganlar ZB, Guclu H, Oztopuz O, Turkon H, Dogan A, Uzun M, et al. the role of melatonin in oxidative stress, DNA damage, apoptosis and angiogenesis in fetal eye under preeclampsia and melatonin deficiency stress. Curr Eye Res. 2019;44(10):1157–69.

    CAS  PubMed  Google Scholar 

  37. Wei S, Smits MG, Tang X, Kuang L, Meng H, Ni S, et al. Efficacy and safety of melatonin for sleep onset insomnia in children and adolescents: a meta-analysis of randomized controlled trials. Sleep Med. 2019;68:1–8.

    Article  PubMed  Google Scholar 

  38. Andersen LP, Gogenur I, Rosenberg J, Reiter RJ. The safety of melatonin in humans. Clin Drug Investig. 2016;36(3):169–75.

    Article  CAS  PubMed  Google Scholar 

  39. de Matos Cavalcante AG, de Bruin PF, de Bruin VM, Nunes DM, Pereira ED, Cavalcante MM, et al. Melatonin reduces lung oxidative stress in patients with chronic obstructive pulmonary disease: a randomized, double-blind, placebo-controlled study. J Pineal Res. 2012;53(3):238–44.

    Article  PubMed  CAS  Google Scholar 

  40. Hansen MV, Andersen LT, Madsen MT, Hageman I, Rasmussen LS, Bokmand S, et al. Effect of melatonin on depressive symptoms and anxiety in patients undergoing breast cancer surgery: a randomized, double-blind, placebo-controlled trial. Breast Cancer Res Treat. 2014;145(3):683–95.

    Article  CAS  PubMed  Google Scholar 

  41. Jain SV, Horn PS, Simakajornboon N, Beebe DW, Holland K, Byars AW, et al. Melatonin improves sleep in children with epilepsy: a randomized, double-blind, crossover study. Sleep Med. 2015;16(5):637–44.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Besag FMC, Vasey MJ, Lao KSJ, Wong ICK. Adverse events associated with melatonin for the treatment of primary or secondary sleep disorders: a systematic review. CNS Drugs. 2019;33(12):1167–86.

    Article  PubMed  Google Scholar 

  43. Balduini W, Weiss MD, Carloni S, Rocchi M, Sura L, Rossignol C, et al. Melatonin pharmacokinetics and dose extrapolation after enteral infusion in neonates subjected to hypothermia. J Pineal Res. 2019;66(4):e12565.

    Article  PubMed  CAS  Google Scholar 

  44. Merchant NM, Azzopardi DV, Hawwa AF, McElnay JC, Middleton B, Arendt J, et al. Pharmacokinetics of melatonin in preterm infants. Br J Clin Pharmacol. 2013;76(5):725–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Salehi B, Sharopov F, Fokou PVT, Kobylinska A, Jonge L, Tadio K, et al. Melatonin in Medicinal and Food Plants: Occurrence, Bioavailability, and Health Potential for Humans. Cells. 2019;8:7.

    Article  CAS  Google Scholar 

  46. Harpsoe NG, Andersen LP, Gogenur I, Rosenberg J. Clinical pharmacokinetics of melatonin: a systematic review. Eur J Clin Pharmacol. 2015;71(8):901–9.

    Article  PubMed  CAS  Google Scholar 

  47. Andersen LP, Werner MU, Rosenkilde MM, Harpsoe NG, Fuglsang H, Rosenberg J, et al. Pharmacokinetics of oral and intravenous melatonin in healthy volunteers. BMC Pharmacol Toxicol. 2016;17:8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Al-Omary FA. Melatonin: comprehensive profile. Prof Drug Subst Excip Relat Methodol. 2013;38:159–226.

    Article  CAS  Google Scholar 

  49. Bellapart J, Roberts JA, Appadurai V, Wallis SC, Nunez-Nunez M, Boots RJ. Pharmacokinetics of a novel dosing regimen of oral melatonin in critically ill patients. Clin Chem Lab Med. 2016;54(3):467–72.

    Article  CAS  PubMed  Google Scholar 

  50. Andersen LP, Gogenur I, Rosenberg J, Reiter RJ. Pharmacokinetics of melatonin: the missing link in clinical efficacy? Clin Pharmacokinet. 2016;55(9):1027–30.

    Article  CAS  PubMed  Google Scholar 

  51. Tordjman S, Chokron S, Delorme R, Charrier A, Bellissant E, Jaafari N, et al. Melatonin: pharmacology, functions and therapeutic benefits. Curr Neuropharmacol. 2017;15(3):434–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. DeMuro RL, Nafziger AN, Blask DE, Menhinick AM, Bertino JS Jr. The absolute bioavailability of oral melatonin. J Clin Pharmacol. 2000;40(7):781–4.

    Article  CAS  PubMed  Google Scholar 

  53. Di WL, Kadva A, Johnston A, Silman R. Variable bioavailability of oral melatonin. New Engl J Med. 1997;336(14):1028–9.

    Article  CAS  PubMed  Google Scholar 

  54. Cavallo A, Ritschel WA. Pharmacokinetics of melatonin in human sexual maturation. J Clin Endocrinol Metabol. 1996;81(5):1882–6.

    CAS  Google Scholar 

  55. Flo A, Cambras T, Diez-Noguera A, Calpena A. Melatonin pharmacokinetics after transdermal administration changes according to the time of the day. Eur J Pharm Sci. 2017;96:164–70.

    Article  CAS  PubMed  Google Scholar 

  56. Rodrigues SC, Pantaleão L, Lellis-Santos C, Veras K, Amaral F, Anhê G, et al. Increased corticosterone levels contribute to glucose intolerance induced by the absence of melatonin. Feder Am Soc Exp Biol. 2013;2013:1161.

    Google Scholar 

  57. Sun H, Wang X, Chen J, Gusdon AM, Song K, Li L, et al. Melatonin treatment improves insulin resistance and pigmentation in obese patients with acanthosis nigricans. Int J Endocrinol. 2018;2018:2304746.

    PubMed  PubMed Central  Google Scholar 

  58. Milosavljević A, Jukić DL, Toljić B, Milašin J, Želetović DB, Brković B, Roganović J. Melatonin levels in human diabetic dental pulp tissue and its effects on dental pulp cells under hyperglycaemic conditions. Int Endodon J. 2018;51(10):1149–58.

    Article  Google Scholar 

  59. de Oliveira AC, Andreotti S, Sertie RAL, Campana AB, de Proença ARG, Vasconcelos RP, et al. Combined treatment with melatonin and insulin improves glycemic control, white adipose tissue metabolism and reproductive axis of diabetic male rats. Life Sci. 2018;199:158–66.

    Article  PubMed  CAS  Google Scholar 

  60. Sartori C, Dessen P, Mathieu C, Monney A, Bloch J, Nicod P, et al. Melatonin improves glucose homeostasis and endothelial vascular function in high-fat diet-fed insulin-resistant mice. Endocrinology. 2009;150(12):5311–7.

    Article  CAS  PubMed  Google Scholar 

  61. da Costa CFP, Gobbo MG, Taboga SR, Pinto-Fochi ME, Góes RM. Melatonin intake since weaning ameliorates steroidogenic function and sperm motility of streptozotocin-induced diabetic rats. Andrology. 2016;4(3):526–41.

    Article  PubMed  CAS  Google Scholar 

  62. Behram KY, Guntekin U, Tosun V, Korucuk N, Bozdemir MN. Melatonin protects against streptozotocin-induced diabetic cardiomyopathy by the phosphorylation of vascular endothelial growth factor-A (VEGF-A). Cell Mol Biol. 2018;64(14):47–52.

    Article  Google Scholar 

  63. Heo JI, Yoon DW, Yu JH, Kim NH, Yoo HJ, Seo JA, et al. Melatonin improves insulin resistance and hepatic steatosis through attenuation of alpha-2-HS-glycoprotein. J Pineal Res. 2018;65(2):e12493.

    Article  PubMed  CAS  Google Scholar 

  64. Xu J, Gao H, Zhang L, Rong S, Yang W, Ma C, et al. Melatonin alleviates cognition impairment by antagonizing brain insulin resistance in aged rats fed a high-fat diet. J Pineal Res. 2019;67(2):e12584.

    PubMed  Google Scholar 

  65. Garfinkel D, Zorin M, Wainstein J, Matas Z, Laudon M, Zisapel N. Efficacy and safety of prolonged-release melatonin in insomnia patients with diabetes: a randomized, double-blind, crossover study. Diabetes, metabolic syndrome and obesity : targets and therapy. 2011;4:307–13.

    CAS  Google Scholar 

  66. Rezvanfar MR, Heshmati G, Chehrei A, Haghverdi F, Rafiee F, Rezvanfar F. Effect of bedtime melatonin consumption on diabetes control and lipid profile. Int J Diabetes Dev Count. 2017;37(1):74–7.

    Article  CAS  Google Scholar 

  67. Kadhim HM, Ismail SH, Hussein KI, Bakir IH, Sahib AS, Khalaf BH, et al. Effects of melatonin and zinc on lipid profile and renal function in type 2 diabetic patients poorly controlled with metformin. J Pineal Res. 2006;41(2):189–93.

    Article  CAS  PubMed  Google Scholar 

  68. Espino J, Rodriguez AB, Pariente JA. Melatonin and oxidative stress in the diabetic state: clinical implications and potential therapeutic applications. Curr Med Chem. 2019;26(22):4178–90.

    Article  CAS  PubMed  Google Scholar 

  69. Ergenc M, Ozacmak HS, Turan I, Ozacmak VH. Melatonin reverses depressive and anxiety like-behaviours induced by diabetes: involvement of oxidative stress, age, rage and S100B levels in the hippocampus and prefrontal cortex of rats. Arch Physiol Biochem. 2019;2019:1–9.

    Article  CAS  Google Scholar 

  70. Doosti-Irani A, Ostadmohammadi V, Mirhosseini N, Mansournia MA, Reiter RJ, Kashanian M, et al. Correction: the effects of melatonin supplementation on glycemic control: a systematic review and meta-analysis of randomized controlled trials. Hormone Metabol Res. 2018;50(11):e6.

    Article  CAS  Google Scholar 

  71. Borghetti G, von Lewinski D, Eaton DM, Sourij H, Houser SR, Wallner M. Diabetic cardiomyopathy: current and future therapies, beyond glycemic control. Front Physiol. 2018;9:1514.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Jia G, Hill MA, Sowers JR. Diabetic cardiomyopathy: an update of mechanisms contributing to this clinical entity. Circ Res. 2018;122(4):624–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Ahire YS, Ghaisas MM, Dandawate P, Gandhi S. Beneficial effects of co-administration of PPAR-γ agonist with melatonin on cardiovascular complications associated with diabetes. Chronic Young Sci. 2013;4(1):59.

    Article  CAS  Google Scholar 

  74. Kandemir Y, Tosun V, Güntekin Ü. Melatonin protects against streptozotocin-induced diabetic cardiomyopathy through the mammalian target of rapamycin (mTOR) signaling pathway. Adv Clin Exp Med. 2019;28(9):1171–7.

    Article  PubMed  Google Scholar 

  75. Hu Y, Suarez J, Fricovsky E, Wang H, Scott BT, Trauger SA, et al. Increased enzymatic O-GlcNAcylation of mitochondrial proteins impairs mitochondrial function in cardiac myocytes exposed to high glucose. J Biol Chem. 2009;284(1):547–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Dunlay SM, Givertz MM, Aguilar D, Allen LA, Chan M, Desai AS, et al. Type 2 Diabetes Mellitus and Heart Failure: a Scientific Statement From the American Heart Association and the Heart Failure Society of America: this statement does not represent an update of the 2017 ACC/AHA/HFSA heart failure guideline update. Circulation. 2019;140(7):e294–324.

    Article  CAS  PubMed  Google Scholar 

  77. Aksoy N, Vural H, Sabuncu T, Aksoy S. Effects of melatonin on oxidative–antioxidative status of tissues in streptozotocin-induced diabetic rats. Cell Biochem Funct. 2003;21(2):121–5.

    Article  CAS  PubMed  Google Scholar 

  78. Amin AH, El-Missiry MA, Othman AI. Melatonin ameliorates metabolic risk factors, modulates apoptotic proteins, and protects the rat heart against diabetes-induced apoptosis. Eur J Pharmacol. 2015;747:166–73.

    Article  CAS  PubMed  Google Scholar 

  79. Yu L, Gong B, Duan W, Fan C, Zhang J, Li Z, et al. Melatonin ameliorates myocardial ischemia/reperfusion injury in type 1 diabetic rats by preserving mitochondrial function: role of AMPK-PGC-1α-SIRT3 signaling. Sci Rep. 2017;7:41337.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Yu L-M, Di W-C, Dong X, Li Z, Zhang Y, Xue X-D, et al. Melatonin protects diabetic heart against ischemia-reperfusion injury, role of membrane receptor-dependent cGMP-PKG activation. BBA Mol Basis Dis. 2018;1864(2):563–78.

    Article  CAS  Google Scholar 

  81. Ding M, Feng N, Tang D, Feng J, Li Z, Jia M, et al. Melatonin prevents D rp1-mediated mitochondrial fission in diabetic hearts through SIRT 1-PGC 1α pathway. J Pineal Res. 2018;65(2):e12491.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  82. Zhou H, Yue Y, Wang J, Ma Q, Chen Y. Melatonin therapy for diabetic cardiomyopathy: a mechanism involving Syk-mitochondrial complex I-SERCA pathway. Cell Signal. 2018;47:88–100.

    Article  CAS  PubMed  Google Scholar 

  83. Xiong FY, Tang ST, Su H, Tang HQ, Jiang P, Zhou Q, et al. Melatonin ameliorates myocardial apoptosis by suppressing endoplasmic reticulum stress in rats with long-term diabetic cardiomyopathy. Mol Med Rep. 2018;17(1):374–81.

    CAS  PubMed  Google Scholar 

  84. Gao L, Zhao YC, Liang Y, Lin XH, Tan YJ, Wu DD, et al. The impaired myocardial ischemic tolerance in adult offspring of diabetic pregnancy is restored by maternal melatonin treatment. J Pineal Res. 2016;61(3):340–52.

    Article  CAS  PubMed  Google Scholar 

  85. Kandemir YB, Guntekin Ü, Tosun V, Korucuk N, Bozdemir MN. Melatonin protects against streptozotocin-induced diabetic cardiomyopathy by the phosphorylation of vascular endothelial growth factor-A (VEGF-A). Cell Mol Biol. 2018;64:14.

    Google Scholar 

  86. Wang S, Zhao Z, Feng X, Cheng Z, Xiong Z, Wang T, et al. Melatonin activates Parkin translocation and rescues the impaired mitophagy activity of diabetic cardiomyopathy through Mst1 inhibition. J Cell Mol Med. 2018;22(10):5132–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Zhang M, Lin J, Wang S, Cheng Z, Hu J, Wang T, et al. Melatonin protects against diabetic cardiomyopathy through Mst1/Sirt3 signaling. J Pineal Res. 2017;63(2):e12418.

    Article  CAS  Google Scholar 

  88. Williams R, Airey M, Baxter H, Forrester J, Kennedy-Martin T, Girach A. Epidemiology of diabetic retinopathy and macular oedema: a systematic review. Eye (London, England). 2004;18(10):963–83.

    Article  CAS  Google Scholar 

  89. Frank RN. Diabetic retinopathy. N Engl J Med. 2004;350(1):48–58.

    Article  CAS  PubMed  Google Scholar 

  90. Maniadakis N, Konstantakopoulou E. Cost effectiveness of treatments for diabetic retinopathy: a systematic literature review. PharmacoEconomics. 2019;37(8):995–1010.

    Article  PubMed  Google Scholar 

  91. Cho N, Shaw J, Karuranga S. International Diabetes Federation (IDF) diabetes atlas: global estimates of diabetes prevalence for 2017 and projections for 2045. DiabetesRes. 2018;138:271–81.

    CAS  Google Scholar 

  92. Tang J, Kern TS. Inflammation in diabetic retinopathy. Prog Retin Eye Res. 2011;30(5):343–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Kowluru RA, Chan PS. Oxidative stress and diabetic retinopathy. Exp Diabetes Res. 2007;2007:43603.

    PubMed  PubMed Central  Google Scholar 

  94. Santiago AR, Boia R, Aires ID, Ambrosio AF, Fernandes R. Sweet stress: coping with vascular dysfunction in diabetic retinopathy. Front Physiol. 2018;9:820.

    Article  PubMed  PubMed Central  Google Scholar 

  95. Al-Kharashi AS. Role of oxidative stress, inflammation, hypoxia and angiogenesis in the development of diabetic retinopathy. Saudi J Ophthalmol. 2018;32(4):318–23.

    Article  PubMed  PubMed Central  Google Scholar 

  96. Shafabakhsh R, Aghadavod E, Mobini M, Heidari-Soureshjani R, Asemi Z. Association between microRNAs expression and signaling pathways of inflammatory markers in diabetic retinopathy. J Cell Physiol. 2019;234(6):7781–7.

    Article  CAS  PubMed  Google Scholar 

  97. Simo-Servat O, Hernandez C, Simo R. Usefulness of the vitreous fluid analysis in the translational research of diabetic retinopathy. Mediat Inflamm. 2012;2012:872978.

    Article  Google Scholar 

  98. Patel JI, Saleh GM, Hykin PG, Gregor ZJ, Cree IA. Concentration of haemodynamic and inflammatory related cytokines in diabetic retinopathy. Eye (London, England). 2008;22(2):223–8.

    Article  CAS  Google Scholar 

  99. Arjamaa O, Nikinmaa M. Oxygen-dependent diseases in the retina: role of hypoxia-inducible factors. Exp Eye Res. 2006;83(3):473–83.

    Article  CAS  PubMed  Google Scholar 

  100. Simão S, Bitoque DB, Calado SM, Silva GA. Oxidative stress modulates the expression of VEGF isoforms in the diabetic retina. N Front Ophthalmol. 2016;2(1):77–83.

    Google Scholar 

  101. Coon SL, Bégay V, Deurloo D, Falcón J, Klein DC. Two arylalkylamine N-acetyltransferase genes mediate melatonin synthesis in fish. J Biol Chem. 1999;274(13):9076–82.

    Article  CAS  PubMed  Google Scholar 

  102. Buonfiglio D, Peliciari-Garcia RA, Amaral FG, Peres R, Nogueira TC, Afeche SC, et al. Early-stage retinal melatonin synthesis impairment in streptozotocin-induced diabetic wistar rats. Investig Ophthalmol Vis Sci. 2011;52(10):7416–22.

    Article  CAS  Google Scholar 

  103. Hikichi T, Tateda N, Miura T. Alteration of melatonin secretion in patients with type 2 diabetes and proliferative diabetic retinopathy. Clin Ophthalmol. 2011;5:655–60.

    Article  PubMed  PubMed Central  Google Scholar 

  104. Gurpinar T, Ekerbicer N, Uysal N, Barut T, Tarakci F, Tuglu MI. The effects of the melatonin treatment on the oxidative stress and apoptosis in diabetic eye and brain. Sci World J. 2012;2012:498489.

    Article  CAS  Google Scholar 

  105. Jiang T, Chang Q, Zhao Z, Yan S, Wang L, Cai J, et al. Melatonin-mediated cytoprotection against hyperglycemic injury in Muller cells. PLoS ONE. 2012;7(12):e50661.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Jiang T, Chang Q, Cai J, Fan J, Zhang X, Xu G. Protective effects of melatonin on retinal inflammation and oxidative stress in experimental diabetic retinopathy. Oxidat Med Cell Long. 2016;2016:3528274.

    Google Scholar 

  107. Ma Y, Zhao Q, Shao Y, Cao MZ, Zhao M, Wang D. Melatonin inhibits the inflammation and apoptosis in rats with diabetic retinopathy via MAPK pathway. Eur Rev Med Pharmacol Sci. 2019;23(3 Suppl):1–8.

    CAS  PubMed  Google Scholar 

  108. Mehrzadi S, Motevalian M, Rezaei Kanavi M, Fatemi I, Ghaznavi H, Shahriari M. Protective effect of melatonin in the diabetic rat retina. Fundam Clin Pharmacol. 2018;32(4):414–21.

    Article  CAS  PubMed  Google Scholar 

  109. Djordjevic B, Cvetkovic T, Stoimenov TJ, Despotovic M, Zivanovic S, Basic J, et al. Oral supplementation with melatonin reduces oxidative damage and concentrations of inducible nitric oxide synthase, VEGF and matrix metalloproteinase 9 in the retina of rats with streptozotocin/nicotinamide induced pre-diabetes. Eur J Pharmacol. 2018;833:290–7.

    Article  CAS  PubMed  Google Scholar 

  110. Ozdemir G, Ergun Y, Bakaris S, Kilinc M, Durdu H, Ganiyusufoglu E. Melatonin prevents retinal oxidative stress and vascular changes in diabetic rats. Eye (London, England). 2014;28(8):1020–7.

    Article  CAS  PubMed Central  Google Scholar 

  111. Biessels G-J, Kappelle A, Bravenboer B, Erkelens D, Gispen W. Cerebral function in diabetes mellitus. Diabetologia. 1994;37(7):643–50.

    Article  CAS  PubMed  Google Scholar 

  112. Li ZG, Zhang W, Sima AA. The role of impaired insulin/IGF action in primary diabetic encephalopathy. Brain Res. 2005;1037(1–2):12–24.

    Article  CAS  PubMed  Google Scholar 

  113. Li ZG, Zhang W, Grunberger G, Sima AA. Hippocampal neuronal apoptosis in type 1 diabetes. Brain Res. 2002;946(2):221–31.

    Article  CAS  PubMed  Google Scholar 

  114. Sima AA. Diabetes underlies common neurological disorders. Ann Neurol. 2004;56(4):459–61.

    Article  PubMed  Google Scholar 

  115. Ristow M. Neurodegenerative disorders associated with diabetes mellitus. J Mol Med. 2004;82(8):510–29.

    Article  PubMed  Google Scholar 

  116. Northam EA, Rankins D, Cameron FJ. Therapy insight: the impact of type 1 diabetes on brain development and function. Nat Clin Pract Neurol. 2006;2(2):78–86.

    Article  PubMed  Google Scholar 

  117. Brismar T, Maurex L, Cooray G, Juntti-Berggren L, Lindstrom P, Ekberg K, et al. Predictors of cognitive impairment in type 1 diabetes. Psychoneuroendocrinology. 2007;32(8–10):1041–51.

    Article  PubMed  Google Scholar 

  118. Kuhad A, Chopra K. Neurobiology of diabetic encephalopathy. Drug. Future. 2008;33:763–75.

    CAS  Google Scholar 

  119. Baydas G, Canatan H, Turkoglu A. Comparative analysis of the protective effects of melatonin and vitamin E on streptozocin-induced diabetes mellitus. J Pineal Res. 2002;32(4):225–30.

    Article  CAS  PubMed  Google Scholar 

  120. Baydas G, Nedzvetskii VS, Tuzcu M, Yasar A, Kirichenko SV. Increase of glial fibrillary acidic protein and S-100B in hippocampus and cortex of diabetic rats: effects of vitamin E. Eur J Pharmacol. 2003;462(1–3):67–71.

    Article  CAS  PubMed  Google Scholar 

  121. Baydas G, Donder E, Kiliboz M, Sonkaya E, Tuzcu M, Yasar A, et al. Neuroprotection by alpha-lipoic acid in streptozotocin-induced diabetes. Biochem Biokhimiia. 2004;69(9):1001–5.

    Article  CAS  Google Scholar 

  122. Tan D-X, Chen L-D, Poeggeler B, Manchester L, Reiter R, Poeggler B. Melatonin a potent endogenous hydroxyl radical scavenger. 1993.

  123. Reiter RJ, Acuna-Castroviejo D, Tan DX, Burkhardt S. Free radical-mediated molecular damage Mechanisms for the protective actions of melatonin in the central nervous system. Ann N Y Acad Sci. 2001;939:200–15.

    Article  CAS  PubMed  Google Scholar 

  124. Tuzcu M, Baydas G. Effect of melatonin and vitamin E on diabetes-induced learning and memory impairment in rats. Eur J Pharmacol. 2006;537(1–3):106–10.

    Article  CAS  PubMed  Google Scholar 

  125. Baydas G, Nedzvetskii VS, Kirichenko SV, Nerush PA. Astrogliosis in the hippocampus and cortex and cognitive deficits in rats with streptozotocin-induced diabetes: effects of melatonin. Neurophysiology. 2008;40(2):91–7.

    Article  CAS  Google Scholar 

  126. Baydas G, Reiter RJ, Yasar A, Tuzcu M, Akdemir I, Nedzvetskii VS. Melatonin reduces glial reactivity in the hippocampus, cortex, and cerebellum of streptozotocin-induced diabetic rats. Free Radical Biol Med. 2003;35(7):797–804.

    Article  CAS  Google Scholar 

  127. Hajam YA, Rai S, Roy A, Basheer M, Ghosh H. Repossession of brain complications in a streptozotocin induced diabetic rat by exogenous melatonin administration. Int J Zool Res. 2017;13:64–73.

    Article  CAS  Google Scholar 

  128. Kahya MC, Naziroğlu M, Çiğ B. Melatonin and selenium reduce plasma cytokine and brain oxidative stress levels in diabetic rats. Brain Inj. 2015;29(12):1490–6.

    Article  PubMed  Google Scholar 

  129. Negi G, Kumar A, Kaundal RK, Gulati A, Sharma SS. Functional and biochemical evidence indicating beneficial effect of Melatonin and Nicotinamide alone and in combination in experimental diabetic neuropathy. Neuropharmacology. 2010;58(3):585–92.

    Article  CAS  PubMed  Google Scholar 

  130. Jangra A, Datusalia AK, Khandwe S, Sharma SS. Amelioration of diabetes-induced neurobehavioral and neurochemical changes by melatonin and nicotinamide: implication of oxidative stress–PARP pathway. Pharmacol Biochem Behav. 2013;114–115:43–51.

    Article  PubMed  CAS  Google Scholar 

  131. Lloyd CE, Roy T, Nouwen A, Chauhan AM. Epidemiology of depression in diabetes: international and cross-cultural issues. J Affect Disord. 2012;142(Suppl):S22–9.

    Article  PubMed  Google Scholar 

  132. Roy T, Lloyd CE. Epidemiology of depression and diabetes: a systematic review. J Affect Disord. 2012;142(Suppl):S8–21.

    Article  PubMed  Google Scholar 

  133. Raisanen S, Lehto SM, Nielsen HS, Gissler M, Kramer MR, Heinonen S. Risk factors for and perinatal outcomes of major depression during pregnancy: a population-based analysis during 2002–2010 in Finland. BMJ Open. 2014;4(11):e004883.

    Article  PubMed  PubMed Central  Google Scholar 

  134. Lancaster CA, Gold KJ, Flynn HA, Yoo H, Marcus SM, Davis MM. Risk factors for depressive symptoms during pregnancy: a systematic review. Am J Obstet Gynecol. 2010;202(1):5–14.

    Article  PubMed  PubMed Central  Google Scholar 

  135. Holt RI, de Groot M, Golden SH. Diabetes and depression. Curr DiabRep. 2014;14(6):491.

    Article  CAS  Google Scholar 

  136. Zanoveli JM, Morais H, Dias IC, Schreiber AK, Souza CP, Cunha JM. Depression associated with diabetes: from pathophysiology to treatment. Curr Diabetes Rev. 2016;12(3):165–78.

    Article  CAS  PubMed  Google Scholar 

  137. Beucher G, Viaris de Lesegno B, Dreyfus M. Maternal outcome of gestational diabetes mellitus. Diabetes Metabol. 2010;36(6 P 2):522–37.

    Article  CAS  Google Scholar 

  138. Korczak DJ, Pereira S, Koulajian K, Matejcek A, Giacca A. Type 1 diabetes mellitus and major depressive disorder: evidence for a biological link. Diabetologia. 2011;54(10):2483–93.

    Article  CAS  PubMed  Google Scholar 

  139. Rebai R, Jasmin L, Boudah A. The antidepressant effect of melatonin and fluoxetine in diabetic rats is associated with a reduction of the oxidative stress in the prefrontal and hippocampal cortices. Brain Res Bull. 2017;134:142–50.

    Article  CAS  PubMed  Google Scholar 

  140. Yang XD, Fang PF, Xiang DX, Yang YY. Topical treatments for diabetic neuropathic pain. Exp Ther Med. 2019;17(3):1963–76.

    CAS  PubMed  PubMed Central  Google Scholar 

  141. Oguntibeju OO. Type 2 diabetes mellitus, oxidative stress and inflammation: examining the links. Int J Physiol Pathophysiol Pharmacol. 2019;11(3):45–63.

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Kim H-G. Cognitive dysfunctions in individuals with diabetes mellitus. Yeungnam Univ J Med. 2019;36(3):183–91.

    Article  PubMed  PubMed Central  Google Scholar 

  143. Azmi S, Petropoulos IN, Ferdousi M, Ponirakis G, Alam U, Malik RA. An update on the diagnosis and treatment of diabetic somatic and autonomic neuropathy. F1000Res. 2019;8:186.

    Article  Google Scholar 

  144. Tutuncu NB, Batur MK, Yildirir A, Tutuncu T, Deger A, Koray Z, et al. Melatonin levels decrease in type 2 diabetic patients with cardiac autonomic neuropathy. J Pineal Res. 2005;39(1):43–9.

    Article  CAS  PubMed  Google Scholar 

  145. O’brien I, Lewin I, O’hare J, Arendt J, Corrall R. Abnormal circadian rhythm of melatonin in diabetic autonomic neuropathy. Clin Endocrinol. 1986;24(4):359–64.

    Article  Google Scholar 

  146. Zangiabadi N, Sheibani V, Asadi-Shekaari M, Shabani M, Jafari M, Asadi AR, et al. Effects of melatonin in prevention of neuropathy in STZ-induced diabetic rats. Am J Pharmacol Toxicol. 2011;6(2):59–67.

    Article  CAS  Google Scholar 

  147. Wang S, Zhang L, Lim G, Sung B, Tian Y, Chou C-W, et al. A combined effect of dextromethorphan and melatonin on neuropathic pain behavior in rats. Brain Res. 2009;1288:42–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Ulugol A, Dokmeci D, Guray G, Sapolyo N, Ozyigit F, Tamer M. Antihyperalgesic, but not antiallodynic, effect of melatonin in nerve-injured neuropathic mice: possible involvements of the l-arginine–NO pathway and opioid system. Life Sci. 2006;78(14):1592–7.

    Article  CAS  PubMed  Google Scholar 

  149. Lopez-Canul M, Palazzo E, Dominguez-Lopez S, Luongo L, Lacoste B, Comai S, et al. Selective melatonin MT2 receptor ligands relieve neuropathic pain through modulation of brainstem descending antinociceptive pathways. Pain. 2015;156(2):305–17.

    Article  CAS  PubMed  Google Scholar 

  150. Posa L, De Gregorio D, Gobbi G, Comai S. Targeting melatonin MT2 receptors: a novel pharmacological avenue for inflammatory and neuropathic pain. Curr Med Chem. 2018;25(32):3866–82.

    Article  CAS  PubMed  Google Scholar 

  151. Lin J-J, Lin Y, Zhao T-Z, Zhang C-K, Zhang T, Chen X-L, et al. Melatonin suppresses neuropathic pain via MT2-dependent and -independent pathways in dorsal root ganglia neurons of mice. Theranostics. 2017;7(7):2015–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Rajchgot T, Thomas SC, Wang J-C, Ahmadi M, Balood M, Crosson T, et al. Neurons and microglia; a sickly-sweet duo in diabetic pain neuropathy. Front Neurosci. 2019;13:13.

    Article  Google Scholar 

  153. Kahya MC, Nazıroğlu M, Övey İS. Modulation of diabetes-induced oxidative stress, apoptosis, and Ca2+ entry through TRPM2 and TRPV1 channels in dorsal root ganglion and hippocampus of diabetic rats by melatonin and selenium. Mol Neurobiol. 2017;54(3):2345–60.

    Article  CAS  PubMed  Google Scholar 

  154. Seyit D, Degirmenci E, Oguzhanoglu A. Evaluation of electrophysiological effects of melatonin and alpha lipoic acid in rats with streptozotocine induced diabetic neuropathy. Exp Clin Endocrinol Diabetes. 2016;124(05):300–6.

    Article  CAS  PubMed  Google Scholar 

  155. Babaei-Balderlou F, Zare S. Melatonin improves spatial navigation memory in male diabetic rats. Vet Res Forum. 2012;3(3):187–92.

    PubMed  PubMed Central  Google Scholar 

  156. Metwally MM, Ebraheim LL, Galal AA. Potential therapeutic role of melatonin on STZ-induced diabetic central neuropathy: a biochemical, histopathological, immunohistochemical and ultrastructural study. Acta Histochem. 2018;120(8):828–36.

    Article  CAS  PubMed  Google Scholar 

  157. Afifi NM. Neuroprotective effect of melatonin in a rat model of streptozotocin-induced diabetic neuropathy: light and electron microscopic study. Egypt J Histol. 2013;36(2):321–35.

    Article  Google Scholar 

  158. Negi G, Kumar A, Sharma SS. Melatonin modulates neuroinflammation and oxidative stress in experimental diabetic neuropathy: effects on NF-κB and Nrf2 cascades. J Pineal Res. 2011;50(2):124–31.

    CAS  PubMed  Google Scholar 

  159. Onphachanh X, Lee HJ, Lim JR, Jung YH, Kim JS, Chae CW, et al. Enhancement of high glucose-induced PINK1 expression by melatonin stimulates neuronal cell survival: involvement of MT2/Akt/NF-κB pathway. J Pineal Res. 2017;63(2):e12427.

    Article  CAS  Google Scholar 

  160. Zhang J-L, Hui Y, Zhou F, Hou J-Q. Neuroprotective effects of melatonin on erectile dysfunction in streptozotocin-induced diabetic rats. Int Urol Nephrol. 2018;50(11):1981–8.

    Article  CAS  PubMed  Google Scholar 

  161. Li X, Zhang M, Tang W. Effects of melatonin on streptozotocin-induced retina neuronal apoptosis in high blood glucose rat. Neurochem Res. 2013;38(3):669–76.

    Article  CAS  PubMed  Google Scholar 

  162. Bherwani S, Saumya A, Sandhya A, Patel S, Ghotekar L. The study of mineral status in type 2 diabetes mellitus with and without diabetic Nephropathy. J Assoc Phys India. 2016;64(1):95.

    Google Scholar 

  163. Adhikary L, Chow F, Nikolic-Paterson DJ, Stambe C, Dowling J, Atkins RC, et al. Abnormal p38 mitogen-activated protein kinase signalling in human and experimental diabetic nephropathy. Diabetologia. 2004;47(7):1210–22.

    Article  CAS  PubMed  Google Scholar 

  164. Dunlop M. Aldose reductase and the role of the polyol pathway in diabetic nephropathy. Kidney Int Suppl. 2000;77:S3–12.

    Article  CAS  PubMed  Google Scholar 

  165. Malgorzewicz S, Skrzypczak-Jankun E, Jankun J. Plasminogen activator inhibitor-1 in kidney pathology (Review). Int J Mol Med. 2013;31(3):503–10.

    Article  CAS  PubMed  Google Scholar 

  166. Hishikawa K, Oemar BS, Nakaki T. Static pressure regulates connective tissue growth factor expression in human mesangial cells. J Biol Chem. 2001;276(20):16797–803.

    Article  CAS  PubMed  Google Scholar 

  167. Ceriello A, Morocutti A, Mercuri F, Quagliaro L, Moro M, Damante G, et al. Defective intracellular antioxidant enzyme production in type 1 diabetic patients with nephropathy. Diabetes. 2000;49(12):2170–7.

    Article  CAS  PubMed  Google Scholar 

  168. Fernyhough P, Huang TJ, Verkhratsky A. Mechanism of mitochondrial dysfunction in diabetic sensory neuropathy. JPNS. 2003;8(4):227–35.

    Article  PubMed  Google Scholar 

  169. Allen DA, Harwood S, Varagunam M, Raftery MJ, Yaqoob MM. High glucose-induced oxidative stress causes apoptosis in proximal tubular epithelial cells and is mediated by multiple caspases. FASEB J. 2003;17(8):908–10.

    Article  CAS  PubMed  Google Scholar 

  170. Simone S, Gorin Y, Velagapudi C, Abboud HE, Habib SL. Mechanism of oxidative DNA damage in diabetes: tuberin inactivation and downregulation of DNA repair enzyme 8-oxo-7,8-dihydro-2′-deoxyguanosine-DNA glycosylase. Diabetes. 2008;57(10):2626–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Adeghate E. Molecular and cellular basis of the aetiology and management of diabetic cardiomyopathy: a short review. Mol Cell Biochem. 2004;261(1–2):187–91.

    Article  CAS  PubMed  Google Scholar 

  172. Zephy D, Ahmad J. Type 2 diabetes mellitus: role of melatonin and oxidative stress. Diabetes Metabol Syndr. 2015;9(2):127–31.

    Article  Google Scholar 

  173. Onk D, Onk OA, Turkmen K, Erol HS, Ayazoglu TA, Keles ON, et al. Melatonin Attenuates Contrast-Induced Nephropathy in Diabetic Rats: the Role of Interleukin-33 and Oxidative Stress. Mediators Inflamm. 2016;2016:9050828.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  174. Shi S, Lei S, Tang C, Wang K, Xia Z. Melatonin attenuates acute kidney ischemia/reperfusion injury in diabetic rats by activation of the SIRT1/Nrf2/HO-1 signaling pathway. Bioscience reports. 2019;39:1.

    CAS  Google Scholar 

  175. Parving HH, Lehnert H, Brochner-Mortensen J, Gomis R, Andersen S, Arner P. The effect of irbesartan on the development of diabetic nephropathy in patients with type 2 diabetes. N Engl J Med. 2001;345(12):870–8.

    Article  CAS  PubMed  Google Scholar 

  176. Motawi TK, Ahmed SA, Hamed MA, El-Maraghy SA, Aziz WM. Combination of melatonin and certain drugs for treatment of diabetic nephropathy in streptozotocin-induced diabetes in rats. Diabetol Int. 2016;7(4):413–24.

    Article  PubMed  PubMed Central  Google Scholar 

  177. Kang M-K, Lim SS, Lee J-Y, Yeo KM, Kang Y-H. Anthocyanin-rich purple corn extract inhibit diabetes-associated glomerular angiogenesis. PLoS ONE. 2013;8(11):e79823.

    Article  PubMed  PubMed Central  Google Scholar 

  178. Peng H, Li Y, Wang C, Zhang J, Chen Y, Chen W, et al. ROCK1 induces endothelial-to-mesenchymal transition in glomeruli to aggravate albuminuria in diabetic nephropathy. Sci Rep. 2016;6:20304.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Liu F, Zhang S, Xu R, Gao S, Yin J. Melatonin attenuates endothelial-to-mesenchymal transition of glomerular endothelial cells via regulating miR-497/ROCK in diabetic nephropathy. Kid Blood Press Res. 2018;43(5):1425–36.

    Article  CAS  Google Scholar 

  180. Rashed LA, Elattar S, Eltablawy N, Ashour H, Mahmoud LM, El-Esawy Y. Mesenchymal stem cells pretreated with melatonin ameliorate kidney functions in a rat model of diabetic nephropathy. Biochem Cell Biol. 2018;96(5):564–71.

    Article  CAS  PubMed  Google Scholar 

  181. Ji ZZ, Xu YC. Melatonin protects podocytes from angiotensin II-induced injury in an in vitro diabetic nephropathy model. Mol Med Rep. 2016;14(1):920–6.

    Article  CAS  PubMed  Google Scholar 

  182. Gumustekin M, Tekmen I, Guneli E, Tugyan K, Topcu A, Ergonen AT, et al. Short-term melatonin treatment improved diabetic nephropathy but did not affect hemorheological changes in diabetic rats. Pharmazie. 2007;62(9):693–8.

    CAS  PubMed  Google Scholar 

  183. Martí-Carvajal AJ, Gluud C, Nicola S, Simancas-Racines D, Reveiz L, Oliva P, et al. Growth factors for treating diabetic foot ulcers. Cochrane Database Syst Rev. 2015;10:CD008548.

    Google Scholar 

  184. Goren I, Müller E, Pfeilschifter J, Frank S. Severely impaired insulin signaling in chronic wounds of diabetic ob/ob mice: a potential role of tumor necrosis factor-alpha. Am J Pathol. 2006;168(3):765–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Galkowska H, Wojewodzka U, Olszewski WL. Chemokines, cytokines, and growth factors in keratinocytes and dermal endothelial cells in the margin of chronic diabetic foot ulcers. Wound Repair Regen. 2006;14(5):558–65.

    Article  PubMed  Google Scholar 

  186. Jeffcoate WJ, Price P, Harding KG. Wound healing and treatments for people with diabetic foot ulcers. Diabetes Metabol Res Rev. 2004;20(Suppl 1):S78–89.

    Article  Google Scholar 

  187. Jeffcoate WJ, Harding KG. Diabetic foot ulcers. Lancet (London, England). 2003;361(9368):1545–51.

    Article  Google Scholar 

  188. Veves A, Akbari CM, Primavera J, Donaghue VM, Zacharoulis D, Chrzan JS, et al. Endothelial dysfunction and the expression of endothelial nitric oxide synthetase in diabetic neuropathy, vascular disease, and foot ulceration. Diabetes. 1998;47(3):457–63.

    Article  CAS  PubMed  Google Scholar 

  189. Acuna-Castroviejo D, Escames G, Venegas C, Diaz-Casado ME, Lima-Cabello E, Lopez LC, et al. Extrapineal melatonin: sources, regulation, and potential functions. CMLS. 2014;71(16):2997–3025.

    Article  CAS  PubMed  Google Scholar 

  190. Pugazhenthi K, Kapoor M, Clarkson AN, Hall I, Appleton I. Melatonin accelerates the process of wound repair in full-thickness incisional wounds. J Pineal Res. 2008;44(4):387–96.

    Article  CAS  PubMed  Google Scholar 

  191. Jin H, Zhang Z, Wang C, Tang Q, Wang J, Bai X, et al. Melatonin protects endothelial progenitor cells against AGE-induced apoptosis via autophagy flux stimulation and promotes wound healing in diabetic mice. Exp Mol Med. 2018;50(11):154.

    Article  PubMed Central  CAS  Google Scholar 

  192. Lee SJ, Jung YH, Oh SY, Yun SP, Han HJ. Melatonin enhances the human mesenchymal stem cells motility via melatonin receptor 2 coupling with Galphaq in skin wound healing. J Pineal Res. 2014;57(4):393–407.

    Article  CAS  PubMed  Google Scholar 

  193. Song R, Ren L, Ma H, Hu R, Gao H, Wang L, et al. Melatonin promotes diabetic wound healing in vitro by regulating keratinocyte activity. Am J Transl Res. 2016;8(11):4682–93.

    CAS  PubMed  PubMed Central  Google Scholar 

  194. Raygan F, Ostadmohammadi V, Bahmani F, Reiter RJ, Asemi Z. Melatonin administration lowers biomarkers of oxidative stress and cardio-metabolic risk in type 2 diabetic patients with coronary heart disease: a randomized, double-blind, placebo-controlled trial. Clin Nutr. 2019;38(1):191–6.

    Article  CAS  PubMed  Google Scholar 

  195. Hussain SA, Khadim HM, Khalaf BH, Ismail SH, Hussein KI, Sahib AS. Effects of melatonin and zinc on glycemic control in type 2 diabetic patients poorly controlled with metformin. Saudi Med J. 2006;27(10):1483–8.

    PubMed  Google Scholar 

  196. Salido EM, Bordone M, De Laurentiis A, Chianelli M, Keller Sarmiento MI, Dorfman D, et al. Therapeutic efficacy of melatonin in reducing retinal damage in an experimental model of early type 2 diabetes in rats. J Pineal Res. 2013;54(2):179–89.

    Article  CAS  PubMed  Google Scholar 

  197. Motawi TK, Ahmed SA, Hamed MA, El-Maraghy SA, Aziz WM. Melatonin and/or rowatinex attenuate streptozotocin-induced diabetic renal injury in rats. J Biomed Res. 2019;33(2):113.

    PubMed  PubMed Central  Google Scholar 

  198. Winiarska K, Dzik JM, Labudda M, Focht D, Sierakowski B, Owczarek A, et al. Melatonin nephroprotective action in Zucker diabetic fatty rats involves its inhibitory effect on NADPH oxidase. J Pineal Res. 2016;60(1):109–17.

    Article  CAS  PubMed  Google Scholar 

  199. Elbe H, Vardi N, Esrefoglu M, Ates B, Yologlu S, Taskapan C. Amelioration of streptozotocin-induced diabetic nephropathy by melatonin, quercetin, and resveratrol in rats. Hum Exp Toxicol. 2015;34(1):100–13.

    Article  CAS  PubMed  Google Scholar 

  200. Zhang H, Zhang HM, Wu LP, Tan DX, Kamat A, Li YQ, et al. Impaired mitochondrial complex III and melatonin responsive reactive oxygen species generation in kidney mitochondria of db/db mice. J Pineal Res. 2011;51(3):338–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Ha H, Yu MR, Kim KH. Melatonin and taurine reduce early glomerulopathy in diabetic rats. Free Radical Biol Med. 1999;26(7–8):944–50.

    Article  CAS  Google Scholar 

  202. Anwar MM, Meki AR. Oxidative stress in streptozotocin-induced diabetic rats: effects of garlic oil and melatonin. Comp Biochem Physiol A. 2003;135(4):539–47.

    Article  CAS  Google Scholar 

  203. Öktem F, Ozguner F, Yilmaz HR, Uz E, Dündar B. Melatonin reduces urinary excretion of N-acetyl-β-d-glucosaminidase, albumin and renal oxidative markers in diabetic rats. Clin Exp Pharmacol Physiol. 2006;33(1–2):95–101.

    Article  PubMed  Google Scholar 

  204. Cam M, Yavuz Ö, Guven A, Ercan F, Bukan N, Üstündag N. Protective effects of chronic melatonin treatment against renal injury in streptozotocin-induced diabetic rats. J Pineal Res. 2003;35(3):212–20.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This research did not receive any specific grant from funding agencies in the public, commercial, or not for-profit sectors.

Funding

Not applicable

Author information

Authors and Affiliations

Authors

Contributions

Conception and design: SM and MHP. Performing the literature search: ED, KH and AH. Drafting the manuscript: all authors. SM is responsible for the integrity of the work as a whole. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Saeed Mehrzadi.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publicatio

Not applicable.

Competing interests

The authors declare that they have no conflict of interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pourhanifeh, M.H., Hosseinzadeh, A., Dehdashtian, E. et al. Melatonin: new insights on its therapeutic properties in diabetic complications. Diabetol Metab Syndr 12, 30 (2020). https://doi.org/10.1186/s13098-020-00537-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13098-020-00537-z

Keywords