TY - JOUR AU - MS(Pharm), Priyanka P. Trivedi, AB - Abstract Ulcerative colitis (UC), an inflammatory bowel disease, affects many people across the globe, and its prevalence is increasing steadily. Inflammation and oxidative stress play a vital role in the perpetuation of inflammatory process and the subsequent DNA damage associated with the development of UC. UC induces not only local but also systemic damage, which involves the perturbation of multiple molecular pathways. Furthermore, UC leads to an increased risk of colorectal cancer, the third most common malignancy in humans. Most of the drugs used for the treatment of UC are unsatisfactory because they are generally mono-targeted, relatively ineffective and unaffordable for many people. Thus, agents that can target multiple molecular pathways and are less expensive have enormous potential to treat UC. Melatonin has beneficial effects against UC in experimental and clinical studies because of its ability to modulate several molecular pathways of inflammation, oxidative stress, fibrosis, and cellular injury. However, many novel targets are yet to be explored on which melatonin may act to exert its favorable effects in UC. It is time to explore improved intervention strategies with melatonin in UC on the basis of studies investigating different molecular targets using proteomic and genomic approaches. This review identifies various molecular targets for melatonin with the intent of providing novel strategies for combating UC and the associated extraintestinal manifestations of this debilitating disease. ulcerative colitis, melatonin, inflammation, oxidative stress, DNA damage, novel targets Melatonin (N-acetyl-5-methoxytryptamine), a pineal gland secretory product and an evolutionarily conserved molecule,1,2 was discovered in 1958 in bovine pineal tissue.3 Evidence regarding the presence of melatonin in the gastrointestinal tract (GIT) was obtained in 1974, when Raikhlin and Kvetnoi4 identified it in the human appendix. Finally, its presence in the GIT was confirmed in 1977, when the indole was found in the mucosal lining of the intestine.5 The content of melatonin is calculated to be 400 to 500 times more than in the pineal gland.6 This suggests that melatonin synthesized in the GIT may have some vital role in the GIT physiology and pathophysiology. In the intestine, melatonin influences physiological effects such as regeneration of the epithelium and regulation of its function, modulation of the immune response and reduction in the tone of GI muscles through specific membrane receptors such as melatonin-1 receptor (MT-1), MT-2, and possibly through MT-3.7,–9 Melatonin is also reported to act as a potent antioxidant, anti-inflammatory and antigenotoxic agent in all organs.10,–16 Because of its lipophilic nature, it enters all cells, and it is considered an important molecule because of its multifactorial role and myriad of beneficial effects. It helps in inducing sleep, treating jet lag, boosting the immune system, preventing free radical damage, and possibly influencing longevity.17,–19 It has an ability to target multiple molecules involved in the pathophysiology of various diseases, which justifies its use for the treatment of a range of disorders such as cataracts, cardiovascular, neurodegenerative, pulmonary, renal, and gastrointestinal (GI) disorders, rheumatoid arthritis, diabetes, and cancer.20,–29 Among the GIT disorders, it is used to treat irritable bowel syndrome, gastroesophageal reflux disease, Crohn's disease, and ulcerative colitis (UC).30,–33 UC is a chronic GI disorder affecting a part of the colon or the entire colon, based on its severity. Inflammation, along with oxidative stress, plays a vital role in the pathogenesis of UC.34 UC causes an elevation in various inflammatory markers including interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), nuclear factor-kappa B (NF-κB), and cyclooxygenase-2 (COX-2).35,–37 Furthermore, the oxidant/antioxidant balance has been reported to play an important role in the recurrence and progression of UC.38 UC is associated with an elevated production of free oxygen radicals accompanied by an increased oxidative damage.39 Moreover, inflammation-associated oxidative stress leads to DNA damage, which may further contribute to the process of carcinogenesis. UC leads to an increased risk of developing colorectal cancer, which is the third most common malignancy observed in humans.40 Apart from colorectal cancer, UC is associated with the development of several extraintestinal manifestations.37,41,42 This involves several molecular processes and global damage. Current treatments for UC are not always effective and often accompanied by serious side effects, and hence, there is considerable interest in finding an alternative treatment for this debilitating disease. Melatonin, with its ability to modulate the multiple molecular pathways involved in the pathogenesis of UC, may have a better therapeutic potential and may be useful in the preventive and therapeutic armamentarium for UC and the associated local and global damage. However, conflicting results have been obtained regarding the use of melatonin for the treatment of experimental colitis.43,44 Hence, well-designed experimental and clinical studies are required before its better clinical application. The present review reveals various molecular targets of melatonin and its subsequent role in potentially diminishing the severity of UC and the associated extraintestinal manifestations. Inflammation: A Roadway to DNA Damage The word “inflammation” is derived from the Latin word “inflammatio”, i.e, to set on fire, which was characterized by a Roman physician, Cornelius Celsus, as the one which consists of heat (calor), redness (rubor), pain (dolor), and swelling (tumor). Inflammation is a short-term friend and a long-term foe, because acute inflammation is the body's most primitive arsenal developed to fight infection; in contrast, chronic inflammation causes several diseases such as GI, cardiovascular, pulmonary, and neurological diseases, diabetes, obesity, and cancer.45,46 One of the means by which inflammation paves the way toward DNA damage is the production of reactive oxygen species (ROS) and reactive nitrogen species by activated neutrophils and macrophages leading to lethal cancer-causing mutations in the epithelial cells.47 Chronic inflammation induces oxidative damage in the DNA of cells of the inflamed tissue. ROS and reactive nitrogen species thus generated can damage both nuclear and mitochondrial DNA, RNA, lipids, and proteins.48,49 Inflammation and DNA damage are associated with UC in mice.50 Oxidative DNA damage in the mucosa of UC increases with disease duration and dysplasia.51 Several reports document that the risk of colorectal cancer is greater in the patients suffering from UC because of inappropriate inflammatory response to a luminal pathogen, abnormal immune response to intestinal bacterial flora, role of cytokines, and oxidative DNA damage at the local site of inflammation in the colon.35,52 Earlier, we reported that UC in mice was associated with not only the local, but also the global damage such as systemic toxicity and hepatotoxicity.37,41,53,54 Moreover, intestinal inflammation also causes DNA damage in extraintestinal tissues and in the spleen, mesenteric and peripheral lymph nodes, and hepatocytes.42 Because, in UC, inflammation and oxidative stress are mainly responsible for the DNA damage in the colon and other tissues, agents combating inflammation and oxidative stress may possess beneficial effects in ameliorating the severity of UC and the associated global damage in patients. Multiple Targets of Melatonin in UC Melatonin is useful in several pathological conditions because of its effect on multiple targets. Melatonin can be classified as a hormone, a tissue factor, an autocoid, a paracoid, and an antioxidant vitamin.55 It acts with overall homeostatic functions, and its pleiotropic nature controls the detoxification and stress response genes, thus conferring protection against a number of xenobiotics produced by acute and chronic lethal stimuli.56 It is present in high concentration in the GIT and is known to be synthesized in the enterochromaffin cells of the intestine, where it plays a vital role in maintaining the GIT physiology.57 Hence, it is likely that melatonin influences inflammation-related GI disorders including UC. UC is a chronic GI disorder associated with elevated levels of various proinflammatory cytokines and ROS, which in turn, activate NF-κB, a proinflammatory transcription factor that has emerged as an important player in the development and progression of UC.58 NF-κB is a transcription factor that resides in the cytoplasm as a heterotrimer consisting of p50, p65, and IκBα in its resting stage. IκBα kinases cause the phosphorylation of IκBα, which results in the release and translocation of NF-κB to the nucleus, where it activates the transcription of target genes that regulate the inflammatory response by the production of cytokines, chemokines, and cell adhesion molecules.59,60 NF-κB is implicated in the development and progression of chronic inflammatory diseases partly through the elevated production of various cytokines such as TNF-α, IL-6, and IL-1β.61 In UC, NF-κB activation occurs both in macrophages and in epithelial cells.62,63 Activation of NF-κB has been reported in the colonic mucosa of patients with collagenous colitis and UC.64 Melatonin ameliorates the severity of colitis by targeting NF-κB. It decreases inflammation-associated injury through the inhibition of NF-κB activation, regulation of macrophages activity, and reduction in the expression of chemokines, bacterial translocation, and apoptosis in rat with trinitrobenzene sulfonic acid (TNBS)-induced colitis.65,–68 It inhibits NF-κB activation and decreases serum levels of pentraxin-3, lipid peroxides, and total thiols in rat with acetic acid–induced colitis.69 By virtue of its powerful anti-inflammatory and antioxidant activities, melatonin treatment resulted in significant improvement of acetic acid–induced colitis in rat by reducing the elevated levels of proinflammatory cytokines (TNF-α, IL- 1β, and IL-6), myeloperoxidase, malondialdehyde, and by increasing the levels of reduced glutathione and superoxide dismutase.34,57 Furthermore, melatonin lowered the immunohistochemical expression of NF-κB in azoxymethane/dextran sodium sulfate (DSS)-induced large bowel oncogenesis in rat.70 Thus, melatonin reduces the levels of various stimuli responsible for the activation of NF-κB and thereby plays a vital role in protecting the gut mucosa by decreasing the expression of NF-κB in UC. In UC, increased expression of NF-κB is accompanied with an elevated expression of inflammation-induced COX-2 enzyme.37 Melatonin has a protective effect on colonic injury induced by both acetic acid and 2,4,6-trinitrobenzene sulfonic acid enemas by reducing inducible nitric oxide synthase (iNOS) and COX-2 expression in the colonic mucosa.43 Melatonin, in combination with erythropoietin, protected pinealectomized rats against dinitrobenzene sulfonic acid–induced colitis by reducing inflammation, cellular damage, and apoptosis.71 Protective effects of melatonin against dinitrobenzene sulfonic acid–induced colitis in rats were due to its ability to decrease the levels of TNF-α, myeloperoxidase, malondialdehyde, nitrotyrosine, poly (ADP-ribose) synthetase, intercellular adhesion molecule 1, P-selectin, COX-2, iNOS, activation of NF-κB, phosphorylation of c-Jun, Fas ligand expression, and apoptosis in the colon.72,73 UC is associated with an increased oxidative stress along with inflammation.37,74 Melatonin and its metabolites have been shown to be highly effective as free radical scavengers and thereby reduce the oxidative stress under a variety of experimental settings, where the free radical generation is elevated.75,76 Interestingly, one report showed that exposure to continuous darkness ameliorates gastric and colonic inflammation in the rat due to excess amount of melatonin synthesized in darkness that reduces the oxidative stress.77 Recently, high levels of melatonin have been found in mitochondria, which are a major source of free radical generation in living organisms, and hence, melatonin has been described as a mitochondrial-targeted antioxidant.78,79 The mitochondrial antioxidant property of melatonin may prove beneficial in the treatment of various inflammatory disorders including UC. Mucosal inflammation in UC leads to collagen deposition, which in turn, results in the induction of matrix metalloproteinases (MMPs). MMPs are implicated in the pathogenesis of colitis because of their influence on the function and migration of inflammatory cells, mucosal ulceration, matrix deposition, and degradation.80,–83 Various clinical and experimental studies have demonstrated an increase in the abundance of these matrix-degrading proteases in inflammatory bowel diseases, and the inhibition of MMP activation has been shown to ameliorate experimental colitis.80,84,–86 Melatonin, an inhibitor of MMP,87,88 prevents experimental colitis in rats by regulating MMP-9 and MMP-2 activity and expression.89 It also alleviates MMP-9 and MMP-3 expression in mice with gastric ulcers through the reduction of activator protein-1 activity.90 Furthermore, it has been reported that reduced MMP-9 and MMP-2 activities are associated with lower expression of TNF-α.89 Moreover, TNF-α is a potent endogenous mutagen, and TNF-α–generated ROS have DNA-damaging effects comparable to those of ionizing radiation.91 Melatonin, because of its anti-inflammatory property, decreases TNF-α level, and may, at least in part, play a role in diminishing TNF-α–induced DNA damage in UC. Recently, we reported that melatonin reduced the local and systemic damage in mice with DSS-induced colitis by reducing the markers of inflammation, oxidative stress, fibrosis, and DNA damage. It also ameliorated the colonic mucosal damage, decreased bacterial translocation to the systemic circulation, and reduced the associated systemic inflammation and DNA damage.92 The protective effect of melatonin against DSS-induced colitis has been reported because of its effect on the smooth muscle of the colon, the blood supply in the mucosa, its capability as an antioxidant and scavenger of free radicals, and its effect on the immune system of the gut.93 Mucosal healing property of melatonin94 may help in ameliorating the severity of UC. Surprisingly enough, few experimental studies reported aggravation of UC symptoms with melatonin treatment. Acutely administered melatonin ameliorated, whereas its chronic administration aggravated the progression of TNBS-induced colitis in rat.44 Melatonin was effective in the prevention and short-term treatment of inflammatory process in acetic acid–induced colitis in rat, whereas the benefit of long-term treatment was ambiguous.69 Similar conflicting results for the use of melatonin in inflammatory bowel diseases exist in clinical conditions. One study reported that the use of melatonin in combined therapy for inflammatory bowel diseases (Crohn's disease and UC) considerably improved the results of treatment and promoted a more complete ultrastructural recovery of the colonic mucosa.32 However, a case report depicted that melatonin triggered Crohn's disease symptoms. A 35-year-old woman diagnosed with Crohn's disease decided by herself to take melatonin capsules before going to sleep during the remission period. After 4 days, symptoms of the disease emerged again. When she stopped taking melatonin, 24 hours later, there was a complete remission of symptoms.95 Furthermore, melatonin used to treat sleep abnormalities in children with neurodevelopmental disabilities surprisingly improved their GI conditions including UC.96,97 A case report showed that self-administration of melatonin as a cure for jet leg on international flights by a 47-year-old man suffering from UC reduced the disease symptoms, and the symptoms recurred within 1 week of running out of the medication.98 Yet in another case report, the symptoms of UC were aggravated in a 56-year-old man on self-administering melatonin.99 The only available pilot randomized, double-blind, placebo-controlled phase 2 clinical trial on the Web site “clinicaltrials.gov” was designed in the year 2009 and sponsored by Emroy University, Atlanta, Georgia, to elucidate the effect of melatonin against UC. However, the status is still incomplete.100 Melatonin in UC: The Possible Reasons for Conflicting Results Conflicting results for melatonin usage in UC could be due to (1) variation in the experimental models used, (2) chemically induced UC in animals versus UC in humans with unknown etiology (genetic, epigenetic, environmental, immunological, microbiological, or lifestyle), (3) variation in the duration of disease onset, (4) difference in the daily time of administration of melatonin, (5) variation in its dose and duration for the treatment, (6) diurnal variations of melatonin and its binding sites in different organs including the GIT, (7) differences in individual sensitivity and their pharmacodynamic and pharmacokinetic profiles, (8) endogenous melatonin level at different stages of the disease (mild, moderate, severe), (9) endogenous melatonin level during different (a) seasons and (b) age. Melatonin level in the GIT peaked at birth and then declined to stable level at the age of 21 days in a postnatal rat.101 Difference in the circadian rhythm and sleeping pattern among individuals may also affect the endogenous melatonin levels, and thereby the disease status and the effect of exogenous melatonin on the disease onset or progression. Sleep deprivation worsens inflammation and delays recovery in experimental models and patients with UC, which might be due to reduced endogenous melatonin level.102,103 Effect of UC on the endogenous melatonin level is also variable among individuals as reported by different studies.93,104 It might be possible that during the initial stage of the disease, GIT may produce more melatonin to overcome the stress. However, as the disease progresses, GI cells might not be efficient enough to produce sufficient melatonin to ameliorate the disease-associated stress, and ultimately its level decreases. Variations in the endogenous melatonin level may further affect the outcome of exogenously administered melatonin in experimental models and patients with UC. Moreover, melatonin level in the lower gut is supposed to be influenced by the luminal contents,8 and therefore, variation in the luminal contents among individuals may affect the role of melatonin in UC. In vitro autoradiography depicted tremendous diversity in the distribution of binding sites of 2-[125I]-iodomelatonin in the GIT of different species substantiating that melatonin may elicit different functions in the gut of diverse species.105 Furthermore, melatonin receptors (MT1, MT2, and MT3) exhibit differences in the regional distribution in the GIT.8 MT1 and MT2 are the G protein-coupled receptors that can couple to different signaling cascades, whose activation can lead to unique cellular responses. MT3 is an enzyme, quinone reductase 2, with potent antioxidant properties.106 Single nucleotide polymorphisms in the genes coding for critical enzymes and receptors involved in the melatonin pathway are known to be associated with several pathological conditions such as multiple sclerosis and type 2 diabetes.107,108 Differences in the susceptibility among individuals toward such single nucleotide polymorphisms due to genetic and epigenetic variations may be one of the causes of variability in their response to melatonin treatment for UC. The concept of personalized melatonin treatment may be beneficial for the management of UC under such conditions. Studies in UC revealed that chronic inflammatory states are accompanied by marked increases in CpG island methylation in normal-appearing tissues, affirming that proinflammatory exposures could account for part of the epigenetic variation in human populations, which in turn, occurs because of difference in age, environment, lifestyle, or genetic factors.109 Melatonin-mediated protection against UC might be due to its anti-inflammatory and antioxidant properties, whereas melatonin-mediated aggravation of UC might be due to its immunostimulatory and proinflammatory properties. Further experimental and clinical studies are needed to elucidate the exact role of various factors on the effect of melatonin against UC and to explore the reasons for contradictory effects in UC. Novel Targets of Melatonin in UC Nrf2 A plethora of evidence points to nuclear factor erythroid 2 (NF-E2)-related factor 2 (Nrf2) as a key regulator of the cellular response to inflammatory cytokines and oxidative stress in multiple tissue and cell types. Nrf2 is a redox-sensitive transcription factor, which is sequestered in the cytoplasm by the cytoskeleton-associated protein, Kelch-like ECH-associated protein 1 (Keap1), under basal resting conditions.110 In response to oxidative stress, Nrf2 is liberated from its repressor Keap1 and translocates into the nucleus, where it forms a heterodimer with a small musculoaponeurotic fibrosarcoma (sMaf) protein.111 The Nrf2–sMaf dimer then binds to antioxidant response elements (ARE) resulting in an activation of various phase II detoxifying or antioxidant enzymes such as NAD(P)H:quinone oxidoreductase (NQO-1), glutathione S-transferase (GST), heme oxygenase-1 (HO-1), glutathione peroxidase (GPx), glutamate cysteine ligase (GCL), and peroxiredoxin I (Prx I) all of which play key roles in cellular defense by removing the cytotoxic electrophiles or ROS.112,113 In this manner, Nrf2 responds to proinflammatory stimuli and rescues cells and tissues from inflammatory injuries.114,–119 UC involves chronic inflammation, wherein various inflammatory cells become activated, and their activation is accompanied by a rise in the release of ROS at the site of inflammation. Nrf2 deficiency leads to elevated inflammation-induced and oxidative stress–induced tissue damage. This notion is substantiated by the fact that in a DSS-mediated mouse colitis model, Nrf2-knockout mice depict an increased production of COX-2, iNOS, IL-1β, IL-6, and TNF-α as compared to the wild-type mice suggesting a beneficial role of Nrf2 in UC. Increased severity of colitis in Nrf2-deficient mice is associated with decreased expression of antioxidant/phase II detoxifying enzymes including HO-1, NQO-1, UDP-glucuronosyltransferase 1A1, and GST Mu-1.120 Furthermore, Nrf2-activated HO-1 mediates protection against TNBS-induced colitis in mice presumably by blocking the TNF-α–induced nuclear translocation of NF-κB.121 Based on this evidence, it can be inferred that Nrf2 plays an important role in the defense against inflammation and oxidative stress possibly by suppression of NF-κB–mediated proinflammatory signaling pathways and activation of cellular antioxidant machinery. Furthermore, Nrf2 genotype plays a major role in modulating inflammation-promoted colorectal tumorigenesis. Increased colonic inflammatory injury and formation of aberrant crypt foci were found in Nrf2-deficient mice on azoxymethane and DSS treatment.122 Nrf2/ARE signaling is involved in attenuating inflammation-associated pathogenesis in UC by increasing gene expression of various cytoprotective enzymes that extinguish the “fires within” (inflammation). Therefore, targeting Nrf2-antioxidative stress signaling is an ideal strategy to prevent or treat inflammation and oxidative stress–related diseases and the associated DNA damage. Interestingly, melatonin targets Nrf2 and thereby protects against tissue damage in experimental and clinical conditions.123,–126 The cell protection signaling of melatonin involves redox-sensitive components, and it regulates the cross talk between NF-κB and Nrf2 pathways.76 By regulating these pathways, melatonin stimulates the expression of antioxidant and detoxification genes and thereby enhances the glutathione system, which is a potent endogenous antioxidant system. Studies demonstrating the protective role of melatonin in UC through an activation of Nrf2-mediated signaling pathway are required to prove its better therapeutic role in UC. Nrf2 could be considered as a novel target for melatonin to ameliorate the severity of UC. Signal Transducer and Activator of Transcription 3 (STAT3) and IL-17 STAT3 belongs to the family of proteins that are intracellular signal transduction molecules involved in the expression of numerous proinflammatory genes in inflammatory cells. It is an important mediator in both human inflammatory bowel diseases and in animal models of colitis. Under resting condition, STAT3 is present in the cytoplasm of most cells. It undergoes sequential tyrosine phosphorylation, dimerization, nuclear translocation, DNA binding, and gene transcription in response to certain inflammatory stimuli and growth factors.45 UC is associated with an elevated level of IL-6, which in turn, induces the transcription factor STAT3 through IL-6/STAT-3 trans-signaling pathway.127,–129 In addition, the expression of IL-24, an activator of the JAK1/STAT3/SOCS3 cascade, is enhanced in inflammatory bowel diseases.130 Furthermore, inhibition of STAT3 by antisense oligonucleotide in TNBS-induced murine colitis had a beneficial effect in the amelioration of colitis.131 STAT3 is upregulated in the colonic mucosa of patients with UC, and it should be explored as a potential therapeutic target.132 Moreover, STAT3, activated by both IL-6 and IL-23, plays a critical role in T helper 17 (Th17) development.133 Th17, also known as inflammatory Th17, is responsible for the synthesis of various cytokines such as IL-17A, IL-17F, IL-21, IL-22, and IL-26.134,–136 IL-17A, IL-17F, IL-22, and IL-26 are excessively produced in the inflamed gut of patients with UC.137,–140 It has been reported that IL-17 functions as a proinflammatory cytokine and acts synergistically with TNF-α and IL-1.141 Moreover, IL-1β and TNF-α may also be involved in promoting Th17 development or in regulating the expression of IL-17.142,143 Both STAT3 and Th-17 cytokines play a role in the perpetuation of the inflammatory process and the subsequent DNA damage associated with the development of UC. Blocking such signaling pathways can open new avenues for the development of therapeutic strategies to ameliorate the severity of UC. Melatonin alleviates high levels of IL-1β, IL-6, and TNF-α, which are involved in regulating the expression of STAT3 and IL-17.144 Another possible target of melatonin against UC could be IL-21, which leads to the expansion of both Th1 and Th17 cell responses in the gut.145,146 Like human inflammatory bowel diseases, mice with acute DSS-colitis and TNBS-relapsing colitis produce elevated levels of IL-21, which in turn, increase the production of Th17-related cytokines.145 Hence, these may serve as potential targets for melatonin to combat inflammation and the associated DNA damage, and thereby reduce the severity of UC. Sirtuins Sirtuins, with either histone deacetylase or mono-ribosyl transferase activity, play a critical role in the regulation of inflammation and the associated pathogenesis. The sirtuin family consists of 7 members (SIRT1 to SIRT7) that localize to the nucleus (SIRT1, 6, and 7), the cytoplasm (SIRT2), and the mitochondria (SIRT3, 4, and 5); however, some of them can shuttle between these compartments.147 The deacetylase activity of these proteins leads to the removal of the lysine-linked acetyl group of the target protein.148 SIRT1 acts as a negative regulator of NF-κB activity through the deacetylation of the p65 lysine 310.149 This NF-κB-SIRT1–negative loop has been established in several experimental models, confirming its biological relevance. Several studies have shown that systemic SIRT1 activation decreases inflammation by reducing NF-κB activity.150,151 Lack of SIRT1 activity results in activation of NF-κB and contributes to the development of cellular inflammation. DSS-induced colitis in mice has been found to be associated with a decrease in SIRT1 gene expression and an increase in NF-κB expression. Colitis was abrogated with the treatment of resveratrol, which led to an induction of SIRT1 and reduction in NF-κB activation.152 This suggests that reduced SIRT1 expression and elevated NF-κB activation are implicated in the development of UC. Several studies have reported the upregulation of SIRT1 and an increased deacetylation of various SIRT1 substrates such as PGC-1a, Foxo1, NF-κB, and p53 by melatonin.153,–155 Recently, melatonin was found to modulate sirtuin activity, which is associated with several pathological conditions, including chronic intestinal inflammation and colon cancer.9 Hence, SIRT1 may be one of the potential targets of melatonin for the treatment of UC. Poly (ADP-ribose) Polymerase 1 (PARP1) PARP1 is a major intracellular NAD-consuming enzyme that competes with SIRT1 for its substrate, and both of their activities depend on the availability of NAD.156 Overactivation of PARP1 depletes NAD and increases the level of nicotinamide, which further inhibits SIRT1 activity.157 PARP1 inhibitors contribute in maintaining high intracellular NAD levels resulting in increased SIRT1 activity.158 Thus, PARP1 inhibitors offer new pharmacological opportunities to activate sirtuins by inhibiting competing NAD-consuming enzymes. PARP1 plays an important role in the colon injury associated with colitis. In UC, the colonic mucosa is infiltrated with neutrophils suggesting the presence of activated T cells and macrophages.159 In macrophages, an ADP-ribosylation reaction mediated by PARP leads to the activation of NF-κB and precedes the upregulation of iNOS activity and the release of proinflammatory cytokines.160 Inhibition of PARP attenuates NF-κB activation in macrophages and prevents the release of cytokines.161 Moreover, inhibition of PARP activation reduced intracellular adhesion molecule-1 and P-selectin expression, leading to an inhibition of neutrophil recruitment and decreased oxidant generation in the lamina propria of mice.162,163 Furthermore, lack of sufficient ATP, which is caused by PARP, results in a damaged cell undergoing necrosis rather than apoptosis.164 PARP inhibition may improve barrier function and suppress inflammation by preventing the necrosis of colonic epithelial cells after oxidant-mediated injury.165 Several studies depict the potential of melatonin to attenuate PARP activation.166,–168 Inflammation and oxidative stress lead to DNA damage, and PARP gets activated in response to DNA damage.169 Thus, an ability of melatonin to reduce PARP activation might be attributed to its anti-inflammatory, antioxidant, and antigenotoxic effects. Because UC is associated with inflammation, oxidative stress, and DNA damage, which in turn, leads to PARP activation; melatonin may decrease PARP activation by modulating inflammation, oxidative stress, and genotoxicity. Therefore, PARP may be another potential target for melatonin to combat inflammation, oxidative stress, and the subsequent DNA damage associated with UC. Death Proteins Death proteins are the cytosolic enzymes that spill out of the cell when the plasma membrane gets ruptured during necrosis and become activated in the presence of high extracellular calcium concentration.170 In the case of fulminating UC, there occurs colonic wall necrosis, which in turn, may result into the leakage of these hydrolytic enzymes from the necrotic cells.171 These death proteins hydrolyze their respective substrates in the plasma membrane of neighboring cells, thereby leading to self-perpetuating tissue injury.170 The evidence for a role of death proteins in the pathogenesis of UC was obtained from a study in which TNBS-induced UC led to an elevation in the level of calpain, a cytosolic enzyme, in the colon mucosa of rat, and calpain inhibitor had been known to reduce the colon injury caused by dinitrobenzene sulfonic acid in rat.172,173 Another cytosolic enzyme, secretory phospholipase A2, increases in the colonic epithelial cells and in serum of the patients with UC.174,175 Melatonin reduces the levels of various proteases such as calpain and caspase-3 in an experimental model of spinal cord injury.176,177 Melatonin, because of its anti-inflammatory and antioxidant properties, reduces UC-associated mucosal damage in experimental models of colitis.34,72 Furthermore, melatonin plays a vital role in the regeneration of mucosal epithelium in the GIT.9 Because of its mucosal healing property, it can ameliorate the colonic mucosal necrosis in UC and lower the levels of hydrolytic enzymes, thereby alleviating the progression of self-perpetuated tissue injury. Hence, elucidating the potential role of melatonin in the reduction of UC-associated elevated death proteins may unravel its prospective as a better interventional agent to ameliorate the severity of UC. Telomere Dysfunction Telomerase enzyme is a key regulator of telomere length that signals cell death. The process of accelerated colon aging has been observed in patients with UC, which is further associated with telomere shortening in the colon mucosa.178,179 In UC, there is a reduction in the level of telomerase and alteration in telomere-binding proteins such as telomere repeat–binding factor 2 (TRF2) and repressor activator protein-1 (RAP1). TRF2 and RAP1 mRNA expression decreases in activated T lymphocytes in the patients with UC, suggesting a possible telomeric deficiency of these proteins, which could lead to telomeric dysfunction.180,181 Chronic inflammation and oxidative stress are associated with shorter leukocyte telomere lengths in the patients with periodontitis and antioxidants have been reported to prevent telomere shortening in women consuming a multivitamin supplement.182,183 Shorter telomeres associated with UC may be due to persistent inflammation and oxidative stress. Melatonin, being a potent anti-inflammatory and antioxidant agent, might play some role in preventing UC-associated telomere shortening by modulating inflammation and oxidative stress. Strategies can be explored to use melatonin alone or in combination with other enteroprotective agents to mitigate the severity of this debilitating disease. Epigenetic Alterations UC is associated not only with genetic, but also with epigenetic alterations, in the colonic mucosa.184,185 UC is characterized by an increased expression of various inflammatory genes in the colonic mucosa.37 Histone acetylation modulates the function of various cytokine receptors, nuclear hormone receptors, intracellular signaling molecules, and transcription factors.186 Increased expression of HDAC9 is associated with colitis in mice, and HDAC9-knockout mice are resistant to developing colitis.187 Apart from alterations in histone acetylation, UC is linked to modifications in DNA methylation.185,188 Studies reported that patients with UC exhibit DNA hypermethylation of tumor suppressor genes such as estrogen receptor gene (ESR-1) and tumor suppressor gene candidate-3 (TUSC3, N-33).189 Furthermore, epigenetic changes such as DNA methylation and histone modification are thought to play an essential role in inflammation-induced carcinogenesis. Histone hyperacetylation ameliorates experimental colitis in mice.190 Melatonin induces histone hyperacetylation and inhibits DNA methyltransferase in experimental studies.191,192 Thus, melatonin may play an important role in the epigenetic regulation and alter UC-associated epigenetic modifications, thereby ameliorating the disease severity. The detailed molecular pathways that can be targeted by melatonin for its better therapeutic intervention in UC have been depicted in Figure 1. Figure 1. View largeDownload slide Diagrammatic representation of the molecular targets, both cytoplasmic and nuclear (genetic and epigenetic), of melatonin for possible protection against UC. “M” represents melatonin. “?” depicts that the targets have not yet been explored for possible protection with melatonin in UC. Figure 1. View largeDownload slide Diagrammatic representation of the molecular targets, both cytoplasmic and nuclear (genetic and epigenetic), of melatonin for possible protection against UC. “M” represents melatonin. “?” depicts that the targets have not yet been explored for possible protection with melatonin in UC. Missing Links and Future Prospects Melatonin has been explored as an interventional agent for ameliorating the severity of UC, both in experimental and clinical conditions, because it acts on various molecular targets. However, there are several other unexplored molecular targets on which melatonin can act to exert its possible beneficial effects in UC. Uncovering previously unexplored targets of melatonin in UC using cutting-edge science with well-designed experimental and clinical trials can provide better opportunity for future research. The effects of melatonin on UC-associated genetic and epigenetic alterations require detailed examination for the design of better clinical strategies. The use of genomic and proteomic techniques could make a substantial contribution in identifying the novel targets for melatonin at the early stages of disease initiation and for more reliable intervention. Interestingly, melatonin-loaded nanomedicines have shown better efficacy in an experimental model of sepsis.193 This approach of therapy should be explored in preclinical and clinical trials for the treatment of UC. Recently, it has been shown that higher accumulation of liposomal drug in inflammatory area and specific release of liposomes by enteric coated capsules provide better option for the treatment of colonic disease.194 Similar concept can be used for the targeted delivery of melatonin-loaded liposomes to the colon in patients with UC. Targeted delivery of hormones such as methylprednisolone and budesonide has already depicted better efficacy in UC.195,196 Melatonin, being a hormone, can also be targeted to colon in a similar manner. Topical melatonin such as melatonin suppositories and enemas could provide better efficacy in colitis. Furthermore, targeted delivery of melatonin to colon could enhance its effectiveness and could be the key for its ability to impact a desired pathway involved in the pathogenesis of UC. For better efficacy, attention must be given to both receptor-mediated and receptor-independent actions of melatonin in UC and its dose and time of administration. This would form the basis for further studies to understand the mechanisms involved in melatonin-mediated protection against UC and the associated extra-intestinal manifestations. It will also provide a clue on how to best take advantage of the potential therapeutic targets of melatonin for the treatment of UC. The targets mentioned in this review need critical validation at both preclinical and clinical situation for better translation. Conclusions UC is a debilitating disease with severe health consequences having considerable impact on the quality of patients' life. Thus, a better understanding of ways to treat UC would be of potential relevance to a great number of people. It has been postulated that inflammation, oxidative stress, and the subsequent DNA damage are hallmarks in the development and progression of UC. Melatonin has the potential to interact with multiple molecular targets and can provide an exciting avenue to explore various novel strategies to reduce the occurrence and to mitigate the severity of UC. However, this cannot be solely based on administering the compound under investigation to the patients with UC. Effects of melatonin on different pathways could be dose dependent and circadian rhythm–dependent, and hence choice of melatonin dose regimens and time of administration to treat patients with UC could be crucial factors. An essential part of investigation should be the study of pharmacogenetic and epigenetic profile of the patients with UC in context of age, disease condition, and dietary profile to explore maximum benefit out of the intervention strategy. Finally, the role of melatonin in targeting the molecular machinery involved in the perpetuation of the pathogenesis of UC is worthy of investigation and exciting both for scientific research and for clinical intervention. References 1. Reiter RJ, Tan DX, Rosales-Corral S, et al.  . The universal nature, unequal distribution and antioxidant functions of melatonin and its derivatives. Mini Rev Med Chem . 2013; 13: 373– 384. Google Scholar PubMed  2. Stehle JH, Saade A, Rawashdeh O, et al.  . A survey of molecular details in the human pineal gland in the light of phylogeny, structure, function and chronobiological diseases. J Pineal Res . 2011; 51: 17– 43. Google Scholar CrossRef Search ADS PubMed  3. Lerner AB, Case JD, Takahashi Y, et al.  . Isolation of melatonin, the pineal gland factor that lightens melanocyteS1. J Am Chem Soc.  1958; 80: 2587. Google Scholar CrossRef Search ADS   4. Raikhlin NT, Kvetnoi IM. Biological identification of melatonin in enterochromaffin cells [in Russian]. Dokl Akad Nauk SSSR . 1974; 215: 731– 732. Google Scholar PubMed  5. Bubenik GA, Brown GM, Grota LJ. Immunohistological localization of melatonin in the rat digestive system. Experientia . 1977; 33: 662– 663. Google Scholar CrossRef Search ADS PubMed  6. Huether G. The contribution of extrapineal sites of melatonin synthesis to circulating melatonin levels in higher vertebrates. Experientia . 1993; 49: 665– 670. Google Scholar CrossRef Search ADS PubMed  7. Bubenik GA. Gastrointestinal melatonin: localization, function, and clinical relevance. Dig Dis Sci . 2002; 47: 2336– 2348. Google Scholar CrossRef Search ADS PubMed  8. Chen CQ, Fichna J, Bashashati M, et al.  . Distribution, function and physiological role of melatonin in the lower gut. World J Gastroenterol . 2011; 17: 3888– 3898. Google Scholar CrossRef Search ADS PubMed  9. Motilva V, Garcia-Maurino S, Talero E, et al.  . New paradigms in chronic intestinal inflammation and colon cancer: role of melatonin. J Pineal Res . 2011; 51: 44– 60. Google Scholar CrossRef Search ADS PubMed  10. Galano A, Tan DX, Reiter RJ. Melatonin as a natural ally against oxidative stress: a physicochemical examination. J Pineal Res . 2011; 51: 1– 16. Google Scholar CrossRef Search ADS PubMed  11. Galano A, Tan DX, Reiter RJ. On the free radical scavenging activities of melatonin's metabolites, AFMK and AMK. J Pineal Res . 2013; 54: 245– 257. Google Scholar CrossRef Search ADS PubMed  12. Hardeland R. Antioxidative protection by melatonin: multiplicity of mechanisms from radical detoxification to radical avoidance. Endocrine . 2005; 27: 119– 130. Google Scholar CrossRef Search ADS PubMed  13. Mauriz JL, Collado PS, Veneroso C, et al.  . A review of the molecular aspects of melatonin's anti-inflammatory actions: recent insights and new perspectives. J Pineal Res . 2013; 54: 1– 14. Google Scholar CrossRef Search ADS PubMed  14. Proietti S, Cucina A, Reiter RJ, et al.  . Molecular mechanisms of melatonin's inhibitory actions on breast cancers. Cell Mol Life Sci . 2013; 70: 2139– 2157. Google Scholar CrossRef Search ADS PubMed  15. Reiter RJ, Paredes SD, Manchester LC, et al.  . Reducing oxidative/nitrosative stress: a newly-discovered genre for melatonin. Crit Rev Biochem Mol Biol . 2009; 44: 175– 200. Google Scholar CrossRef Search ADS PubMed  16. Tan DX, Chen LD, Poeggeler B, et al.  . Melatonin: a potent, endogenous hydroxyl radical scavenger. Endocr J . 1993; 1: 57– 60. 17. Arguelles S, Munoz MF, Cano M, et al.  . In vitro and in vivo protection by melatonin against the decline of elongation factor-2 caused by lipid peroxidation: preservation of protein synthesis. J Pineal Res . 2012; 53: 1– 10. Google Scholar CrossRef Search ADS PubMed  18. Laliena A, San Miguel B, Crespo I, et al.  . Melatonin attenuates inflammation and promotes regeneration in rabbits with fulminant hepatitis of viral origin. J Pineal Res . 2012; 53: 270– 278. Google Scholar CrossRef Search ADS PubMed  19. Song N, Kim AJ, Kim HJ, et al.  . Melatonin suppresses doxorubicin-induced premature senescence of A549 lung cancer cells by ameliorating mitochondrial dysfunction. J Pineal Res . 2012; 53: 335– 343. Google Scholar CrossRef Search ADS PubMed  20. Abe M, Reiter RJ, Orhii PB, et al.  . Inhibitory effect of melatonin on cataract formation in newborn rats: evidence for an antioxidative role for melatonin. J Pineal Res . 1994; 17: 94– 100. Google Scholar CrossRef Search ADS PubMed  21. Cardinali DP, Srinivasan V, Brzezinski A, et al.  . Melatonin and its analogs in insomnia and depression. J Pineal Res . 2012; 52: 365– 375. Google Scholar CrossRef Search ADS PubMed  22. de Matos Cavalcante AG, de Bruin PF, de Bruin VM, 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: 238– 244. Google Scholar CrossRef Search ADS PubMed  23. Gitto E, Pellegrino S, Gitto P, et al.  . Oxidative stress of the newborn in the pre- and postnatal period and the clinical utility of melatonin. J Pineal Res . 2009; 46: 128– 139. Google Scholar CrossRef Search ADS PubMed  24. Koppisetti S, Jenigiri B, Terron MP, et al.  . Reactive oxygen species and the hypomotility of the gall bladder as targets for the treatment of gallstones with melatonin: a review. Dig Dis Sci . 2008; 53: 2592– 2603. Google Scholar CrossRef Search ADS PubMed  25. Kotlarczyk MP, Lassila HC, O'Neil CK, et al.  . Melatonin osteoporosis prevention study (MOPS): a randomized, double-blind, placebo-controlled study examining the effects of melatonin on bone health and quality of life in perimenopausal women. J Pineal Res . 2012; 52: 414– 426. Google Scholar CrossRef Search ADS PubMed  26. Maldonado MD, Murillo-Cabezas F, Terron MP, et al.  . The potential of melatonin in reducing morbidity-mortality after craniocerebral trauma. J Pineal Res . 2007; 42: 1– 11. Google Scholar CrossRef Search ADS PubMed  27. Peschke E, Hofmann K, Ponicke K, et al.  . Catecholamines are the key for explaining the biological relevance of insulin-melatonin antagonisms in type 1 and type 2 diabetes. J Pineal Res . 2012; 52: 389– 396. Google Scholar CrossRef Search ADS PubMed  28. Rosales-Corral SA, Acuna-Castroviejo D, Coto-Montes A, et al.  . Alzheimer's disease: pathological mechanisms and the beneficial role of melatonin. J Pineal Res . 2012; 52: 167– 202. Google Scholar CrossRef Search ADS PubMed  29. Santhi N, Thorne HC, van der Veen DR, et al.  . The spectral composition of evening light and individual differences in the suppression of melatonin and delay of sleep in humans. J Pineal Res . 2012; 53: 47– 59. Google Scholar CrossRef Search ADS PubMed  30. Konturek PC, Brzozowska I, Targosz A, et al.  . Esophagoprotection mediated by exogenous and endogenous melatonin in an experimental model of reflux esophagitis. J Pineal Res . 2013; 55: 46– 57. Google Scholar CrossRef Search ADS PubMed  31. Mozaffari S, Rahimi R, Abdollahi M. Implications of melatonin therapy in irritable bowel syndrome: a systematic review. Curr Pharm Des . 2010; 16: 3646– 3655. Google Scholar CrossRef Search ADS PubMed  32. Rakhimova O. Use of melatonin in combined treatment for inflammatory bowel diseases [in Russian]. Ter Arkh . 2010; 82: 64– 68. Google Scholar PubMed  33. Werbach MR. Melatonin for the treatment of gastroesophageal reflux disease. Altern Ther Health Med . 2008; 14: 54– 58. Google Scholar PubMed  34. Tahan G, Gramignoli R, Marongiu F, et al.  . Melatonin expresses powerful anti-inflammatory and antioxidant activities resulting in complete improvement of acetic-acid-induced colitis in rats. Dig Dis Sci . 2011; 56: 715– 720. Google Scholar CrossRef Search ADS PubMed  35. Okada Y, Tsuzuki Y, Miyazaki J, et al.  . Propionibacterium freudenreichii component 1.4-dihydroxy-2-naphthoic acid (DHNA) attenuates dextran sodium sulphate induced colitis by modulation of bacterial flora and lymphocyte homing. Gut . 2006; 55: 681– 688. Google Scholar CrossRef Search ADS PubMed  36. Rosillo MA, Sanchez-Hidalgo M, Cardeno A, et al.  . Dietary supplementation of an ellagic acid-enriched pomegranate extract attenuates chronic colonic inflammation in rats. Pharmacol Res . 2012; 66: 235– 242. Google Scholar CrossRef Search ADS PubMed  37. Trivedi PP, Jena GB. Dextran sulfate sodium-induced ulcerative colitis leads to increased hematopoiesis and induces both local as well as systemic genotoxicity in mice. Mutat Res . 2012; 744: 172– 183. Google Scholar CrossRef Search ADS PubMed  38. Yao J, Wang JY, Liu L, et al.  . Anti-oxidant effects of resveratrol on mice with DSS-induced ulcerative colitis. Arch Med Res . 2010; 41: 288– 294. Google Scholar CrossRef Search ADS PubMed  39. Bitiren M, Karakilcik AZ, Zerin M, et al.  . Protective effects of selenium and vitamin E combination on experimental colitis in blood plasma and colon of rats. Biol Trace Elem Res . 2010; 136: 87– 95. Google Scholar CrossRef Search ADS PubMed  40. Saleh M, Trinchieri G. Innate immune mechanisms of colitis and colitis-associated colorectal cancer. Nat Rev Immunol . 2011; 11: 9– 20. Google Scholar CrossRef Search ADS PubMed  41. Trivedi PP, Jena GB. Ulcerative colitis-induced hepatic damage in mice: studies on inflammation, fibrosis, oxidative DNA damage and GST-P expression. Chem Biol Interact . 2013; 201: 19– 30. Google Scholar CrossRef Search ADS PubMed  42. Westbrook AM, Wei B, Braun J, et al.  . Intestinal inflammation induces genotoxicity to extraintestinal tissues and cell types in mice. Int J Cancer . 2011; 129: 1815– 1825. Google Scholar CrossRef Search ADS PubMed  43. Dong WG, Mei Q, Yu JP, et al.  . Effects of melatonin on the expression of iNOS and COX-2 in rat models of colitis. World J Gastroenterol . 2003; 9: 1307– 1311. Google Scholar CrossRef Search ADS PubMed  44. Marquez E, Sanchez-Fidalgo S, Calvo JR, et al.  . Acutely administered melatonin is beneficial while chronic melatonin treatment aggravates the evolution of TNBS-induced colitis. J Pineal Res . 2006; 40: 48– 55. Google Scholar CrossRef Search ADS PubMed  45. Aggarwal BB, Sethi G, Ahn KS, et al.  . Targeting signal-transducer-and-activator-of-transcription-3 for prevention and therapy of cancer: modern target but ancient solution. Ann N Y Acad Sci . 2006; 1091: 151– 169. Google Scholar CrossRef Search ADS PubMed  46. Aggarwal BB, Shishodia S, Sandur SK, et al.  . Inflammation and cancer: how hot is the link? Biochem Pharmacol.  2006; 72: 1605– 1621. Google Scholar CrossRef Search ADS PubMed  47. Pollard JW. Tumour-educated macrophages promote tumour progression and metastasis. Nat Rev Cancer . 2004; 4: 71– 78. Google Scholar CrossRef Search ADS PubMed  48. Ohshima H, Tatemichi M, Sawa T. Chemical basis of inflammation-induced carcinogenesis. Arch Biochem Biophys . 2003; 417: 3– 11. Google Scholar CrossRef Search ADS PubMed  49. Sawa T, Ohshima H. Nitrative DNA damage in inflammation and its possible role in carcinogenesis. Nitric Oxide . 2006; 14: 91– 100. Google Scholar CrossRef Search ADS PubMed  50. Jin Y, Kotakadi VS, Ying L, et al.  . American ginseng suppresses inflammation and DNA damage associated with mouse colitis. Carcinogenesis . 2008; 29: 2351– 2359. Google Scholar CrossRef Search ADS PubMed  51. D'Inca R, Cardin R, Benazzato L, et al.  . Oxidative DNA damage in the mucosa of ulcerative colitis increases with disease duration and dysplasia. Inflamm Bowel Dis . 2004; 10: 23– 27. Google Scholar CrossRef Search ADS PubMed  52. Westbrook AM, Wei B, Braun J, et al.  . Intestinal mucosal inflammation leads to systemic genotoxicity in mice. Cancer Res . 2009; 69: 4827– 4834. Google Scholar CrossRef Search ADS PubMed  53. Jena GB, Trivedi PP, Sandala B. Oxidative stress in ulcerative colitis: an old concept but a new concern. Free Radic Res . 2012; 46: 1339– 1345. Google Scholar CrossRef Search ADS PubMed  54. Trivedi PP, Jena GB. Role of alpha-lipoic acid in dextran sulfate sodium-induced ulcerative colitis in mice: studies on inflammation, oxidative stress, DNA damage and fibrosis. Food Chem Toxicol . 2013; 59: 339– 355. Google Scholar CrossRef Search ADS PubMed  55. Tan DX, Manchester LC, Hardeland R, et al.  . Melatonin: a hormone, a tissue factor, an autocoid, a paracoid, and an antioxidant vitamin. J Pineal Res . 2003; 34: 75– 78. Google Scholar CrossRef Search ADS PubMed  56. Luchetti F, Canonico B, Betti M, et al.  . Melatonin signaling and cell protection function. FASEB J . 2010; 24: 3603– 3624. Google Scholar CrossRef Search ADS PubMed  57. Nosal'ova V, Zeman M, Cerna S, et al.  . Protective effect of melatonin in acetic acid induced colitis in rats. J Pineal Res . 2007; 42: 364– 370. Google Scholar CrossRef Search ADS PubMed  58. Zhao ZJ, Xiang JY, Liu L, et al.  . Parthenolide, an inhibitor of the nuclear factor-kappaB pathway, ameliorates dextran sulfate sodium-induced colitis in mice. Int Immunopharmacol . 2012; 12: 169– 174. Google Scholar CrossRef Search ADS PubMed  59. Ghosh S, Hayden MS. New regulators of NF-kappaB in inflammation. Nat Rev Immunol . 2008; 8: 837– 848. Google Scholar CrossRef Search ADS PubMed  60. Sun SC, Ganchi PA, Ballard DW, et al.  . NF-kappa B controls expression of inhibitor I kappa B alpha: evidence for an inducible autoregulatory pathway. Science . 1993; 259: 1912– 1915. Google Scholar CrossRef Search ADS PubMed  61. Lappas M, Permezel M, Georgiou HM, et al.  . Nuclear factor kappa B regulation of proinflammatory cytokines in human gestational tissues in vitro. Biol Reprod . 2002; 67: 668– 673. Google Scholar CrossRef Search ADS PubMed  62. Rogler G, Brand K, Vogl D, et al.  . Nuclear factor kappaB is activated in macrophages and epithelial cells of inflamed intestinal mucosa. Gastroenterology . 1998; 115: 357– 369. Google Scholar CrossRef Search ADS PubMed  63. Schreiber S, Nikolaus S, Hampe J. Activation of nuclear factor kappa B inflammatory bowel disease. Gut . 1998; 42: 477– 484. Google Scholar CrossRef Search ADS PubMed  64. Andresen L, Jorgensen VL, Perner A, et al.  . Activation of nuclear factor kappaB in colonic mucosa from patients with collagenous and ulcerative colitis. Gut . 2005; 54: 503– 509. Google Scholar CrossRef Search ADS PubMed  65. Akcan A, Kucuk C, Sozuer E, et al.  . Melatonin reduces bacterial translocation and apoptosis in trinitrobenzene sulphonic acid-induced colitis of rats. World J Gastroenterol . 2008; 14: 918– 924. Google Scholar CrossRef Search ADS PubMed  66. Li JH, Yu JP, Yu HG, et al.  . Melatonin reduces inflammatory injury through inhibiting NF-kappaB activation in rats with colitis. Mediators Inflamm . 2005; 2005: 185– 193. Google Scholar CrossRef Search ADS PubMed  67. Li JH, Zhou W, Liu K, et al.  . Melatonin reduces the expression of chemokines in rat with trinitrobenzene sulfonic acid-induced colitis. Saudi Med J . 2008; 29: 1088– 1094. Google Scholar PubMed  68. Mei Q, Yu JP, Xu JM, et al.  . Melatonin reduces colon immunological injury in rats by regulating activity of macrophages. Acta Pharmacol Sin . 2002; 23: 882– 886. Google Scholar PubMed  69. Sayyed HG, Jaumdally RJ, Idriss NK, et al.  . The effect of melatonin on plasma markers of inflammation and on expression of nuclear factor-kappa beta in acetic acid-induced colitis in the rat. Dig Dis Sci . 2013; 58: 3156– 3164. Google Scholar CrossRef Search ADS PubMed  70. Tanaka T, Yasui Y, Tanaka M, et al.  . Melatonin suppresses AOM/DSS-induced large bowel oncogenesis in rats. Chem Biol Interact . 2009; 177: 128– 136. Google Scholar CrossRef Search ADS PubMed  71. Tasdemir S, Parlakpinar H, Vardi N, et al.  . Effect of endogen-exogenous melatonin and erythropoietin on dinitrobenzene sulfonic acid-induced colitis. Fundam Clin Pharmacol . 2013; 27: 299– 307. Google Scholar CrossRef Search ADS PubMed  72. Cuzzocrea S, Mazzon E, Serraino I, et al.  . Melatonin reduces dinitrobenzene sulfonic acid-induced colitis. J Pineal Res . 2001; 30: 1– 12. Google Scholar CrossRef Search ADS PubMed  73. Mazzon E, Esposito E, Crisafulli C, et al.  . Melatonin modulates signal transduction pathways and apoptosis in experimental colitis. J Pineal Res . 2006; 41: 363– 373. Google Scholar CrossRef Search ADS PubMed  74. Yang XL, Guo TK, Wang YH, et al.  . Therapeutic effect of ginsenoside Rd in rats with TNBS-induced recurrent ulcerative colitis. Arch Pharm Res . 2012; 35: 1231– 1239. Google Scholar CrossRef Search ADS PubMed  75. Reiter RJ, Tan DX, Mayo JC, et al.  . Melatonin as an antioxidant: biochemical mechanisms and pathophysiological implications in humans. Acta Biochim Pol . 2003; 50: 1129– 1146. Google Scholar PubMed  76. Tripathi DN, Jena GB. Effect of melatonin on the expression of Nrf2 and NF-kappaB during cyclophosphamide-induced urinary bladder injury in rat. J Pineal Res . 2010; 48: 324– 331. Google Scholar CrossRef Search ADS PubMed  77. Cevik H, Erkanli G, Ercan F, et al.  . Exposure to continuous darkness ameliorates gastric and colonic inflammation in the rat: both receptor and non-receptor-mediated processes. J Gastroenterol Hepatol . 2005; 20: 294– 303. Google Scholar CrossRef Search ADS PubMed  78. Tan DX, Manchester LC, Liu X, et al.  . Mitochondria and chloroplasts as the original sites of melatonin synthesis: a hypothesis related to melatonin's primary function and evolution in eukaryotes. J Pineal Res . 2013; 54: 127– 138. Google Scholar CrossRef Search ADS PubMed  79. Venegas C, Garcia JA, Escames G, et al.  . Extrapineal melatonin: analysis of its subcellular distribution and daily fluctuations. J Pineal Res . 2012; 52: 217– 227. Google Scholar CrossRef Search ADS PubMed  80. Di Sebastiano P, di Mola FF, Artese L, et al.  . Beneficial effects of Batimastat (BB-94), a matrix metalloproteinase inhibitor, in rat experimental colitis. Digestion . 2001; 63: 234– 239. Google Scholar CrossRef Search ADS PubMed  81. Parks WC, Shapiro SD. Matrix metalloproteinases in lung biology. Respir Res . 2001; 2: 10– 19. Google Scholar CrossRef Search ADS PubMed  82. Parks WC, Wilson CL, Lopez-Boado YS. Matrix metalloproteinases as modulators of inflammation and innate immunity. Nat Rev Immunol . 2004; 4: 617– 629. Google Scholar CrossRef Search ADS PubMed  83. Sternlicht MD, Werb Z. How matrix metalloproteinases regulate cell behavior. Annu Rev Cell Dev Biol . 2001; 17: 463– 516. Google Scholar CrossRef Search ADS PubMed  84. Medina C, Videla S, Radomski A, et al.  . Increased activity and expression of matrix metalloproteinase-9 in a rat model of distal colitis. Am J Physiol Gastrointest Liver Physiol . 2003; 284: G116– G122. Google Scholar CrossRef Search ADS PubMed  85. Naito Y, Takagi T, Kuroda M, et al.  . An orally active matrix metalloproteinase inhibitor, ONO-4817, reduces dextran sulfate sodium-induced colitis in mice. Inflamm Res . 2004; 53: 462– 468. Google Scholar CrossRef Search ADS PubMed  86. Naito Y, Yoshikawa T. Role of matrix metalloproteinases in inflammatory bowel disease. Mol Aspects Med . 2005; 26: 379– 390. Google Scholar CrossRef Search ADS PubMed  87. Rudra DS, Pal U, Maiti NC, et al.  . Melatonin inhibits matrix metalloproteinase-9 activity by binding to its active site. J Pineal Res . 2013; 54: 398– 405. Google Scholar CrossRef Search ADS PubMed  88. Swarnakar S, Paul S, Singh LP, et al.  . Matrix metalloproteinases in health and disease: regulation by melatonin. J Pineal Res . 2011; 50: 8– 20. Google Scholar CrossRef Search ADS PubMed  89. Esposito E, Mazzon E, Riccardi L, et al.  . Matrix metalloproteinase-9 and metalloproteinase-2 activity and expression is reduced by melatonin during experimental colitis. J Pineal Res . 2008; 45: 166– 173. Google Scholar CrossRef Search ADS PubMed  90. Ganguly K, Swarnakar S. Chronic gastric ulceration causes matrix metalloproteinases-9 and -3 augmentation: alleviation by melatonin. Biochimie . 2012; 94: 2687– 2698. Google Scholar CrossRef Search ADS PubMed  91. Yan B, Peng Y, Li CY. Molecular analysis of genetic instability caused by chronic inflammation. Methods Mol Biol . 2009; 512: 15– 28. Google Scholar CrossRef Search ADS PubMed  92. Trivedi PP, Jena GB. Melatonin reduces ulcerative colitis-associated local and systemic damage in mice: investigation on possible mechanisms. Dig Dis Sci . 2013; 58: 3460– 3474. Google Scholar CrossRef Search ADS PubMed  93. Pentney PT, Bubenik GA. Melatonin reduces the severity of dextran-induced colitis in mice. J Pineal Res . 1995; 19: 31– 39. Google Scholar CrossRef Search ADS PubMed  94. Ham M, Kaunitz JD. Gastroduodenal defense. Curr Opin Gastroenterol . 2007; 23: 607– 616. Google Scholar CrossRef Search ADS PubMed  95. Calvo JR, Guerrero JM, Osuna C, et al.  . Melatonin triggers Crohn's disease symptoms. J Pineal Res . 2002; 32: 277– 278. Google Scholar CrossRef Search ADS PubMed  96. Jan JE, Freeman RD. Re: Mann–melatonin for ulcerative colitis? Am J Gastroenterol.  2003; 98: 1446. Google Scholar PubMed  97. Jan JE, O'Donnell ME. Use of melatonin in the treatment of paediatric sleep disorders. J Pineal Res . 1996; 21: 193– 199. Google Scholar CrossRef Search ADS PubMed  98. Mann S. Melatonin for ulcerative colitis? Am J Gastroenterol.  2003; 98: 232– 233. Google Scholar CrossRef Search ADS PubMed  99. Maldonado MD, Calvo JR. Melatonin usage in ulcerative colitis: a case report. J Pineal Res . 2008; 45: 339– 340. Google Scholar CrossRef Search ADS PubMed  100. Available at: http://clinicaltrials.gov/ct2/show/NCT00790478?term=melatonin+colitis&rank=1. Accessed September 10, 2013. 101. Bubenik GA. Localization of melatonin in the digestive tract of the rat. Effect of maturation, diurnal variation, melatonin treatment and pinealectomy. Horm Res . 1980; 12: 313– 323. Google Scholar CrossRef Search ADS PubMed  102. Ranjbaran Z, Keefer L, Farhadi A, et al.  . Impact of sleep disturbances in inflammatory bowel disease. J Gastroenterol Hepatol . 2007; 22: 1748– 1753. Google Scholar CrossRef Search ADS PubMed  103. Tang Y, Preuss F, Turek FW, et al.  . Sleep deprivation worsens inflammation and delays recovery in a mouse model of colitis. Sleep Med . 2009; 10: 597– 603. Google Scholar CrossRef Search ADS PubMed  104. Chen M, Mei Q, Xu J, et al.  . Detection of melatonin and homocysteine simultaneously in ulcerative colitis. Clin Chim Acta . 2012; 413: 30– 33. Google Scholar CrossRef Search ADS PubMed  105. Poon AM, Chow PH, Mak AS, et al.  . Autoradiographic localization of 2[125I]iodomelatonin binding sites in the gastrointestinal tract of mammals including humans and birds. J Pineal Res . 1997; 23: 5– 14. Google Scholar CrossRef Search ADS PubMed  106. Witt-Enderby PA, Bennett J, Jarzynka MJ, et al.  . Melatonin receptors and their regulation: biochemical and structural mechanisms. Life Sci . 2003; 72: 2183– 2198. Google Scholar CrossRef Search ADS PubMed  107. Natarajan R, Einarsdottir E, Riutta A, et al.  . Melatonin pathway genes are associated with progressive subtypes and disability status in multiple sclerosis among Finnish patients. J Neuroimmunol . 2012; 250: 106– 110. Google Scholar CrossRef Search ADS PubMed  108. Xia Q, Chen ZX, Wang YC, et al.  . Association between the melatonin receptor 1B gene polymorphism on the risk of type 2 diabetes, impaired glucose regulation: a meta-analysis. PLoS One.  2012; 7: e50107. Google Scholar CrossRef Search ADS PubMed  109. Issa JP. Epigenetic variation and human disease. J Nutr . 2002; 132: 2388S– 2392S. Google Scholar PubMed  110. Itoh K, Wakabayashi N, Katoh Y, et al.  . Keap1 represses nuclear activation of antioxidant responsive elements by Nrf2 through binding to the amino-terminal Neh2 domain. Genes Dev . 1999; 13: 76– 86. Google Scholar CrossRef Search ADS PubMed  111. Forrest CM, Mackay GM, Stoy N, et al.  . Inflammatory status and kynurenine metabolism in rheumatoid arthritis treated with melatonin. Br J Clin Pharmacol . 2007; 64: 517– 526. Google Scholar CrossRef Search ADS PubMed  112. Kensler TW, Wakabayashi N, Biswal S. Cell survival responses to environmental stresses via the Keap1-Nrf2-ARE pathway. Annu Rev Pharmacol Toxicol . 2007; 47: 89– 116. Google Scholar CrossRef Search ADS PubMed  113. Lee JM, Johnson JA. An important role of Nrf2-ARE pathway in the cellular defense mechanism. J Biochem Mol Biol . 2004; 37: 139– 143. Google Scholar PubMed  114. Arisawa T, Tahara T, Shibata T, et al.  . The relationship between Helicobacter pylori infection and promoter polymorphism of the Nrf2 gene in chronic gastritis. Int J Mol Med . 2007; 19: 143– 148. Google Scholar PubMed  115. Braun S, Hanselmann C, Gassmann MG, et al.  . Nrf2 transcription factor, a novel target of keratinocyte growth factor action which regulates gene expression and inflammation in the healing skin wound. Mol Cell Biol . 2002; 22: 5492– 5505. Google Scholar CrossRef Search ADS PubMed  116. Chen XL, Dodd G, Thomas S, et al.  . Activation of Nrf2/ARE pathway protects endothelial cells from oxidant injury and inhibits inflammatory gene expression. Am J Physiol Heart Circ Physiol . 2006; 290: H1862– H1870. Google Scholar CrossRef Search ADS PubMed  117. Chen XL, Varner SE, Rao AS, et al.  . Laminar flow induction of antioxidant response element-mediated genes in endothelial cells. A novel anti-inflammatory mechanism. J Biol Chem . 2003; 278: 703– 711. Google Scholar CrossRef Search ADS PubMed  118. Cho HY, Jedlicka AE, Reddy SP, et al.  . Role of NRF2 in protection against hyperoxic lung injury in mice. Am J Respir Cell Mol Biol . 2002; 26: 175– 182. Google Scholar CrossRef Search ADS PubMed  119. Papp D, Lenti K, Modos D, et al.  . The NRF2-related interactome and regulome contain multifunctional proteins and fine-tuned autoregulatory loops. FEBS Lett . 2012; 586: 1795– 1802. Google Scholar CrossRef Search ADS PubMed  120. Khor TO, Huang MT, Kwon KH, et al.  . Nrf2-deficient mice have an increased susceptibility to dextran sulfate sodium-induced colitis. Cancer Res . 2006; 66: 11580– 11584. Google Scholar CrossRef Search ADS PubMed  121. Lee SH, Sohn DH, Jin XY, et al.  . 2′,4′,6′-tris(methoxymethoxy) chalcone protects against trinitrobenzene sulfonic acid-induced colitis and blocks tumor necrosis factor-alpha-induced intestinal epithelial inflammation via heme oxygenase 1-dependent and independent pathways. Biochem Pharmacol . 2007; 74: 870– 880. Google Scholar CrossRef Search ADS PubMed  122. Osburn WO, Karim B, Dolan PM, et al.  . Increased colonic inflammatory injury and formation of aberrant crypt foci in Nrf2-deficient mice upon dextran sulfate treatment. Int J Cancer . 2007; 121: 1883– 1891. Google Scholar CrossRef Search ADS PubMed  123. Crespo I, Miguel BS, Laliena A, et al.  . Melatonin prevents the decreased activity of antioxidant enzymes and activates nuclear erythroid 2-related factor 2 signaling in an animal model of fulminant hepatic failure of viral origin. J Pineal Res . 2010; 49: 193– 200. Google Scholar PubMed  124. de Haan JB. Nrf2 activators as attractive therapeutics for diabetic nephropathy. Diabetes . 2011; 60: 2683– 2684. Google Scholar CrossRef Search ADS PubMed  125. Harvey CJ, Thimmulappa RK, Sethi S, et al.  . Targeting Nrf2 signaling improves bacterial clearance by alveolar macrophages in patients with COPD and in a mouse model. Sci Transl Med.  2011; 3: 78ra32. Google Scholar CrossRef Search ADS PubMed  126. Jung KH, Hong SW, Zheng HM, et al.  . Melatonin downregulates nuclear erythroid 2-related factor 2 and nuclear factor-kappaB during prevention of oxidative liver injury in a dimethylnitrosamine model. J Pineal Res . 2009; 47: 173– 183. Google Scholar CrossRef Search ADS PubMed  127. Atreya R, Neurath MF. Involvement of IL-6 in the pathogenesis of inflammatory bowel disease and colon cancer. Clin Rev Allergy Immunol . 2005; 28: 187– 196. Google Scholar CrossRef Search ADS PubMed  128. Lin WW, Karin M. A cytokine-mediated link between innate immunity, inflammation, and cancer. J Clin Invest . 2007; 117: 1175– 1183. Google Scholar CrossRef Search ADS PubMed  129. Mudter J, Neurath MF. Il-6 signaling in inflammatory bowel disease: pathophysiological role and clinical relevance. Inflamm Bowel Dis . 2007; 13: 1016– 1023. Google Scholar CrossRef Search ADS PubMed  130. Andoh A, Shioya M, Nishida A, et al.  . Expression of IL-24, an activator of the JAK1/STAT3/SOCS3 cascade, is enhanced in inflammatory bowel disease. J Immunol . 2009; 183: 687– 695. Google Scholar CrossRef Search ADS PubMed  131. Bai A, Hu P, Chen J, et al.  . Blockade of STAT3 by antisense oligonucleotide in TNBS-induced murine colitis. Int J Colorectal Dis . 2007; 22: 625– 635. Google Scholar CrossRef Search ADS PubMed  132. Li F, Zou Y, Li X. Up-regulation of signal transducer and activator of transcription-3 is associated with aggravation of ulcerative colitis. Surgeon . 2010; 8: 262– 266. Google Scholar CrossRef Search ADS PubMed  133. Yang XO, Panopoulos AD, Nurieva R, et al.  . STAT3 regulates cytokine-mediated generation of inflammatory helper T cells. J Biol Chem . 2007; 282: 9358– 9363. Google Scholar CrossRef Search ADS PubMed  134. Beriou G, Bradshaw EM, Lozano E, et al.  . TGF-beta induces IL-9 production from human Th17 cells. J Immunol . 2010; 185: 46– 54. Google Scholar CrossRef Search ADS PubMed  135. Bettelli E, Korn T, Oukka M, et al.  . Induction and effector functions of T(H)17 cells. Nature . 2008; 453: 1051– 1057. Google Scholar CrossRef Search ADS PubMed  136. Korn T, Bettelli E, Oukka M, et al.  . IL-17 and Th17 Cells. Annu Rev Immunol . 2009; 27: 485– 517. Google Scholar CrossRef Search ADS PubMed  137. Brand S, Beigel F, Olszak T, et al.  . IL-22 is increased in active Crohn's disease and promotes proinflammatory gene expression and intestinal epithelial cell migration. Am J Physiol Gastrointest Liver Physiol . 2006; 290: G827– G838. Google Scholar CrossRef Search ADS PubMed  138. Dambacher J, Beigel F, Zitzmann K, et al.  . The role of the novel Th17 cytokine IL-26 in intestinal inflammation. Gut . 2009; 58: 1207– 1217. Google Scholar CrossRef Search ADS PubMed  139. Fujino S, Andoh A, Bamba S, et al.  . Increased expression of interleukin 17 in inflammatory bowel disease. Gut . 2003; 52: 65– 70. Google Scholar CrossRef Search ADS PubMed  140. Seiderer J, Elben I, Diegelmann J, et al.  . Role of the novel Th17 cytokine IL-17F in inflammatory bowel disease (IBD): upregulated colonic IL-17F expression in active Crohn's disease and analysis of the IL17F p.His161Arg polymorphism in IBD. Inflamm Bowel Dis . 2008; 14: 437– 445. Google Scholar CrossRef Search ADS PubMed  141. Miossec P, Korn T, Kuchroo VK. Interleukin-17 and type 17 helper T cells. N Engl J Med . 2009; 361: 888– 898. Google Scholar CrossRef Search ADS PubMed  142. Sutton C, Brereton C, Keogh B, et al.  . A crucial role for interleukin (IL)-1 in the induction of IL-17-producing T cells that mediate autoimmune encephalomyelitis. J Exp Med . 2006; 203: 1685– 1691. Google Scholar CrossRef Search ADS PubMed  143. Veldhoen M, Hocking RJ, Atkins CJ, et al.  . TGFbeta in the context of an inflammatory cytokine milieu supports de novo differentiation of IL-17-producing T cells. Immunity . 2006; 24: 179– 189. Google Scholar CrossRef Search ADS PubMed  144. Wu WS, Chou MT, Chao CM, et al.  . Melatonin reduces acute lung inflammation, edema, and hemorrhage in heatstroke rats. Acta Pharmacol Sin . 2012; 33: 775– 782. Google Scholar CrossRef Search ADS PubMed  145. Fina D, Sarra M, Fantini MC, et al.  . Regulation of gut inflammation and th17 cell response by interleukin-21. Gastroenterology . 2008; 134: 1038– 1048. Google Scholar CrossRef Search ADS PubMed  146. Monteleone G, Monteleone I, Fina D, et al.  . Interleukin-21 enhances T-helper cell type I signaling and interferon-gamma production in Crohn's disease. Gastroenterology . 2005; 128: 687– 694. Google Scholar CrossRef Search ADS PubMed  147. Haigis MC, Mostoslavsky R, Haigis KM, et al.  . SIRT4 inhibits glutamate dehydrogenase and opposes the effects of calorie restriction in pancreatic beta cells. Cell . 2006; 126: 941– 954. Google Scholar CrossRef Search ADS PubMed  148. Sauve AA, Wolberger C, Schramm VL, et al.  . The biochemistry of sirtuins. Annu Rev Biochem . 2006; 75: 435– 465. Google Scholar CrossRef Search ADS PubMed  149. Yeung F, Hoberg JE, Ramsey CS, et al.  . Modulation of NF-kappaB-dependent transcription and cell survival by the SIRT1 deacetylase. EMBO J . 2004; 23: 2369– 2380. Google Scholar CrossRef Search ADS PubMed  150. Escande C, Chini CC, Nin V, et al.  . Deleted in breast cancer-1 regulates SIRT1 activity and contributes to high-fat diet-induced liver steatosis in mice. J Clin Invest . 2010; 120: 545– 558. Google Scholar CrossRef Search ADS PubMed  151. Pfluger PT, Herranz D, Velasco-Miguel S, et al.  . Sirt1 protects against high-fat diet-induced metabolic damage. Proc Natl Acad Sci U S A . 2008; 105: 9793– 9798. Google Scholar CrossRef Search ADS PubMed  152. Singh UP, Singh NP, Singh B, et al.  . Resveratrol (trans-3,5,4′-trihydroxystilbene) induces silent mating type information regulation-1 and down-regulates nuclear transcription factor-kappaB activation to abrogate dextran sulfate sodium-induced colitis. J Pharmacol Exp Ther . 2010; 332: 829– 839. Google Scholar CrossRef Search ADS PubMed  153. Chang HM, Wu UI, Lan CT. Melatonin preserves longevity protein (sirtuin 1) expression in the hippocampus of total sleep-deprived rats. J Pineal Res . 2009; 47: 211– 220. Google Scholar CrossRef Search ADS PubMed  154. Gutierrez-Cuesta J, Tajes M, Jimenez A, et al.  . Evaluation of potential pro-survival pathways regulated by melatonin in a murine senescence model. J Pineal Res . 2008; 45: 497– 505. Google Scholar CrossRef Search ADS PubMed  155. Tajes M, Gutierrez-Cuesta J, Ortuno-Sahagun D, et al.  . Anti-aging properties of melatonin in an in vitro murine senescence model: involvement of the sirtuin 1 pathway. J Pineal Res . 2009; 47: 228– 237. Google Scholar CrossRef Search ADS PubMed  156. Zhang J. Are poly(ADP-ribosyl)ation by PARP-1 and deacetylation by Sir2 linked? Bioessays.  2003; 25: 808– 814. Google Scholar CrossRef Search ADS PubMed  157. Pillai JB, Isbatan A, Imai S, et al.  . Poly(ADP-ribose) polymerase-1-dependent cardiac myocyte cell death during heart failure is mediated by NAD+ depletion and reduced Sir2alpha deacetylase activity. J Biol Chem . 2005; 280: 43121– 43130. Google Scholar CrossRef Search ADS PubMed  158. Hwang JW, Chung S, Sundar IK, et al.  . Cigarette smoke-induced autophagy is regulated by SIRT1-PARP-1-dependent mechanism: implication in pathogenesis of COPD. Arch Biochem Biophys . 2010; 500: 203– 209. Google Scholar CrossRef Search ADS PubMed  159. Jijon HB, Churchill T, Malfair D, et al.  . Inhibition of poly(ADP-ribose) polymerase attenuates inflammation in a model of chronic colitis. Am J Physiol Gastrointest Liver Physiol . 2000; 279: G641– G651. Google Scholar CrossRef Search ADS PubMed  160. Lennie TA, McCarthy DO, Keesey RE. Body energy status and the metabolic response to acute inflammation. Am J Physiol . 1995; 269: R1024– R1031. Google Scholar PubMed  161. Le Page C, Sanceau J, Drapier JC, et al.  . Inhibitors of ADP-ribosylation impair inducible nitric oxide synthase gene transcription through inhibition of NF kappa B activation. Biochem Biophys Res Commun . 1998; 243: 451– 457. Google Scholar CrossRef Search ADS PubMed  162. Zingarelli B, Salzman AL, Szabo C. Genetic disruption of poly (ADP-ribose) synthetase inhibits the expression of P-selectin and intercellular adhesion molecule-1 in myocardial ischemia/reperfusion injury. Circ Res . 1998; 83: 85– 94. Google Scholar CrossRef Search ADS PubMed  163. Zingarelli B, Szabo C, Salzman AL. Blockade of Poly(ADP-ribose) synthetase inhibits neutrophil recruitment, oxidant generation, and mucosal injury in murine colitis. Gastroenterology . 1999; 116: 335– 345. Google Scholar CrossRef Search ADS PubMed  164. Los M, Mozoluk M, Ferrari D, et al.  . Activation and caspase-mediated inhibition of PARP: a molecular switch between fibroblast necrosis and apoptosis in death receptor signaling. Mol Biol Cell . 2002; 13: 978– 988. Google Scholar CrossRef Search ADS PubMed  165. Watson AJ. Review article: manipulation of cell death–the development of novel strategies for the treatment of gastrointestinal disease. Aliment Pharmacol Ther . 1995; 9: 215– 226. Google Scholar CrossRef Search ADS PubMed  166. Esposito E, Paterniti I, Mazzon E, et al.  . Melatonin reduces hyperalgesia associated with inflammation. J Pineal Res . 2010; 49: 321– 331. Google Scholar CrossRef Search ADS PubMed  167. Fischer TW, Zmijewski MA, Wortsman J, et al.  . Melatonin maintains mitochondrial membrane potential and attenuates activation of initiator (casp-9) and effector caspases (casp-3/casp-7) and PARP in UVR-exposed HaCaT keratinocytes. J Pineal Res . 2008; 44: 397– 407. Google Scholar CrossRef Search ADS PubMed  168. Yoo YM, Yim SV, Kim SS, et al.  . Melatonin suppresses NO-induced apoptosis via induction of Bcl-2 expression in PGT-beta immortalized pineal cells. J Pineal Res . 2002; 33: 146– 150. Google Scholar CrossRef Search ADS PubMed  169. Aguilar-Quesada R, Munoz-Gamez JA, Martin-Oliva D, et al.  . Interaction between ATM and PARP-1 in response to DNA damage and sensitization of ATM deficient cells through PARP inhibition. BMC Mol Biol.  2007; 8: 29. Google Scholar CrossRef Search ADS PubMed  170. Mehendale HM. Once initiated, how does toxic tissue injury expand? Trends Pharmacol Sci.  2012; 33: 200– 206. Google Scholar CrossRef Search ADS PubMed  171. Hickey RC, Tidrick RT, Layton JM. Fulminating ulcerative colitis with colonic wall necrosis. Arch Surg . 1963; 86: 764– 771. Google Scholar CrossRef Search ADS PubMed  172. Cuzzocrea S, McDonald MC, Mazzon E, et al.  . Calpain inhibitor I reduces colon injury caused by dinitrobenzene sulphonic acid in the rat. Gut . 2001; 48: 478– 488. Google Scholar CrossRef Search ADS PubMed  173. D'Argenio G, Mazzone G, Tuccillo C, et al.  . Apple polyphenols extract (APE) improves colon damage in a rat model of colitis. Dig Liver Dis . 2012; 44: 555– 562. Google Scholar CrossRef Search ADS PubMed  174. Haapamaki MM, Gronroos JM, Nurmi H, et al.  . Gene expression of group II phospholipase A2 in intestine in ulcerative colitis. Gut . 1997; 40: 95– 101. Google Scholar CrossRef Search ADS PubMed  175. Yamaguchi O, Sugimura K, Ishizuka K, et al.  . Correlation between serum phospholipase A(2) IIA levels and histological activity in patients with ulcerative colitis. Int J Colorectal Dis . 2002; 17: 311– 316. Google Scholar CrossRef Search ADS PubMed  176. Ray SK, Samantaray S, Smith JA, et al.  . Inhibition of cysteine proteases in acute and chronic spinal cord injury. Neurotherapeutics . 2011; 8: 180– 186. Google Scholar CrossRef Search ADS PubMed  177. Samantaray S, Sribnick EA, Das A, et al.  . Melatonin attenuates calpain upregulation, axonal damage and neuronal death in spinal cord injury in rats. J Pineal Res . 2008; 44: 348– 357. Google Scholar CrossRef Search ADS PubMed  178. Kinouchi Y, Hiwatashi N, Chida M, et al.  . Telomere shortening in the colonic mucosa of patients with ulcerative colitis. J Gastroenterol . 1998; 33: 343– 348. Google Scholar CrossRef Search ADS PubMed  179. Risques RA, Lai LA, Brentnall TA, et al.  . Ulcerative colitis is a disease of accelerated colon aging: evidence from telomere attrition and DNA damage. Gastroenterology . 2008; 135: 410– 418. Google Scholar CrossRef Search ADS PubMed  180. Da-Silva N, Arasaradnam R, Getliffe K, et al.  . Altered mRNA expression of telomere binding proteins (TPP1, POT1, RAP1, TRF1 and TRF2) in ulcerative colitis and Crohn's disease. Dig Liver Dis . 2010; 42: 544– 548. Google Scholar CrossRef Search ADS PubMed  181. Usselmann B, Newbold M, Morris AG, et al.  . Deficiency of colonic telomerase in ulcerative colitis. Am J Gastroenterol . 2001; 96: 1106– 1112. Google Scholar CrossRef Search ADS PubMed  182. Masi S, Salpea KD, Li K, et al.  . Oxidative stress, chronic inflammation, and telomere length in patients with periodontitis. Free Radic Biol Med . 2011; 50: 730– 735. Google Scholar CrossRef Search ADS PubMed  183. Xu Q, Parks CG, DeRoo LA, et al.  . Multivitamin use and telomere length in women. Am J Clin Nutr . 2009; 89: 1857– 1863. Google Scholar CrossRef Search ADS PubMed  184. Aust DE, Haase M, Dobryden L, et al.  . Mutations of the BRAF gene in ulcerative colitis-related colorectal carcinoma. Int J Cancer . 2005; 115: 673– 677. Google Scholar CrossRef Search ADS PubMed  185. Takeshima H, Ikegami D, Wakabayashi M, et al.  . Induction of aberrant trimethylation of histone H3 lysine 27 by inflammation in mouse colonic epithelial cells. Carcinogenesis . 2012; 33: 2384– 2390. Google Scholar CrossRef Search ADS PubMed  186. Halili MA, Andrews MR, Sweet MJ, et al.  . Histone deacetylase inhibitors in inflammatory disease. Curr Top Med Chem . 2009; 9: 309– 319. Google Scholar CrossRef Search ADS PubMed  187. de Zoeten EF, Wang L, Sai H, et al.  . Inhibition of HDAC9 increases T regulatory cell function and prevents colitis in mice. Gastroenterology . 2010; 138: 583– 594. Google Scholar CrossRef Search ADS PubMed  188. Hasler R, Feng Z, Backdahl L, et al.  . A functional methylome map of ulcerative colitis. Genome Res . 2012; 22: 2130– 2137. Google Scholar CrossRef Search ADS PubMed  189. Arasaradnam RP, Khoo K, Bradburn M, et al.  . DNA methylation of ESR-1 and N-33 in colorectal mucosa of patients with ulcerative colitis (UC). Epigenetics . 2010; 5: 422– 426. Google Scholar CrossRef Search ADS PubMed  190. Glauben R, Batra A, Fedke I, et al.  . Histone hyperacetylation is associated with amelioration of experimental colitis in mice. J Immunol . 2006; 176: 5015– 5022. Google Scholar CrossRef Search ADS PubMed  191. Korkmaz A, Reiter RJ. Epigenetic regulation: a new research area for melatonin? J Pineal Res.  2008; 44: 41– 44. Google Scholar PubMed  192. Niles LP, Pan Y, Kang S, et al.  . Melatonin induces histone hyperacetylation in the rat brain. Neurosci Lett . 2013; 541: 49– 53. Google Scholar CrossRef Search ADS PubMed  193. Li Volti G, Musumeci T, Pignatello R, et al.  . Antioxidant potential of different melatonin-loaded nanomedicines in an experimental model of sepsis. Exp Biol Med (Maywood) . 2012; 237: 670– 677. Google Scholar CrossRef Search ADS PubMed  194. Gupta AS, Kshirsagar SJ, Bhalekar MR, et al.  . Design and development of liposomes for colon targeted drug delivery. J Drug Target . 2013; 21: 146– 160. Google Scholar CrossRef Search ADS PubMed  195. Kong H, Lee Y, Hong S, et al.  . Sulfate-conjugated methylprednisolone as a colon-targeted methylprednisolone prodrug with improved therapeutic properties against rat colitis. J Drug Target . 2009; 17: 450– 458. Google Scholar CrossRef Search ADS PubMed  196. Varshosaz J, Ahmadi F, Emami J, et al.  . Colon delivery of budesonide using solid dispersion in dextran for the treatment and secondary prevention of ulcerative colitis in rat. Int J Prev Med . 2010; 1: 115– 123. Google Scholar PubMed  Copyright © 2013 Crohn's & Colitis Foundation of America, Inc. TI - A Review of the Use of Melatonin in Ulcerative Colitis: Experimental Evidence and New Approaches JO - Inflammatory Bowel Diseases DO - 10.1097/01.MIB.0000436962.32164.6e DA - 2014-03-01 UR - https://www.deepdyve.com/lp/oxford-university-press/a-review-of-the-use-of-melatonin-in-ulcerative-colitis-experimental-NZmUOp49m0 SP - 553 EP - 563 VL - 20 IS - 3 DP - DeepDyve ER -