Please cite this paper as:

Pal, P.K., Chattopadhyay, A. and Bandyopadhyay, D. 2021. Functional interplay of melatonin in the bile duct and gastrointestinal tract to mitigate disease development: An overview. Melatonin Research. 4, 1 (Jan. 2021), 118-140. DOI:https://doi.org/https://doi.org/10.32794/mr11250086.


Review

 

Functional interplay of melatonin in the bile duct and gastrointestinal tract to mitigate disease development: An overview

Palash Kumar Pala, Aindrila Chattopadhyayb, Debasish Bandyopadhyaya*

aOxidative Stress and Free Radical Biology Laboratory, Department of Physiology, University of Calcutta, 92, APC Road, Kolkata-700009

bDepartment of Physiology, Vidyasagar College, 39, Sankar Ghosh Lane, Kolkata-700006

*Correspondence:debasish63@gmail.com,Tel: +91 9433072066

Running title: Biliary and gastrointestinal melatonin

Received: October 12, 2020; Accepted: December 14, 2020


ABSTRACT

     Prevalence of bile duct and gastrointestinal (GI) tract associated diseases is increasing globally. Commonly, the bile duct epithelial cell (cholangiocytes) malfunction and its uncontrolled proliferation often cause liver fibrosis and tumorigenesis, particularly the cholangiocarcinoma. Specifically, GI tract is constantly under diverse endogenous and exogenous stressors which interrupt GI physiological functions and promote inflammation, tissue damage, ulceration, gastrointestinal bleeding, gastroesophageal reflux disease (GERD), irritable bowel disease (IBD) and gastritis. On the other hand, melatonin exhibits important functions in both cholangiocyte and GI tract. The abundance of melatonin generated in the GI tract and its widely distributed receptors facilitate its protective effects in GI tissues. In the most of the cases, the disease progression in GI tract, particularly in bile duct, is associated with endogenous melatonergic system suppression. Therefore, to increase the endogenous melatonin production appears a suitable strategy to retard the disease development in these tissues. Melatonin administration or, exposure to prolonged darkness not only reverse the detrimental biochemical alterations, but also inhibit cholangiocyte proliferation as well as ulceration in the GI tract. Thus, use of melatonin as a natural therapeutic agent is beneficial and exhibits advantages over other contemporary drugs in prevention and treatment of bile duct and gastrointestinal tract associated diseases.

Key words: Bile duct, cholangiocytes, gastrointestinal tract, diseases, melatonin, prevention.

_____________________________________________________________________________________________________________ 

1. INTRODUCTION

     In humans, interruption of bile metabolism leads to cholestasis that is considered as a hallmark of different liver disorders (1). Over production of particular bile acids will cause structural damage, inflammation, fibrosis and tumorigenesis in hepatic tissue, and induce bile duct proliferation as well (2). Thus, it is important to understand the functionality of the biliary system during any pathogenesis. It is well known that the response of cholangiocytes, epithelial cells lining the intra- and extrahepatic bile duct, plays an important role during the development of various biliary system associated disorders, such as cholestatic liver diseases (3, 4). Although the exact mechanism of cholangiopathy is not understood to date, a preliminary ductular response induced biliary hyperplasia is known to trigger apoptosis, ductopenia and fibrosis (5). Normally, cholangiocytes remain quiescent but any sort of stress or tissue injury in the liver will activate the proliferative characteristics of the cholangiocytes. This response is regulated by a complex network of different proteins and transcription factors (6, 7). This ductular reaction is actually a critical interplay between cholangiocytes and different liver cells during the development of cirrhosis (4). Physiologically, gastrointestinal (GI) tract is the primary battle ground since pathogens directly interact with its epithelial lining to trigger inflammatory reaction (8). The reactive oxygen species (ROS) generated by pathogens or internal stressors can cause numerous pathophysiological alterations including malignancy (8). In addition, GI damage can also be caused by several other factors such as heavy metals (9, 10), ischemia-reperfusion (11), alcohol consumption (12), non-steroidal anti-inflammatory drugs (NSAIDs) (13, 14) and high fat diet (8, 15). Notably, high level of bile acids can cause gastroesophageal reflux disease (GERD) by the mechanism of elevated ROS (16). Bile acids are also the strong cyclooxygenase-2 (COX-2) inducers in case of Barrett’s esophagus and esophageal adenocarcinoma with increased ROS production and activated PA3K/AKT and ERK1/2 pathways (17). These important observations clearly suggest the possible functional interplay between the biliary system and GI tract during pathogenesis.

     Several approaches have been adopted so far to overcome these pathophysiological conditions related to the biliary system in GI tract. These include use of some neuro-endocrine hormones, neurotransmitters, and growth factors which show promising effects in inhibiting the detrimental changes induced by the stressors (18-20). Among these, melatonin is considered as the most promising agent due to its lipophilic nature, paracrine actions, broad spectrum antioxidant and direct free radical scavenging properties (21-26). High levels of melatonin presence in bile (27) and GI tract (25, 28, 29) of mammals including humans (30) provide a unique advantage to overcome many pathological conditions in these tissues. In this review, we will focus on the beneficial effects of melatonin in combating against biliary and GI pathogeneses in mammals.

 

2. STRUCTURAL FEATURES OF BILIARY EPITHELIUM AND ITS DUCTAL SYSTEM

     Among the different hepatic epithelial cells, hepatocytes and cholangiocytes play the critical roles in the regulation of biliary secretion and intra-hepatic functional homeostasis (3, 31). Anatomically, the intra-hepatic bile ductal apparatus forms the tubular structures that are interconnected and lined by diverse cholangiocytes (3, 32, 33). In humans, the intra-hepatic biliary epithelium has been classified into six categories (33) based on the diameter of the bile duct- (i) small bile ductules (<15 μm), (ii) intra-lobular ducts (15-100 μm), (iii) septal ducts (100-300 μm), (iv) area ducts (300-400 μm), (v) segmental ducts (400-800 μm) and (vi) hepatic ducts (<800 μm). However, the biliary epithelium of rodent has been classified into two types- (a) large (>15 μm) bile ducts that are lined by large cholangiocytes while (b) small (<15 μm) bile ducts that are lined by smaller ones (34-36). The intra-hepatic cholangiocytes comprise 3-5% of the entire liver cell population (3). In addition to cholangiocyte-regulated autocrine loop, the branches of hepatic artery form a complex mesh of minute vessels known as the peri-biliary vascular plexus (PBP) that secretes numerous growth factors, such as vascular endothelial growth factor (VEGF), are responsible for the regulation of the bile duct system (37, 38). Hepatic sinusoids are directly supplied through the lobular branches or, indirectly via the pre-lobular portal vein branches (37, 39). Usually, PBP is profoundly distributed around the large bile ducts but is present in lesser number in the smaller bile ducts (37).

     At this point it is important to discuss the path of progression of the secreted bile. Initially, bile is secreted into the canalicular region from where it travels through the Hering canals to reach to the small ductules (31). Eventually, large bile ducts collect the canalicular bile by re-absorption and secretion (35, 40, 41) that are under the control of different parasympathetic, sympathetic and dopaminergic innervations as well as numerous gastrointestinal hormones (for example secretin, gastrin and somatostatin) (35, 40-45). Interestingly, the properties of such re-absorption and secretion related to the biliary epithelium vary according to the size of the bile ducts and its position in the biliary tree (34, 35, 41, 46). For instance, among the diverse hepatic cell types, secretin receptor (SR), cystic fibrosis trans-membrane conductance regulator (CFTR), somatostatin receptor subtype 2 (SSTR2), and Cl-/HCO3- anion exchanger AE2 are solely found in the large colangiocytes (34, 35, 42, 47); this explains the possible cause of why large cholangiocytes can only respond to secretin mediated stimuli to regulate the release of water and electrolytes (34, 35, 42, 47). In contrast, small cholangiocytes within the small ducts does not express any of these receptors or transporter proteins and thereby remains inactive to secretin and somatostatin stimulations (34, 35, 42, 47).

 

3. FUNCTIONAL DIVERSITY OF CHOLANGIOCYTES

     The size variation of cholangiocytes actually reflects its functional diversity in mammals (34, 48). The large cholangiocytes which are responsible for the secretion of water and bicarbonate are dependent on 3', 5'-cyclic adenosine monophosphate (cAMP) regulation (34). In contrast, small cholangiocytes are inositol trisphosphate/Ca2+ dependent ones. Under normal condition, these small cholangiocytes serve as a silent progenitor which then, can differentiate into large cholangiocytes after cellular injury (48). The active transportations of glutathione and organic anions such as bilirubin glucuronide into the canaliculus, a phenomenon known as bile acid-independent bile flow (49), occur in large cholangiocytes since they possess secretin specific receptors (SRs) on their baso-lateral membranes (40, 50). Secretin binds to SR to elevate the intracellular levels of cAMP which in turn, activates cystic fibrosis trans-membrane conductance regulator (CFTR). These events result in the leakage of Cl- to extracellular side causing activation of the Cl-/HCO3- exchanger AE2, thus triggering bicarbonate-rich choleresis; however, Cl- is restored in cholangiocytes (40).

Apart from its crucial physiological roles, cholangiocytes also exhibit potent protective effects against the diverse biliary disorders including primary sclerosing cholangitis (3), and symptoms associated with fibrosis and cirrhosis (4). Otherwise, the malfunctioned cholangiocytes will promote biliary hyperplasia which in turn causes apoptosis, ductopenia, and fibrosis (5).

 

4. INJURY INDUCED MODULATION OF CHOLANGIOCYTE’S FUNCTIONS

     During the initial stages of injury, sensitivity of cholangiocytes to a variety of neuroendocrine peptides (secretin), neurotransmitters (histamine), growth factors [nerve growth factor and vascular endothelial growth factor (VEGF)] and hormones [follicle-stimulating hormone (FSH), estrogen and progesterone] increases in a neuroendocrine like manner that eventually triggers biliary proliferation, liver inflammation and fibrosis (6, 7, 38, 51-55). Some of these transdifferentiation inducing factors are discussed herein. In case of cholestatic hepatic injury, proliferative response of cholangiocytes occurs due to the suppression of the hypothalamic-pituitary-adrenal axis. This is indicated by the increased hypothalamic bile acid signalling, suggesting the involvement of other neural hormones in the regulation of cholangiocyte mediated actions (56, 57).

     Abundance of GnRH receptors (GnRHR1 and GnRHR2) found in cholangiocytes indicates regulatory role of GnRH in these cells (58). This is also supported by the observation that bile duct ligation (BDL) increases the levels of GnRH in the cholangiocytes and triggers their proliferation (58). Generally, GnRH level varies between 0.05-0.06 ng/mL in the supernatant of normal mouse cholangiocyte cell line (58). Treatment with recombinant GnRH enhanced intrahepatic bile duct mass due to increasing cholangiocyte proliferation (57). Several molecules can modify the proliferation of cholangiocytes. For example, melatonin can regulate cholangicyte proliferation by suppressing the expression of hypothalamic GnRH through different protein kinase pathways (59-60). Sex hormones such as FSH are also involved in biliary hyperplasia possibly by triggering proliferation of cholangiocyte, increasing ductal mass and intracellular levels of cAMP, inducing phosphorylation of both ETS domain-containing protein and membrane bound kinases (61). Another important factor is pancreatic duodenal homeobox-1 which is over expressed in the isolated cholangiocytes of BDL rat. In pancreatic duodenal homeobox-1 deficient mice, their bile duct mass, VEGF and insulin-like growth factor 1 are reduced which are biomarkers of cholangiocyte proliferation (62). In addition, secretin/secretin receptor axis also plays potent roles in regulating cholangiocyte responses (63). Physical damage to cholangiocyte suppresses the expression of SR in large cholangiocyte, whereas basal cholangiocyte proliferation is completely inhibited by SR knockdown (54). These findings indicate the importance of secretin/SR as an essential tropic regulator of biliary growth in mammals (54). Histamine receptors are other regulators for cholangiocyte proliferation. All four types of histamine receptors (H1HR, H2HR, H3HR and H4HR) are present in cholangiocyte. H3HR and H4HR exhibit inhibitory while H1HR and H2HR exhibit stimulatory actions to maintain the balance of cholangiocyte proliferation (64-67). Notably, application of H3HR agonist decreased BDL-induced cholagiocyte hyperplasia in rats by targeting the cAMP signalling along with phosphorylation of ERK1/2 and transcription factor E1k1 pathway (64). In contrast, activation of H1HR induces proliferation of small cholangiocytes through IP3/Ca2+/PKC/CaMKI/CREB-dependent pathway (64). Application of H4HR agonist (clobenpropit) restricts cholangiocarcinoma growth and metastatic potential through activation of a Ca2+/PKC-dependent pathway and by reducing the expression of matrix metalloproteinases (67). Noteworthy, VEGF and its receptors (VEGFR-2 and VEGFR-3) are present in cholangiocytes. In BDL animals, both VEGF level and VEGF receptor expression are enhanced with increased proliferation which are mediated by activation of IP3/Ca2+/PKCα and Src/ERK1/2 signalling (68). Both subtypes of estrogen receptor (ER-α and ER-β) are also expressed in cholangiocytes to facilitate the proliferative and secretory activities of cholangiocytes. In BDL animals, expressions of ER-a and ER-b were upregulated from 3 to 30 folds, respectively, compared to the controls (69). This is also observed in the in vitro study, in which 17β estradiol stimulates cholangiocyte proliferation (69). Usually, the nuclear (PR-B) and membrane (PRGMC1, PRGMC2, and mPRα) progesterone receptors are expressed in cholangiocytes. Progesterone exerts its autocrine/paracrine actions on cholangiocyte proliferation (70). In BDL rat, antiprogesterone antibodies efficiently restricted the proliferation of cholangiocytes (70).

 

5. BILIARY SYSTEM ASSOCIATED DISORDERS

     In diverse cholangiopathy models, extrahepatic bile duct ligation (BDL), acute carbon tetrachloride (CCl4) treatment and chronic gamma-aminobutyric acid (GABA) administration are a few of them which alter the secretory and proliferative activities of cholangiocytes (48, 61, 71). Under these treatments, cholangiocytes usually lose their differentiation capability and reduce sensitivity to hormone intervention with increased biliary apoptosis (48, 61, 71). When an injury occurs in large bile ducts, the small cholangiocytes amplifies Ca2+-dependent signalling pathways to restore the biliary epithelium followed by Ca2+/CaMK I-dependent adenylyl cyclase 8 activation and appearance of large cholangiocyte phenotype (48, 61, 71). On the other hand, cholangiopathies such as primary biliary cholangitis (PBC), primary sclerosing cholangitis (PSC) and cholangiocarcinoma (CCA), if not handled properly, will cause liver inflammation, ultimately hepatic fibrosis (72, 73). For example, PBC and PSC associated bile duct proliferation, commonly termed as the ductular reaction (DR), is an essential step of disease development where non-functional truncated bile ducts invade into the parenchymal region of liver through the portal areas (52, 74, 75). Suppression of DR is linked with reduced liver fibrosis. This might be a collective resultant of declined response in biliary secretory phenotypes and hepatic stellate cell activation factors (76). Bile acid malabsorption (BAM) induced chronic diarrhoea is another area of concern since the absorbed chenodeoxycholic and deoxycholic acids, major components of dihydroxy bile acid, are known to restrain sodium absorption in the colon but triggers chloride secretion, ultimately leading to diarrhoea (77).

     Several other physiological factors are also involved in biliary system complications. For example, excess production of mitochondrial reactive oxygen species (mROS) in extrahepatic cholestatic patients causes oxidative damage to different mitochondrial components, such as unsaturated fatty acids, proteins and DNA (78). Glycochenodeoxycholic acid (GCDCA), the predominant compound in the bile, seems to be responsible for the development of extrahepatic cholestasis (79) where lipid peroxidation of the mitochondrial membrane plays the pivotal role (79, 80).

    

6. GASTROINTESTINAL TRACT ASSOCIATED DISORDERS

     As mentioned above, GI tract is always under the endogenous and exogenous insults (23, 25, 26, 81). For example, the endogenously produced adrenaline, due to its rapid pro-oxidant and auto-oxidation properties (82-84), promotes oxidative stress and proinflammatory cytokines including necrosis factor-kappa beta (NF-kβ), tumour necrosis factor-α (TNF-α), interleukin-1 beta (IL-1β) and interleukin-6 (IL6) in the stomach, duodenum and colon tissues (25). The environmentally extracted heavy metals such as arsenic (As), lead (Pb) and mercury (Hg) also cause detrimental changes in the GI tract (10, 85, 86). These metals bind to sulfhydryl group of enzymes to interrupt their functions and increase the production of superoxide anion, hydrogen peroxide and hydroxyl radical which in turn elevates peroxidation of membrane lipids and apoptosis (86). The monomethylarginine (MMA) compounds, the products of interaction of As and thiol group of nitric oxide synthases, cause diverse GI symptoms termed as arsenicosis (86). Similarly, Hg causes gastric ulceration and hemorrhage (85), abdominal pain, vomiting and bloody diarrhoea (87). Pb is another disease promoting agent in the GI tract (88, 89). Pb mimics some co-factors, such as calcium, iron and zinc, to catalyze different intracellular enzymatic reactions and activities of varied pro-oxidant enzymes (90) to increase the threat of stomach cancer in humans (10).

     The non-steroidal anti-inflammatory drugs (NSAIDs) can also cause diverse pathogenesis in the GI tract (8, 91). Basically, NSAIDs promote generation of ROS and inhibit mucosal blood flow, collectively to trigger the gastric ulceration and inflammation (92, 93). NSAIDs, such as aspirin, indomethacin and ibuprofen inhibit prostaglandin production and cellular differentiation possibly by exhaustion of ATP supply due to disruption of mitochondrial transmembrane potential and increasing permeability of transition pore. The dysfunctional mitochondria generate more hydrogen peroxide and hydroxyl radical leading to oxidative damage in the gastric mucosa (92, 94-97). Indomethacin and piroxicam cause irreversible inactivation of gastric peroxidases, downregulate antioxidant enzymes and increase lipid peroxidation, thus promoting over production of ROS and ulcerative symptoms in the GI tissues (98, 99). Ischemia reperfusion (I/R) also induces severe inflammation and tissue injury in the mammalian GI tract (11). Profound accumulation of activated neutrophils and over production of ROS during reperfusion further exacerbates the GI injuries (100, 101). The sepsis and ulceration are also noted in patients with ischemic colitis (102). Noteworthy, consumption of high fat diet (HFD) induces inflammation in mammalian GI tract (15) possibly by altering the intestinal levels of chylomicrons and gut microbiota sub-colonies (103). Similarly, alcohol consumption also affects the intestinal microbiome and inhibits bile acid metabolism (104), triggers over production of hydrogen peroxide and carbon-centred radicals which in turn lead to endothelial dysfunction, microcirculatory disturbance and ischemia of gastric mucosa (21). Moreover, some bacteria, such as Helicobacter pylori, are the culprits of peptic ulcer disease, gastroesophageal reflux disease (GERD), irritable Bowel disease (IBD) and gastritis. These pathogens activate non-phagocytic NADPH oxidase to promote proinflammatory responses, oxidative damage to the gastric epithelial pit cells (8, 105, 106). The development of duodenal ulcer and severe duodenitis have been observed in humans infected by H. pylori (107).

 

7. MELATONIN: A POTENT THERAPEUTIC MOLECULE

     The protective effect of melatonin on biliary system disorders were observed in the BDL study in which pinealectomy and prolonged exposure to light exacerbate biliary damage and liver fibrosis in rats. Both treatments reduce melatonin level, and thus, increase ROS and inflammation with upregulation of expression of CLOCK, ARNTL, Cry1, and Per1, and miR-200b clock genes in cholangiopathies (108). The potent antioxidant and cellular permissible properties lead melatonin having advantages over other antioxidants in protecting cholangiocytes oxidative damage (21, 109, 110). Melatonin upregulates antioxidant enzymes and downregulates pro-oxidant enzyme expressions to exhibit its indirect antioxidant actions (22, 24, 25). This action is mediated by the activation of its membrane bound receptors (MT1R, MT2R and MT3R) (111). In addition, its metabolites Nl-acetyl-N2-formyl-5-methoxykynuramineAFMK, N1-acetyl-5-methoxy-kynuramine (AMK) and cyclic 3-hydroxylmelatonin (C3HM) are also known to exert strong antioxidant and anti-inflammatory activities (112). Melatonin also maintains the cellular energy homeostasis by targeting mitochondria and prevents bile-acid induced oxidative damage in the hepatocytes through modulation of the AMPK-SIRT3-SOD2 pathway in liver (25, 113-116). All these contribute to the beneficial effects of melatonin on cholestatic liver injury (117, 118).

 

8. MELATONINAND ITS METABOLISM

     Melatonin is mainly metabolized in the liver through a two-step mechanism. First, cytochrome P450 mono-oxygenases hydroxylates C6 position forming 6-hydroxymelatonin; then, 6-hydroxymelatonin conjugates with (i) sulphate catalyzed by the sulphotransferase to form 6-sulphomelatonin or, (ii) glucuronic acid catalyzed by UDP-glucuronosultransferase to form 6-hydroxymelatonin glucuronide (119). In the second catabolic pathway, indolamine-2,3-dioxygenase (2,3-IOD) or myeloperoxidase catalyze melatonin forming AFMK which is later transformed to a more stable compound AMK (120, 121). Noteworthy, melatonin is also demonstrated to be metabolized non-enzymatically by the interaction with different ROS and RNS (21).

 

9. PRESENCE OF MELATONIN IN THE GI TRACT AND BILIARY SYSTEM

     Concentration of melatonin in the bile is much higher than that in other organs/tissues. Melatonin levels in bile vary (ranging from 2,000 to 11,000 pg/ml) among the different mammalian species (27). Highest melatonin levels in bile were detected in rabbit (>10,000 pg/ml) followed by monkey (>8,000 pg/ml), human (>6,000 pg/ml) and rat (~4,000 pg/ml) (27). Such high level of melatonin in the human bile has protective effects on liver damage (122). Melatonin concentrations in the gastrointestinal tract are also relatively high. For example, the level is 467 ± 99 pg/g tissue in human (30) and 461± 29 pg/g tissue in rat (25). There are some variations in different tissues including 136 ± 27 pg/100 mg in the GI mucosa and 243 ± 37 pg/100 mg in the stomach and descending colon, respectively (123). A study has reported a relative low melatonin level in bile (85 ± 45 pg/ml) and this may be due to its excretion into the GI tract via the bile duct (123). Immunolocalization and autoradiographic studies revealed the presence of melatonin in the enterochromaffin cells (EC) of the mucosa and villi of the gastrointestinal tract (28, 29, 124, 125). The discovery of melatonin synthesis rate-limiting enzymes, hydroxyindole-O-methyltransferase (HIOMT) and aralkylamine N-acetyltransferase (AANAT) in the EC cells confirm the de novo melatonin synthesis in the EC cells of the GI tract (25, 80, 110, 126, 127).

 

10. FUNCTIONAL RELEVANCE OF MELATONIN IN THE BILIARY TRACT

     As yet cholangiopathies are considered as the prime cue for the development of hepatic failure and death in humans (128). Melatonin seems to be a suitable agent in the management of chronic cholestatic liver diseases because of its regulatory capability to maintain biliary homeostasis and to inhibit collagen deposition in the hepatic tissue (129).

     Melatonin at a dose of 750 μg/kg BW/day suppressed the levels of transaminases, lipid peroxidation and glutathione in BDL rats and its protective efficiency was found to be superior to S-adenosyl-methionine (SAME, 10 mg/kg BW/day) (130). Melatonin receptor MT1 expression increased in the cholangiocytes isolated from the bile ducts of cholestatic rodents and melatonin (20 mg/L in drinking water) reduced serum transaminase and bilirubin and ductular reaction in the cholangiocytes (20). Melatonin treatment also inhibited the expression of clock genes, and levels of protein kinase A (PKA) phosphorylation and cAMP and these effects are probably mediated by its membrane receptor MT1 (20). In a separate experiment, upregulation of AANAT and increase in melatonin secretion were observed in isolated cholangiocytes of BDL rats (131).

On the other hand, natural light regimen was also applied to evaluate the role of melatonin through brain-liver axis in preventing the development of different cholangiopathies (76, 132). In order to upregulate the pineal secretion of melatonin, BDL rats and Mdr2-/- [possesses some characteristics of human primary sclerosing cholangitis (PSC)] mice are kept under complete darkness for 1 week. The results indicate that this treatment not only increases the melatonin levels in the pineal and circulation, but also improves the morphology of the hepatic tissue by decreasing collagen deposition and diminishes ductular reaction as well. Moreover, expressions of miR-200b, different clock genes, PER1, BMAL1, CLOCK and Cry1 are also declined (76, 133). The in vivo and in vitro studies on human PSC samples reveal similar responses when the studies are carried out in the complete darkness or, with melatonin treatment (76).

In contrast, circulating level of melatonin is found to be diminished in BDL cholestatic rats when they are subjected to pinealectomy or prolonged light exposure, accordantly, serum level of different hepatic enzymes, ductular reaction, and tissue levels of inflammation, angiogenesis, fibrosis, miR-200b and ROS generation are significantly increased (108). If the BDL rats are given intracerebroventricular (ICV) infusion of melatonin, ductular reaction and fibrosis of hepatic tissue are significantly reduced. These may relate to the suppressive effects of melatonin on GnRH expression and its secretion from the cholangiocytes along with a reduction in its receptor expression (133). Such observations are important as both biliary proliferation and fibrosis of liver are known to be triggered through GnRH and its receptor mediated pathway (134). Based on these evidences it may be assumed that enhancing pineal level of melatonin is beneficial in resisting the progression of cholestatic liver diseases.

 

11. FUNCTIONAL INTERPLAY BETWEEN BILIARY AND GASTROINTESTINAL MELATONI

     EC cells synthesize melatonin. A fraction of this melatonin is metabolized by cytochrome CYP1B1 in the enterocytes (135), a maximum amount (90%) of this melatonin enters the hepatic portal vein (136, 137) to reach the liver where it is metabolized (138) by hepatocytes by the cytochrome P-450 enzymes forming 6-sulfatoxymelatonin and 6-hydroxymelatonin glucuronide to be eliminated from the body through urine (139-141). However, some melatonin is released in the bile (27, 123) to prevent the epithelial lining of the bile and GI tracts from unwanted injuries induced by bile acids. Hepatic metabolic disturbances are also found to modulate the circulating levels of melatonin and its circadian rhythm (142-144). Many other instances have been reported to date that clearly suggested the possible functional correlation between the melatonin levels in the bile and GI tract (145).

 

12. MELATONIN PREVENTS GASTROINTESTINAL DISEASES

     Gastrointestinal tract seems to be the richest source of endogenous melatonin in mammals where it regulates numerous local physiological actions mostly through its paracrine action (25, 26). In case of heavy metal toxicity, such as Cd-induced neurotoxicity melatonin exerts its protective role by activating transcription factor EB associated autophagic pathway (146) and restoring the equilibrium between mitochondrial fusion and fission (78, 147). The beneficial effects of melatonin against numerous heavy metal induced tissue damages have been frequently reported (148) and quite a few are dealt with GI tract. For example, exogenous administration of melatonin in rat prevents stomach and duodenum against Pb-induced oxidative injuries (149). Similar ameliorative actions of melatonin have also been reported against different NSAIDs, such as indomethacin, aspirin and piroxicam, induced intestinal tissue injuries (93, 119, 150-152). In case of diclofenac induced gastric damages, application of melatonin also restores mitochondrial membrane potential and energy metabolism, thus decreases intestinal permeability and maintains mucosal integrity (153, 154). The protection lies in the potentiality of melatonin in activating complex I and IV and inhibiting electron leakage, ultimately promoting mitochondrial respiration and ATP synthesis (97, 155, 156). Generally, melatonin is known to exert its gastro-protective efficacy by upregulating activities of different antioxidant enzymes, such as gastric peroxidise, SOD and catalase (157, 158). During GI ischemia-reperfusion, activation of MT2 receptor upregulates matrix metalloproteinase-2 (MMP-2) but downregulates MMP-9 and tissue inhibitor of metalloproteinases-2 (TIMP-2) to inhibit the oxidative tissue injury (159, 160). Apart from it, melatonin also prohibits gastric acid secretion and neutrophil infiltration and increases mucosal blood flow, duodenal level of bicarbonate secretion and prostaglandin production to inhibit intestinal tissue damage (161-163).

     In GI disorders, such as GERD, use of melatonin and L-tryptophan (precursor of melatonin synthesis) as dietary supplement leads to regression of GERD symptoms (164). In water immersion and restrain stressed animal models, both melatonin and L-tryptophan protect GI tissue injury by increasing nitric oxide production and mucosal microcirculation (150, 165). Intragastric administration of melatonin (20 mg/kg) or tryptophan (100 mg/kg) caused a reduction in the lipid peroxidation level, while increasing the levels of SOD and GSH that clearly indicated the antioxidant role of melatonin (92). Melatonin also exerts its protective role against a number of stressors that induce serious gastric ulcers in mammals. In pig, application of melatonin (2.5 mg/kg/feed) in diet profoundly decreased the incidence of gastric ulcer (166). Since administration of melatonin or L-tryptophan upregulates MT2 receptor gene expression; therefore, the protective effects are attributed to its MT2 mediated pathways and the enhancement of melatonin in ulcer bed speeds up the healing process (167). Combination of melatonin and ranitidine or omeprazole can produce synergic effects for the treatment of diverse types of gastric ulcers (157, 167, 168). Use of melatonin (20 mg per day) or tryptophan (500 mg per day) in H. Pylori induced gastric ulcer patients promoted their healing process (167). These may contribute to melatonin’s antioxidant activity (22, 109, 111, 169, 170).

     Interestingly, the metabolites of melatonin interaction with ROS also exert strong antioxidant functions thus amplifying the restoration process of the damages in GI tissues (171-173). Melatonin can also act on the melatonin receptors of the mast cells to produce its anti-inflammatory and immunomodulatory actions through inhibition of TNFα release, prostaglandin production and NF-κB expression (174-176). Similar anti-inflammatory (177) and antioxidant (178) roles of melatonin (25 mg/kg) are also observed in cerulein-induced acute pancreatitis in rats. The detailed mechanistic pathways have been described in Figure 1.

Figure 1.jpg

Fig. 1. The protective pathways of melatonin against GI disorders.

     TF: Transcription factor; AOG: Antioxidant genes; IAG: Inflammation associated genes; MTR: Melatonin receptor (MT1 and MT2). ROS; Reactive oxygen species.

 

13. MELATONIN PREVENTS BILIARY TRACT DISORDERS

     Current studies related to beneficial effects of melatonin on cholangiocytes are focused on the endogenous melatonergic system (130, 179). Decrease in AANAT expression and melatonin secretion in cholangiocytes of BDL rat causes biliary proliferation and increased activity of ductal secretion (130). In contrast, overexpression of AANAT restricts the proliferation of cholangiocytes (130). The α-naphthylisothiocyanate (ANIT), a potent biliary tissue toxin, induces hepatotoxicity which is prevented by oral melatonin (180, 181). The downregulation of AANAT/ASMT in patients with cholangiocarcinoma strongly suggests use of melatonin as a possible remedial agent (179, 182). As expected, treatment with melatonin inhibits the growth of Opisthorchis viverrini and N-nitrosodimethylamine-induced cholangiocarcinoma (179, 182). The potential mechanisms are summarized in Figure 2.

Figure2.jpg

Fig. 2. The protective pathways of melatonin on disorders of bile duct epithelial cell (cholangiocyte).

     MTR: Melatonin receptor (MT1 and MT2); ROS: Reactive oxygen species; GnRH: Gonadotrophin releasing hormone.

     In addition to its peripheral effect, the role of melatonin on hypothalamic GnRH is important to combat cholangiocyte associated diseases since (i) GnRH secretion by cholangiocyte induces biliary proliferation and fibrosis (58, 131), (ii) melatonin can inhibit the activity of GnRH and suppress its secretion (183, 184), and (iii) exposing BDL rats to prolong darkness increases melatonin level in the cholangiocyte and reduces its proliferation (20, 131). Thus, the central suppressive effect of melatonin on secretion and expression of GnRH in the cholangiocyte will inhibit biliary proliferation and disorders. Actually, melatonin can inhibit GnRH secretion of hypothalamic cell in rat through modulation of different intracellular signalling pathways such as protein kinase A (PKA), protein kinase C (PKC) and mitogen-activated protein kinase (MAPK) (59, 60). Similar antiproliferative action of melatonin in the cholangiocytes is also observed in different human CCA cell lines (Mz-ChA-1, HuH-28, TFK-1, CCLP1, SG231, and HUCC-T1) (179, 185). Study in rat hepatoma 7288CTC cells revealed that melatonin administration inhibited mitogenic signal for linoleic acid-dependent tumor growth by suppressing the absorption of linoleic acid and its further metabolism (186). Similar anticarcinogenic properties of melatonin are also documented in several human studies (187, 188).

     The protective effects of melatonin on hepatotoxic compounds (30, 189, 190), ionizing radiation (191), cold storage and reperfusion (192-194), and carbon tetrachloride (195) are well documented. These protections are mediated either by its receptor dependent or receptor independent anti-inflammatory and antioxidant actions (196-200).

    

14. FUTURE PERSPECTIVES

     Based on the above discussion it is clear that the functional relationship between melatonin levels in the bile duct and GI tract are important to prevent GI disorders. Thereby, use of melatonin as a natural therapeutic agent is beneficial and has advantages to other contemporary drugs as to treat biliary disorders since it lacks obvious side effects. Biliary proliferation induced by GnRH seems to be a key step in initiation of biliary pathology; therefore, the inhibition of GnRH secretion will be a target for future drug development. Melatonin is a molecule to inhibit the GnRH secretion. Thus, melatonin alone or, as co-treatment with other medicines may be a future selection to combat diverse GI disorders.


ACKNOWLEDGEMENTS

     Dr. Palash Kumar Pal gratefully acknowledges the receipt of UGC Dr. D. S. Kothari Post Doctoral Fellowship (BL/16-17/0502), Govt. of India. Dr. Aindrila Chattopadhyay is supported by funds available to her from Department of Science and Technology, Govt. of West Bengal. Prof. DB is also supported from departmental BI grant of University of Calcutta. DB also gratefully acknowledges the support he received from DST-PURSE Program awarded to University of Calcutta. Prof. DB gratefully acknowledges the contribution of the Editor-In-Chief of Melatonin Research, Dr. Dun-Xian Tan in critically reading and editing of the manuscript which has definitely improved the scientific and readership quality of the article.


AUTHORSHIP

     Dr. DB contributed to conception, revised the manuscript critically and approved it. Dr. AC revised the manuscript critically and approved it. Dr. PKP contributed to conception, prepared figures, drafted the manuscript and edited it.


CONFLICT OF INTEREST

     Authors declare no conflict of interest.

 

REFERENCES

 

  1. Trauner M, Meier PJ, Boyer JL (1998) Molecular pathogenesis of cholestasis. N. Engl. J. Med. 339 (17): 1217-27. doi: 10.1056/NEJM199810223391707.

  2. Pollheimer MJ, Fickert P, Stieger B (2014) Chronic cholestatic liver diseases: clues from histopathology for pathogenesis. Mol. Aspects Med. 37: 35-56. doi: 10.1016/j.mam.2013. 10.001.

  3. Alpini G, McGill JM, Larusso NF (2002) The pathobiology of biliary epithelia. Hepatology 35: 1256–1268. doi:10.1053/jhep.2002.33541.

  4. Williams MJ, Clouston AD, Forbes SJ (2014) Links between hepatic fibrosis, ductular reaction, and progenitor cell expansion. Gastroenterology 146: 349–356. doi:10.1053/j.gastro.2013.11.034.

  5. Lazaridis KN, Strazzabosco M, Larusso NF (2004) The cholangiopathies: disorders of biliary epithelia. Gastroenterology 127: 1565–1577. doi:10.1053/j.gastro.2004.08.006.

  6. Franchitto A, Onori P, Renzi A, et al. (2013) Recent advances on the mechanisms regulating cholangiocyte proliferation and the significance of the neuroendocrine regulation of cholangiocyte pathophysiology. Ann. Transl. Med. 1: 27. doi:10.3978/ j.issn.2305-5839.2012.10.03.

  7. Maroni L, Haibo B, Ray D, et al. (2015) Functional and structural features of cholangiocytes in health and disease. Cell. Mol. Gastroenterol. Hepatol. 1: 368–380. doi:10.1016/j.jcmgh.2015.05.005.

  8. Bhattacharyya A, Chattopadhyay R, Mitra S, et al. (2014) Oxidative stress: an essential factor in the pathogenesis of gastrointestinal mucosal diseases. Physiol. Rev. 94: 329–354. doi:10.1152/physrev.00040.2012.

  9. Zalups RK (2000) Molecular interactions with mercury in the kidney. Pharmacol. Rev. 52: 113-143.

  10. Koh DH, Bhatti P, Coble JB, et al. (2014) Calibrating a population-based job-exposure matrix using inspection measurements to estimate historical occupational exposure to lead for a population-based cohort in Shanghai, China. J. Exp. Sci. Env. Epid. 24: 9–16. doi: 10.1038/jes.2012.86.

  11. Victoni T, Coelho FR, Soares AL, et al. (2010) Local and remote tissue injury upon intestinal ischemia and reperfusion depends on the TLR/MyD88 signaling pathway. Med. Microbiol. Immunol. 199: 35–42.

  12. Beckman JS, Koppenol WH (1996) Nitric oxide, superoxide and peroxynitrite: the good, the bad and the ugly. Am. J. Physiol. 271: C1424–C1437. doi: 10.1152/ajpcell.1996. 271.5.c1424.

  13. Pal PK, Bhattacharjee B, Chattopadhyay A, et al. (2019) Melatonin as an armament against non-steroidal anti-inflammatory drug (NSAID) induced gastric injury: An overview. Melatonin Res. 2: 116-138. doi: 10.32794/mr11250015.

  14. Wongrakpanich S, Wongrakpanich A, Melhado K, et al. (2018) A comprehensive review of non-steroidal anti-inflammatory drug use in the elderly. Aging Dis. 9: 143-150. http://dx.doi.org/10.14336/AD.2017.0306.

  15. Fritsche KL (2015) The science of fatty acids and inflammation. Adv. Nutr. 6: 293S–301S. doi: 10.3945/an.114.006940.

  16. Wild CP, Hardie LJ (2003) Reflux, Barrett’s oesophagus and adenocarcinoma: burning questions. Nat. Rev. Cancer 3: 676–684.

  17. Song S, Guha S, Liu K, et al. (2007) COX-2 induction by unconjugated bile acids involves reactive oxygen species-mediated signalling pathways in Barrett’s oesophagus and oesophageal adenocarcinoma. Gut 56: 1512–1521.doi: 10.1136/gut.2007.121244.

  18. DeMorrow S, Francis H, Gaudio E, et al. (2008) The endocannabinoid anandamide inhibits cholangiocarcinoma growth via activation of the noncanonical Wnt signaling pathway. Am. J. Physiol. Gastrointest. Liver Physiol. 295: G1150–G1158. doi:10.1152/ajpgi.90455.2008.

  19. Marzioni M, Glaser S, Francis H, et al. (2004) Autocrine/paracrine regulation of the growth of the biliary tree by the neuroendocrine hormone serotonin. Gastroenterology 128: 121–137. doi:10.1053/j.gastro.2004.10.002.

  20. Renzi A, Glaser S, Demorrow S, et al. (2011) Melatonin inhibits cholangiocyte hyperplasia in cholestatic rats by interaction with MT1 but not MT2 melatonin receptors. Am. J. Physiol. Gastrointest. Liver. Physiol. 301: G634–G643. doi:10.1152/ajpgi.00206. 2011.

  21. Tan DX, Chen LD, Poeggeler B, et al. (1993) Melatonin: a potent, endogenous hydroxyl radical scavenger. Endocr. J. 1:57–60.

  22. Rodriguez C, Mayo JC, Sainz RM, et al. (2004) Regulation of antioxidant enzymes: A significant role for melatonin. J. Pineal Res. 36: 1–9.

  23. Pal PK, Hasan NK, Maitra SK (2016) Gut melatonin response to microbial infection in carp Catla catla. Fish Physiol. Biochem. 42: 579–592. doi: 10.1007/s10695-015-0161-7.

  24. Pal PK, Maitra SK (2018) Response of gastrointestinal melatonin, antioxidants, and digestive enzymes to altered feeding conditions in carp (Catla catla). Fish Physiol. Biochem. 44: 1061-1073. https://doi.org/10.1007/s10695-018-0494-0.

  25. Pal PK, Bhattacharjee B, Ghosh A, et al. (2019) Adrenaline induced disruption of endogenous melatonergic system, antioxidant and inflammatory responses in the gastrointestinal tissues of male Wistar rat: an in vitro study. Melatonin Res. 1: 109–131. doi: 10.32794/mr11250007.

  26. Pal PK, Bhattacharjee B, Chattopadhyay A, et al. (2019) Pleiotropic roles of melatonin against oxidative stress mediated tissue injury in the gastrointestinal tract: An overview. Melatonin Res. 2: 158-184. DOI: 10.32794/mr11250027.

  27. Tan DX, Manchester LC, Reiter RJ, et al. (1999) High physiological levels of melatonin in the bile of mammals. Life Sci. 65 (23): 2523-2529.

  28. Raikhlin NT, Kvetnoy IM (1974) Lightening effect of the extract of human appendix mucosa on frog skin melanophores. Bull. Exp. Biol. Med. 8: 114–116.

  29. Bubenik GA (1999) Localization and physiological significance of gastrointestinal melatonin. In: R Watson (eds.) Melatonin in health promotion. CRC press, Boca Raton, Florida, pp. 21–39.

  30. Chen CQ, Fichna J, Bashashati M, et al. (2011) Distribution, function and physiological role of melatonin in the lower gut. World J. Gastroenterol. 17: 3888-3898. doi: 10.3748/wjg.v17.i34.3888.

  31. Nathanson MH, Boyer JL (1991) Mechanisms and regulation of bile secretion. Hepatology 14: 551-566.

  32. Kanno N, LeSage G, Glaser S, et al. (2000) Functional heterogeneity of the intrahepatic biliary epithelium. Hepatology 31: 555-561.

  33. Ludwig J (1987) New concepts in biliary cirrhosis. Semin. Liver Dis.7: 293-301.

  34. Alpini G, Roberts S, Kuntz SM, et al. (1996) Morphological, molecular, and functional heterogeneity of cholangiocytes from normal rat liver. Gastroenterology 110: 1636-1643. doi: 10.1053/gast.1996.v110.pm8613073.

  35. Alpini G, Glaser S, Robertson W, et al. (1997) Large but not small intrahepatic bile ducts are involved in secretin-regulated ductal bile secretion. Am. J. Physiol. 272: G1064-G1074. doi: 10.1152/ajpgi.1997.272.5.G1064.

  36. Glaser SS, Gaudio E, Rao A, et al. (2009) Morphological and functional heterogeneity of the mouse intrahepatic biliary epithelium. Lab. Invest. 89: 456-469. doi: 10.1038/labinvest.2009.6.

  37. Gaudio E, Onori P, Pannarale L, et al. (1996) Hepatic microcirculation and peribiliary plexus in experimental biliary cirrhosis: a morphological study. Gastroenterology 111: 1118-124. doi: 10.1016/s0016-5085(96)70081-1.

  38. Gaudio E, Barbaro B, Alvaro D, et al. (2006) Administration of r-VEGF-A prevents hepatic artery ligation-induced bile duct damage in bile duct ligated rats. Am. J. Physiol. Gastrointest. Liver. Physiol. 291: G307-G317. doi: 10.1152/ajpgi.00507.2005.

  39. Terada T, Ishida F, Nakanuma Y (1989) Vascular plexus around intrahepatic bile ducts in normal livers and portal hypertension. J. Hepatol. 8: 139-149. doi: 10.1016/0168-8278(89)90001-9.

  40. Alpini G, Lenzi R, Sarkozi L, et al. (1988) Biliary physiology in rats with bile ductular cell hyperplasia. Evidence for a secretory function of proliferated bile ductules. J. Clin. Invest. 81: 569-578. doi: 10.1172/JCI113355.

  41. Kanno N, LeSage G, Glaser S, et al. (2001) Regulation of cholangiocyte bicarbonate secretion. Am. J. Physiol. Gastrointest. Liver. Physiol. 281: G612-G625. doi: 10.1152/ajpgi.2001.281.3.G612.

  42. Tietz PS, Alpini G, Pham LD, et al. (1995) Somatostatin inhibits secretin-induced ductal hypercholeresis and exocytosis by cholangiocytes. Am. J. Physiol. 269: G110-G118. doi: 10.1152/ajpgi.1995.269.1.G110.

  43. Alvaro D, Alpini G, Jezequel AM, et al. (1997) Role and mechanisms of action of acetylcholine in the regulation of rat cholangiocyte secretory functions. J. Clin. Invest. 100: 1349-1362. doi: 10.1172/JCI119655.

  44. LeSage GD, Alvaro D, Glaser S, et al. (2004) Alpha-1 adrenergic receptor agonists modulate ductal secretion of BDL rats via Ca(2+)- and PKC-dependent stimulation of cAMP. Hepatology 40: 1116-1127. doi: 10.1002/hep.20424.

  45. Glaser S, Alvaro D, Francis H, et al. (2006) Adrenergic receptor agonists prevent bile duct injury induced by adrenergic denervation by increased cAMP levels and activation of Akt. Am. J. Physiol. Gastrointest. Liver Physiol. 290: G813-G826. doi: 10.1152/ajpgi.00306.2005.

  46. Han Y, Glaser S, Meng F, et al. (2013) Recent advances in the morphological and functional heterogeneity of the biliary epithelium. Exp. Biol. Med. (Maywood) 238: 549-565. doi: 10.1177/1535370213489926.

  47. Banales JM, Arenas F, Rodríguez-Ortigosa CM, et al. (2006) Bicarbonate-rich choleresis induced by secretin in normal rat is taurocholate-dependent and involves AE2 anion exchanger. Hepatology 43: 266-275. doi: 10.1002/hep.21042.

  48. LeSage GD, Glaser SS, Marucci L, et al. (1999) Acute carbon tetrachloride feeding induces damage of large but not small cholangiocytes from BDL rat liver. Am. J. Physiol. 276: G1289-G1301. doi: 10.1152/ajpgi.1999.276.5.G1289.

  49. Lanzini A (2003) Bile. In: Trugo L and Finglas PM (Eds.) Encyclopedia of food sciences and nutrition (2nd edition). pp. 471-478. https://doi.org/10.1016/B0-12-227055-X/00095-X.

  50. Alpini G, Ulrich CD, 2nd, Phillips JO, et al. (1994) Upregulation of secretin receptor gene expression in rat cholangiocytes after bile duct ligation. Am. J. Physiol. Gastrointest. Liver Physiol. 266: G922-G928. doi: 10.1152/ajpgi.1994.266.5.G922.

  51. Gigliozzi A, Alpini G, Baroni GS, et al. (2004) Nerve growth factor modulates the proliferative capacity of the intrahepatic biliary epithelium in experimental cholestasis. Gastroenterology 127: 1198–1209. doi:10.1053/j.gastro.2004.06.023.

  52. Alvaro D, Mancino MG, Glaser S, et al. (2007) Proliferating cholangiocytes: a neuroendocrine compartment in the diseased liver. Gastroenterology 132: 415-431. https://doi.org/10.1053/j.gastro.2006.07.023.

  53. Mancinelli R, Onori P, Gaudio E, et al. (2009) Follicle-stimulating hormone increases cholangiocyte proliferation by an autocrine mechanism via cAMP-dependent phosphorylation of ERK1/2 and Elk-1. Am. J. Physiol. Gastrointest. Liver Physiol. 297: G11-G26. doi: 10.1152/ajpgi.00025.2009.

  54. Glaser S, Lam IP, Franchitto A, et al. (2010) Knockout of secretin receptor reduces large cholangiocyte hyperplasia in mice with extrahepatic cholestasis induced by bile duct ligation. Hepatology 52: 204–214. doi:10.1002/hep.23657.

  55. Francis HL, Demorrow S, Franchitto A, et al. (2012) Histamine stimulates the proliferation of small and large cholangiocytes by activation of both IP3/Ca2+ and cAMP-dependent signaling mechanisms. Lab. Invest. 92: 282–294. doi:10.1038/ labinvest.2011.158.

  56. Quinn M, Ueno Y, Pae HY, et al. (2012) Suppression of the HPA axis during extrahepatic biliary obstruction induces cholangiocyte proliferation in the rat. Am. J. Physiol. Gastrointest. Liver Physiol. 302: G182–G193. doi:10.1152/ajpgi.00205.2011.

  57. McMillin M, Frampton G, Quinn M, et al. (2015) Suppression of the HPA axis during cholestasis can be attributed to hypothalamic bile acid signaling. Mol. Endocrinol. 29: 1720–1730. doi:10.1210/me.2015-1087.

  58. Ray D, Han Y, Franchitto A, et al. (2015) Gonadotropin-releasing hormone stimulates biliary proliferation by paracrine/autocrine mechanisms. Am. J. Pathol. 185: 1061– 1072. doi:10.1016/j.ajpath.2014.12.004.

  59. Roy D, Belsham DD (2002) Melatonin receptor activation regulates GnRH gene expression and secretion in GT1-7GnRH neurons. Signal transduction mechanisms. J. Biol. Chem. 277: 251–258. doi:10.1074/ jbc.M108890200.

  60. Schomerus C, Korf HW (2005) Mechanisms regulating melatonin synthesis in the mammalian pineal organ. Ann. NY. Acad. Sci. 1057: 372–383. doi:10.1196/annals. 1356.028.

  61. Mancinelli R, Franchitto A, Gaudio E, et al. (2010) After damage of large bile ducts by gamma-aminobutyric acid, small ducts replenish the biliary tree by amplification of calcium-dependent signaling and de novo acquisition of large cholangiocyte phenotypes. Am. J. Pathol. 176: 1790-1800. https://doi.org/10.2353/ajpath.2010.090677.

  62. Marzioni M, Saccomanno S, Candelaresi C, et al. (2010) Pancreatic Duodenal Homeobox-1 de novo expression drives cholangiocyte neuroendocrine-like trans differentiation. J. Hepatol. 53: 663–670. doi:10.1016/j.jhep.2010.04.022.

  63. Sato K, Meng F, Giang T, et al. (2018) Mechanisms of cholangiocyte responses to injury. Biochim. Biophys. Acta Mol. Basis Dis. 1864: 1262-1269. doi: 10.1016/j.bbadis.2017.06.017.

  64. Francis H, Franchitto A, Ueno Y, et al. (2007) H3 histamine receptor agonist inhibits biliary growth of BDL rats by downregulation of the cAMP-dependent PKA/ ERK1/2/ELK-1 pathway. Lab. Invest. 87: 473–487. https://doi.org/10.1038/labinvest. 3700533.

  65. Francis H, Glaser S, DeMorrow S, et al. (2008) Small mouse cholangiocytes proliferate in response to H1 histamine receptor stimulation by activation of the IP3/CAMK I/CREB pathway. Am. J. Physiol. Cell. Physiol. 295: C499–C513. doi: 10.1152/ajpcell. 00369.2007.

  66. Francis H, Onori P, Gaudio E, et al. (2009) H3 histamine receptor mediated activation of protein kinase C alpha inhibits the growth of cholangiocarcinoma in vitro and in vivo. Mol. Cancer Res. 7: 1704–1713. doi: 10.1158/1541-7786.MCR-09-0261.

  67. Meng F, Han Y, Staloch D, et al. (2011) The H4HR agonist, clobenpropit, suppresses human cholangiocarcinoma progression by disruption of EMT and tumor metastasis. Hepatology. 54 (5): 1718-1728. doi: 10.1002/hep.24573.

  68. Gaudio E, Barbaro B, Alvaro D, et al. (2006) Vascular endothelial growth factor stimulates rat cholangiocyte proliferation via an autocrine mechanism. Gastroenterology. 130 (4): 1270-1282. doi: 10.1053/j.gastro.2005.12.034.

  69. Alvaro D, Alpini G, Onori P, et al. (2000) Estrogens stimulate proliferation of intrahepatic biliary epithelium in rats. Gastroenterology 119 (6): 1681-1691.  https://doi.org/10.1053/gast.2000.20184.

  70. Glaser S, DeMorrow S, Francis H (2008) Progesterone stimulates the proliferation of female and male cholangiocytes via autocrine/paracrine mechanisms. Am. J. Physiol. Gastrointest. Liver Physiol. 295 (1): G124–G136. doi: 10.1152/ajpgi.00536.2007.

  71. Mancinelli R, Franchitto A, Glaser S, et al. (2013) GABA induces the differentiation of small into large cholangiocytes by activation of Ca(2+) /CaMK I-dependent adenylyl cyclase 8. Hepatology 58: 251-263. doi: 10.1002/hep.26308.

  72. Chapman R, Fevery J, Kalloo A, et al. (2010) Diagnosis and management of primary sclerosing cholangitis. Hepatology 51: 660-678. https://doi.org/10.1002/hep.23294.

  73. Lindor KD, Bowlus CL, Boyer J, et al. (2019) Primary biliary cholangitis: 2018 Practice guidance from the american association for the study of liver diseases. Hepatology 69: 394-419. https://doi.org/10.1002/hep.30145.

  74. Carpino G, Cardinale V, Folseraas T, et al. (2018) Hepatic stem/progenitor cell activation differs between primary sclerosing and primary biliary cholangitis. Am. J. Pathol. 188: 627-639. doi: 10.1016/j.ajpath.2017.11.010.

  75. Sato K, Marzioni M, Meng F, et al. (2019) Ductular reaction in liver diseases: pathological mechanisms and translational significances. Hepatology 69: 420-430. doi: 10.1002/hep.30150.

  76. Wu N, Kyritsi K, Invernizzi P, et al. (2017) Differential impact of MT1 and MT2 melatonin receptor deletion on biliary proliferation, senescence, and liver fibrosis duringcholestatic liver injury. Hepatology 66: A330.

  77. Mekjian HS, Phillips SF, Hofmann AF (1971) Colonic secretion of water and electrolytes induced by bile acids: perfusion studies in man. J. Clin. Invest. 50: 1569–1577. doi: 10.1172/JCI106644.

  78. Xu S, Pi H, Zhang L, et al. (2016) Melatonin prevents abnormal mitochondrial dynamics resulting from the neurotoxicity of cadmium by blocking calcium-dependent translocation of Drp1 to the mitochondria. J. Pineal Res. 60: 291-302. doi: 10.1111/jpi.12310.

  79. Perez MJ, Briz O (2009) Bile-acid-induced cell injury and protection. World J. Gastroenterol. 15(14): 1677-1689. doi: 10.3748/wjg.15.1677.

  80. Chen Y, Lv L, Jiang Z, et al. (2013) Mitofusin 2 protects hepatocyte mitochondrial function from damage induced by GCDCA. PLoS One 8 (6): e65455. https://doi.org/10.1371/journal.pone.0065455.

  81. Pal PK, Hasan NK, Maitra SK (2016) Temporal relationship between the daily profiles of gut melatonin, oxidative status and major digestive enzymes in carp Catla catla. Biol. Rhythm Res. 47: 755–771. doi: 10.1080/09291016.2016.1191697.

  82. Liu X, Wu W-K, Yu L, et al. (2008) Epinephrine stimulates esophageal squamous-cell carcinoma cell proliferation via beta adrenoceptor-dependent transactivation of extracellular signal-regulated kinase/cyclooxygenase-2 pathway. J. Cell Biochem. 105: 53–60. DOI: 10.1002/jcb.21802.

  83. Yao H, Duan Z, Wang M, et al. (2009) Adrenaline induces chemoresistance in HT-29 colon adenocarcinoma cells. Cancer Genet. Cytogenet. 190: 81–87. doi: 10.1016/j.cancergencyto.2008.12.009.

  84. Rudra S, Mukherjee D, Dutta M, et al. (2014) Orally administered melatonin protects against adrenaline-induced oxidative stress in rat liver and heart: Involvement of antioxidant mechanism(s). J. Pharm. Res. 8: 303–320. http://jprsolutions.info/files/final-file-56bff9adee9ad8.28733292.pdf.

  85. Langford NJ, Ferner RE (1999) Toxicity of mercury. J. Hum. Hypertens. 13: 651–656. https://doi.org/10.1038/sj.jhh.1000896.

  86. Shi H, Shi X, Liu KJ (2004) Oxidative mechanism of arsenic toxicity and carcinogenesis. Mol. Cell Biochem. 255: 67-78.

  87. Barnes JL, McDowell EM, McNeil JS (1980) Studies on the pathophysiology of acute renal failure. V. Effect of chronic saline loading on the progression of proximal tubular injury and functional impairment following administration of mercuric chloride in the rat. Virchows Arch. B Cell Pathol. Incl. Mol. Pathol. 32: 233–260.

  88. Henning SJ, Leeper LL (1984) Duodenal uptake of lead by suckling and weanling rats. Bioi. Neonate 46: 27-35. doi: 10.1159/000242029.

  89. Conrad ME, Barton JC (1978) Factors affecting the absorption and excretion of lead in the rat. Gastroenterol. 74: 731-740. doi: https://doi.org/10.1016/0016-5085(78)90253-6.

  90. Valko M, Morris H, Cronin MTD (2005) Metals, toxicity and oxidative stress. Curr. Med. Chem. 12: 1161–1208. doi: 10.2174/0929867053764635.

  91. Awtry EH, Loscalzo J (2000) Cardiovascular drugs. aspirin. Circulation 101: 1206-1218.

  92. Yoshikawa T, Naito Y, Kishi A, et al. (1993) Role of active oxygen, lipid peroxidation, and antioxidants in the pathogenesis of gastric mucosal injury induced by indomethacin in rats. Gut 34: 732-737. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1374252.

  93. Brzozowski T, Konturek PC, Konturek SJ, et al. (1997) The role of melatonin and L-tryptophan in prevention of acute gastric lesions induced by stress, ethanol, ischemia, and aspirin. J. Pineal Res. 23: 79-89. https://doi.org/10.1111/j.1600-079X.1997. tb00339.x.

  94. Banerjee RK (1990) Nonsteroidal anti-inflammatory drugs inhibit gastric peroxidase activity. Biochim. Biophys. Acta 1034: 275-280.

  95. Das D, Bandyopadhyay D, Bhattacharjee M, et al. (1997) Hydroxyl radical is the major causative factor in stress-induced gastric ulceration. Free Radic. Biol. Med. 23: 8-18. https://doi.org/10.1016/S0891-5849(96)00547-3.

  96. Naito Y, Yoshikawa T (2006) Oxidative stress Involvement and gene expression in indomethacin-induced gastropathy. Redox Rep. 11: 243–253. doi: 10.1179/135100006X 155021.

  97. Tan DX, Reiter RJ (2019) Mitochondria: the birth place, battle ground and the site of melatonin metabolism in cells. Melatonin Res. 2: 44-66. doi: https://doi.org/10.32794/ mr11250011.

  98. Bandyopadhyay D, Biswas K, Bandyopadhyay U, et al. (2000) Melatonin protects against stress-induced lesions by scavenging the hydroxyl radical. J. Pineal Res. 29: 143–151.

  99. Chattopadhyay I, Bandyopadhyay U, Biswas K, et al. (2006) Indomethacin inactivates gastric peroxidase to induce reactive-oxygen-mediated gastric mucosal injury and curcumin protects it by preventing peroxidise inactivation and scavenging reactive oxygen. Free Radic. Biol. Med. 40: 1397–1408. doi: 10.1016/j.freeradbiomed.2005. 12.016.

  100. Phull PS, Green CJ, Jacyna MR (1995) A radical view of the stomach: the role of oxygen-derived free radicals and anti-oxidants in gastroduodenal disease. Eur. J. Gastroenterol. Hepatol. 7: 265–274.

  101. Mallick IH, Yang W, Winslet MC, et al. (2004) Ischemia-reperfusion injury of the intestine and protective strategies against injury. Dig. Dis. Sci. 49: 1359–1377.

  102. Jarry A, Bach-Ngohou K, Masson D, et al. (2006) Human colonic myocytes are involved in post-ischemic inflammation through ADAM17-dependent TNF alpha production. Br. J. Pharmacol. 147: 64–72. doi: 10.1038/sj.bjp.0706449.

  103. de La Serre CB, Ellis CL, Lee J, et al. (2010) Propensity to high-fat diet induced obesity in rats is associated with changes in the gut microbiota and gut inflammation. Am. J. Physiol. Gastrointest. Liver Physiol. 299: G440–G448. doi: 10.1152/ajpgi.00098.2010.

  104. Schnabl B, Brenner DA (2014) Interactions between the intestinal microbiome and liver diseases. Gastroenterol. 146: 1513–1524. doi: 10.1053/j.gastro.2014.01.020.

  105. O’Hara AM, Bhattacharya A, Bai J, et al. (2006) Interleukin-8 induction by Helicobacter pylori in human gastric epithelial cells is dependent on a purinic/ apyrimidinic endonuclease-1/redoxfactor-1. J. Immunol. 177: 7990–7999.

  106. Ding SZ, Minohara Y, Fan XJ, et al. (2007) Helicobacter pylori infection induces oxidative stress and programmed cell death in human gastric epithelial cells. Infect. Immun. 75: 4030–4039. doi: 10.1128/IAI.00172-07.

  107. Davies GR, Simmonds NJ, Stevens TR, et al. (1992) Mucosal reactive oxygen metabolite production in duodenal ulcer disease. Gut 33: 1467–1472. doi: 10.1136/gut.33.11.1467.

  108. Chen L, Zhou T, Wu N, et al. (2019) Pinealectomy or light exposure exacerbates biliary damage and liver fibrosis in cholestatic rats through decreased melatonin synthesis. Biochim. Biophys. Acta Mol. Basis. Dis. 1865: 1525-1539. doi: 10.1016/j.bbadis. 2019.03.002.

  109. Reiter RJ, Mayo JC, Tan DX, et al. (2016) Melatonin as an antioxidant: under promises but over delivers. J. Pineal Res. 61: 253–278. doi: 10.1111/jpi.12360.

  110. Galano A, Tan DX, Reiter RJ (2018) Melatonin: a versatile protector against oxidative DNA damage. Molecules 27: 23. doi: 10.3390/molecules23030530.

  111. Dobocovich M, Mankowsha M (2005) Functional MP1 and MP2 melatonin receptors in mammals. Endocrine 27: 101–110. doi: 10.1385/ENDO:27:2:101.

  112. Tan DX, Manchester LC, Terron MP, et al. (2007) One molecule, many derivatives: a never-ending interaction of melatonin with reactive oxygen and nitrogen species? J. Pineal Res. 42 (1): 28-42. DOI: 10.1111/j.1600-079X.2006.00407.x.

  113. Li Y, Yang Y, Feng Y, et al. (2014) A review of melatonin in hepatic ischemia/reperfusion injury and clinical liver disease. Ann. Med. 46(7): 503-511. doi: 10.3109/07853890.2014.934275.

  114. Guo P, Pi H, Xu S, et al. (2014) Melatonin Improves mitochondrial function by promoting MT1/SIRT1/PGC-1 alpha-dependent mitochondrial biogenesis in cadmium-induced hepatotoxicity in vitro. Toxicol. Sci.142 (1):182-195. doi: 10.1093/toxsci/ kfu164.

  115. Mitra E, Bhattacharjee B, Pal PK, et al. (2019) Melatonin protects against cadmium-induced oxidative damage in different tissues of rat: a mechanistic insight. Melatonin Res. 2: 1-21. doi: 10.32794/mr11250018.

  116. Chen Y, Qing W, Sun M, et al. (2015) Melatonin protects hepatocytes against bile acid-induced mitochondrial oxidative stress via the AMPK-SIRT3-SOD2 pathway. Free Radic. Res. 49: 1275-1284. doi: 10.3109/10715762.2015.1067806.

  117. Reiter RJ, Rosales-Corral SA, Manchester LC, et al. (2014) Melatonin in the biliary tract and liver: health implications. Curr. Pharm. Des. 20 (30): 4788-4801.

  118. Tahan G, Akin H, Aydogan F, et al. (2010) Melatonin ameliorates liver fibrosis induced by bile-duct ligation in rats. Can. J. Surg. 53 (5): 313-318.

  119. Pandi-Perumal SR, Srinivasan V, Maestroni GJ, et al. (2006) Melatonin: Nature's most versatile biological signal? FEBS J. 273: 2813–2838. doi: 10.1111/j.1742-4658.2006.05322.x.

  120. Tan DX, Manchester LC, Reiter RJ, et al. (1998) Ischemia/reperfusion‐induced arrhythmias in the isolated rat heart: Prevention by melatonin. J. Pineal Res. 25: 184-191. https://doi.org/10.1111/j.1600-079X.1998.tb00558.x.

  121. Ressmeyer AR, Mayo JC, Zelosko V, et al. (2003) Antioxidant properties of the melatonin metabolite N1-acetyl-5-methoxykynuramine (AMK): scavenging of free radicals and prevention of protein destruction. Redox Rep. 8 (4): 205-213. doi: 10.1179/135100003225002709.

  122. Chojnacki C, Walecka-Kapica E, Klupińska G, et al. (2012) Serotonin and melatonin secretion and metabolism in patients with hepatic cirrhosis. Pol. Arch. Med. Wewn. 122: 392-397.

  123. Messner M, Huether G, Lorf T, et al. (2001) Presence of melatonin in the human hepatobiliary-gastrointestinal tract. Life Sci. 69: 543–551.

  124. Lee PPN, Shiu SYU, Chow PH, et al. (1995) Regional and diurnal studies on melatonin binding sites in the duck gastrointestinal tract. Biol. Signals 4: 212–224.

  125. Chow PH, Lee PN, Poon AMS, et al. (1996) The gastrointestinal system: A site of melatonin paracrine action. In: Tang PL, Pang SF, Reiter RJ (eds.) Melatonin: A universal photoperiodic signal with diverse action. Frontiers of hormone research, Basel, Karger, 21: 123–132.

  126. Quay WB, Ma YH (1976) Demonstration of gastro-intestinal hydroxyindole-O-methyltransferase. IRCS Med. Sci. 4: 563.

  127. Stefulj J, Hörtner M, Ghosh M, et al. (2001) Gene expression of the key enzymes of melatonin synthesis in extrapineal tissues of the rat. J. Pineal Res. 30: 243–247.

  128. Glaser S, Han Y, Francis H, et al. (2014) Melatonin regulation of biliary functions. Hepatobiliary Surg. Nutr. 3: 35-43. doi: 10.3978/j.issn.2304-3881.2013.10.04.

  129. Chascsa D, Carey EJ, Lindor KD (2017) Old and new treatments for primary biliary 469 cholangitis. Liver Int. 37: 490-499. doi: 10.1111/liv.13294.

  130. Lopez PM, Finana IT, De Agueda MC, et al. (2000) Protective effect of melatonin against oxidative stress induced by ligature of extra-hepatic biliary duct in rats: comparison with the effect of S-adenosyl-L-methionine. J. Pineal Res. 28: 143-149. https://doi.org/10.1034/j.1600-079X.2001.280303.x.

  131. Renzi A, DeMorrow S, Onori P, et al. (2013) Modulation of the biliary expression of arylalkylamine N-acetyltransferase alters the autocrine proliferative responses of cholangiocytes in rats. Hepatology 57: 1130-1141. doi: 10.1002/hep.26105.

  132. Han Y, Onori P, Meng F, et al. (2014) Prolonged exposure of cholestatic rats to complete dark inhibits biliary hyperplasia and liver fibrosis. Am. J. Physiol. Gastrointest. Liver Physiol. 307: G894–G904. doi:10.1152/ajpgi. 00288.2014.

  133. McMillin M, DeMorrow S, Glaser S, et al. (2017) Melatonin inhibits hypothalamic gonadotropin-releasing hormone release and reduces biliary hyperplasia and fibrosis in cholestatic rats. Am. J. Physiol. Gastrointest. Liver Physiol. 313: G410-G418. https://doi.org/10.1152/ajpgi.00421.2016.

  134. Kyritsi K, Meng F, Zhou T, et al. (2017) Knockdown of hepatic gonadotropin-releasing hormone by vivo- morpholino decreases liver fibrosis in multidrug resistance gene 2 knockout mice by down- regulation of mir-200b. Am. J. Pathol. 187: 1551-1565. doi: 10.1016/j.ajpath.2017.03.013.

  135. Gibson P, Gill JH, Khanetal PA (2003) Cytochrome P450 1B1 (CYP1B1) is overexpressed in human colon adenocarcinomas relative to normal colon: implications for drug development. Mol. Cancer Ther. 2:527–534.

  136. Kvetnoy IM, Ingel IE, Kvetnaia TV et al. (2002) Gastrointestinal melatonin: cellular identification and biological role. Neuro. Endocrinol. Lett. 23:121–132.

  137. Reiter RJ, Tan DX, Mayo JC, et al. (2003) Neurally-mediated and neurally-independent beneficial actions of melatonin in the gastrointestinal tract. J. Physiol. Pharmacol. 54: 113–125.

  138. Bubenik GA, Pang SF, Cockshutetal JR (2000) Circadian variation of portal, arterial and venous blood levels of melatonin in pigs and its relationship to food intake and sleep. J. Pineal Res. 28: 9-15. doi: 10.1034/j.1600-079x.2000.280102.x.

  139. Lane EA, Noss HB (1985) Pharmacokinetics of melatonin in man: first pass hepatic metabolism. J. Clin. Endocrinol. Metab. 61: 1214-1216.

  140. Facciolá G, Hidestrand M, vonBahr C, et al. (2001) Cytochrome P450 isoforms involved in melatonin metabolism in human liver microsomes. Eur. J. Clin. Pharmacol. 56: 881–888. doi: 10.1007/s002280000245.

  141. Ma X, Idle JR, Krausz KW, et al. (2005) Metabolism of melatonin by human cytochromes P450. Drug Metab. Dispos. 33 (4): 489-494. doi: 10.1124/dmd.104.002410.

  142. Steindl PE, Ferenci P, Marktl W (1997) Impaired hepatic catabolism of melatonin in cirrhosis. Ann. Intern. Med. 127: 494. https://doi.org/10.7326/0003-4819-127-6-199709150-00025.

  143. Ardizzi A, Grugni G, Saglietti G, et al. (1998) Circadian rhythm of melatonin (aMT) in liver cirrhosis. Minerva Medica 89: 1–4.

  144. Montagnese S, Middleton B, Mani AR, et al. (2010) On the origin and the consequences of circadian abnormalities in patients with cirrhosis. Am. J. Gastroenterol. 105: 1773-1781. doi: 10.1038/ajg.2010.86.

  145. Celinski K, Konturek PC, Slomkaetal M (2009) Altered basal and postprandial plasma melatonin, gastrin, ghrelin, leptin and insulin in patients with liver cirrhosis and portal hypertension without and with oral administration of melatonin or tryptophan. J. Pineal Res. 46: 408– 414. doi: 10.1111/j.1600-079X.2009.00677.x.

  146. Li M, Pi H, Yang Z, Reiter RJ, et al. (2016) Melatonin antagonizes cadmium-induced neurotoxicity by activating the transcription factor EB-dependent autophagy-lysosome machinery in mouse neuroblastoma cells. J. Pineal Res. 61: 353-69. doi: 10.1111/jpi.12353.

  147. León J, Acuña-Castroviejo D, Escames G, et al. (2005) Melatonin mitigates mitochondrial malfunction. J. Pineal Res. 38: 1–9. doi: 10.1111/j.1600-079x.2004.00181.x.

  148. Sener G, Sehirli AO, Ayanoglu-Dülger G (2003) Melatonin protects against mercury (II)-induced oxidative tissue damage in rats. Pharmacol. Toxicol. 93: 290-296.

  149. Mishra S, Ghosh D, Dutta M, et al. (2013) Melatonin protects against lead-induced oxidative stress in stomach, duodenum and spleen of male Wistar rats. J. Pharm. Res. 1: 997-1004.

  150. Konturek PC, Konturek SJ, Majka J, et al. (1997) Melatonin affords protection against gastric lesions induced by ischemia-reperfusion possibly due to its antioxidant and mucosal microcirculatory effects. Eur. J. Pharmacol. 322: 73–77. https://doi.org/10.1016/S0014-2999(97)00051-4.

  151. Cabeza J, Alarcón-de-la-Lastra C, Jiménez D, et al. (2003) Melatonin modulates the effects of gastric injury in rats: role of prostaglandins and nitric oxide. Neurosignals 12: 71–77.doi: 10.1159/000071816.

  152. Ozturk H, Oztürk H, Yagmur Y, et al. (2006) Effects of melatonin administration on intestinal adaptive response after massive bowel resection in rats. Dig. Dis. Sci. 51: 333–337. doi: 10.1007/s10620-006-3134-y.

  153. Hardeland R, Tan DX, Reiter RJ (2009) Kynuramines, metabolites of melatonin and other indoles: the resurrection of an almost forgotten class of biogenic amines. J. Pineal Res. 47: 109–124. doi: 10.1111/j.1600-079x.2009.00701.x.

  154. Gitto E, Pellegrino S, Gitto P, et al. (2009) Oxidative stress of the newborn in the pre-and postnatal period and the clinical utility of melatonin. J. Pineal Res. 44: 128–139. doi: 10.1111/j.1600-079x.2008.00649.x.

  155. Martín M, Macías M, León J, et al. (2002) Melatonin increases the activity of the oxidative phosphorylation enzymes and the production of ATP in rat brain and liver mitochondria. Int. J. Biochem. Cell. Biol. 34: 348–357.

  156. Acuña-Castroviejo D, Escames G, León J, et al. (2003) Mitochondrial regulation by melatonin and its metabolites. Adv. Exp. Med. Biol. 527: 549–557.

  157. Bandyopadhyay D, Ghosh G, Bandyopadhyay A, et al. (2004) Melatonin protects against piroxicam-induced gastric ulceration. J. Pineal Res. 36: 195–203. https://doi.org/10.1111/j.1600-079X.2004.00118.x.

  158. Jaworek J, Brzozowski T, Konturek SJ (2005) Melatonin as an organoprotector in the stomach and the pancreas. J. Pineal Res. 38: 73–83. doi: 10.1111/j.1600-079x.2004. 00179.x.

  159. Ganguly K, Maity P, Reiter RJ, et al. (2005) Effect of melatonin on secreted and induced matrix metalloproteinase -9 and -2 activity during prevention of indomethacin-induced gastric ulcer. J. Pineal Res. 39: 307–315. doi: 10.1111/j.1600-079x.2005.00250.x.

  160. Ganguly K, Kundu P, Banerjee A, et al. (2006) Hydrogen peroxide-mediated down regulation of matrixmetalloprotease-2 in indomethacin-induced acute gastric ulceration is blocked by melatonin and other antioxidants. Free Radic. Biol. Med. 41: 911–925. https://doi.org/10.1016/j.freeradbiomed.2006.04.022.

  161. Konturek PC, Konturek SJ, Burnat G, et al. (2008) Dynamic physiological and molecular changes in gastric ulcer healing achieved by melatonin and its precursor L-tryptophan in rats. J. Pineal Res. 45: 180–190. doi: 10.1111/j.1600-079x.2008.00574.x.

  162. Brzozowska I, Konturek PC, Brzozowski T, et al. (2002) Role of prostaglandins, nitric oxide, sensory nerves and gastrin in acceleration of ulcer healing by melatonin and its precursor, L-tryptophan. J. Pineal Res. 32: 149–162. https://doi.org/10.1034/j.1600-079x.2002.1o811.x.

  163. Brzozowski T, Konturek PC, ZwirskaKorczala K, et al. (2005) Importance of the pineal gland, endogenous prostaglandins and sensory nerves in the gastroprotective actions of central and peripheral melatonin against stress-induced damage. J. Pineal Res. 39: 375–385. https://doi.org/10.1111/j.1600-079X.2005.00264.x.

  164. Ricardo Rde S (2006) Regression of gastroesophageal reflux disease symptoms using dietary supplementation with melatonin, vitamins and aminoacids: comparison with omeprazole. J. Pineal Res. 41 (3): 195‐200. doi:10.1111/j.1600-079X.2006.00359.x.

  165. Konturek PC, Konturek SJ, Brzozowski T (2006) Gastric cancer and Helicobacter pylori infection. J. Physiol. Pharmacol. 57 (Suppl 3): 51-65.

  166. Bubenik GA, Ayles HL, Ball RO, et al. (1998) Relationship between melatonin levels in plasma and gastrointestinal tissues and the incidence and severity of gastric ulcers in pigs. J. Pineal Res. 24: 62–66. https://doi.org/10.1111/j.1600-079X.1998.tb00367.x.

  167. Konturek SJ, Konturek PC, Brzozowska I, et al. (2007) Localization and biological activities of melatonin in intact and diseased gastrointestinal tract (GIT). J. Physiol. Pharmacol. 58 (3): 381-405.

  168. Bandyopadhyay D, Bandyopadhyay A, Das PK, et al. (2002) Melatonin protects against gastric ulceration and increases the efficacy of ranitidine and omeprazole in reducing gastric damage. J. Pineal Res. 33: 1-7. https://doi.org/10.1034/j.1600-079X.2002. 01107.x.

  169. Bandyopadhyay D, Chattopadhyay A (2006) Reactive oxygen species-induced gastric ulceration: protection by melatonin. Curr. Med. Chem. 13: 1187–1202. doi: 10.2174/ 092986706776360842.

  170. Witt-Enderby PA, Radio NM, Doctor JS, et al. (2006) Therapeutic treatments potentially mediated by melatonin receptors: potential clinical uses in the prevention of osteoporosis, cancer and as an adjuvant therapy. J. Pineal Res. 41: 297–305. doi: 10.1111/j.1600-079x.2006.00369.x.

  171. Lopez-Burillo S, Tan DX, Rodriguez-Gallego V, et al. (2003) Melatonin and its derivatives cyclic 3-hydroxymelatonin, N1-acetyl-N2-formyl-5- methoxykynuramine, and 6-hydroxymelatonin reduced oxidative damage induced by Fenton reagents. J. Pineal Res. 34: 237–256. doi: 10.1034/j.1600-079x.2003.00025.x.

  172. Nosál'ová V, Zeman M, Černá S, et al. (2007) Protective effect of melatonin in acetic acid induced colitis in rats. J. Pineal Res. 42: 364–370. doi: 10.1111/j.1600-079X.2007.00428.x.

  173. Srinivasan V, Lauterbach EC, Ho KY, et al. (2012) Melatonin in antinociception: Its therapeutic applications. Curr. Neuropharmacol. 10: 167–178. doi: 10.2174/157015912800604489.

  174. Cuzzocrea S, Reiter JR (2002) Pharmacological action of melatonin in acute and chronic inflammation. Curr. Top. Med. Chem. 2:153–165.

  175. Carrillo-Vico A, Lardone PJ, Alvarez-Sánchez N, et al. (2013) Melatonin: buffering the immune system. Int. J. Mol. Sci. 14: 8638–8683. doi: 10.3390/ijms14048638.

  176. Maldonado MD, Reiter RJ, Pérez‐San‐Gregorio MA (2016) Melatonin as a potential therapeutic agent in psychiatric illness. J. Pineal Res. 24: 391-400. https://doi.org/ 10.1002/ hup.1032.

  177. Carrasco C, Marchena AM, Holguín-Arévalo MS, et al. (2013) Anti-inflammatory effects of melatonin in a rat model of caerulein-induced acute pancreatitis. Cell. Biochem. Funct. 31 (7): 585-590. doi: 10.1002/cbf.2942.

  178. Carrasco C, Rodriguez AB, Pariente JA (2014) Effects of melatonin on the oxidative damage and pancreatic antioxidant defenses in cerulein-induced acute pancreatitis in rats. Hepatobiliary Pancreat. Dis. Int. 13 (4): 442-446. doi: 10.1016/s1499-3872(14)60271-x. 

  179. Han Y, DeMorrow S, Invernizzi P, et al. (2011) Melatonin exerts by an autocrine loop antiproliferative effects in cholangiocarcinoma: its synthesis is reduced favoring cholangiocarcinoma growth. Am. J. Physiol. Gastrointest. Liver Physiol. 301: G623-G633. doi: 10.1152/ajpgi.00118.2011.

  180. Lesage G, Glaser S, Ueno Y, et al. (2001) Regression of cholangiocyte proliferation after cessation of ANIT feeding is coupled with increased apoptosis. Am. J. Physiol. Gastrointest. Liver Physiol. 281: G182-G190. doi: 10.1152/ajpgi.2001.281.1.G182.

  181. Ohta Y, Kongo-Nishimura M, Imai Y, et al. (2006) alpha- Tocopherol protects against alpha-naphthylisothiocyanate-induced hepatotoxicity in rats less effectively than melatonin. Chem. Biol. Interact. 161: 115-124. https://doi.org/10.1016/j.cbi.2006.03.006.

  182. Laothong U, Pinlaor P, Boonsiri P, et al. (2013) Melatonin inhibits cholangiocarcinoma and reduces liver injury in Opisthorchis viverrini-infected and N-nitrosodimethylamine-treated hamsters. J. Pineal Res. 55: 257-266. doi: 10.1111/jpi.12068.

  183. Sumova A, Vanecek J (1997) Melatonin inhibits GnRH-induced increase of cFOS immunoreactivity in neonatal rat pituitary. J. Neuroendocrinol. 9: 135–139. https://doi.org/10.1046/j.1365-2826.1997.d01-1076.x.

  184. Vanecek J (1999) Inhibitory effect of melatonin on GnRH-induced LH release. Rev. Reprod. 4: 67-72. doi:10.1530/ror.0.0040067.

  185. Han Y, Meng F, Venter J, et al. (2016) miR-34a-dependent 535 overexpression of Per1 decreases cholangiocarcinoma growth. J. Hepatol. 64: 1295-1304. doi: 10.1016/j.jhep.2016.02.024.

  186. Blask DE, Dauchy RT, Sauer LA, et al. (2004) Melatonin uptake and growth prevention in rat hepatoma 7288CTC in response to dietary melatonin: melatonin receptor-mediated inhibition of tumor linoleic acid metabolism to the growth signaling molecule 13-hydroxyoctadecadienoic acid and the potential role of phytomelatonin. Carcinogenesis 25: 951-960. doi: 10.1093/carcin/bgh090.

  187. Lissoni P, Barni S, Tancini G, et al. (1993) Immunotherapy with subcutaneous low-dose interleukin-2 and the pineal indole melatonin as a new effective therapy in advanced cancers of the digestive tract. Br. J. Cancer 67: 1404-1407. doi: 10.1038/bjc.1993.260.

  188. Yan JJ, Shen F, Wang K, et al. (2002) Patients with advanced primary hepatocellular carcinoma treated by melatonin and transcatheter arterial chemoembolization: a prospective study. Hepatobiliary Pancreat. Dis. Int. 1: 183-186.

  189. Tahan V, Ozaras R, Canbakan B, et al. (2004) Melatonin reduces dimethylnitrosamine-induced liver fibrosis in rats. J. Pineal Res. 37: 78-84. doi: 10.1111/j.1600-079X.2004.00137.x.

  190. Cruz A, Padillo FJ, Torres E, et al. (2005) Melatonin prevents experimental liver cirrhosis induced by thioacetamide in rats. J. Pineal Res. 39: 143-150. https://doi.org/10.1111/j.1600-079X.2005.00227.x.

  191. Koc M, Taysi S, Buyukokuroglu ME (2003) Melatonin protects rat liver against irradiation-induced oxidative injury. J. Radiat. Res. 44 (3): 211-215.

  192. Sewerynek E, Reiter RJ, Melchiorri D, et al. (1996) Oxidative damage in the liver induced by ischemia-reperfusion: protection by melatonin. Hepato-Gastroenterology 43 (10): 898-905.

  193. Vairetti M, Ferrigno A, Bertoneetal R (2005) Exogenous melatonin enhances bile flow and ATP levels after cold storage and reperfusion in rat liver: implications for liver transplantation. J. Pineal Res. 38 (4): 223-230. doi: 10.1111/j.1600-079X.2004.00193.x.

  194. Zaoualí MA, Reiter RJ, Padrissa-Altés S, et al. (2011) Melatonin protects steatotic and nonsteatotic liver grafts against cold ischemia and reperfusion injury. J. Pineal Res. 50 (2): 213–221. doi: 10.1111/j.1600-079X.2010.00831.x.

  195. Daniels WM, Reiter RJ, Melchiorri D, et al. (1995) Melatonin counteracts lipid peroxidation induced by carbon tetrachloride but does not restore glucose-6 phosphatase activity. J. Pineal Res.19 (1): 1-6. doi: 10.1111/j.1600-079x.1995.tb00164.x.

  196. Sahna E, Parlakpinar H, Vardi N, et al. (2004) Efficacy of melatonin as protectant against oxidative stress and structural changes in liver tissue in pinealectomized rats. Acta Histochemica. 106 (5): 331-336. https://doi.org/10.1016/j.acthis.2004.07.006.

  197. Reiter RJ, Paredes SD, Manchester LC, et al. (2009) Reducing oxidative/nitrosative stress: a newly-discovered genre for melatonin. Crit. Rev. Biochem. 44 (4): 175-200. doi: 10.1080/10409230903044914.

  198. Mathes AM (2010) Hepatoprotective actions of melatonin: possible mediation by melatonin receptors. World J. Gastroenterol. 16 (48): 6087-6097. doi: 10.3748/wjg.v16. i48.6087.

  199. Hong RT, Xu JM, Mei Q (2009) Melatonin ameliorates experimental hepatic fibrosis induced by carbon tetrachloride in rats. World J. Gastroenterol. 15 (12): 1452-1458. doi: 10.3748/wjg.15.1452.

  200. Rosa DP, Bona S, Simonetto D, et al. (2010) Melatonin protects the liver and erythrocytes against oxidative stress in cirrhotic rats. Arq. Gastroenterol. 47: 72-78.

 CCBY.png

 

This work is licensed under a Creative Commons Attribution 4.0 International License