Abstract
Circadian rhythms and clock gene expressions are regulated by the suprachiasmatic nucleus in the hypothalamus, and melatonin is produced in the pineal gland. Although the brain detects the light through retinas and regulate rhythms and melatonin secretion throughout the body, the liver has independent circadian rhythms and expressions as well as melatonin production. Previous studies indicate the association between circadian rhythms with various liver diseases, and disruption of rhythms or clock gene expression may promote liver steatosis, inflammation, or cancer development. It is well known that melatonin has strong antioxidant effects. Alcohol drinking or excess fatty acid accumulation produces reactive oxygen species and oxidative stress in the liver leading to liver injuries. Melatonin administration protects these oxidative stress-induced liver damage and improves liver conditions. Recent studies have demonstrated that melatonin administration is not limited to antioxidant effects and it has various other effects contributing to the management of liver conditions. Accumulating evidence suggests that restoring circadian rhythms or expressions as well as melatonin supplementation may be promising therapeutic strategies for liver diseases. This review summarizes recent findings for the functional roles and therapeutic potentials of circadian rhythms and melatonin in liver diseases.
Keywords: melatonin, circadian rhythms, clock genes, liver fibrosis, liver steatosis, reactive oxygen species, non-alcoholic fatty liver disease
1. Introduction
Circadian rhythms are an internal cycle, which is a biological process with oscillation of approximately 24 hr. Almost all essential physiological events and metabolisms are under control of circadian rhythms1. For example, in humans, circadian rhythms control sleep-wake cycle, and cognitive activity is upregulated at the wake phase in daytime and the preparation for the sleep phase occurs in nighttime, which is important for memory consolidation2. The suprachiasmatic nucleus (SCN) in the hypothalamus is the central pacemaker of the rhythms and synchronizes circadian clocks throughout the body according to light-dark cycle. Synchronization of circadian clocks in different organs controlled by the SCN is mediated by autoregulatory expression of clock genes, such as circadian locomotor output cycles kaput (CLOCK) and brain and muscle ARNT-like 1 (BMAL1) as activators, and period circadian protein homolog 1 (PER1), PER2, cryptochrome circadian regulator 1 (CRY1), and CRY2 as repressors3. Although the brain detects light through retinas and synchronize circadian clocks throughout the body, specific organs, such as the liver, may have their own circadian rhythms or clock gene expression patterns. A previous study has demonstrated that expression levels of clock genes including PER1 and PER2 differ before and after feeding as well as in different tissues, such as the hypothalamus, the skeletal muscle, and the liver, in swine4. Koronowski et al. have generated transgenic mice, which have a stop cassette inserted between exon 5 and 6 of BMAL1 resulting in global BMAL1 knockout phenotypes5. The stop cassette was flanked by loxP sites and crossing those global BMAL1−/− mice with mice that express Cre recombinase under the control of α-fetoprotein enhancer (Alfp-Cre mice) allowed the removal of the stop cassette specifically in Alfp-positive hepatic cells5. Cre expression is observed selectively in hepatocytes and cholangiocytes in Alfp-Cre mice6. As a result, crossed mice expressed BMAL1 only in the liver (hepatocytes and cholangiocytes)5. Global BMAL1−/− mice lost circadian rhythms and expressions, but mice with hepatic BMAL1 expression had rhythmic circadian expressions and metabolisms in the liver without BMAL1 expression and circadian rhythms in the brain and other organs5. This liver-specific rhythm was dependent on light-dark cycle, indicating that the liver may have independent circadian rhythms and functions5. Since circadian rhythms regulate various physiological events throughout the body, the association of disrupted circadian rhythms with human disorders including liver diseases has been suggested7,8.
Melatonin (N-acetyl-5-methoxytryptamine) is a hormone associated with circadian rhythms especially in the dark phase. Melatonin synthesis is facilitated at night and inhibited by light at daytime detected by retinas9. Melatonin is produced primarily by the pineal gland from the amino acid tryptophan10. Tryptophan is converted to 5-hydroxytryptophan followed by serotonin (5-hydroxytryptamine). Serotonin is converted to N-acetylserotonin by aralkylamine N-acetyltransferase (AANAT), and finally N-acetylserotonin is converted to melatonin by N-acetylserotonin O-methyltransferase (ASMT) in the pineal gland. Melatonin has a strong antioxidant activity and is an endogenous free radical scavenger. The liver produces melatonin as well as the pineal gland. A study using goldfish identified expressions of AANAT and ASMT in the liver, and expression rhythms of those genes were altered depending on lighting conditions11. Melatonin binds to G protein-coupled receptors MT1 and MT2 in humans and rodents, and various hepatic and gastrointestinal cells, which include hepatocytes, gallbladder epithelia, and bile duct epithelia, express these melatonin receptors and can be regulated by melatonin12–14. Melatonin has protective effects against oxidative stress followed by inflammation caused by reactive oxygen species (ROS) or reactive nitrogen species15. Therefore, melatonin administration shows therapeutic effects in various disorders including liver diseases16,17. Melatonin has potentials for novel treatments of liver diseases by decreasing oxidative stress or restoring circadian rhythms and functions. This review summarizes current understandings of the functional roles of circadian rhythms and melatonin signaling in the pathophysiology of liver diseases as well as the potentials of melatonin administration as therapeutic treatments.
2. Functional roles and therapeutic potentials of melatonin and circadian rhythms in liver diseases
2.1. Alcoholic liver disease
Excessive alcohol consumption causes alcoholic liver disease (ALD), which is characterized by liver steatosis, hepatitis, and cirrhosis. Alcohol drinking disrupts intestinal permeability leading to elevated serum levels of lipopolysaccharide (LPS), which is a bacterial endotoxin causing inflammatory responses18,19. A previous study has analyzed the effects of moderate alcohol drinking on circadian rhythms using 11 daytime workers and 11 nighttime workers. In this study, all subjects were told to avoid alcohol drinking and have usual sleep schedule for 1 week20. After an alcohol-free week, subjects were kept in dim light and blood samples were collected and serum melatonin levels were analyzed in every hour for 24 hr (baseline samples)20. In a following week, all subjects were given red wine (0.5 g of ethanol per kg body weight) every day after finishing work and before bedtime for 1 week20. Blood samples were collected with same procedures to baseline samples (post-alcohol samples)20. This study has demonstrated that nighttime workers but not daytime workers have delayed melatonin secretion peak after 1 week alcohol period, and alcohol consumption increases gut permeability only in nighttime workers, suggesting that nighttime workers are more susceptible for alcohol drinking than daytime workers20. This study compared clock gene expression patterns before and after alcohol period in isolated peripheral blood mononuclear cells from daytime and nighttime workers20. Alcohol drinking elevated expression levels of PER1 and decreased for CRY1, and changed circadian oscillation patterns for CRY2 regardless of working time20. Another study has performed acute or chronic alcohol feeding in mice and has demonstrated that both acute and chronic alcohol drinking changes profiles of hepatic circadian transcriptome, especially in sterol regulatory element-binding protein (SREBP) pathways in the liver, indicating the change of circadian rhythms by alcohol drinking21. Binge alcohol drinking increases serum levels of alcohol as well as endotoxin (LPS) in healthy individuals19. Zeitgeber time (ZT) is the standard notation in studies of the circadian cycle. ZT0 indicates the start of the light phase and ZT12 indicates the start of the dark phase. Alcohol drinking may cause different effects depending on the time of drinking, and binge alcohol gavage in different ZT showed different effects in mice22. Mice with binge alcohol drinking at ZT12 had higher serum alcohol levels than ZT0, and serum LPS levels were highest when gavage was performed at ZT4 or ZT822. Exposure of zebrafish larvae to 5% ethanol for 1 hr can be fatal, and a previous study found that mortality was highest with exposure at ZT2 and lowest at ZT18, indicating the time-dependent effects of alcohol23. Mice with albumin promoter-driven Cre recombinase (Alb-Cre mice) have Cre expression selectively in hepatocytes and hepatic stellate cells (HSCs)24. Zhang et al. generated Alb-Cre-mediated liver-specific BMAL1 knockout mice and found that these mice had higher serum aminotransferase (ALT) levels and liver steatosis compared to control mice after chronic and binge alcohol feeding25. Hepatic BMAL1 deficiency decreased expression levels of genes associated with de novo lipogenesis and β-oxidation in the liver, indicating the association between clock genes and liver steatosis25. Adenovirus-mediated BMAL1 overexpression in the liver attenuated alcohol-induced liver damage and steatosis in vivo, indicating the protective effects of BMAL1 on ALD25. Another study has demonstrated that Alb-Cre-mediated hepatic BMAL1 knockout mice had higher serum ALT levels after chronic 3% alcohol feeding for 5 weeks by decreasing expression levels of genes associated with glycogen metabolism26.
Alcohol consumption leads to the production of ROS during alcohol metabolism, which induces liver damage27. Therefore, melatonin can protect the liver against ALD with its antioxidant effects28. Melatonin administration (10 mg/kg body weight daily for 10 days via intraperitoneal [i.p.] injection) along with ethanol i.p. injection increased circulating white blood cells in serum as well as antioxidant capacity in the liver, the skeletal muscle, and the kidney in mice29. Another study administered alcohol into pregnant rats by gavage with or without melatonin (10 or 15 mg/kg, i.p. injection during gestational period)30. In this model, melatonin administration decreased levels of alcohol-induced liver damage not only in mothers but also in newborn pups30. Although further studies are required to develop novel treatments for ALD targeting circadian rhythms and clock gene expressions, melatonin administration has shown promising therapeutic effects in these previous studies.
2.2. Non-alcoholic fatty liver disease
Patients with non-alcoholic fatty liver disease (NAFLD) have similar symptoms to ALD, such as liver steatosis, inflammation, and cirrhosis without alcohol consumption. A study of 4,740 male workers has found that night shift workers have higher serum ALT levels than daytime workers with the increase in night shift years, indicating the association between circadian disruption and liver malfunctions31. Exposure to light for 2 hr during night time induces glucose intolerance in rats, which is associated with NAFLD32. Shimizu et al. have developed a rat model with 4 hr delayed feeding during ZT16-ZT4 compared to control feeding during ZT12-ZT2433. In this model, rats with delayed feeding gained increased body weights and perirenal adipose tissues compared to rats with control feeding, and circadian oscillations of hepatic clock and fatty acid synthesis genes were delayed 2–4 hr due to shifted feeding time33. Another study fed mice with high-fat diet (HFD) during active phase (nighttime) or sleep phase (daytime) and found that mice with sleep phase feeding had increased body weights as well as hepatic concentrations of free fatty acids (FFAs) and triglycerides compared to mice with active phase feeding34. Arrhythmic feeding of mice with one-eighth portion of daily food intake given every 3 hr disrupted rhythmic expression of over 70% of transcriptomes in the liver35. Circadian disruption induces leptin resistance in mice, which may lead to obesity or metabolic syndromes, indicating different impacts of food intake in the liver depending on the timing of intake36. A previous study has demonstrated that Alb-Cre-mediated liver specific BMAL1 knockout mice have elevated mRNA methylation especially in peroxisome proliferator-activated receptor alpha (PPARα) in the liver37. PPARα promotes β-oxidation of FFAs and lipid metabolism38, and methylation of PPARα mRNA causes mRNA degradation and inhibition of PPARα expression leading to accumulation of triglycerides in the liver37. Although detailed mechanisms are undefined, circadian rhythms and rhythmic clock gene expressions may be critical for liver homeostasis and lipid metabolism.
Mitochondrial β-oxidation breaks down FFAs but produces ROS, which causes liver damage. FFAs also induce endoplasmic reticulum (ER) stress, which is caused by accumulation of unfolded and misfolded proteins, and ER stress is another potent source of ROS39. Melatonin has strong antioxidant effects and can be utilized to protect the liver from NAFLD or fatty acid-induced liver damage. Melatonin administration (500 μg/kg daily for 4 weeks, i.p. injection) increased antioxidant activities and decreased lipid peroxidation as well as ER stress in the liver of obese (ob/ob) mice40. Another study has demonstrated that melatonin (100 mg/kg daily for 10 weeks, dissolved in drinking water) attenuates ER stress leading to improved liver steatosis and insulin resistance in mice fed with HFD41. HFD induced elevated hepatic de novo lipogenesis and triglyceride concentrations in hamsters, but melatonin administration (10, 20, or 50 mg/kg daily for 8 weeks via gavage) decreased enzyme activities of acetyl-CoA carboxylase (ACC) and fatty acid synthase (FAS), which are associated with de novo lipogenesis, leading to attenuated liver steatosis42. Activation of brown adipose tissues leads to body weight loss, improved glucose tolerance, and decreased lipid accumulation in the liver, and may be a therapeutic target for NAFLD43. Halpern et al. administered melatonin (3 mg) to patients with melatonin deficiency due to radiotherapy or surgical removal of pineal gland for 3 months and analyzed brown adipose tissue mass44. Melatonin supplementation increased mass and activity of brown adipose tissues and decreased blood levels of total cholesterol and triglyceride44. Pre-treatment with melatonin (0.1–0.3 mM) inhibited accumulation of cholesterol and triglyceride in hepatocellular carcinoma (HCC) line, HepG2 cells, incubated with high concentrations of oleic acid in vitro45. Oleic acid elevated expression levels of genes associated with de novo lipogenesis including SREBP-1c, FAS, and stearoyl-CoA desaturase 1 (SCD1) and decreased β-oxidation gene expression such as PPARα and carnitine palmitoyltransferase I (CPT1), but melatonin treatment abolished those effects leading to inhibited lipid accumulation in HepG2 cells45.
A previous study has demonstrated that melatonin administration (30 mg/kg daily for 7 weeks, i.p. injection) attenuates HFD-induced liver steatosis, inflammation, and fibrosis in mice46. HFD elevated activation of apoptosis signal-regulating kinase 1 (ASK1) and downstream signaling pathways including mitogen-activated protein kinase kinase 3/6 (MKK3/6), MKK4/7, p38, and c-Jun N-terminal kinase (JNK) leading to liver damage, but melatonin ameliorated HFD-induced liver damage by inhibiting activation of these pathways in vivo46. Intragastric melatonin administration (10 mg/kg daily for 12 weeks) in another study decreased serum ALT levels, liver steatosis, proinflammatory cytokine expression including interleukin 6 (IL-6) and tumor necrosis factor alpha (TNFα), and activation of mitogen-activated protein kinase (MAPK), JNK, and p38 signaling pathways in HFD mice, indicating that melatonin inhibits signaling pathways associated with cytokine production, inflammation, and apoptosis47. Mitochondria play a vital role in β-oxidation of FFAs, and mitochondrial dysfunctions, such as extensive mitochondrial permeability transition pore (mPTP) opening and impaired respiratory functions, lead to excess ROS production causing apoptotic cell deaths48. Das et al. have demonstrated that palmitic acid induces mitochondrial fragmentation, calcium overload, and mPTP opening, but melatonin treatment (1 mM for 30 min) inhibits these effects in HepG2 cells in vitro49. HFD decreased mitochondrial enzyme activities in mice, such as cytochrome C oxidase and pyruvate dehydrogenase, but melatonin administration (10 or 20 mg/kg daily for 4 weeks, i.p. injection) restored these activities in vivo49. Another study also demonstrated mitochondrial dysfunction including calcium overload and mPTP opening caused by HFD in mice, and melatonin administration (20 mg/kg daily for 12 weeks, i.p. injection) reversed these effects by inhibiting expression of nuclear receptor subfamily 4 group A member 1 (NR4A1) and activation of p53, suggesting that melatonin restores mitochondrial functions leading to improved liver conditions in NAFLD50. Gut microbiota is associated with the pathophysiology of NAFLD and gut bacteria transplantation has potential therapeutic effects to improve liver conditions51. Yin et al. have demonstrated that HFD decreases gut microbiota diversity in mice, and melatonin administration (108 mg/kg daily for 2 weeks, supplemented in drinking water) improved this diversity as well as liver steatosis in HFD mice52. In this model, HFD altered the mouse gut microbiota balance by elevating abundance of Lactobacillus and decreasing abundance of Bacteroides, which was also observed in cholangiopathies and may be associated with liver damage and fibrosis53, and melatonin reversed these effects52. Although further studies are required, melatonin administration may be a promising therapeutic tool for NAFLD by improving mitochondrial activities, inhibiting lipid accumulation in the liver, and maintaining gut microbiota diversity.
2.3. Cholangiopathies
Cholangiopathies or bile duct disorders include primary sclerosing cholangitis (PSC), primary biliary cholangitis (PBC), and biliary atresia. Cholangiopathies are characterized by cholestasis, ductular reaction, biliary inflammation, and liver fibrosis54. These disorders are slow but progressive and liver conditions can be severe and cirrhotic especially in the later stages55. It has been suggested that circadian rhythm and melatonin synthesis is associated with the pathophysiology of cholangiopathies and liver cirrhosis. A previous study using 74 PBC patients, 60 cirrhosis patients, and 79 healthy individuals has found that sleep timing is significantly delayed in patients with PBC or cirrhosis, and delayed sleep timing is associated with impaired sleep quality in PBC patients56. Another study using 13 PBC patients, 7 cirrhosis patients, and 6 healthy individuals found that patients with PBC or cirrhosis had poor sleep quality assessed by the Pittsburgh Sleep Quality Index57. This study has demonstrated that 15-day morning bright light treatment for PBC patients using 10,000 lux light box for 45 minutes improves sleep quality and rhythmic secretion of 6-sulphatoxymelatonin in urine, which is a metabolite of melatonin, indicating that cholangiopathies and liver cirrhosis are associated with disrupted circadian rhythms and melatonin production, and restoring rhythms by morning light treatment may be a therapeutic approach for cholangiopathies57.
Bile duct ligation (BDL) is a surgical ligation of the common bile duct to mimic cholestasis in rodents, and causes ductular reaction, biliary inflammation, liver damage and fibrosis54. BDL elevates expression levels of MT1 and MT2 melatonin receptors and clock genes including CLOCK, BMAL1, PER1, and CRY1 in cholangiocytes in vivo14,58. BDL also elevates expression of AANAT in cholangiocytes, and inhibition of AANAT leads to cholangiocyte proliferation via enhanced expression of vascular endothelial growth factor (VEGF)-A and VEGF-C in BDL rats59,60. Overexpression of AANAT inhibits VEGF-A and VEGF-C expression as well as cell proliferation in murine cultured cholangiocytes, indicating local melatonin synthesis followed by regulations of cholangiocyte functions during cholestatic liver injury59,60. A previous study has performed melatonin administration (1 mg/kg per day for 7 days) via intracerebroventricular-implanted cannulas for BDL rats and found that melatonin ameliorated ductular reaction and liver fibrosis by inhibition of gonadotropin releasing hormone (GnRH) secretion in vivo61. Elevated GnRH secretion activates HSCs leading to robust liver fibrosis62. Dark therapy, which maintains animals in complete dark 24 hr for 1 week, facilitates AANAT expression and melatonin secretion form the pineal gland in rats63. Dark therapy for BDL rats attenuated ductular reaction and liver fibrosis by decreasing cAMP levels in cholangiocytes63. A previous study has performed pinealectomy surgery or light treatment, which maintains animals in complete light 24 hr for 1 week, for BDL rats64. In this model, both pinealectomy and light treatment decreased serum melatonin levels and AANAT expression in cholangiocytes compared to control BDL rats64. Pinealectomy or light treatment exacerbated ductular reaction and liver fibrosis in BDL rats via elevated VEGF-A and proinflammatory cytokine production, such as IL-1β and IL-6, as well as increased ROS levels in the liver64. Mdr2−/− mice are the most common mouse model for human PSC, which have similar conditions such as cholestasis, biliary inflammation, ductular reaction, and liver fibrosis54. Dark therapy (complete dark for 1 week) or melatonin administration (2 mg/g body weight per day for 1 week via drinking water) for Mdr2−/− mice improved liver conditions compared to control Mdr2−/− mice with 12:12 hr light dark cycle by inhibiting expression of VEGF-A/C in cholangiocytes via decreased levels of microRNA (miRNA) miR-200b65. Thioacetamide (TAA) induces biliary damage in rodents and utilized as a model of cholestatic liver injury54. A previous study has demonstrated that melatonin administration (5 mg/kg body weight per day for 1 week via i.p. injection) improved liver conditions with lower serum alanine ALT and aspartate aminotransferase (AST) levels, decreased proinflammatory cytokine expression, such as IL-1β and TNFα, as well as profibrogenic factors such as transforming growth factor beta 1 (TGF-β1) and collagen type I in the liver by attenuating oxidative stress during TAA-induced liver injury in rats66. Melatonin supplementation may be a promising approach to regulate cholangiocyte functions and maintain liver homeostasis.
2.4. Toxin-induced liver injury
Toxins or carcinogens, such as TAA and carbon tetrachloride (CCl4), are commonly administered into rodents to cause liver damage and understand the pathophysiology of liver injury54. Acute or chronic CCl4 administration causes oxidative stress-induced liver damage and fibrosis, and melatonin can protect the liver with its antioxidant characteristics. Melatonin administration (2.5–20 mg/kg per day for 6 weeks, i.p. injection) for CCl4-administered rats decreased serum ALT and AST levels, as well as liver fibrosis by inhibiting expression of TGF-β1 and VEGF-A in the liver67,68. Although both melatonin treatments before or after CCl4 administration have protective effects on CCl4-induced liver injury69, a study demonstrated that post-treatment had better therapeutic effects compared to co-treatment in CCl4-administered rats70. Another study demonstrated therapeutic effects of melatonin administration (5 or 10 mg/kg per day, i.p. injection) against CCl4-induced liver injury in mice with decreased liver fibrosis levels71. This study found that administration of CCl4 decreased expression of clock genes, such as BMAL1, CLOCK, PER1/2, CRY1/2, and melatonin treatment restored expression levels of these genes in the liver71. Stem cell therapy performs transplantation of stem cells to facilitate liver regeneration and improve conditions in various liver diseases including cirrhosis72. A previous study treated bone marrow-derived mesenchymal stem cells with or without 5 μM melatonin for 24 hr and infused them into CCl4-administered rats via tail vein73. Stem cell transplantation decreased serum ALT and AST levels as well as liver fibrosis, and melatonin pre-treatment of stem cells enhanced these therapeutic effects73. Melatonin treatment elevated expression levels of MT1 and MT2 melatonin receptors in stem cells, which may be associated with those effects73.
Melatonin shows therapeutic effects on liver damage caused by not only TAA and CCl4 but also other toxins, indicating its universal effects. α-naphthylisothiocyanate (ANIT) is a chemical compound that causes cholestasis and liver damage. ANIT elevated serum ALT and AST levels, and melatonin supplementation (100 mg/kg in drinking water for 12 hr following the ANIT injection) decreased those levels in rats74. Valproic acid (VPA) is a mood-stabilizing agent used for psychiatric disorders, but also causes liver damage. VPA elevated expression of proinflammatory cytokines including IL-1β and TNFα as well as hepatocyte apoptosis, and melatonin administration (10 mg/kg via gavage, 1 hr prior to VPA treatment for 14 days) inhibited those effects75. Dexamethasone (DEX) is a type of corticosteroids used to decrease inflammatory responses, but it can cause liver steatosis. A previous study administered DEX to pregnant rats at gestational day 14–21, and rats were sacrificed at postnatal day 12076. In this model, prenatal DEX exposure elevated liver steatosis, IL-6, TNFα and TGF-β1 expression, and apoptosis in the liver, and melatonin supplementation (1 mg/kg per day in drinking water, from gestational day 14–21 to postnatal day 120) reversed this DEX-induced liver conditions76. Melatonin has strong protective effects on toxin-induced liver damage especially caused by oxidative stress.
2.5. Hepatocellular carcinoma
HCC is the most common malignancy of primary liver cancers. Previous studies suggest the association of disruption of circadian rhythms with cancer development in various organs including the liver77. Kettner et al. generated Cry1−/−Cry2−/− mice, Per1−/−Per2−/− mice, and Alb-Cre-mediated liver-specific BMAL1 knockout mice and maintained them for 90 weeks78. These transgenic mice had lower survival rates with various disorders including neurodegeneration, cystic renal dysplasia, liver steatosis, and HCC compared to wild-type mice78. This study also maintained wild-type mice with steady 12:12 hr light-dark cycle or weekly transfer to another room with 8 hr difference in light-dark cycle for 4–90 weeks to generate “jet-lagged” mice78. Jet-lagged mice had higher lipid deposition in the liver followed by HCC development and poor survival rates compared to mice with steady cycle78. Chronic jet lag caused disruption of hepatic cholesterol, bile acid, and xenobiotic metabolism via elevation of constitutive androstane receptor (CAR), which is a regulator of xenobiotic metabolism and associated with hepatocarcinogenesis, indicating the association of circadian rhythms with liver metabolism and HCC development78. Another study has found negative correlation of expression levels between BMAL1 and HNF4α, which is important for hepatocyte fate determination and functions79. HNF4α had circadian activities with rhythmic expression of downstream targets such as cyclin B1 and D1 in murine hepatocyte line AML12 cells79. Human HCC lines, HepG2, Hep3B, and Huh7 cells, expressed high levels of HNF4α and low levels of BMAL1, and overexpression of BMAL1 in HepG2 cells caused elevated apoptosis and decreased tumor growth in xenograft mouse models, indicating the roles of circadian rhythms in hepatocyte carcinogenesis79.
HCC tumors often have genetic mutations in various genes or their promoters such as telomerase reverse transcriptase (TERT), tumor protein p53 (TP53), and catenin beta 1 (CTNNB1)80. A previous study using 335 HCC patients and 1196 control individuals has identified single nucleotide polymorphisms (SNPs) in MT1 melatonin receptor, which are associated with the elevated risk of HCC81. A specific haplotype of four MT2 SNPs was also associated with HCC development, indicating that functions of receptors and melatonin signaling may be associated with HCC development81. During 12:12 light-dark cycle, exposure to blue-enriched light (increased transmittance of 462–484 nm and decreased red light greater than 640 nm) at daytime using blue-tinted cages increased plasma levels of melatonin at nighttime in rats82. This blue light treatment followed by promotion of melatonin secretion at night inhibited tumor growth in xenograft rat models with Morris 7288CTC hepatoma although detailed mechanisms are undefined83. Diethylnitrosamine (DEN) administration is utilized to induce HCC in rodents as an animal model of HCC84. A previous study administered melatonin (5 or 10 mg/kg per day for 10, 20, 30, or 40 weeks via i.p. injection) to DEN mice and demonstrated that melatonin inhibited HCC development and improved liver conditions during DEN-induced liver injury85. DEN elevated gene expression of CLOCK and BMAL1, and decreased expression of PER1, PER2, and CRY1, but melatonin injection inhibited these gene expression changes85. Another study analyzed the efficacy of the combination of stem cell therapy and melatonin administration in DEN rat models86. Although both transplantation of bone marrow-derived MSCs isolated from young rats as well as melatonin administration (20 mg/kg, twice a week for 5 weeks, i.p. injection) attenuated DEN-induced liver damage and HCC development in rat liver, the combination of stem cell transplantation and melatonin administration had the highest therapeutic effects by decreasing expression of anti-apoptosis genes including B-cell lymphoma 2 (Bcl-2) and survivin and by increasing apoptotic gene expression such as caspase3 and Bcl-2-associated X protein (BAX) in this model86.
Chemotherapy is commonly performed especially for advanced HCC, and sorafenib has been widely utilized as the standard agent87. Sorafenib treatment induced apoptosis by decreasing the Bcl-2/BAX ratio as well as ER stress followed by autophagy via activation of protein kinase R-like endoplasmic reticulum kinase (PERK)/activating transcription factor 4 (ATF4) pathways in HepG2 cells881. Melatonin increased the sensitivity of HepG2 cells to sorafenib leading to enhanced cell death and inhibition of proliferation by inhibiting PERK/ATF4-mediated autophagy in vitro88. Melatonin itself suppressed cell proliferation, migration, and invasion of HepG2 and Huh7 cells, and the combination of melatonin and etoposide (VP16) had better anti-cancer effects compared to administrations of melatonin only or VP16 only in xenograft mouse models with Huh7 tumors by inhibiting expression of DNA repair protein RAD51 via long non-coding RNA (lncRNA) RAD51-AS1 in vivo, indicating effective anti-cancer effects of melatonin against HCC89.
2.6. Cholangiocarcinoma
Cholangiocarcinoma (CCA) is a type of hepatic malignancy that emerges from the biliary tree and is the second most common primary liver cancer after HCC. Although the association of circadian rhythms with CCA development is still not fully elucidated, a previous study found upregulated and downregulated expression levels of BMAL1 and PER1, respectively in human CCA biopsies or human CCA cell lines, such as Mz-ChA-1 and HuH28 cells90. Normal human cholangiocyte line, H69 cells, but not CCA cell lines had circadian expression rhythms for clock genes including BMAL1, CLOCK, PER1, and CRY190. Overexpression of PER1 inhibited cell proliferation in Mz-ChA-1 cells and decreased tumor size in xenograft mouse models, indicating potentials of circadian rhythms and clock genes as therapeutic targets for CCA90.
Han et al. has demonstrated that CCA tumor tissues from patients express lower levels of AANAT and ASMT compared to normal liver tissues, and human CCA cell lines including Mz-ChA-1 and HuH28 cells express significantly lower levels of AANAT and ASMT compared to H69 cells91. Overexpression of AANAT inhibited cell proliferation in Mz-ChA-1 cells, and melatonin administration (4 mg/kg, three times a week for 43 days, i.p. injection) decreased tumor size in the xenograft mouse models with Mz-ChA-1 tumors91. Melatonin also inhibited cell proliferation in human CCA cell lines KKU-M055 and KKU-M214 cells by elevation of ROS-induced DNA damage and decreased expression of Bcl-2 leading to apoptosis in vitro92. Liver fluke infection caused by parasites such as Opisthorchis viverrini is a common risk factor for CCA in Asia93. O. viverrini infection plus carcinogen N-nitrosodimethylamine (NDMA) administration developed CCA in hamsters, and melatonin administration (10 or 50 mg/kg, daily for 120 days, orally) elevated Bcl-2 expression and decreased BAX expression resulting in attenuated CCA development and improved survival rates in this model94. CCA tumors are often accompanied with the tumor microenvironment in dense stromal tissues containing fibroblasts and lymphocytes promoting CCA progression95. Melatonin inhibited lymphocyte infiltration such as Th17+ cells and decreased inflammatory responses including expression of IL-1β and NF-κB in hamsters with fluke infestation and NDMA treatment96. Although further evidence is required, melatonin administration may be used for CCA to inhibit tumor growth and regulate functions of CCA microenvironment.
2.7. Liver transplantation and ischemia-reperfusion injury
Liver transplantation is often the sole treatment for end-stage liver diseases, such as liver cirrhosis, late stage PSC and PBC, and liver cancers. However, liver transplantation has a risk of ischemia-reperfusion injury (IRI), which is characterized by oxidative stress, inflammation, and liver damage, and IRI may lead to graft rejection97. A previous study monitored blood pressure of 107 liver transplant recipients and found that approximately 90% of patients had arterial hypertension and abnormal circadian blood pressure pattern after operation98. Another study analyzed 147 patients who underwent liver transplantation in one institution and found that 45 patients who had transplantation at night suffered higher incidence of intraoperative blood loss and other complications, postoperative abdominal infection, longer operation time, and longer time to restore liver functions after transplantation compared to 102 patients with daytime operation99. Although night shift could influence performances of medical workers, circadian rhythms ant the timing of operation may affect conditions of patients after liver transplantation.
Deng et al. clamped first porta hepatis and released it after 35 minutes to recover the blood flow to induce IRI in rats100. In this model, pretreatment of melatonin (10 mg/kg, 35 and 70 min prior to ischemia, i.p. injection) decreased serum ALT and AST levels showing therapeutic potentials of melatonin against IRI liver damage100. Living donor liver transplantation is performed using a portion of the liver from a healthy living individual and may compensate the shortage of available liver grafts. However, the donated liver graft from the living person is generally small, and small grafts may cause small-for-size syndrome, which leads to graft dysfunction or complications101. A previous study has performed liver transplantation in mice with small grafts (30% of total liver) and found that melatonin treatments (20 μg/mL in cold saline to perfuse the donor liver and 10 mg/kg 15 min before operation for the recipient, i.p. injection) improved survival rates of recipient mice102. The combination of IRI and partial hepatectomy (60 min hepatic ischemia and reperfusion combined with 70% or 80% liver excision) caused severe liver damage in mice, and 10 mg/kg melatonin pretreatment decreased serum ALT and AST levels and promoted hepatocyte proliferation and liver regeneration via elevation of IL-6 and TNFα expression102. Since IRI is caused by oxidative stress during reperfusion, melatonin treatment shows strong antioxidant effects to protect liver damage.
3. Therapeutic potentials of extracellular vesicles
Extracellular vesicles (EVs) are small membrane-bound vesicles secreted from various cells. There are several types EVs including exosomes, microvesicles or microparticles, and apoptotic bodies according to their biogenesis and particle size. EVs contain cargo mediators inside, such as proteins, DNAs, and RNAs, and donor cells can transfer those mediators into recipient cells by secreting EVs. This EV-mediated cell-to-cell communication plays a vital role in the pathophysiology of liver diseases103. For example, hepatocytes secrete elevated numbers of EVs during lipid-induced damage, and these EVs contain tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)104. Lipotoxic hepatocyte-derived EVs induced inflammatory responses such as IL-1β and IL-6 production in macrophages by delivering cargo TRAIL104. Another study has demonstrated that cholangiocytes secrete EVs that contain elevated levels of lncRNA H19 during cholestatic liver injury, and these H19-enriched EVs activates HSCs leading to liver fibrosis in cholangiopathies105.
EVs mediate inflammatory or fibrogenic responses in hepatic cells by delivering cargo mediators, which means that EVs could be used as therapeutic tools or drug carriers to regulate functions of cells by delivering mediators to maintain homeostasis during liver diseases106. For example, EVs secreted from liver stem cells contained elevated levels of miRNA let-7a, and injection of stem cell-derived EVs ameliorated ductular reaction and liver fibrosis in Mdr2−/− mice by delivering cargo let-7a107. Tunicamycin is a drug that causes hepatic ER stress followed by steatosis in mice and used as a drug-induced NAFLD model. Rong et al. isolated EVs secreted from cultured primary adipocytes harvested from mice treated with melatonin (20 mg/kg daily for 2 weeks, i.p. injection) and injected those EVs into tunicamycin-treated mice108. Tunicamycin caused elevated hepatic triglyceride levels, ER stress gene expression, such as C/EBP homologous protein (CHOP), 78-kDa glucose-regulated protein (GRP78), and inositol-requiring enzyme 1 (IRE1), elevated de novo lipogenesis gene expression including ACC and SREBP-1c, and decreased β-oxidation gene expression such as CPT1108. EVs isolated from mice with melatonin administration ameliorated tunicamycin-induced gene expression changes and hepatic steatosis in vivo108. This study has demonstrated that resistin, which is an adipocytokine associated with obesity, is a key cargo mediator and causes hepatic steatosis via EVs delivered from adipose tissues into the liver108. Although current studies are limited and further experimental evidence is required, EVs may have the great potentials as drug/mediator carriers to deliver melatonin or melatonin-induced elevated levels of proteins or RNAs leading to the regulations of hepatic cells and improvements of liver conditions.
4. Conclusions and future perspectives
Current studies suggest the association between circadian rhythms as well as melatonin secretion with pathophysiological events in various liver diseases. Table 1 summarizes effects of disruption of circadian rhythms leading to liver diseases. Melatonin administration has numbers of therapeutic effects in liver disorders, which are not limited to antioxidant effects. Figure 1 and Table 2 summarize therapeutic effects of melatonin administration shown in recent studies. Disruption of circadian rhythms and altered clock gene expressions have been identified in various liver diseases, and melatonin administration can change expression levels of clock genes. Although detailed mechanisms are still not fully elucidated, expression changes of clock genes and circadian rhythms may contribute to the therapeutic effects of melatonin. Table 3 summarizes clock gene expression in liver diseases and effects of melatonin administration. Melatonin treatments mainly focus on the regulation of functions or cell events in target cells (e.g., restoration of mitochondrial functions in hepatocytes to inhibit lipid deposition during NAFLD49). However, hepatic cells communicate with each other and contribute in orchestration to the pathophysiology of liver diseases109. Melatonin administration in the future may target cell-to-cell communication between hepatic cells to inhibit pathological cellular communication during liver diseases (e.g., improved EV-mediated communication between adipose tissue and the liver during NAFLD108). Current studies of melatonin therapies have limitations. A number of studies use various doses and administration methods (intraperitoneal injection or via drinking water), and there are no gold-standard methods for melatonin treatments. Although a majority of current studies use 10 mg/kg/day for rats or mice, this dose may not be reasonable for human patients because melatonin tablets that can be purchased over the counter are 3 or 5 mg. Daily melatonin supplementation (3 mg) for 3 months increased brown adipose tissue mass in patients with melatonin deficiency44. Although 3 or 5 mg melatonin may be effective in humans, current studies using human subjects are limited and detailed effects and optimal doses of melatonin are still undefined. In addition, melatonin regulates circadian rhythms and melatonin secretion is rhythmic in healthy individuals, but current studies merely supplement melatonin and do not consider the timing of supplementation or rhythms. Melatonin administration via drinking water does not mimic rhythmic melatonin secretion. It is not fully elucidated whether melatonin has therapeutic effects regardless of circadian rhythms or rhythmic melatonin supplementation restores disrupted rhythms and has better effects. Further studies are required to understand the roles of melatonin and circadian rhythms in liver diseases and to develop effective and feasible melatonin treatments for patients with liver diseases.
Table 1.
Selected effects of circadian disruption leading to liver diseases.
| Effects | Associated disease | Model |
|---|---|---|
| Elevation of gut permeability | ALD | Nighttime workers with moderate alcohol drinking20 |
| Elevated liver damage and steatosis | ALD | Liver-specific BMAL1 knockout mice with alcohol feeding25,26 |
| Increased glucose tolerance | NAFLD | Rats with light exposure at night32 |
| Increased body weight and adipose tissue | NAFLD | Rats with delayed feeding33 |
| Increased hepatic lipid accumulation | NAFLD | Mice with HFD at daytime34 |
| Impaired lipid metabolism | NAFLD | Liver-specific BMAL1 knockout mice37 |
| Elevated liver damage | Cholestasis | BDL rats with complete light64 |
| High HCC development | HCC | Jet-lagged mice78 |
| High incidence of complications | IRI | Patients with liver transplantation at night99 |
Figure 1. Therapeutic effects of melatonin in liver diseases.

Current studies have represented various therapeutic effects of melatonin administration that are not limited to antioxidant effects. Melatonin decreases de novo lipogenesis in the liver and increases the mass and activities of brown adipose tissues leading to attenuated liver steatosis in NAFLD. Melatonin can increase functions and enzyme activities in mitochondria, which promotes β-oxidation of FFAs and inhibits lipid accumulation in the liver. Melatonin also increases circulating white blood cells and diversity of gut microbiota contributing to improved liver conditions. Proliferation of cholangiocytes can be regulated by melatonin and melatonin administration could be utilized to inhibit ductular reaction and liver fibrosis in cholangiopathies such as PSC. Melatonin inhibits proliferation and tumor growth of HCC and CCA and could be utilized to increase the effects of chemotherapy.
Table 2.
Selected therapeutic effects of melatonin administration in liver diseases.
| Effects | Dose | Associated disease | Model |
|---|---|---|---|
| Increase of circulating white blood cells | 10 mg/kg/day | ALD | Mouse with ethanol injection29 |
| Decreased liver damage | 10 or 15 mg/kg/day | ALD | Pregnant rat with ethanol gavage30 |
| Decreased oxidative and ER stress | 500 μg/kg/day | NAFLD | Ob/ob mice40 |
| Improved steatosis and insulin resistance | 100 mg/kg/day | NAFLD | HFD mice41 |
| Decreased de novo lipogenesis | 10–50 mg/kg | NAFLD | HFD mice42 |
| Increase of brown adipose tissue mass and activity | 3 mg/day | NAFLD | Patients with melatonin deficiency44 |
| Restored mitochondria functions | 10 or 20 mg/kg/day | NAFLD | HFD mice49,50 |
| Improved gut microbiota diversity | 108 mg/kg/day | NAFLD | HFD mice52 |
| Improved liver conditions | 2 mg/g/day | PSC | Mdr2−/− mice65 |
| Improved liver conditions | 5 mg/kg/day | Bile duct injury | Rats with TAA administration66 |
| Decreased liver damage and steatosis | 2.5–20 mg/kg/day | Toxin-induced liver damage | Rats with CCl4 administration67,68 |
| Improved clock gene expression | 5 or 10 mg/kg/day | Toxin-induced liver damage | Mice with CCl4 administration71 |
| Inhibition of tumor development | 5 or 10 mg/kg/day | HCC | Mice with DEN administration85 |
| Increased sensitivity to chemotherapy | 40 mg/kg, 5 days per week | HCC | Xenograft mice with etoposide administration89 |
| Decreased tumor size | 4 mg/kg, 3 times per week | CCA | Xenograft mice91 |
| Inhibition of tumor development | 10 or 50 mg/kg/day | CCA | Hamsters with O. viverrini infection and NDMA administration94 |
| Reduced liver damage | 10 mg/kg | IRI | Rats with porta hepatis clamping and reperfusion100 |
| Promoted liver regeneration | 10 mg/kg | Partial liver transplantation | Mice with right lobe clamping and partial hepatectomy102 |
Table 3.
Expression changes of clock genes in liver diseases and effects of melatonin.
| Disease/model | Clock gene expression | Effects of melatonin |
|---|---|---|
| Alcohol drinking |
PER1↑, CRY1↓ in humans20 CLOCK↑, PER2↓ in mice21 |
Undefined |
| Four hour-shifted feeding mice | Shifted expression oscillation for BMAL1, CLOCK, PER1/2, CRY1/233 | Undefined |
| Daytime or nighttime feeding mice | Different expression pattern for PER1 and PER234 | Undefined |
| BDL rats | BMAL1↑, CLOCK↑, PER1↑, CRY1↑14 | BMAL1↓, CLOCK↓, PER1↓, CRY1↓14 |
| Human PSC patients | BMAL1↑, CLOCK↑, PER1↑, CRY1↑64 | Undefined |
| CCl4-treated mice | BMAL1↓, CLOCK↓, PER1/2/3↓, CRY1/2↓71 | BMAL1↑, CLOCK↑, PER1/2/3↑, CRY1/2↑71 |
| DEN-induced HCC mice | BMAL1↑, CLOCK↑, PER1/2/3↓, CRY1↓85 | BMAL1↓, CLOCK↓, PER1/2/3↑, CRY1↑85 |
| CCA |
BMAL1↑, PER1↓ in CCA tumors90 Lost rhythmic expression for BMAL1, CLOCK, PER1, CRY1 in CCA cell lines90 |
Undefined |
In conclusion, melatonin has protective effects against various liver diseases, and circadian rhythms and clock gene expressions are promising therapeutic targets for the management of liver conditions.
Acknowledgments
This work was supported by: The Senior Research Career Scientist Award to Dr. Alpini and the VA Merit awards to Dr. Meng (1I01BX001724), Dr. Glaser (5I01BX002192), Dr. Francis (1I01BX003031), and Dr. Alpini (5I01BX000574) from the United States Department of Veteran’s Affairs Biomedical Laboratory Research and Development Service; U.S. National Institutes of Health (NIH) National Institute of Diabetes and Digestive and Kidney Diseases Grants DK108959, DK119421, DK115184, DK054811, DK076898, DK107310, DK110035, and DK062975 to Drs. Meng, Glaser, Francis, and Alpini and NIH National Institute on Alcohol Abuse and Alcoholism Grants AA025997 and AA025157 to Drs. Meng, Glaser, and Alpini; The Hickam Endowed Chair, Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine to Dr. Alpini; Development Service by University of Rome “La Sapienza” to Dr. Onori. The project described was supported by the Indiana University Health - Indiana University School of Medicine Strategic Research Initiative. Dr. Alpini acknowledges the support from PSC Partners Seeking a Cure. This material is the result of work supported by resources at the Central Texas Veterans Health Care System, Temple, TX, Richard L. Roudebush VA Medical Center, Indianapolis, IN, and Medical Physiology, Medical Research Building, Temple, TX. The views expressed in this article are those of the authors and do not necessarily represent the views of the Department of Veterans Affairs.
Abbreviations
- AANAT
aralkylamine N-acetyltransferase
- ACC
acetyl-CoA carboxylase
- Alb
albumin
- ALD
alcoholic liver disease
- Alfp
α-fetoprotein
- ALT
aminotransferase
- ANIT
α-naphthylisothiocyanate
- ASK1
apoptosis signal-regulating kinase 1
- ASMT
N-acetylserotonin O-methyltransferase
- AST
aminotransferase
- ATF4
activating transcription factor 4
- BAX
Bcl-2-associated X protein
- Bcl-2
B-cell lymphoma 2
- BDL
bile duct ligation
- BMAL1
brain and muscle ARNT-like 1
- CAR
constitutive androstane receptor
- CCA
cholangiocarcinoma
- CCl4
carbon tetrachloride
- CHOP
C/EBP homologous protein
- CLOCK
circadian locomotor output cycles kaput
- CPT1
carnitine palmitoyltransferase I
- CRY1
cryptochrome circadian regulator 1
- CTNNB1
catenin beta 1
- DEN
diethylnitrosamine
- DEX
dexamethasone
- ER
endoplasmic reticulum
- FAS
fatty acid synthase
- FFAs
free fatty acids
- GnRH
gonadotropin releasing hormone
- GRP78
78-kDa glucose-regulated protein
- HCC
hepatocellular carcinoma
- HFD
high-fat diet
- HSCs
hepatic stellate cells
- IL
interleukin
- IRE1
inositol-requiring enzyme 1
- IRI
ischemia-reperfusion injury
- JNK
c-Jun N-terminal kinase
- lncRNA
long non-coding RNA
- LPS
lipopolysaccharide
- MAPK
mitogen-activated protein kinase
- miRNA
microRNA
- MKK
mitogen-activated protein kinase kinase
- mPTP
mitochondrial permeability transition pore
- NAFLD
non-alcoholic fatty liver disease
- NDMA
N-nitrosodimethylamine
- NR4A1
nuclear receptor subfamily 4 group A member 1
- PBC
primary biliary cholangitis
- PER1
period circadian protein homolog 1
- PERK
protein kinase R-like endoplasmic reticulum kinase
- PPARα
peroxisome proliferator-activated receptor alpha
- PSC
primary sclerosing cholangitis
- ROS
reactive oxygen species
- SCD1
stearoyl-CoA desaturase 1
- SCN
suprachiasmatic nucleus
- SNPs
single nucleotide polymorphisms
- SREBP
sterol regulatory element-binding protein
- TAA
thioacetamide
- TERT
telomerase reverse transcriptase
- TGF-β1
transforming growth factor beta 1
- TNFα
tumor necrosis factor alpha
- TRAIL
tumor necrosis factor-related apoptosis-inducing ligand
- TP53
tumor protein p53
- VEGF
vascular endothelial growth factor
- VPA
valproic acid
- ZT
zeitgeber time
Footnotes
Conflict of interest:
The authors declare no conflict of interest.
References
- 1.Hastings MH, Maywood ES, Brancaccio M. Generation of circadian rhythms in the suprachiasmatic nucleus. Nat Rev Neurosci. 2018;19(8):453–469. [DOI] [PubMed] [Google Scholar]
- 2.Kyriacou CP, Hastings MH. Circadian clocks: genes, sleep, and cognition. Trends Cogn Sci. 2010;14(6):259–267. [DOI] [PubMed] [Google Scholar]
- 3.Takahashi JS. Transcriptional architecture of the mammalian circadian clock. Nat Rev Genet. 2017;18(3):164–179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Cardoso TF, Quintanilla R, Castello A, et al. Analysing the expression of eight clock genes in five tissues from fasting and fed sows. Front Genet. 2018;9:475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Koronowski KB, Kinouchi K, Welz PS, et al. Defining the independence of the liver circadian clock. Cell. 2019;177(6):1448–1462 e1414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Muller M, Wetzel S, Kohn-Gaone J, et al. A disintegrin and metalloprotease 10 (ADAM10) is a central regulator of murine liver tissue homeostasis. Oncotarget. 2016;7(14):17431–17441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Roenneberg T, Merrow M. The circadian clock and human health. Curr Biol. 2016;26(10):R432–443. [DOI] [PubMed] [Google Scholar]
- 8.Mukherji A, Bailey SM, Staels B, Baumert TF. The circadian clock and liver function in health and disease. J Hepatol. 2019;71(1):200–211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Zisapel N New perspectives on the role of melatonin in human sleep, circadian rhythms and their regulation. Br J Pharmacol. 2018;175(16):3190–3199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Zhao D, Yu Y, Shen Y, et al. Melatonin synthesis and function: Evolutionary history in animals and plants. Front Endocrinol (Lausanne). 2019;10:249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Velarde E, Cerda-Reverter JM, Alonso-Gomez AL, Sanchez E, Isorna E, Delgado MJ. Melatonin-synthesizing enzymes in pineal, retina, liver, and gut of the goldfish (Carassius): mRNA expression pattern and regulation of daily rhythms by lighting conditions. Chronobiol Int. 2010;27(6):1178–1201. [DOI] [PubMed] [Google Scholar]
- 12.Poon AM, Choy EH, Pang SF. Modulation of blood glucose by melatonin: a direct action on melatonin receptors in mouse hepatocytes. Biol Signals Recept. 2001;10(6):367–379. [DOI] [PubMed] [Google Scholar]
- 13.Aust S, Thalhammer T, Humpeler S, et al. The melatonin receptor subtype MT1 is expressed in human gallbladder epithelia. J Pineal Res. 2004;36(1):43–48. [DOI] [PubMed] [Google Scholar]
- 14.Renzi A, Glaser S, DeMorrow S, et al. Melatonin inhibits cholangiocyte hyperplasia in cholestatic rats by interaction with MT1 but not MT2 melatonin receptors. Am J Physiol Gastrointest Liver Physiol. 2011;301(4):G634–643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Reiter RJ, Mayo JC, Tan DX, Sainz RM, Alatorre-Jimenez M, Qin L. Melatonin as an antioxidant: under promises but over delivers. J Pineal Res. 2016;61(3):253–278. [DOI] [PubMed] [Google Scholar]
- 16.Zhang JJ, Meng X, Li Y, et al. Effects of melatonin on liver injuries and diseases. Int J Mol Sci. 2017;18(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Baiocchi L, Zhou T, Liangpunsakul S, et al. Possible application of melatonin treatment in human diseases of the biliary tract. Am J Physiol Gastrointest Liver Physiol. 2019;317(5):G651–G660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Zhou Z, Zhong W. Targeting the gut barrier for the treatment of alcoholic liver disease. Liver Res. 2017;1(4):197–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Bala S, Marcos M, Gattu A, Catalano D, Szabo G. Acute binge drinking increases serum endotoxin and bacterial DNA levels in healthy individuals. PLoS One. 2014;9(5):e96864. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Swanson GR, Gorenz A, Shaikh M, et al. Night workers with circadian misalignment are susceptible to alcohol-induced intestinal hyperpermeability with social drinking. Am J Physiol Gastrointest Liver Physiol. 2016;311(1):G192–201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Gaucher J, Kinouchi K, Ceglia N, et al. Distinct metabolic adaptation of liver circadian pathways to acute and chronic patterns of alcohol intake. Proceedings of the National Academy of Sciences of the United States of America. 2019;116(50):25250–25259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Voigt RM, Forsyth CB, Shaikh M, et al. Diurnal variations in intestinal barrier integrity and liver pathology in mice: implications for alcohol binge. Am J Physiol Gastrointest Liver Physiol. 2018;314(1):G131–G141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Vera LM, Bello C, Paredes JF, Carmona-Antonanzas G, Sanchez-Vazquez FJ. Ethanol toxicity differs depending on the time of day. PLoS One. 2018;13(1):e0190406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Newberry EP, Xie Y, Lodeiro C, et al. Hepatocyte and stellate cell deletion of liver fatty acid binding protein reveals distinct roles in fibrogenic injury. FASEB J. 2019;33(3):4610–4625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Zhang D, Tong X, Nelson BB, et al. The hepatic BMAL1/AKT/lipogenesis axis protects against alcoholic liver disease in mice via promoting PPARalpha pathway. Hepatology. 2018;68(3):883–896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Udoh US, Valcin JA, Swain TM, et al. Genetic deletion of the circadian clock transcription factor BMAL1 and chronic alcohol consumption differentially alter hepatic glycogen in mice. Am J Physiol Gastrointest Liver Physiol. 2018;314(3):G431–G447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Ceni E, Mello T, Galli A. Pathogenesis of alcoholic liver disease: role of oxidative metabolism. World J Gastroenterol. 2014;20(47):17756–17772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Hu S, Yin S, Jiang X, Huang D, Shen G. Melatonin protects against alcoholic liver injury by attenuating oxidative stress, inflammatory response, and apoptosis. Eur J Pharmacol. 2009;616(1–3):287–292. [DOI] [PubMed] [Google Scholar]
- 29.Kurhaluk N, Sliuta A, Kyriienko S, Winklewski PJ. Melatonin restores white blood cell count, diminishes glycated haemoglobin level and prevents liver, kidney and muscle oxidative stress in mice exposed to acute ethanol intoxication. Alcohol Alcohol. 2017;52(5):521–528. [DOI] [PubMed] [Google Scholar]
- 30.Sousa Coelho IDD, Lapa Neto CJC, Souza T, et al. Protective effect of exogenous melatonin in rats and their offspring on the genotoxic response induced by the chronic consumption of alcohol during pregnancy. Mutat Res Genet Toxicol Environ Mutagen. 2018;832–833:52–60. [DOI] [PubMed] [Google Scholar]
- 31.Wang F, Zhang L, Wu S, et al. Night shift work and abnormal liver function: is non-alcohol fatty liver a necessary mediator? Occup Environ Med. 2019;76(2):83–89. [DOI] [PubMed] [Google Scholar]
- 32.Opperhuizen AL, Stenvers DJ, Jansen RD, Foppen E, Fliers E, Kalsbeek A. Light at night acutely impairs glucose tolerance in a time-, intensity- and wavelength-dependent manner in rats. Diabetologia. 2017;60(7):1333–1343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Shimizu H, Hanzawa F, Kim D, et al. Delayed first active-phase meal, a breakfast-skipping model, led to increased body weight and shifted the circadian oscillation of the hepatic clock and lipid metabolism-related genes in rats fed a high-fat diet. PLoS One. 2018;13(10):e0206669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Okauchi H, Hashimoto C, Nakao R, Oishi K. Timing of food intake is more potent than habitual voluntary exercise to prevent diet-induced obesity in mice. Chronobiol Int. 2019;36(1):57–74. [DOI] [PubMed] [Google Scholar]
- 35.Greenwell BJ, Trott AJ, Beytebiere JR, et al. Rhythmic food intake drives rhythmic gene expression more potently than the hepatic circadian clock in mice. Cell Rep. 2019;27(3):649–657 e645. [DOI] [PubMed] [Google Scholar]
- 36.Kettner NM, Mayo SA, Hua J, Lee C, Moore DD, Fu L. Circadian dysfunction induces leptin resistance in mice. Cell Metab. 2015;22(3):448–459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Zhong X, Yu J, Frazier K, et al. Circadian clock regulation of hepatic lipid metabolism by modulation of m6A mRNA methylation. Cell Rep. 2018;25(7):1816–1828 e1814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Pawlak M, Lefebvre P, Staels B. Molecular mechanism of PPARα action and its impact on lipid metabolism, inflammation and fibrosis in non-alcoholic fatty liver disease. J Hepatol. 2015;62(3):720–733. [DOI] [PubMed] [Google Scholar]
- 39.Masarone M, Rosato V, Dallio M, et al. Role of oxidative stress in pathophysiology of nonalcoholic fatty liver disease. Oxid Med Cell Longev. 2018;2018:9547613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.de Luxan-Delgado B, Potes Y, Rubio-Gonzalez A, et al. Melatonin reduces endoplasmic reticulum stress and autophagy in liver of leptin-deficient mice. J Pineal Res. 2016;61(1):108–123. [DOI] [PubMed] [Google Scholar]
- 41.Heo JI, Yoon DW, Yu JH, et al. Melatonin improves insulin resistance and hepatic steatosis through attenuation of alpha-2-HS-glycoprotein. J Pineal Res. 2018;65(2):e12493. [DOI] [PubMed] [Google Scholar]
- 42.Ou TH, Tung YT, Yang TH, Chien YW. Melatonin improves fatty liver syndrome by inhibiting the lipogenesis pathway in hamsters with high-fat diet-induced hyperlipidemia. Nutrients. 2019;11(4). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Poekes L, Gillard J, Farrell GC, Horsmans Y, Leclercq IA. Activation of brown adipose tissue enhances the efficacy of caloric restriction for treatment of nonalcoholic steatohepatitis. Lab Invest. 2019;99(1):4–16. [DOI] [PubMed] [Google Scholar]
- 44.Halpern B, Mancini MC, Bueno C, et al. Melatonin increases brown adipose tissue volume and activity in patients with melatonin deficiency: A proof-of-concept study. Diabetes. 2019;68(5):947–952. [DOI] [PubMed] [Google Scholar]
- 45.Mi Y, Tan D, He Y, Zhou X, Zhou Q, Ji S. Melatonin modulates lipid metabolism in HepG2 cells cultured in high concentrations of oleic acid: AMPK pathway activation may play an important role. Cell Biochem Biophys. 2018;76(4):463–470. [DOI] [PubMed] [Google Scholar]
- 46.Li DJ, Tong J, Li YH, et al. Melatonin safeguards against fatty liver by antagonizing TRAFs-mediated ASK1 deubiquitination and stabilization in a beta-arrestin-1 dependent manner. J Pineal Res. 2019;67(4):e12611. [DOI] [PubMed] [Google Scholar]
- 47.Sun H, Wang X, Chen J, et al. Melatonin improves non-alcoholic fatty liver disease via MAPK-JNK/P38 signaling in high-fat-diet-induced obese mice. Lipids Health Dis. 2016;15(1):202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Panel M, Ghaleh B, Morin D. Mitochondria and aging: A role for the mitochondrial transition pore? Aging Cell. 2018;17(4):e12793. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Das N, Mandala A, Naaz S, et al. Melatonin protects against lipid-induced mitochondrial dysfunction in hepatocytes and inhibits stellate cell activation during hepatic fibrosis in mice. J Pineal Res. 2017;62(4). [DOI] [PubMed] [Google Scholar]
- 50.Zhou H, Du W, Li Y, et al. Effects of melatonin on fatty liver disease: The role of NR4A1/DNA-PKcs/p53 pathway, mitochondrial fission, and mitophagy. J Pineal Res. 2018;64(1). [DOI] [PubMed] [Google Scholar]
- 51.Campo L, Eiseler S, Apfel T, Pyrsopoulos N. Fatty Liver Disease and Gut Microbiota: A Comprehensive Update. J Clin Transl Hepatol. 2019;7(1):56–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Yin J, Li Y, Han H, et al. Melatonin reprogramming of gut microbiota improves lipid dysmetabolism in high-fat diet-fed mice. J Pineal Res. 2018;65(4):e12524. [DOI] [PubMed] [Google Scholar]
- 53.Sato K, Meng F, Fava G, Glaser S, Alpini G. Functional roles of gut bacteria imbalance in cholangiopathies. Liver Res. 2019;3(1):40–45. [Google Scholar]
- 54.Sato K, Glaser S, Kennedy L, et al. Preclinical insights into cholangiopathies: Disease modeling and emerging therapeutic targets. Expert Opin Ther Targets. 2019;23(6):461–472. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Lazaridis KN, LaRusso NF. Primary sclerosing cholangitis. N Engl J Med. 2016;375(12):1161–1170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Montagnese S, Nsemi LM, Cazzagon N, et al. Sleep-Wake profiles in patients with primary biliary cirrhosis. Liver Int. 2013;33(2):203–209. [DOI] [PubMed] [Google Scholar]
- 57.Turco M, Cazzagon N, Franceschet I, et al. Morning bright light treatment for sleep-wake disturbances in primary biliary cholangitis: A pilot study. Front Physiol. 2018;9:1530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Wu N, Meng F, Zhou T, et al. The secretin/secretin receptor axis modulates ductular reaction and liver fibrosis through changes in transforming growth factor-beta1-mediated biliary senescence. Am J Pathol. 2018;188(10):2264–2280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Renzi A, DeMorrow S, Onori P, et al. Modulation of the biliary expression of arylalkylamine N-acetyltransferase alters the autocrine proliferative responses of cholangiocytes in rats. Hepatology. 2013;57(3):1130–1141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Renzi A, Mancinelli R, Onori P, et al. Inhibition of the liver expression of arylalkylamine N-acetyltransferase increases the expression of angiogenic factors in cholangiocytes. Hepatobiliary Surg Nutr. 2014;3(1):4–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.McMillin M, DeMorrow S, Glaser S, et al. Melatonin inhibits hypothalamic gonadotropin-releasing hormone release and reduces biliary hyperplasia and fibrosis in cholestatic rats. Am J Physiol Gastrointest Liver Physiol. 2017;313(5):G410–G418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Kyritsi K, Meng F, Zhou T, et al. 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. 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Han Y, Onori P, Meng F, et al. Prolonged exposure of cholestatic rats to complete dark inhibits biliary hyperplasia and liver fibrosis. Am J Physiol Gastrointest Liver Physiol. 2014;307(9):G894–904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Chen L, Zhou T, Wu N, et al. Pinealectomy or light exposure exacerbates biliary damage and liver fibrosis in cholestatic rats through decreased melatonin synthesis. Biochim Biophys Acta Mol Basis Dis. 2019;1865(6):1525–1539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Wu N, Meng F, Zhou T, et al. Prolonged darkness reduces liver fibrosis in a mouse model of primary sclerosing cholangitis by miR-200b down-regulation. FASEB J. 2017;31(10):4305–4324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Lebda MA, Sadek KM, Abouzed TK, Tohamy HG, El-Sayed YS. Melatonin mitigates thioacetamide-induced hepatic fibrosis via antioxidant activity and modulation of proinflammatory cytokines and fibrogenic genes. Life Sci. 2018;192:136–143. [DOI] [PubMed] [Google Scholar]
- 67.Wang YR, Hong RT, Xie YY, Xu JM. Melatonin ameliorates liver fibrosis induced by carbon tetrachloride in rats via inhibiting TGF-beta1/Smad signaling pathway. Curr Med Sci. 2018;38(2):236–244. [DOI] [PubMed] [Google Scholar]
- 68.Bona S, Rodrigues G, Moreira AJ, et al. Antifibrogenic effect of melatonin in rats with experimental liver cirrhosis induced by carbon tetrachloride. JGH Open. 2018;2(4):117–123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Nickovic VP, Novakovic T, Lazarevic S, et al. Pre- vs. post-treatment with melatonin in CCl4-induced liver damage: Oxidative stress inferred from biochemical and pathohistological studies. Life Sci. 2018;202:28–34. [DOI] [PubMed] [Google Scholar]
- 70.Mortezaee K, Majidpoor J, Daneshi E, Abouzaripour M, Abdi M. Post-treatment of melatonin with CCl4 better reduces fibrogenic and oxidative changes in liver than melatonin co-treatment. J Cell Biochem. 2018;119(2):1716–1725. [DOI] [PubMed] [Google Scholar]
- 71.Gonzalez-Fernandez B, Sanchez DI, Crespo I, et al. Melatonin attenuates dysregulation of the circadian clock pathway in mice with CCl4-induced fibrosis and human hepatic stellate cells. Front Pharmacol. 2018;9:556. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 72.Kwak KA, Cho HJ, Yang JY, Park YS. Current perspectives regarding stem cell-based therapy for liver cirrhosis. Can J Gastroenterol Hepatol. 2018;2018:4197857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Mortezaee K, Khanlarkhani N, Sabbaghziarani F, et al. Preconditioning with melatonin improves therapeutic outcomes of bone marrow-derived mesenchymal stem cells in targeting liver fibrosis induced by CCl4. Cell Tissue Res. 2017;369(2):303–312. [DOI] [PubMed] [Google Scholar]
- 74.Li Y, Yu H, Xu Z, et al. Melatonin ameliorates ANIT-induced cholestasis by activating Nrf2 through a PI3K/Akt-dependent pathway in rats. Mol Med Rep. 2019;19(2):1185–1193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Oztopuz O, Turkon H, Buyuk B, et al. Melatonin ameliorates sodium valproate-induced hepatotoxicity in rats. Mol Biol Rep. 2019. [DOI] [PubMed] [Google Scholar]
- 76.Tsai CC, Lin YJ, Yu HR, et al. Regulation of leptin methylation not via apoptosis by melatonin in the rescue of chronic programming liver steatosis. Int J Mol Sci. 2018;19(11). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Masri S, Sassone-Corsi P. The emerging link between cancer, metabolism, and circadian rhythms. Nat Med. 2018;24(12):1795–1803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Kettner NM, Voicu H, Finegold MJ, et al. Circadian homeostasis of liver metabolism suppresses hepatocarcinogenesis. Cancer Cell. 2016;30(6):909–924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Fekry B, Ribas-Latre A, Baumgartner C, et al. Incompatibility of the circadian protein BMAL1 and HNF4alpha in hepatocellular carcinoma. Nat Commun. 2018;9(1):4349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Niu ZS, Niu XJ, Wang WH. Genetic alterations in hepatocellular carcinoma: An update. World J Gastroenterol. 2016;22(41):9069–9095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Su SC, Ho YC, Liu YF, et al. Association of melatonin membrane receptor 1A/1B gene polymorphisms with the occurrence and metastasis of hepatocellular carcinoma. Oncotarget. 2017;8(49):85655–85669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Dauchy RT, Hoffman AE, Wren-Dail MA, et al. Daytime blue light enhances the nighttime circadian melatonin inhibition of human prostate cancer growth. Comp Med. 2015;65(6):473–485. [PMC free article] [PubMed] [Google Scholar]
- 83.Dauchy RT, Wren-Dail MA, Dupepe LM, et al. Effect of daytime blue-enriched LED light on the nighttime circadian melatonin inhibition of hepatoma 7288CTC warburg effect and progression. Comp Med. 2018;68(4):269–279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Friemel J, Frick L, Unger K, et al. Characterization of HCC mouse models: Towards an etiology-oriented subtyping approach. Mol Cancer Res. 2019;17(7):1493–1502. [DOI] [PubMed] [Google Scholar]
- 85.Sanchez DI, Gonzalez-Fernandez B, Crespo I, et al. Melatonin modulates dysregulated circadian clocks in mice with diethylnitrosamine-induced hepatocellular carcinoma. J Pineal Res. 2018;65(3):e12506. [DOI] [PubMed] [Google Scholar]
- 86.El-Magd MA, Mohamed Y, El-Shetry ES, et al. Melatonin maximizes the therapeutic potential of non-preconditioned MSCs in a DEN-induced rat model of HCC. Biomed Pharmacother. 2019;114:108732. [DOI] [PubMed] [Google Scholar]
- 87.Le Grazie M, Biagini MR, Tarocchi M, Polvani S, Galli A. Chemotherapy for hepatocellular carcinoma: The present and the future. World J Hepatol. 2017;9(21):907–920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Zhou B, Lu Q, Liu J, et al. Melatonin increases the sensitivity of hepatocellular carcinoma to sorafenib through the PERK-ATF4-Beclin1 pathway. Int J Biol Sci. 2019;15(9):1905–1920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Chen CC, Chen CY, Wang SH, et al. Melatonin sensitizes hepatocellular carcinoma cells to chemotherapy through long non-coding RNA RAD51-AS1-mediated suppression of DNA repair. Cancers (Basel). 2018;10(9). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Han Y, Meng F, Venter J, et al. miR-34a-dependent overexpression of Per1 decreases cholangiocarcinoma growth. J Hepatol. 2016;64(6):1295–1304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Han Y, DeMorrow S, Invernizzi P, et al. Melatonin exerts by an autocrine loop antiproliferative effects in cholangiocarcinoma: its synthesis is reduced favoring cholangiocarcinoma growth. Am J Physiol Gastrointest Liver Physiol. 2011;301(4):G623–633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Laothong U, Hiraku Y, Oikawa S, Intuyod K, Murata M, Pinlaor S. Melatonin induces apoptosis in cholangiocarcinoma cell lines by activating the reactive oxygen species-mediated mitochondrial pathway. Oncol Rep. 2015;33(3):1443–1449. [DOI] [PubMed] [Google Scholar]
- 93.Khan AS, Dageforde LA. Cholangiocarcinoma. Surg Clin North Am. 2019;99(2):315–335. [DOI] [PubMed] [Google Scholar]
- 94.Laothong U, Pinlaor P, Boonsiri P, et al. Melatonin inhibits cholangiocarcinoma and reduces liver injury in Opisthorchis viverrini-infected and N-nitrosodimethylamine-treated hamsters. J Pineal Res. 2013;55(3):257–266. [DOI] [PubMed] [Google Scholar]
- 95.Fabris L, Perugorria MJ, Mertens J, et al. The tumour microenvironment and immune milieu of cholangiocarcinoma. Liver Int. 2019;39:63–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Wongsena W, Charoensuk L, Dangtakot R, Pinlaor P, Intuyod K, Pinlaor S. Melatonin suppresses eosinophils and Th17 cells in hamsters treated with a combination of human liver fluke infection and a chemical carcinogen. Pharmacol Rep. 2018;70(1):98–105. [DOI] [PubMed] [Google Scholar]
- 97.Dar WA, Sullivan E, Bynon JS, Eltzschig H, Ju C. Ischaemia reperfusion injury in liver transplantation: Cellular and molecular mechanisms. Liver Int. 2019;39(5):788–801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Hryniewiecka E, Pilecki T, Zieniewicz K, Paczek L. Circadian and short-term blood pressure abnormalities after liver transplantation. Clin Exp Hypertens. 2018;40(8):730–733. [DOI] [PubMed] [Google Scholar]
- 99.Ren SS, Xu LL, Wang P, et al. Circadian rhythms have effects on surgical outcomes of liver transplantation for patients with hepatocellular carcinoma: A retrospective analysis of 147 cases in a single center. Transplant Proc. 2019;51(6):1913–1919. [DOI] [PubMed] [Google Scholar]
- 100.Deng WS, Xu Q, Liu YE, Jiang CH, Zhou H, Gu L. Effects of melatonin on liver function and lipid peroxidation in a rat model of hepatic ischemia/reperfusion injury. Exp Ther Med. 2016;11(5):1955–1960. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Kim PT, Testa G. Living donor liver transplantation in the USA. Hepatobiliary Surg Nutr. 2016;5(2):133–140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Song Z, Humar B, Gupta A, et al. Exogenous melatonin protects small-for-size liver grafts by promoting monocyte infiltration and releases interleukin-6. J Pineal Res. 2018;65(1):e12486. [DOI] [PubMed] [Google Scholar]
- 103.Sato K, Meng F, Glaser S, Alpini G. Exosomes in liver pathology. J Hepatol. 2016;65(1):213–221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Hirsova P, Ibrahim SH, Krishnan A, et al. Lipid-induced signaling causes release of inflammatory extracellular vesicles from hepatocytes. Gastroenterology. 2016;150(4):956–967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Liu R, Li X, Zhu W, et al. Cholangiocyte-derived exosomal long noncoding RNA H19 promotes hepatic stellate cell activation and cholestatic liver fibrosis. Hepatology. 2019;70(4):1317–1335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Sato K, Meng F, Venter J, Giang T, Glaser S, Alpini G. The role of the secretin/secretin receptor axis in inflammatory cholangiocyte communication via extracellular vesicles. Sci Rep. 2017;7(1):11183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.McDaniel K, Wu N, Zhou T, et al. Amelioration of ductular reaction by stem cell derived extracellular vesicles in MDR2 knockout mice via lethal-7 microRNA. Hepatology. 2019;69(6):2562–2578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Rong B, Feng R, Liu C, Wu Q, Sun C. Reduced delivery of epididymal adipocyte-derived exosomal resistin is essential for melatonin ameliorating hepatic steatosis in mice. J Pineal Res. 2019;66(4):e12561. [DOI] [PubMed] [Google Scholar]
- 109.Sato K, Kennedy L, Liangpunsakul S, et al. Intercellular communication between hepatic cells in liver diseases. Int J Mol Sci. 2019;20(9). [DOI] [PMC free article] [PubMed] [Google Scholar]
