Skip to main content
Erschienen in: Cancer Cell International 1/2021

Open Access 01.12.2021 | Review

An updated review of mechanistic potentials of melatonin against cancer: pivotal roles in angiogenesis, apoptosis, autophagy, endoplasmic reticulum stress and oxidative stress

verfasst von: Saeed Mehrzadi, Mohammad Hossein Pourhanifeh, Alireza Mirzaei, Farid Moradian, Azam Hosseinzadeh

Erschienen in: Cancer Cell International | Ausgabe 1/2021

Abstract

Cancers are serious life-threatening diseases which annually are responsible for millions of deaths across the world. Despite many developments in therapeutic approaches for affected individuals, the rate of morbidity and mortality is high. The survival rate and life quality of cancer patients is still low. In addition, the poor prognosis of patients and side effects of the present treatments underscores that finding novel and effective complementary and alternative therapies is a critical issue. Melatonin is a powerful anticancer agent and its efficiency has been widely documented up to now. Melatonin applies its anticancer abilities through affecting various mechanisms including angiogenesis, apoptosis, autophagy, endoplasmic reticulum stress and oxidative stress. Regarding the implication of mentioned cellular processes in cancer pathogenesis, we aimed to further evaluate the anticancer effects of melatonin via these mechanisms.
Hinweise
Saeed Mehrzadi and Mohammad Hossein Pourhanifeh contributed equally to this work

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
JNK
C-Jun N-terminal kinase
ROS
Reactive oxygen species
VEGF
Vascular endothelial growth factor
TNF-α
Tumor necrosis factor-α
IL-2
Interleukin-2
Nrf2
Nuclear factor erythroid 2-related factor 2
Apaf-1
Apoptotic protease activating factor-1
COX-2
Cyclooxygenase-2
Sirt1
Sirtuin
HIF
Hypoxia-inducible factor
TRAIL
TNF-related apoptosis-inducing ligand
PARP-1
Poly [ADP-ribose] polymerase 1
ATG
Autophagy related genes
IRE1
Inositol-requiring enzyme 1
ATF6
Activating transcription factor 6
ERK
Extracellular signal-regulated kinase
MEK
Mitogen-activated protein kinase kinase
MAPK
Mitogen-activated protein kinase
CHOP
CCAAT-enhancer-binding proteins homologous protein
PUMA
P53-upregulated modulator of apoptosis
Bcl-2
B cell lymphoma-2
Bim
Bcl-2-interacting mediator of cell death
PI3K
Phosphatidylinositol-3-kinase
Akt
Protein kinase B
UPR
Unfolded protein response
ATF6α
Activating transcription factor 6α
ER
Endoplasmic reticulum
PERK
Protein kinase RNA-like ER kinase
ERK
Extracellular signal-regulated kinase
SPHK1
Sphingosine kinase 1
IFN-γ
Interferon-gamma
PGC1A
Peroxisome proliferator-activated receptor gamma coactivator 1A
NQO-1
NAD(P)H:quinone oxidoreductase
HO-1
Heme oxygenase-1
FADD
Fas-associated death domain
tBid
Truncated Bid
ASK1
Apoptosis signal-regulating kinase 1
TRAF2
IRE1α-TNF receptor-associated factor 2
ERKs
Extracellular-regulated protein kinases
CaMKIIα
Ca2+/calmodulin-dependent protein kinase II alpha
DEN
Diethylnitrosamine
HCC
Hepatocellular carcinoma
MSCs
Mesenchymal stem cells
NEDD9
Neural precursor cell expressed developmentally downregulated protein 9
p90RSK
90-KDa ribosomal S6 kinase
HSP27
Heat shock protein 27

Introduction

As the second cause of mortality worldwide, new cases of cancer have recently been reported to increase by 2025 (approximately 19.3 million annually) [1]. Cancer growth control, complete eradication and preventing its incidence are main purposes for cancer-associated investigations. Chemotherapy, radiotherapy and surgery are the major conventional anticancer treatments. The restricted efficiency of these treatments as well as their dangerous side effects have forced researchers to find novel effective anticancer therapies based on herbal extracts and natural compounds as single or combined therapies [24].
Melatonin, a multifunctional pleiotropic neurohormone secreted by the pineal gland and other organs including bone marrow, retina, and skin. It is an immune regulatory agent and powerful antioxidant with a capability of preventing cell death in oxidative stress situations. [5, 6]. Moreover, melatonin interrupts cell death mechanisms, inflammation, and redox activity probably resulting in cancer cells sensitization to chemotherapy and radiation [7].
Furthermore, in addition to diverse therapeutic potentials for several diseases [8, 9], melatonin has been shown to possess anticancer abilities against skin cancer [10], glioma [11], lung cancer [12], gastrointestinal cancers [13], gynecological cancers [14, 15], and hematological cancers [16, 17]. Although mechanistic impacts of melatonin on various cancers have been widely demonstrated, in the present review we discuss anticancer effects of melatonin with focusing on molecular pathways including angiogenesis, apoptosis, autophagy, endoplasmic reticulum stress, and oxidative stress.

Melatonin, a neurohormone with a broad spectrum functions

Monitoring of circadian rhythm is one of the several properties of melatonin, which also possesses oncostatic, vasoregulation, antioxidant, anti-inflammatory, and immunomodulatory abilities [18, 19]. It has been demonstrated that the normally enhanced melatonin levels at night help in the organization of homeostatic metabolic rhythms of targeted organs and systems [20]. Of note, disruption of circadian rhythm has been shown as one of the contributing factors in cancer progression and development [21].
Melatonin, as an antioxidant agent, scavenges free radicals. Melatonin has protective effects on neurodegenerative disorders, epilepsy, and cancer through inhibiting oxidative stress in vitro and in vivo [22, 23]. Melatonin increases the activity and expression of enzymes, including catalase, superoxide dismutase and glutathione peroxidase, implicated in antioxidant abilities [24, 25]. Melatonin also has anti-inflammatory impacts and attenuates pathogenic inflammation through modulating different pathways, including reducing the secretion of tumor necrosis factor-alpha (TNF-α), interleukin-1 (IL-2) and interferon-gamma (IFN-γ), and enhancing the amounts of IL-4, IL-10 and IL-27. Melatonin alleviates pro-inflammatory cytokines secretion via suppressing nuclear factor kappa B (NF-κB) [2628]. In addition, in neurodegenerative disorders, melatonin blocks cyclooxygenase-2 (COX-2) expression, a pro-inflammatory mediator [29]. Melatonin inhibits apoptosis through regulating Bax/Bcl2 and decreasing caspase-3 activity and expression, proposing that melatonin modulates apoptotic functions in the protection against malignancy and neurodegenerative disorders [3032].
Melatonin regulates multiple physiological and neural functions (Fig. 1). Among of them, effects on blood lipid profile, glycemic control, gestation, reproduction, and fetal development, neural protection, immune system, and cardiovascular system have been widely documented [33, 34]. Melatonin prevents the growth and promotion of spontaneous and chemically mediated breast tumors [35, 36]. Moreover, at physiological concentrations, melatonin suppresses cell invasiveness and proliferation in breast cancer cells [37].

Melatonin and cancer: effect on different molecular mechanisms and cellular pathways

In this section we describe the effect of melatonin on oxidative stress and endoplasmic reticulum stress, and various signaling pathways including angiogenesis, apoptosis, autophagy affected by melatonin in different cancer cells (Fig. 2).

Melatonin and angiogenesis

Angiogenesis is a crucial event implicated in the progression of tumor as well as its metastasis [38]. Hypoxia in the central areas of solid tumor is a leading cause of angiogenesis via activation of angiogenic mediators [38, 39]. Vascular endothelial growth factor (VEGF), the specific mitogen of endothelial cells and the most active pro-angiogenic agent, is a powerful angiogenesis enhancer which increases vascular permeability. Numerous data suggest that, in tumor development, anti-VEGF therapy has important roles in the suppression of tumor cell growth, leading to a considerable amelioration in progression-free survival [40]. Hypoxia-inducible factor-1 (HIF-1) is another key factor in angiogenesis, which modulates hypoxia-activated genes transcription and consists of HIF-1α and HIF-1β heterodimer. The α subunit of HIF-1 is stabilized under hypoxia and degraded under normoxic situations, however, HIF-1β is expressed constitutively [41].
Melatonin has been shown to have regulatory role in angiogenesis process [42]. In other words, melatonin possesses various impacts on neovascularization under diverse pathological and physiological situations. In skin lesions, gastric ulcers, and some physiologic events, melatonin promotes angiogenesis, while in a hypoxic environment, in age-related ocular diseases, and in tumors melatonin suppresses neovascularization in tissues [43].
Melatonin exerts its antitumor potentials via inhibiting HIF-1-induced angiogenesis [44]. Furthermore, melatonin inhibits the accumulation of HIF-1α through suppressing the formation of ROS and the sphingosine kinase 1 (SPHK1) pathway in prostate cancer cells under hypoxic conditions [45]. Melatonin plays an important role in the paracrine interaction between proximal endothelial cells and malignant epithelial cells by a downmodulatory effect on the expression of VEGF in breast tumor cells, which reduces VEGF levels around endothelial cells [46].
Of note, anti-angiogenic potential of melatonin is a key factor resulting in the inhibition of proliferation of cancer cells, as demonstrated in various investigations. For instance, melatonin attenuates proliferation of prostate cancer cells triggered by epidermal growth factor [47]. Melatonin also hampers vasculogenic mimicry of oral cancer cells via inhibition of ROS-activated Akt and ERKs signaling pathway implicating the HIF-α pathway [48]. Melatonin up-regulates TGF-β1 expression in tumor tissues during the inhibition of gastric cancer tumor growth process [49]. Furthermore, apoptotic and anti-proliferative effects of melatonin on breast cancer cells are mediated by the simultaneous activation of the Apaf-1/caspase-dependent apoptotic pathway and the inhibition of PI3K/Akt, p300/NF-κB, and COX-2/PGE2 signaling pathways [32].
Endothelin-1 is a peptide acting as a survival factor in colon cancer, promoting angiogenesis and mediating cell proliferation. Melatonin suppresses endothelin-1 mRNA expression. Also, melatonin blocks the activity of endothelin-1 promoter modulated by NF-κβ and FoxO1 [50]. Melatonin represses ROCK-1, VEGF and HIF-1α genes expressions in oral cancer [51]. Melatonin alters the expression of inflammatory and angiogenic proteins in both co-culture and monoculture of cancer cells and cancer-associated fibroblasts [52]. Melatonin suppresses tumor angiogenesis and the growth of gastric cancer cells in tumor-bearing nude mice. Moreover, melatonin decreases the expression of VEGF and HIF-1α at translational and transcriptional levels within gastric cancer cells during tumorigenesis [53]. Reduced serum levels of VEGF have been reported in cancer subjects treated with melatonin [54]. Vimalraj et al. [55] showed that melatonin upregulates miR-424-5p expression in osteosarcoma cells suppressing VEGFA. Furthermore, it inhibits tumor angiogenesis, regulating surrounding endothelial cells migration and proliferation, and angiogenic growth factors and the morphology of blood vessels E (Table 1).
Table 1
Melatonin fights against different cancers trough angiogenesis modulation
Type of malignancy
Melatonin dose/concentration
Angiogenesis-related targets
Key findings
Model
Cell line
Refs.
Breast cancer
1 mM
VEGF, ANG-1, ANG-2
Downregulated angiopoietins with a decrease in VEGF
In vitro
MCF-7
[125]
Dalton's lymphoma
1 and 5 mM
VEGF, FGF, TIMP3
Decreased the Dalton’s lymphoma ascites–mediated angiogenesis
In vivo, in vitro
A549 and SiHa
[126]
serous papillary
ovarian cancer
200 µg/100 g b.w
VEGF, HIF-1α
Significantly decreased angiogenesis-associated markers, ovarian cancer size and microvessel density
In vivo
-
[127]
canine mammary tumor cells
1 mM
VEGF
Decreased cell viability, enhanced caspase-3 cleaved and proteins implicated
in the apoptotic pathway and diminished pro-angiogenic VEGFA
In vitro
CF-41, CMT-U229
[128]
Neuroblastoma
1 mM or 1 nM
VEGF
Inhibited proliferation and migration of cancer cells
In vitro
SH-SY5Y
[129]
Gastric cancer
3 mM
100, 150 mg/kg
VEGF, HIF-1α, RZR/RORγ, SENP1
Suppressed gastric cancer growth and blockaded tumor angiogenesis
Decreased the expression of melatonin nuclear receptor RZR/RORγ
In vitro, in vivo
SGC-7901
[130]
Breast cancer
1 mM
-
Regulated inflammation
Decreased cancer cell viability and cancer associated fibroblasts
In vitro
MDA-MB-231
[52]
Breast cancer
1 mM
HIF-1α, VEGF, EGFR, angiogenin,
Reduced protein and gene expression of angiogenesis markers and also decreased cancer cell viability
In vitro
MCF-7, MDA-MB-231
[131]
Breast cancer
10 mg/kg
VEGF
The combination of melatonin and P. acnes cured forty percent of treated mice, suppressed metastasisand decreased angiogenesis and mediated apoptosis
In vivo
EMT6/P
[132]
Prostate cancer
1 mM
HIF-1α, VEGF
Upregulationn of miRNA374b and miRNA3195 mediated melatonin-induced anti-angiogenic properties
In vitro
PC-3
[133]
Oral cancer
1 mM
HIF-1α, VEGF
Decreased cancer cell viability, inhibited metastasis and angiogenesis
In vitro
SCC9, SCC25
[51]
Hepatocellular carcinoma
1 mM
VEGF, HIF-1α, STAT3
Melatonin exerted its anti-angiogenic effects through interfering with the transcriptional activation of mentioned markers
In vitro
HepG2
[134]
Breast cancer
1 mM
VEGF
Inhibited stimulatory impacts on the proliferation of human umbilical vein endothelial cells (HUVECs) as well as VEGF protein levels
In vitro
MCF-7
[46]
Renal cancer
20 mg/kg
10 µM
HIF-1α
Inhibited tumor growth, blocked tumor angiogenesis and diminished HIF-1α protein expression within the tumor mass during tumorigenesis
In vivo, in vivo
RENCA
[135]
Breast cancer
1 mM
40 mg/kg
VEGFR2, micro-vessel density (MVD)
Inhibited tumor growth and proliferation
In vivo, in vitro
MDA-MB-231
[136]
Colon cancer
1 mM
VEGF, HIF-1α
Suppressed invasion and migration
In vitro
HCT116
[44]
Breast cancer
40 mg
0.001 mM, 0.01 mM, 0.1 mM
and 1 mM
VEGF, IGF-IR, HIF-1α
Increased miR-152-3p expression leading to suppress breast cancer
In vitro
MDA-MB-468
[137]
Breast cancer
-
IGF-1R, VEGF
Inhibited survival, migration and invasion of breast cancer cells
Increased the gene level of miR-148a-3p
In vivo, in vitro
MDA-MB-231
[138]
Advanced cancer patients (CRC, HCC, RCC, NSCLC)
20 mg
VEGF
Controlled tumor growth by anti-angiogenic roles
Human
-
[54]

Melatonin and oxidative stress

In normal cellular condition, there is a balance between the production of oxidants, so called reactive oxygen species (ROS), and their neutralizing compounds, named antioxidants. The state of excess ROS, in which the oxidant content of the cells dominates the neutralizing capacity of antioxidants, is defined as oxidative stress [56, 57]. Sustained oxidative stress increases the risk of cancer development either through inducing mutagenesis or by promoting the expression of proto-oncogenes such as cyclin D1. It also plays a signaling role in the activation of several genes involved in the cancer progression including the mitogen-activated protein kinase (MAPK), extracellular signal-regulated kinase (ERK) and JUN N-terminal kinase (JNK) [58, 59].
Melatonin role as a natural ally against oxidative stress has been revealed in many in vitro and in vivo studies. Detoxification of oxidants by melatonin is triggered by several direct or indirect mechanisms. In direct scenario, melatonin neutralizes the oxidants by its nonreceptor‐mediated free radical scavenging capacity. As indirect scenario, melatonin reduces the oxidative content through several mechanisms such as activating anti-oxidative enzymes and suppressing pro‐oxidative enzymes. It also stabilizes the mitochondrial inner membrane, thereby maintaining mitochondrial integrity leading to a reduced electron leakage and ROS generation [60, 61].
The inducing role of oxidative stress in cancer progression and preventive role of melatonin in the production and function of oxidants indicated a possible oncostatic property for melatonin [62]. Subsequently, it was revealed that melatonin reduces the oxidative damage to cellular components under conditions where toxic oxygen derivatives are acknowledged to be released [63, 64]. Moreover, in vitro studies demonstrated that melatonin treatment reduces the amount of oxidative contents in a variety of cancer cells, which was further supported by in vivo studies (Table 2).
Table 2
Melatonin acts as an antioxidant in cancer models
Cancer
Melatonin dose/concentration
Key findings
Model
Cell line/animal
Refs.
Breast cancer
1 μM
5, 10, 50 mg/kg
Limited paclitaxel-mediated mitochondrial dysfunction and protected against paclitaxel-mediated neuropathic pain
In vitro, in vivo
MCF-7
Male and female Sprague Dawley rats
[198]
Neuroblastoma
10 μM
Reduced oxaliplatin-induced neurotoxicity
In vitro
SH-SY5Y
[199]
Breast cancer
0.3 mM
Supported doxorubicin effects by apoptosis and TRPV1activation, and through mediating cancer cell death
In vitro
MCF-7
[200]
Cervical cancer
1 mM
Enhanced cisplatin-mediated cytotoxicity and apoptosis
In vitro
HeLa
[163]
Lung cancer
1 nm, 1 μm, 1 mm
Exerted immunomodulatory effects
In vitro
SK‐LU‐1
[201]
Pancreatic cancer
26.8 mg
capecitabine and melatonin provided an amelioration in antioxidant status and synergistic antitumoral effects
In vivo
Male Syrian hamsters
[202]
Leukemia
1 mM
Protected healthy cells from chemotherapy-mediated ROS production and induced tumor cell death
In vitro
HL-60
[180]
Hepatocellular carcinoma
1, 100 μM
The responses of angiogenic chemokine genes to melatonin were determined by the characteristics of cancer cells
In vitro
HCC24/KMUH,
[203]
Pancreatic cancer
53.76 mg
Exerted more potent beneficial effects than celecoxib on the decrease in tumor nodules, oxidative stress and death
In vivo
Male Syrian hamsters
[204]
Breast cancer
2.5 mg/kg
Antioxidant effects
In vivo
Female Sprague Dawley rats
[205]
Pancreatic cancer
26.88, 53.76 mg
Decreased oxidative damage and cancer nodules mediated by BOP in the pancreas
In vivo
Male Syrian hamsters
[206]
Cervical cancer
10–1000 μM
This study showed melatonin effects on radiotherapy is dose-dependent
In vitro
HeLa
[207]
Hepatocellular carcinoma
20 mg/kg
Fostered the survival and therapeutic potential of MSCs in HCC, by inhibition of oxidative stress and inflammation as well as apoptosis induction
In vivo
Adult female rats
[120]
Cervical cancer
10 μM
Enhanced TNF-α-mediated cervical cancer cells mitochondrial apoptosis
In vitro
HeLa
[14]
Bladder cancer
1 mM
100 mg/kg
Inhibited the growth, migration, and invasion of cancer cells
In vivo, in vitro
HT1197, HT1376, T24, RT4
Male C57B/L6 mice
[208]
Lung cancer
0.25–2.5 mM
Enhanced palladium-nanoparticle-induced cytotoxicity and apoptosis
In vitro
A549, H1299
[209]
Lymphoma, cervical cancer, hepatoblastoma, gastric cancer, breast, colon and lung adenocarcinoma,
0–2 mM
Sensitizees shikonin-mediated cancer cell death induced by oxidative stress
In vitro
U937, HeLa, Hep-G2, AGS, MCF-7, SW480, A549
[210]

Melatonin and endoplasmic reticulum stress

Endoplasmic reticulum (ER) is an entry site for secretory proteins and most integral membrane proteins for proper folding and covalent modifications to assemble into a functional complex. In addition to the processing of proteins, ER is involved in various cellular functions including lipid synthesis, fatty acid turnover, detoxification, Ca2+ homeostasis. The ER network extends into all cell compartments to sense intrinsic and extrinsic perturbations and integrate the stress signals for maintenance of cellular homeostasis to preserve proper cellular and organismal function [65, 66]. However, the ER function can be impacted and disturbed by a multitude of exogenous and endogenous factors, leading to the accumulation of mis/unfolded proteins in the ER. This causes the imbalance between the client proteins load in the lumen of ER and the folding capacity of this organelle leading to the failure of the ER to cope with unusual high protein folding load, which is termed 'ER stress' [67]. To restore protein homeostasis, an adaptive signal transduction pathway called the unfolded protein response (UPR) is activated to induce compensatory responses to stressors for recovering normal ER function [68]. Signaling proteins sensing UPR include inositol-requiring protein-1α (IRE1α), activating transcription factor 6α (ATF6α) and protein kinase RNA (PKR)-like ER kinase (PERK). In nonstressed cells, UPR stress sensors are maintained in an inactive state through direct binding to the ER chaperone proteins, Bip (78-kDa glucose regulated protein, GRP78). Upon ER stress, aggregation of misfolded proteins leads to the dissociation of UPR sensors from Bip, which causes activation of UPR signals [69]. Although activation of UPR signaling pathways is a cellular strategy to increase survival, this pathway will instead activate cell death signaling pathways when the intensity or duration of cellular stress increases. Therefore, certain anti-cancer patterns may activate ER stress/UPR pathway to induce apoptosis in cancer cells [70].
Melatonin induces mitochondria-mediated apoptosis in colorectal cancer cells through reducing the expression of PrPC and PINK1 resulting in the enhancement of superoxide production and induction of ER stress [71]. Melatonin also ameliorates ER-stress mediated insulin resistance. ER stress induces autophagy in pancreatic β cells, which this pathway plays an important role in insulin production and secretion. In the glucose analog 2-DG-treated rat insulinoma INS-1E cells, melatonin reduces insulin production via ER stress-induced autophagy [72]. Combination of melatonin with ER stress inducer tunicamycin increases the sensitivity of cancer cells to apoptosis through inhibiting the expression of COX-2 and increasing the Bax/Bcl-2 ratio and CHOP levels [73]. Selective inhibition of ATF-6 by melatonin results in the suppression of COX-2 production and enhancement of cancer cells to ER-stress induced apoptosis [74].
Melatonin increases apoptosis through enhancing caspase-3, -8 and -9 activities, Bax/Bcl-2 ratio, PARP cleavage and cytochrome c, p53 and Fas-L proteins concentrations in hepatocellular carcinoma, which this effect is mediated by the elevation of ER stress characterized by up-regulation of ATF6, CHOP and Bip [75]. Furthermore, melatonin increases the sensitivity of hepatocellular carcinoma cells to sorafenib through targeting the PERK-ATF4-Beclin1 pathway [76]. The same results have been reported in gastric cancer; melatonin inhibits cell proliferation through inducing activation of the IRE/JNK/Beclin1 signaling [77].
Melatonin in combination with the ER stressor thapsigargin increases the expression level of nuclear mammalian RNA-binding protein (HuD) resulting in the reduction of intracellular biosynthesis of insulin. Suppression of AKT/PI3K pathway and induction of nuclear mTOR (Ser2481, Ser2448) expressions by melatonin sensitizes rat insulinoma INS-1E cells to insulin through increasing the expression of insulin receptor substrate [78]. In contrast with these reports, melatonin has been reported to inhibit tunicamycin-induced ER stress in human hepatocellular carcinoma cells and increase the response of these cells to cytotoxic effects of doxorubicin; this is accompanied by inhibition of the PI3K/AKT pathway, elevation of CHOP and reduction of Survivin [79]. These evidences suggest that melatonin could improve the toxic effect of anti-cancer agents on cancer cells through regulating ER stress in cells (Table 3).
Table 3
Melatonin suppressive effects on various cancers via regulation of ER stress
Cancer
Melatonin dose/concentration
Effect on ER stress
Key findings
Model
Cell line/animal
Refs.
Gastric cancer
1, 2, 3 mM
50 mg/kg
Activate
Melatonin-mediated inhibition of cancer cell proliferation is induced by the IRE/JNK/Beclin1 signaling activation
In vitro, in vivo
AGS, SGC-7901 cells
Male BALB/c nude mice
[77]
Lung, liver and cervical cancer
2 mM
Activate
Induced apoptosis by ROS generation and JNK activation
In vitro
HepG2, A549, HeLa
[211]
Hepatocellular carcinoma
10–5 M
-
enhanced HCC sensitivity to sorafenib through suppressing autophagy
In vitro
HepG2, 7721, Huh7, LO2
[76]
Colorectal cancer
0–1 mM
Activate
Induced mitochondria-induced cellular apoptosis
In vitro
SNUC5/
WT
[71]
Hepatoma
10–7-10–3 mM
-
Melatonin was shown as a novel selective ATF-6 inhibitor that can sensitize human hepatoma cells to ER stress inducing apoptosis
In vitro
HepG2
[74]
Insulinoma
100 μM
Activate
Melatonin-induced insulin synthesis involved autophagy and EDC3 protein in rat insulinoma cells and subsequently resulted in a resuction in intracellular production of insulin
In vitro
INS-1E
[72]
Hepatocellular carcinoma
1 mg/kg/d
Activate
Activated ER stress and apoptosis
In vivo
Male Wistar rats
[75]
Insulinoma
10, 50 μM
-
Decreased nuclear and cellular expressions of p85α
Decreased cellular expression of HuD and led to a reduction in cellular insulin level and rise in insulin secretion
In vitro
INS-1E
[78]
Hepatocellular carcinoma
10–3 M
Inhibit
Attenuated ER stress-mediated resistance to doxorubicin by downregulating the PI3K/AKT pathway, enhancing CHOP levels and reducing Survivin levels
In vitro
HepG2, SMMC-7721
[79]
Hepatoma
10–9, 10–7, 10–5, 10–3 μM
Activate
Sensitized cancer cells to ER stress-mediated apoptosis by downregulating COX-2 expression, enhancing the levels of CHOP and reducing the Bcl-2/Bax ratio
In vitro
HepG2, HL-7702
[73]

Melatonin and autophagy

Autophagy is a complicated process maintaining intracellular homeostasis by eliminating degraded proteins and organelles during cellular stress. Autophagy is principally considered as a pro-survival process, but, excessive or inappropriate autophagy contributes to the cell death, a process known as autophagic cell death or type II programmed cell death [80].
Autophagy is a complicated process, which consists of five sequential steps, including: (a) initiation complex formation and double-membrane phagophore (nucleation) maturation; (b) membrane elongation and autophagosome formation sequestering cargo; (c) fusion with lysosome; (d) inner membrane disruption leading to degradation of cargo by hydrolases; and (e) macromolecular component utilization [81]. These steps of the autophagy pathway are regulated by more than 35 autophagy related genes (ATGs) and proteins most of which function in complexes. The initiation phase is regulated by Unc-51-like kinase1 (mammalian homologues of Atg1, ULK1)–Atg13–Atg101–FIP200 (mammalian homologues of Atg17) protein complex. Unc-51-like kinase1 phosphorylates and activates Beclin-1 (mammalian homologue of Atg6). Beclin-1 is a part of multiprotein-complex, class III PI3-kinase Vps34–p150 (mammalian homolog of Vps15)–Atg14-like protein (Atg14L)–Beclin-1, promoting nucleation [8183]. The elongation phase is regulated by two ubiquitin-like conjugation systems, Atg12 and LC3 (mammalian homologue of Atg8). In the first conjugation system, Atg12 is activated by Atg7 (E1-like enzyme), transferred to Atg10 (E2-like enzyme) and conjugated to Atg5. The Atg12–Atg5 conjugates further couples with Atg16 (Atg16L in mammals) to form the E3-like complex. In the LC3 conjugation system, LC3 is cleaved by a cysteine protease, Atg4, forming LC3-I. Thereafter, LC3-I is activated by Atg7 (E1-like enzyme), transferred to Atg3 (E1-like enzyme) and conjugated to phosphatidylethanolamine (PE) to form LC3-II; this process is facilitated by the E3-like complex. This lipidated form of LC3, LC3-II, is recruited to the autophagosome membrane. Finally, the Atg9 dependent pathway promotes autophagosome membrane expansion [8183].
Cargo sequestration can be selective or non-selective; the selectivity is based on autophagy receptors such as P62/SQSTM1, NBR1, NDP52, NIX/BNIP3L, BNIP3 and FUNDC1 [82]. Fusion of autophagosome with lysosome is the next step. The inner vesicle is degraded by lysosomal hydrolases, including cathepsin B, D (a homolog of proteinase A), and L. The degradation products are released to the cytosol and used in different anabolic pathways [84]. ER stress-induced activation of UPR pathways promotes induction of autophagy [85]. Activated PERK/ATF4 pathway up-regulates the expression of ATG genes including ATG5, ATG7, and ATG10 [86]. The conversion of LC3-I conversion to LC3-II is also induced by PERK pathway [87]. Activation of IRE1α pathway induces the expression of Beclin1 and the phosphorylation of Bcl-2 by JNK, which subsequently results in the Bcl-2–Beclin 1 dissociation [8890]. The release of Ca2+ from ER to cytosol triggers autophagy pathway through activating several mechanisms including (I) inhibition of mTOR by Ca2+/calmodulin dependent kinase kinase-β-mediated activation of AMP-activated protein kinase (AMPK) [91], and (II) dissociation of Bcl-2–Beclin 1 by inducing death-associated protein kinase (DAPK) 3-mediated Beclin 1 phosphorylation [92].
Melatonin has a modulatory effect on autophagy in various cell types and different conditions. Melatonin indirectly modulates autophagy through affecting oxidative stress, ER stress and inflammation [69]. Melatonin enhances the effectiveness of cisplatin and radiotherapy in head and neck squamous cell carcinoma, which this effect is mediated by the excessive activation of mitochondria leading to the over-production of ROS and subsequent induction of autophagy and apoptosis [93]. Melatonin also increases cytotoxic effects of rapamycin in cancer cells. Combination of rapamycin and melatonin suppresses AKT/mTOR pathway activation, which this effect leads to the enhancement of mitochondrial function and ROS production resulting in the induction of apoptosis and mitophagy [94]. Melatonin induces autophagy in clear cell renal cell carcinoma through activating transcriptional coactivator peroxisome proliferator-activated receptor gamma coactivator 1A (PGC1A) and uncoupling protein 1 (UCP1); this is associated with the elimination of lipid deposits without generating ATP, which subsequently leads to the tumor size reduction [95].
Melatonin reduces the viability liver cancer cells through transient induction autophagy by up-regulating JNK phosphorylation. However, ATG5 silencing sensitizes cancer cells to melatonin-induced apoptosis. This suggests that modulation of autophagy by melatonin has dual effect on cell death [96]. Similarly, disruption of autophagy sensitizes glioblastoma cells and tongue squamous cell carcinoma to melatonin-induced apoptosis [97]. Melatonin-induced autophagy is suggested to be mediated by activation of melatonin membrane receptor in tongue squamous cell carcinoma and suppression of melatonin membrane receptor-dependent autophagy may be strategy for treatment of tongue squamous cell carcinoma [98].
Several studies indicate that melatonin may induce apoptosis in cancer cells through inhibiting autophagy pathway. Melatonin down-regulates Beclin-1 and p62 expressions and LC3B-II/LC3B-I ratio in colitis-associated colon carcinogenesis in mice; this effect is associated with the increased level of Nrf2 and its downstream antioxidant enzymes including NAD(P)H:quinone oxidoreductase (NQO-1) and heme oxygenase-1 (HO-1). These suggest that the ameliorative effect of melatonin on inflammation and oxidative stress results in the reduction of autophagy [99]. Induction of ER stress is associated with the activation of autophagy in sorafenib-treated hepatocellular carcinoma cells, which this contributes to the resistance of cancer cells to apoptosis. Combination of melatonin with sorafenib inhibits ER stress-related autophagy through suppressing the PERK-ATF4-Beclin1 pathway leading to the sensitivity of hepatocellular carcinoma cells to sorafenib [76]. Co-stimulation of cancer cells with cisplatin and melatonin induce apoptosis in HeLa cells, which this effect is accompanied by inactivating mitophagy via blockade of JNK/Parkin pathway [100]. In contrast with this report, melatonin has been found to reversed the effects of cisplatin in HepG2 cells through suppression of mTOR and DNA excision repair cross complementary 1 (ERCC1) proteins expressions and up-regulation of Beclin-1 and LC3II expressions [101]. Taken together, different effects of melatonin on autophagy may be related to type of cancer cells, the stage of cancer and dose of melatonin (Table 4).
Table 4
The effect of melatonin on autophagy machinery in recently reported findings
Cancer
Melatonin dose/concentration
Autophagy-related targets
Effect on autophagy
Key findings
Model
Cell line
Refs.
Lung, liver and cervical cancer
2 mM
LC3
Activate
Induced apoptosis by ROS generation and JNK activation
In vitro
HepG2, A549, HeLa
[124]
Glioblastoma
1 mM
Beclin 1, LC3-II
Activate
Autophagy disruption stimulated the melatonin-mediated apoptosis in cancer cells
In vitro
A172, U87-MG
[97]
Uterine leiomyoma
25 mg/kg
0.1, 0.5, 1, 1.5, 2 mM
Beclin1 and LC3
Activate
Reduced tumor growth and proliferation
In vivo, in vitro
ELT3 cells, orthotopic uterine leiomyoma mouse model
[192]
Hepatocellular carcinoma
10−5_ 10–3 M
PERK-ATF4-Beclin1 pathway
Inhibit
enhanced HCC sensitivity to sorafenib through suppressing autophagy
Human
-
[76]
Colorectal cancer
10 μM
LC3-II
Activate
Induced interplay of apoptosis, autophagy, and senescence
In vitro
HCT116
[171]
Clear cell renal cell carcinoma
200 mg/kg
0.5, 1, 2 μM
PGC1A, UCP1, LC3‐II
Activate
Melatonin/PGC1A/UCP1 promoted tumor slimming and repressed tumor progression through initiating autophagy and lipid browning
In vivo, in vitro
HK2, 786‐O, A498, Caki‐1, and ACHN cells
Mice
[95]
Neuroblastoma
0.1‐ 10 nM
40‐80 mg/kg
LC3II
Activate
Promoted cancer cell differentiation through activation of hyaluronan synthase 3-mediated mitophagy
In vivo, in vitro
N2a
N2a‐allografted nude mice
[193]
Head and neck squamous cell carcinoma
0.1, 0.5, 1, 1.5 mM
ATG12-ATG5
Activate
Induced intracellular ROS
In vitro
Cal-27, SCC-9
[93]
Hepatocellular carcinoma
1 mM
mTOR, Beclin-1
Activate
Decreased cisplatin-mediated cell death by a counter-balance between the roles of apoptotic- and autophagy-related proteins
In vitro
HepG2
[101]
Hepatocellular carcinoma
2 mM
Beclin-1, p62, LC3II, LAMP-2
Activate
Ceramide metabolism regulated apoptotic and autophagy cell death mediated by melatonin
In vitro
HepG2
[96]
Neuroblastoma
1 μM
Beclin‐1, LC3‐II
Activate
Enhanced autophagic activity by the SIRT1 signaling
In vitro
SH‐SY5Y
[194]
Gastric cancer
10−4 M
LC3
Activate
Hyperbaric oxygen sensitized cancer cells to melatonin-mediated apoptosis
In vitro
SGC7901
[151]
Colon cancer
1 mg/kg
Beclin-1, LC3B-II/LC3B-I ratio, p62
Inhibit
Decreased autophagy by improving oxidative stress and inflammation
In vivo
Male Swiss Albino mice
[99]
Glioblastoma
1 mM
LC3, Beclin-1
Activate
Inhibited tumor bulk proliferation, and enhanced chemotherapy effects
In vitro
Glioblastoma-initiating cells
[195]
Oral cancer
0.5–2 mM
LC3-II
Activate
Decreased drug resistance, and induced autophagy and apoptosis
In vitro
SAS, SCC9, SASV16, SASV32, SCC9V16, SCC9V32
[139]
Gastric cancer
50 mg/kg
1, 2, 3 mM
p62, Beclin-1, LC3A/B-II
Activate
Melatonin-mediated inhibition of cancer cell proliferation is induced by the IRE/JNK/Beclin1 signaling activation
In vivo, in vitro
AGS, SGC-7901
Male BALB/c nude mice
[77]
Hepatocellular carcinoma
10, 20 mg/kg
100 μM
Beclin-1, LC3-I/LC3-II
Activate
Induced protective autophagy preventing hepatoma cells from undergoing apoptosis
In vitro, in vivo
H22
[196]
Insulinoma
100 μM
LC3II
Activate
Melatonin-induced insulin synthesis involved autophagy and EDC3 protein in rat insulinoma cells and subsequently resulted in a resuction in intracellular production of insulin
In vitro
INS-1E
[72]
Chriocarcinoma
1 mM
LC3B
Inhibit
Modulated autophagy and the Nrf2 pathway in normal vs. tumor trophoblast cells, being cytoprotective in normal cells whilst enhancing apoptosis in tumoral trophoblast cells
In vitro
BeWo
[197]
Cervical cancer
1 mM
JNK/Parkin
Inhibit
Sensitized cancer cells to cisplatin-mediated apoptosis by suppression of JNK/Parkin/mitophagy pathways
In vitro
HeLa
[100]
Head and neck squamous cell carcinoma
0.1, 0.5 or 1 mM
300 mg/kg
LC3-II, Nix
Activate
Enhanced ROS production, increased apoptosis and mitophagy, and could be used as an adjuvant agent with rapamycin
In vitro, in vivo
Cal-27, SCC-9
Harlan Sprague–Dawley mice
[94]
Tongue squamous cell carcinoma
0, 0.5, 1, 2 mM
100 mg/kg
LC3, ATG7
Activate
Suppression of MT2-TFE3-dependent autophagy enhanced melatonin-mediated apoptosis
In vitro, in vivo
Cal27, SCC9
Male athymic nude mice
[98]

Melatonin and apoptosis

The balance between cell proliferation and death in tissues is maintained by apoptosis, a classical form of programmed cell death. Apoptosis is associated with the disassembly of apoptotic cells into membrane-enclosed vesicles, which are removed by macrophages without inducing inflammatory responses. Apoptosis is mediated by two principle signaling pathways, including extrinsic and intrinsic pathways [102]. The extrinsic apoptotic signaling pathways, defined as death receptor pathways, are initiated by the interaction of transmembrane death receptors (Fas, TNFR1, DR4 and DR5) with extracellular ligands (FasL, TNFα, TRAIL, and TNFSF10) resulting in the activation of adaptor proteins such as Fas-associated death domain (FADD). Activated FADD recruits initiator caspases (caspase 8 and caspase 10) to form the death-inducing signal complex (DISC). Formation of DISC leads to the proteolytic activation of caspase 8, which is the main initiator caspase of the extrinsic apoptotic signaling pathway. Caspase 8 activates executioner caspases (caspase 3, caspase 6, and caspase 7) and cleaves Bid, a BH3-only domain member of the B cell lymphoma-2 (Bcl-2) family. Truncated Bid (tBid) translocates to mitochondria and activates other proapoptotic Bcl-2 family members including Bak or Bax [102, 103].
The intrinsic apoptosis pathway, defined as mitochondrial-mediated apoptotic pathway, is activated by exogenous and endogenous stimuli such as DNA damage, oxidative stress, chemotherapy and radiotherapy. This apoptosis pathway is mediated by insertion of pro-apoptotic Bcl-2 family members (Bax/Bak) into mitochondrial membrane leading to the mitochondrial outer membrane permeabilization and release of pro-apoptotic factors such as cytochrome c, Smac/DIABLO, the nuclease EndoG, the oxidoreductase AIF, and the protease HtrA2/Omi [104]. Therefore, activation of pro-apoptotic Bcl-2 family members (Bax/Bak) is essential for cancer therapy. In contrast, elevation of anti-apoptotic Bcl-2 family proteins inhibits apoptosis in cancer cells through heterodimerization with Bax/Bak preventing the release of pro-apoptotic factors from mitochondria; this could result in the resistance of cancer cells to immune-surveillance [105, 106]. Once in the cytosol, cytochrome c combines with Apaf–1 and procaspase-9 to drive the assembly of the apoptosome; this molecular platform activates caspase 9, which this is followed by the activation of caspase-3 cascade of apoptosis [107]. Smac/DIABLO and HtrA2/Omi induce apoptosis through degrading inhibitor of apoptosis protein (IAP) family, neutralizing the inhibitory effect of IAPs on caspases [108]. The nuclease EndoG and the oxidoreductase AIF translocate to the nucleus, where they trigger internucleosomal DNA fragmentation independently of caspases [109].
As mentioned earlier, UPR signaling may promote the apoptotic pathways. Upon ER stress, apoptosis signal-regulating kinase 1 (ASK1) is recruited by IRE1α-TNF receptor-associated factor 2 (TRAF2) complex, causing activation of ASK1 and the downstream JNK pathway. Activation of JNK results in the phosphorylation of Bcl-2 and Bax; phosphorylation of Bcl-2 family suppresses antiapoptotic activity of Bcl-2, while induces mitochondrial translocation of Bax and activation of apoptosis pathway. Activated JNK also activates C/EBP homologous protein (CHOP), a stress-induced transcription factor inducing the expression of pro-apoptotic Bcl-2 family members. Furthermore, IRE1α-TRAF2 complex triggers the activation of caspase-12, which this caspase translocates from the ER to the cytosol, where it activates caspase-9, independent from the apoptosome pathway [110]. Furthermore, Activated PERK phosphorylates eIF2α, promoting the expression of activating transcriptional factor 4 (ATF4); ATF4 translocates to the nucleus where it induces CHOP expression [111].
Melatonin is reported to restrict tumor growth and cancer cell proliferation through inducing apoptosis in cancer cells (Table 5). As a powerful antioxidant melatonin inhibits ROS-induced activation of extracellular-regulated protein kinases (ERKs) and Akt pathways which are involved in the cancer cell survivor; inactivation of ROS-dependent Akt signaling contributes to the down-regulation of cyclin D1, PCNA, and Bcl-2 and up-regulation of Bax in cancer cells [48]. Inhibition of MDM2 expression is a mechanism by which melatonin induces apoptosis through upregulating the activity of caspase-3 and -9; MDM2 is an E3 ubiquitin ligase, which negatively regulates the p53 tumor suppressor [112, 113]. Under hypoxic conditions, tumor cells become resistant to TRAIL-induced cell apoptosis; this contributes to the up-regulation of anti-apoptotic protein expression and reduction of pro-apoptotic protein expression. Treatment with melatonin blocks hypoxic responses leading to the induction of apoptosis in TRAIL resistance tumor cells by the regulation of mitochondrial transmembrane potential and induction of Bax translocation [114]. Melatonin inhibits cancer cell growth by increasing cell cycle arrest in the G2/M phase, which this effect is coincident with the induction of apoptosis through up-regulating the expression of p53, p21, caspase-3/8/9, PARP, cytochrome c, Bax, JNK 1,-2 and -3 and p38 MAPKs in cancer cells [115]. Melatonin triggers two distinct apoptotic processes including TGFβ1 and caspase-independent early apoptosis and TGFβ1 and caspases-dependent late apoptosis. Early apoptosis is associated with the elevation level of p53/MDM2 ratio and up-regulation of AIF release; this process is independent to caspase activity or cleavage of PARP. Late apoptosis is associated with elevation of caspases-9 and -7 activity and cleaved-PARP level as well as reduction of Bcl-2/Bax ratio [116]. Melatonin also induces apoptosis through simultaneous suppression of COX-2/PGE2, p300/NF-κB, and PI3K/Akt signaling pathway. Inhibition of these pathways leads to the induction of Apaf-1 expression triggering cytochrome c release, and caspase-3 and -9 activation and cleavage [32].
Table 5
Anticancer effects of melatonin by apoptosis induction in experimental investigations
Cancer
Melatonin dose/concentration
Apoptosis-related targets
Key findings
Model
Cell line
Refs.
Oral cancer
0.5–2 mM
caspase-3, caspase-9, PARP
Decreased drug resistance, and induced autophagy and apoptosis
In vitro
SCC9V32, SCC9V16, SASV32, SASV16,
SAS, SCC9
[139]
Lung cancer
Hepatocellular carcinoma
Cervical cancer
2 mM
caspase-3, PARP, Bax, Bcl-2
Decreased cell viability and increased LDH release
In vitro
Hela
A549
HepG2
[124]
Glioblastoma
1 mM
Bax, Bcl-2
Induced apoptosis and autophagy
In vitro
A172
U87-MG
[97]
Colorectal cancer
0.5, 1 mM
caspase-3, PARP, NEDD9, SOX9, Bcl-xL, SOX10
Enhanced apoptosis through miR-25-5p induced NEDD9 suppression in cancer cells
In vitro
CCD-18Co, HT29, SW480, HCT116
[121]
Breast cancer
3.5–20 mM
2 mg/kg
caspase-3
Repressed drug resistance through apoptosis induction and angiogenesis inhibition
In vitro, in vivo
EMT6/CPR, EMT6/VCR/R
[140]
Lung cancer
2, 4, 6 mM
HDAC9
HDAC9 knockdown increased the anticancer potentials of melatonin
In vitro, in vivo
A549, H838, H1299, and Calu-1
[118]
Ehrlich
carcinoma
20 mg/kg
Bcl‐2, caspase-3, caspase-9,
Inhibited the proliferation and growth of tumor via inducing apoptosis and through suppressing tumor vascularization
In vivo
EAC
[141]
Head and neck squamous cell carcinoma
0.1, 0.5, 1, and 1.5 mM
Bax, Bcl-2
Potentiated the cytotoxic impacts of radiotherapy and CDDP, and induced intracellular ROS leading to mitochondria-induced autophagy and apoptosis
In vitro
SCC-9, Cal-27
[93]
Hepatocellular carcinoma
20 mg/kg
Caspase-3, Bax, Bcl-2, survivin
Fostered the survival and therapeutic potential of MSCs in HCC, by inhibition of oxidative stress and inflammation as well as apoptosis induction
In vivo
-
[120]
Cervical cancer
10 μM
CaMKII/Parkin/mitophagy, caspase-3, caspase-9
Enhanced TNF-α-mediated cervical cancer cells mitochondrial apoptosis
In vitro
HeLa
[119]
Gastric cancer
3 mmol/L
Caspase 9, Caspase 3, AKT, MDM2
Promoted apoptosis through downregulation of MDM2and AKT
In vitro
AGS, MGC803
[112]
Melanoma
1 M
25 mg/kg
cytochrome c, caspase-3, caspase-9, Bcl-2
Synergized the antitumor effects of vemurafenib through suppressing cell proliferation and cancer-stem cell traits by targeting NF-κB/iNOS/hTERT signaling
In vitro, in vitro
G361, A431, A375, SK-Mel-28
[142]
Breast cancer
1 mM
caspase-3
Increased apoptosiss and decreased proliferation in cancer cells
In vitro
MDA-MB-231, MCF-7
[143]
Pancreatic cancer
10–10, 10–12 M
Bax, Bcl-2, caspase-3, caspase-9
Improved the anti-tumor effects of gemcitabine through apoptosis regulation
In vitro
PANC-1
[144]
Breast cancer
25 µM
Bax, Bcl-2
Decreased the cell proliferation and increased apoptosis and differentiation in cancer cells
In vitro
MCF-7, HEK293
[145]
Leukemia
1 mM
Bcl-2, Bcl-xL
Synergistic effect on chemotherapeutic agent
In vitro
HL-60
[146]
Breast cancer
0.1–5 mm
1 mg/kg
-
Melatonin caused apoptosis induction, angiogenesis inhibition, and activation of T helper 1
In vitro, in vivo
EMT6/P
[147]
Colorectal cancer
1 mM
BAX, caspase3, PARP1
Induced mitochondria-induced cellular apoptosis
In vitro
SNUC5/WT
[71]
Breast cancer
1 nM and 100 nM
c-IAP1, XIAP, survivin, MCL-1, BCL-2,
Enhanced cytotoxic effects of arsenic trioxide and apoptosis induction
In vitro
MCF-7
[148]
Pancreatic cancer
0.1, 1, or 2 mM
40 mg/kg
cytochrome c
XIAP, Mcl-1, Survivin, Bcl-2, PARP
Reinforced the anticancer effect of sorafenib via downregulation of PDGFR-β/STAT3 signaling
In vitro, in vivo
MIAPaCa-2, PANC-1
[149]
Glioblastoma
1 mM, 3 mM
-
Delayed cell cycle progression and potentiated the decrease of cell survival due to treatment with temozolomide
In vitro
U87MG
[150]
Oral cancer
1 mM
40 mg/kg
cyclin D1, PCNA, Bcl-2, Bax
Suppressed the invasion and migration of cancer cells through repressing ROS-activated Akt signaling
Hampered vasculogenic mimicry and retarded tumorigenesis of cancer cells
In vitro, in vivo
SCC25, SCC9, Tca8113, Cal27, and FaDu
[48]
Gastric cancer
10−4 mol/L
Bcl-2, Bax, p53, caspase3,
Hyperbaric oxygen sensitized cancer cells to melatonin-mediated apoptosis
In vitro
SGC7901
[151]
Thyroid cancer
1, 2, 4, 8, 15 mM
25 mg/kg
caspase 3/7, PARP, cytochrome c
Reduced cell viability, inhibited cell migration and induced apoptosis
Synergized with irradiation to induce cytotoxicity to thyroid cancer cells
In vitro, in vivo
8505c, ARO
[152]
Gastric cancer
1, 2, 3, 4 or 5 mM
Bax, Bcl-xL, caspase-9, caspase-3
Induced cell cycle arrest and induced apoptosis
In vitro
SGC-7901
[153]
Neural cancer
0.5, 1 mM
Bax, Bcl-2, caspase-9, cytochrome c
Mitochondrial cytochrome P450 1B1 is responsible for melatonin-induced apoptosis
In vitro
U118, SH-SY5Y, U87, U251, A172
[154]
Gastric cancer
1, 5 µM
-
Inhibited the proliferation of cancer cells by regulating the miR-16-5p-Smad3 pathway
In vitro
BSG823, SGC-7901
[155]
Head and neck squamous cell carcinoma
0.1, 0.5, or 1 mM
Bax, Bcl-2
Enhanced ROS production, increased apoptosis and mitophagy, and could be used as an adjuvant agent with rapamycin
In vitro
Cal-27, SCC-9
[94]
Ovarian cancer, colorectal cancer
0.1, 1.0, and 10 μM
-
Induced apoptosis and showed antioxidant effects
In vitro
DLD1, A2780
[156]
Cervical cancer
1 mM
JNK/Parkin/mitophagy, caspase-9
Sensitized cancer cells to cisplatin-mediated apoptosis by suppression of JNK/Parkin/mitophagy pathways
In vitro
HeLa
[100]
Melanoma
Breast cancer
Melatonin: 10−5 − 10−3 M
Melatonin analogues (UCM 1037):
10−6 − 10−4 M and 16 mg/Kg
Bcl-2, Bax, caspase-3
Inactivated mitophagy by suppression of JNK/Parkin, leading to the inhibition of anti-apoptotic mitophagy
Sensitized cervical cancer cells to cisplatin-mediated apoptosis
In vitro, in vivo
DX3, WM-115, MCF-7, MDA-MB231
[157]
Bladder cancer
10 mg/kg
1.0 mM
caspase-3, Bcl-2, BAX
Synergized the inhibitory effects of curcumin against the growth of bladder cancer through increasing the anti-proliferation, anti-migration, and pro-apoptotic properties
In vivo, in vitro
T24, UMUC3, 5637
[158]
Colorectal cancer
1 mM
caspase-3
Increased the sensitivity of cancer cells to 5-FU
In vitro
HT-29
[159]
Lung cancer
25 mg/kg
1 mM
caspse-9, Bcl-2, PARP, cytochrome C
Increased antitumor activities of berberine through activating caspase/Cyto C and suppressing AP-2β/hTERT, NF-κB/COX-2 and Akt/ERK pathways
In vitro, in vivo
H1299, A549
[160]
Gastric cancer
1, 2 mM
caspase-3, Bcl-2, BAX
Suppressed cell viability, clone formation, cell migration and invasion and induced apoptosis
In vitro
AGS
[161]
Ovarian cancer
200 μg/100 g b.w
p53, BAX, caspase-3, Bcl-2, survivin
Promoted apoptosis
In vivo
-
[162]
Cervical cancer
1 mM
Caspase-3
Enhanced cisplatin-mediated cytotoxicity and apoptosis
In vitro
HeLa
[163]
Rhabdomyosarcoma
0.01, 0.1, 1, 2 mM
Bax, Bcl-2, caspase-3
Enhanced the sensitivity of cancer cells to apoptosis
In vitro
U57810, U23674
[164]
Hepatocellular carcinoma
2 mM
PARP, Bax
Ceramide metabolism regulated apoptotic and autophagy cell death mediated by melatonin
In vitro
HepG2
[96]
Neuroblastoma
0.25, 0.5, 1, 2 mM
-
Exerted cytotoxic potentials against cancer cells
In vitro
SH-SY5Y
[165]
Colorectal cancer
0.1–2.0 mM
HDAC4, Bcl-2, CaMKIIα
Melatonin-induced apoptosis depends on the nuclear import of HDAC4 and subsequent H3 deacetylation by CaMKIIα inactivation
In vitro
LoVo
[117]
RCC, CRC, Head and neck cancer, Prostate cancer, breast cancer
1 mM
PUMA, Mcl-1, Bcl-xL, Bim, COX-2
Enhanced antitumor effects by COX-2 downregulation and Bim up-regulation
In vitro
MDA-MB-231, Caki, HN4,
HCT116, PC3
[123]
Cholangiocarcinoma
1 nM, 1 μM
0.5, 1, 2 mM
Caspase-3/7, cytochrome c
Functioned as a pro-oxidant through activating ROS-dependent DNA damage and hence leading to the apoptosis of cancer cells
In vitro
KKU-M055, KKU-M214
[166]
Lung cancer
1–5 mM
caspases-3/7
Increased cisplatin-induced cytotoxicity and apoptosis in lung cancer cells
In vitro
SK-LU-1
[167]
Gastric cancer
25, 50, 100 mg/kg
Bcl-2, Bax, p21, p53
Inhibited tumor growth by apoptosis induction
In vivo
MFC
[168]
Lung cancer
1, 5, 10 mM
caspase-3/7
Showed anticancer impacts by changing biomolecular structure of lipids, nucleic acids and proteins
In vitro
SK-LU-1
[169]
Lung cancer
10−13 M (subphysiological), 10−10 M
(physiological)
10−7, 10−4, 10− 3 M
(Cytotoxic)
CCAR2
Cell cycle and apoptosis regulator 2 (CCAR2) is critical for maintaining cell survival in the presence of melatonin
In vitro
A549, A427
[170]
Lung cancer
500 μM
Bcl-2, Bcl-xL, TRAIL
Induced apoptosis in TRAIL-resistant hypoxic tumor cells trough diminishing the anti-apoptotic signals induced by hypoxia
In vitro
A549
[114]
Breast cancer
1 nM
p53, MDM2/MDMX/p300
Enhanced p53 acetylation by regulating the MDM2/MDMX/p300 pathway
In vitro
MCF-7
[113]
Colorectal cancer
10 μM
Bax, Bcl-xL,
Activated cell death programs early and induced G1-phase arrest at the advanced phase
In vitro
HCT116
[171]
Renal cancer
0.1, 0.5,1 mM
Bim
Induced apoptosis by the upregulation of Bim expression
In vitro
Caki
[172]
Leukemia
1 mM
Bax, cytochrome c
Induced apoptosis by a caspase-dependent but ROS-independent manner
In vitro
Molt-3
[173]
Gastric cancer
10–4 mol/l
Caspase-3
Inhibited tumor cell proliferation and reduced the metastatic potential of cancer cells
In vitro
SGC7901
[174]
Colorectal cancer
1 mM
caspase-3/9, PARP
Potentiated the anti-proliferative and pro-apoptotic impacts of Ursolic acid in cancer cells
In vitro
SW480, LoVo
[175]
Pancreatic cancer
1.5 mmol/L
20 mg/kg
Bax, Bcl-2
Melatonin may be a pro-apoptotic and pro-necrotic molecule for cancer cells by its regulation of Bcl-2/Bax balance
In vitro, in vivo
SW-1990
[176]
Breast cancer
10–3 M
COX-2/PGE2, p300/NF-κB, PI3K/Akt, Apaf-1/caspase-3/9
Inhibited cell proliferation and induced apoptosis
In vitro
MDA-MB-361
[32]
Hepatocellular carcinoma
10–9, 10–7, 10–5, 10–3 μM
CHOP, Bcl-2, Bax, COX-2
Sensitized cancer cells to ER stress-mediated apoptosis by downregulating COX-2 expression, enhancing the levels of CHOP and reducing the Bcl-2/Bax ratio
In vitro
HepG2
[73]
Ovarian cancer
0, 0.5, 1, 2 mM
ERK/p90RSK/HSP27
Enhanced cisplatin-mediated apoptosis through the inactivation of ERK/p90RSK/HSP27 pathway
In vitro
SK-OV-3
[122]
Gastric cancer
2 mM
NF-κB, MAPK
Conflicting growth signals in cells may suppress melatonin efficacy in the treatment of gastric cancer
In vitro
SGC7901
[177]
Hepatocellular carcinoma
10–3, 10–5, 10–7, 10–9 mmol/L
COX-2, Bcl-2, Bax
Melatonin was shown as a novel selective ATF-6 inhibitor that can sensitize human hepatoma cells to ER stress inducing apoptosis
In vitro
HepG2
[74]
Glioma
1 μM
-
Inhibited miR-155 expression and hence repressed glioma cell proliferation, invasion and migration
In vitro
U87, U373, U251
[178]
Breast cancer
1 mM
caspase-3, hTRA, XIAP, TNFRII, P53, P21, Livin, IGF-1R, IGF-1, IGFPB-6, IGFBP-5, IGFBP-3, DR6, CYTO-C
Showed pro-apoptotic, anti-angiogenic and oncostatic properties
In vitro
MDA-MB-231, MCF-7
[179]
Leukemia
1 mM
ROS, caspase-3/8/9
Enhanced apoptotic effects of hydrogen peroxide
In vitro
HL-60
[180]
Renal cancer
1 nM
CHOP, PUMA
PUMA up-regulation contributed to the sensitizing impact of melatonin plus kahweol on apoptosis
In vitro
Caki
[181]
Pancreatic cancer
10−8 –10−12 M
Bcl-2, Bax, caspase-9
Induced pro-apoptotic pathways by interaction with the Mel-1 A/B receptors
In vitro
PANC-1
[182]
Ewing sarcoma
50 μM-1 mM
caspase-3/8/9, Bid
Showed cytoprotective effects on noncancer cells and induced apoptosis
In vitro
SK-N-MC
[183]
Glioma
1 mM
Survivin, Bcl-2
Increased cell sensitivity to TRAIL-mediated cell apoptosis
In vitro
A172, U87
[184]
Leukemia
1 mM
250 mg/kg
Bax, Bcl-2, p53
Enhanced radiation-mediated apoptosis in cancer cells, while decreasin radiation-meditated apoptosis in normal cells
In vitro, in vivo
Jurkat
[185]
Breast cancer
1 nM
Caspase-7/9, p53, MDM2, PARP, Bcl-2, Bax
Induced apoptosis in cancer cells
In vitro
MCF-7
[116]
Hepatocellular carcinoma
1000–10,000 μM
caspase-3/8/9, PARP, cytochrome c, Bax, p53, p21
Induced cell cycle arrest and apoptosis
In vitro
HepG2
[115]
Pheochromocytoma
100 μM
GSH
Apoptotic and antioxidant effects
In vitro
PC12
[186]
Neuroblastoma
100 μM
Caspase-3
Induced apoptosis
In vitro
SK-N-MC
[187]
Leukemia
Cervical cancer
50 μM
Caspase-3
Protectted normal and cancer cells against genotoxic treatment and apoptosis induced by idarubicin
In vitro
K562,
HeLa
[188]
Colorectal cancer
1 mM
Caspase-3
Potentiated flavone-mediated apoptosis in cancer cells
In vitro
HT-29
[189]
Breast cancer
1 nM
Bax, p53, p21, WAF1, bcl-XL, bcl-2
Decreased cancer cell proliferation through regulating cell-cycle length by the control of the p53-p21 pathway
In vitro
MCF-7
[190]
Esophageal cancer
0–5 mM
25 mg/kg
PARP, caspase-3/7/8
Increased cytotoxicity of 5-Fu
In vivo, in vitro
KYSE30, KYSE150, KYSE410, KYSE520
[191]
Melatonin induces dephosphorylation and nuclear import of histone deacetylase 4 (HDAC4) in cancer cells; melatonin exerts this effect through inactivation of Ca2+/calmodulin-dependent protein kinase II alpha (CaMKIIα), leading to the H3 acetylation on Bcl-2 promoter and subsequent reduction of Bcl-2 expression [117]. Furthermore, inhibition of HDAC9 expression is a mechanism of melatonin to promote apoptosis in non-small cell lung cancer; the increased level of HDAC9 in patients with non-small cell lung cancer is correlated with worse overall survival and poor prognosis [118]. Melatonin promotes TNF-α-mediated apoptosis via inhibiting mitophagy in tumor cells. Since activation of mitophagy suppresses mitochondrial apoptosis, inhibition of mitophagy by melatonin results in the repression of mitochondrial potential, elevation of ROS generation, augmentation of mPTP opening rate and up-regulation of cytochrome c expression and caspases activity. Melatonin inhibits autophagy in tumor cells through inhibiting CaMKII activity leading to the suppression of Parkin expression [119]. In diethylnitrosamine (DEN)-induced hepatocellular carcinoma (HCC), melatonin increases therapeutic potential of mesenchymal stem cells (MSCs) through reduction of oxidative stress and inflammation, and induction of apoptosis [120].
Melatonin has been reported to increase therapeutic potential of anti-cancer agents, which this effect may result from its stimulatory effect on apoptosis. Co-treatment of melatonin and pterostilbene in colorectal cancer cells synergically enhances ROS production and apoptosis. Combination of these two agents upregulates the mRNA level of miR-25-5p, which this results in the activation of PARP and sex-determining region Y-Box10 (SOX10), and attenuation of Bcl-xL, neural precursor cell expressed developmentally downregulated protein 9 (NEDD9), and SOX9 expressions [121]. Melatonin synergically enhances anticancer potential of cisplatin through inducing apoptosis; melatonin increases the effect of cisplatin to the inhibition of ERK phosphorylation and induction of 90-kDa ribosomal S6 kinase (p90RSK) and heat shock protein 27 (HSP27) dephosphorylation [122]. Treatment with melatonin enhances ER stress–mediated apoptosis in tunicamycin-treated cancer cells; this effect is associated with the down-regulation of COX-2 and Bcl-2 expressions and up-regulation of Bim, CHOP and Bax expressions [73]. Melatonin inhibits tunicamycin-induced COX-2 activation in tumor cells through inhibiting NF-ĸB and p38 MAPK activation and p65 nuclear translocation [123]. Combination of melatonin with phenylarsine oxide also induces endoplasmic reticulum stress-induced cell death, accompanied by JNK activation, PARP cleavage, ROS generation and caspase-3 activation [124].

Conclusions

This review summarizes the anti-carcinogenic potentials of melatonin by evaluating various signaling pathways. Melatonin inhibits proliferation of cancer cells through triggering cell cycle arrest and causes cell death by induction of apoptosis. Melatonin suppresses metastasis angiogenesis, and proliferation of cancer cells through affecting various signaling pathways in tumor cells. Melatonin also regulates autophagy pathway in cancer cell by affecting oxidative stress condition in tumor cells. These findings suggest that melatonin may increase the sensitivity of cancer cells to anti-cancer agents and may be a potential treatment for cancers either alone or in combination with other anti-cancer drugs. However, further clinical studies are needed to clarify the effect of this molecule in different cancers and obtain affective dose of melatonin for patients with cancer.

Acknowledgements

Not applicable.

Declarations

Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Block KI, Gyllenhaal C, Lowe L, Amedei A, Amin A, Amin A, Aquilano K, Arbiser J, Arreola A, Arzumanyan A, et al. Designing a broad-spectrum integrative approach for cancer prevention and treatment. Seminars Cancer Biol. 2015;35 Suppl(Suppl):S276–304.CrossRef Block KI, Gyllenhaal C, Lowe L, Amedei A, Amin A, Amin A, Aquilano K, Arbiser J, Arreola A, Arzumanyan A, et al. Designing a broad-spectrum integrative approach for cancer prevention and treatment. Seminars Cancer Biol. 2015;35 Suppl(Suppl):S276–304.CrossRef
2.
Zurück zum Zitat Alobaedi OH, Talib WH, Basheti IA. Antitumor effect of thymoquinone combined with resveratrol on mice transplanted with breast cancer. Asian Pac J Trop Med. 2017;10(4):400–8.PubMedCrossRef Alobaedi OH, Talib WH, Basheti IA. Antitumor effect of thymoquinone combined with resveratrol on mice transplanted with breast cancer. Asian Pac J Trop Med. 2017;10(4):400–8.PubMedCrossRef
3.
Zurück zum Zitat Talib WH. Consumption of garlic and lemon aqueous extracts combination reduces tumor burden by angiogenesis inhibition, apoptosis induction, and immune system modulation. Nutrition (Burbank, Los Angeles County, Calif). 2017;43–44:89–97.CrossRef Talib WH. Consumption of garlic and lemon aqueous extracts combination reduces tumor burden by angiogenesis inhibition, apoptosis induction, and immune system modulation. Nutrition (Burbank, Los Angeles County, Calif). 2017;43–44:89–97.CrossRef
4.
Zurück zum Zitat Falah RR, Talib WH, Shbailat SJ. Combination of metformin and curcumin targets breast cancer in mice by angiogenesis inhibition, immune system modulation and induction of p53 independent apoptosis. Therapeutic Adv Med Oncol. 2017;9(4):235–52.CrossRef Falah RR, Talib WH, Shbailat SJ. Combination of metformin and curcumin targets breast cancer in mice by angiogenesis inhibition, immune system modulation and induction of p53 independent apoptosis. Therapeutic Adv Med Oncol. 2017;9(4):235–52.CrossRef
5.
Zurück zum Zitat Slominski A, Fischer TW, Zmijewski MA, Wortsman J, Semak I, Zbytek B, Slominski RM, Tobin DJ. On the role of melatonin in skin physiology and pathology. Endocrine. 2005;27(2):137–48.PubMedPubMedCentralCrossRef Slominski A, Fischer TW, Zmijewski MA, Wortsman J, Semak I, Zbytek B, Slominski RM, Tobin DJ. On the role of melatonin in skin physiology and pathology. Endocrine. 2005;27(2):137–48.PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Pourhanifeh MH, Mehrzadi S, Hosseinzadeh A. Melatonin and regulation of miRNAs: novel targeted therapy for cancerous and noncancerous disease. Epigenomics. 2020;13:65–81.PubMedCrossRef Pourhanifeh MH, Mehrzadi S, Hosseinzadeh A. Melatonin and regulation of miRNAs: novel targeted therapy for cancerous and noncancerous disease. Epigenomics. 2020;13:65–81.PubMedCrossRef
7.
Zurück zum Zitat Pourhanifeh MH, Hosseinzadeh A, Juybari KB, Mehrzadi S. Melatonin and urological cancers: a new therapeutic approach. Cancer Cell Int. 2020;20(1):444.CrossRef Pourhanifeh MH, Hosseinzadeh A, Juybari KB, Mehrzadi S. Melatonin and urological cancers: a new therapeutic approach. Cancer Cell Int. 2020;20(1):444.CrossRef
8.
Zurück zum Zitat Bahrampour Juybari K, Pourhanifeh MH, Hosseinzadeh A, Hemati K, Mehrzadi S. Melatonin potentials against viral infections including COVID-19: Current evidence and new findings. Virus Res. 2020;287:198108.PubMedPubMedCentralCrossRef Bahrampour Juybari K, Pourhanifeh MH, Hosseinzadeh A, Hemati K, Mehrzadi S. Melatonin potentials against viral infections including COVID-19: Current evidence and new findings. Virus Res. 2020;287:198108.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Mehrzadi S, Karimi MY, Fatemi A, Reiter RJ, Hosseinzadeh A. SARS-CoV-2 and other coronaviruses negatively influence mitochondrial quality control: beneficial effects of melatonin. Pharmacol Ther. 2021;224:107825–107825.PubMedPubMedCentralCrossRef Mehrzadi S, Karimi MY, Fatemi A, Reiter RJ, Hosseinzadeh A. SARS-CoV-2 and other coronaviruses negatively influence mitochondrial quality control: beneficial effects of melatonin. Pharmacol Ther. 2021;224:107825–107825.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Pourhanifeh MH, Mahdavinia M, Reiter RJ, Asemi Z. Potential use of melatonin in skin cancer treatment: a review of current biological evidence. J Cell Physiol. 2019;234(8):12142–8.PubMedCrossRef Pourhanifeh MH, Mahdavinia M, Reiter RJ, Asemi Z. Potential use of melatonin in skin cancer treatment: a review of current biological evidence. J Cell Physiol. 2019;234(8):12142–8.PubMedCrossRef
11.
Zurück zum Zitat Anderson G. The effects of melatonin on signaling pathways and molecules involved in glioma: melatonin and glioblastoma: pathophysiology and treatment. Fundam Clin Pharmacol. 2020;34(2):189–91.PubMedCrossRef Anderson G. The effects of melatonin on signaling pathways and molecules involved in glioma: melatonin and glioblastoma: pathophysiology and treatment. Fundam Clin Pharmacol. 2020;34(2):189–91.PubMedCrossRef
12.
Zurück zum Zitat Pourhanifeh MH, Sharifi M, Reiter RJ, Davoodabadi A, Asemi Z. Melatonin and non-small cell lung cancer: new insights into signaling pathways. Cancer Cell Int. 2019;19:131.PubMedPubMedCentralCrossRef Pourhanifeh MH, Sharifi M, Reiter RJ, Davoodabadi A, Asemi Z. Melatonin and non-small cell lung cancer: new insights into signaling pathways. Cancer Cell Int. 2019;19:131.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Zhao Q, Wang W, Cui J. Melatonin enhances TNF-α-mediated cervical cancer HeLa cells death via suppressing CaMKII/Parkin/mitophagy axis. Cancer Cell Int. 2019;19:58.PubMedPubMedCentralCrossRef Zhao Q, Wang W, Cui J. Melatonin enhances TNF-α-mediated cervical cancer HeLa cells death via suppressing CaMKII/Parkin/mitophagy axis. Cancer Cell Int. 2019;19:58.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Bu S, Wang Q, Sun J, Li X, Gu T, Lai D. Melatonin suppresses chronic restraint stress-mediated metastasis of epithelial ovarian cancer via NE/AKT/β-catenin/SLUG axis. Cell Death Dis. 2020;11(8):644.PubMedPubMedCentralCrossRef Bu S, Wang Q, Sun J, Li X, Gu T, Lai D. Melatonin suppresses chronic restraint stress-mediated metastasis of epithelial ovarian cancer via NE/AKT/β-catenin/SLUG axis. Cell Death Dis. 2020;11(8):644.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Yan G, Lei H, He M, Gong R, Wang Y, He X, Li G, Pang P, Li X, Yu S, et al. Melatonin triggers autophagic cell death by regulating RORC in Hodgkin lymphoma. Biomed Pharmacother. 2020;123:109811.PubMedCrossRef Yan G, Lei H, He M, Gong R, Wang Y, He X, Li G, Pang P, Li X, Yu S, et al. Melatonin triggers autophagic cell death by regulating RORC in Hodgkin lymphoma. Biomed Pharmacother. 2020;123:109811.PubMedCrossRef
17.
Zurück zum Zitat Tian T, Li J, Li Y, Lu YX, Tang YL, Wang H, Zheng F, Shi D, Long Q, Chen M, et al. Melatonin enhances sorafenib-induced cytotoxicity in FLT3-ITD acute myeloid leukemia cells by redox modification. Theranostics. 2019;9(13):3768–79.PubMedPubMedCentralCrossRef Tian T, Li J, Li Y, Lu YX, Tang YL, Wang H, Zheng F, Shi D, Long Q, Chen M, et al. Melatonin enhances sorafenib-induced cytotoxicity in FLT3-ITD acute myeloid leukemia cells by redox modification. Theranostics. 2019;9(13):3768–79.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Reiter RJ, Tan DX, Fuentes-Broto L. Melatonin: a multitasking molecule. Prog Brain Res. 2010;181:127–51.PubMedCrossRef Reiter RJ, Tan DX, Fuentes-Broto L. Melatonin: a multitasking molecule. Prog Brain Res. 2010;181:127–51.PubMedCrossRef
19.
Zurück zum Zitat Bonnefont-Rousselot D, Collin F. Melatonin: action as antioxidant and potential applications in human disease and aging. Toxicology. 2010;278(1):55–67.PubMedCrossRef Bonnefont-Rousselot D, Collin F. Melatonin: action as antioxidant and potential applications in human disease and aging. Toxicology. 2010;278(1):55–67.PubMedCrossRef
20.
Zurück zum Zitat Slominski RM, Reiter RJ, Schlabritz-Loutsevitch N, Ostrom RS, Slominski AT. Melatonin membrane receptors in peripheral tissues: distribution and functions. Mol Cell Endocrinol. 2012;351(2):152–66.PubMedPubMedCentralCrossRef Slominski RM, Reiter RJ, Schlabritz-Loutsevitch N, Ostrom RS, Slominski AT. Melatonin membrane receptors in peripheral tissues: distribution and functions. Mol Cell Endocrinol. 2012;351(2):152–66.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Stevens RG, Brainard GC, Blask DE, Lockley SW, Motta ME. Breast cancer and circadian disruption from electric lighting in the modern world. CA Cancer J Clin. 2014;64(3):207–18.PubMedCrossRef Stevens RG, Brainard GC, Blask DE, Lockley SW, Motta ME. Breast cancer and circadian disruption from electric lighting in the modern world. CA Cancer J Clin. 2014;64(3):207–18.PubMedCrossRef
22.
Zurück zum Zitat He R, Cui M, Lin H, Zhao L, Wang J, Chen S, Shao Z. Melatonin resists oxidative stress-induced apoptosis in nucleus pulposus cells. Life Sci. 2018;199:122–30.PubMedCrossRef He R, Cui M, Lin H, Zhao L, Wang J, Chen S, Shao Z. Melatonin resists oxidative stress-induced apoptosis in nucleus pulposus cells. Life Sci. 2018;199:122–30.PubMedCrossRef
23.
Zurück zum Zitat Reiter RJ, Tan DX, Erren TC, Fuentes-Broto L, Paredes SD. Light-mediated perturbations of circadian timing and cancer risk: a mechanistic analysis. Integr Cancer Ther. 2009;8(4):354–60.PubMedCrossRef Reiter RJ, Tan DX, Erren TC, Fuentes-Broto L, Paredes SD. Light-mediated perturbations of circadian timing and cancer risk: a mechanistic analysis. Integr Cancer Ther. 2009;8(4):354–60.PubMedCrossRef
24.
Zurück zum Zitat Pourhanifeh MH, Hosseinzadeh A, Dehdashtian E, Hemati K, Mehrzadi S. Melatonin: new insights on its therapeutic properties in diabetic complications. Diabetol Metab Syndr. 2020;12:1–20.CrossRef Pourhanifeh MH, Hosseinzadeh A, Dehdashtian E, Hemati K, Mehrzadi S. Melatonin: new insights on its therapeutic properties in diabetic complications. Diabetol Metab Syndr. 2020;12:1–20.CrossRef
25.
Zurück zum Zitat Dehdashtian E, Pourhanifeh MH, Hemati K, Mehrzadi S, Hosseinzadeh A. Therapeutic application of nutraceuticals in diabetic nephropathy: current evidence and future implications. Diabetes Metab Res Rev. 2020;36:e3336.PubMedCrossRef Dehdashtian E, Pourhanifeh MH, Hemati K, Mehrzadi S, Hosseinzadeh A. Therapeutic application of nutraceuticals in diabetic nephropathy: current evidence and future implications. Diabetes Metab Res Rev. 2020;36:e3336.PubMedCrossRef
26.
Zurück zum Zitat Rosales-Corral S, Tan DX, Reiter RJ, Valdivia-Velázquez M, Martínez-Barboza G, Acosta-Martínez JP, Ortiz GG. Orally administered melatonin reduces oxidative stress and proinflammatory cytokines induced by amyloid-beta peptide in rat brain: a comparative, in vivo study versus vitamin C and E. J Pineal Res. 2003;35(2):80–4.PubMedCrossRef Rosales-Corral S, Tan DX, Reiter RJ, Valdivia-Velázquez M, Martínez-Barboza G, Acosta-Martínez JP, Ortiz GG. Orally administered melatonin reduces oxidative stress and proinflammatory cytokines induced by amyloid-beta peptide in rat brain: a comparative, in vivo study versus vitamin C and E. J Pineal Res. 2003;35(2):80–4.PubMedCrossRef
27.
Zurück zum Zitat Hosseinzadeh A, Javad-Moosavi SA, Reiter RJ, Hemati K, Ghaznavi H, Mehrzadi S. Idiopathic pulmonary fibrosis (IPF) signaling pathways and protective roles of melatonin. Life Sci. 2018;201:17–29.PubMedCrossRef Hosseinzadeh A, Javad-Moosavi SA, Reiter RJ, Hemati K, Ghaznavi H, Mehrzadi S. Idiopathic pulmonary fibrosis (IPF) signaling pathways and protective roles of melatonin. Life Sci. 2018;201:17–29.PubMedCrossRef
28.
Zurück zum Zitat Daryani A, Montazeri M, Pagheh AS, Sharif M, Sarvi S, Hosseinzadeh A, Reiter RJ, Hadighi R, Joghataei MT, Ghaznavi H. The potential use of melatonin to treat protozoan parasitic infections: a review. Biomed Pharmacother. 2018;97:948–57.PubMedCrossRef Daryani A, Montazeri M, Pagheh AS, Sharif M, Sarvi S, Hosseinzadeh A, Reiter RJ, Hadighi R, Joghataei MT, Ghaznavi H. The potential use of melatonin to treat protozoan parasitic infections: a review. Biomed Pharmacother. 2018;97:948–57.PubMedCrossRef
29.
Zurück zum Zitat Yokota O, Terada S, Ishizu H, Ishihara T, Ujike H, Nakashima H, Nakashima Y, Kugo A, Checler F, Kuroda S. Cyclooxygenase-2 in the hippocampus is up-regulated in Alzheimer’s disease but not in variant Alzheimer’s disease with cotton wool plaques in humans. Neurosci Lett. 2003;343(3):175–9.PubMedCrossRef Yokota O, Terada S, Ishizu H, Ishihara T, Ujike H, Nakashima H, Nakashima Y, Kugo A, Checler F, Kuroda S. Cyclooxygenase-2 in the hippocampus is up-regulated in Alzheimer’s disease but not in variant Alzheimer’s disease with cotton wool plaques in humans. Neurosci Lett. 2003;343(3):175–9.PubMedCrossRef
30.
Zurück zum Zitat Juybari KB, Hosseinzadeh A, Ghaznavi H, Kamali M, Sedaghat A, Mehrzadi S, Naseripour M. Melatonin as a modulator of degenerative and regenerative signaling pathways in injured Retinal Ganglion Cells. Curr Pharm Des. 2019;25(28):3057–73.PubMedCrossRef Juybari KB, Hosseinzadeh A, Ghaznavi H, Kamali M, Sedaghat A, Mehrzadi S, Naseripour M. Melatonin as a modulator of degenerative and regenerative signaling pathways in injured Retinal Ganglion Cells. Curr Pharm Des. 2019;25(28):3057–73.PubMedCrossRef
31.
Zurück zum Zitat Menéndez-Menéndez J, Hermida-Prado F, Granda-Díaz R, González A, García-Pedrero JM, Del-Río-Ibisate N, González-González A, Cos S, Alonso-González C, Martínez-Campa C. Deciphering the molecular basis of melatonin protective effects on breast cells treated with doxorubicin: TWIST1 a transcription factor involved in EMT and metastasis, a novel target of melatonin. Cancers. 2019;11(7):1011.PubMedCentralCrossRef Menéndez-Menéndez J, Hermida-Prado F, Granda-Díaz R, González A, García-Pedrero JM, Del-Río-Ibisate N, González-González A, Cos S, Alonso-González C, Martínez-Campa C. Deciphering the molecular basis of melatonin protective effects on breast cells treated with doxorubicin: TWIST1 a transcription factor involved in EMT and metastasis, a novel target of melatonin. Cancers. 2019;11(7):1011.PubMedCentralCrossRef
32.
Zurück zum Zitat Wang J, Xiao X, Zhang Y, Shi D, Chen W, Fu L, Liu L, Xie F, Kang T, Huang W, et al. Simultaneous modulation of COX-2, p300, Akt, and Apaf-1 signaling by melatonin to inhibit proliferation and induce apoptosis in breast cancer cells. J Pineal Res. 2012;53(1):77–90.PubMedCrossRef Wang J, Xiao X, Zhang Y, Shi D, Chen W, Fu L, Liu L, Xie F, Kang T, Huang W, et al. Simultaneous modulation of COX-2, p300, Akt, and Apaf-1 signaling by melatonin to inhibit proliferation and induce apoptosis in breast cancer cells. J Pineal Res. 2012;53(1):77–90.PubMedCrossRef
33.
Zurück zum Zitat Cipolla-Neto J, Amaral FGD. Melatonin as a hormone: new physiological and clinical insights. Endocr Rev. 2018;39(6):990–1028.PubMedCrossRef Cipolla-Neto J, Amaral FGD. Melatonin as a hormone: new physiological and clinical insights. Endocr Rev. 2018;39(6):990–1028.PubMedCrossRef
34.
Zurück zum Zitat Pourhanifeh MH, Dehdashtian E, Hosseinzadeh A, Sezavar SH, Mehrzadi S. Clinical application of melatonin in the treatment of cardiovascular diseases: current evidence and new insights into the cardioprotective and cardiotherapeutic properties. Cardiovasc Drugs Ther. 2020;14:1–25. Pourhanifeh MH, Dehdashtian E, Hosseinzadeh A, Sezavar SH, Mehrzadi S. Clinical application of melatonin in the treatment of cardiovascular diseases: current evidence and new insights into the cardioprotective and cardiotherapeutic properties. Cardiovasc Drugs Ther. 2020;14:1–25.
35.
Zurück zum Zitat Sánchez-Barceló EJ, Cos S, Fernández R, Mediavilla MD. Melatonin and mammary cancer: a short review. Endocr Relat Cancer. 2003;10(2):153–9.PubMedCrossRef Sánchez-Barceló EJ, Cos S, Fernández R, Mediavilla MD. Melatonin and mammary cancer: a short review. Endocr Relat Cancer. 2003;10(2):153–9.PubMedCrossRef
36.
Zurück zum Zitat Cos S, Sánchez-Barceló EJ. Melatonin and mammary pathological growth. Front Neuroendocrinol. 2000;21(2):133–70.PubMedCrossRef Cos S, Sánchez-Barceló EJ. Melatonin and mammary pathological growth. Front Neuroendocrinol. 2000;21(2):133–70.PubMedCrossRef
37.
Zurück zum Zitat Cos S, Fernández R, Güézmes A, Sánchez-Barceló EJ. Influence of melatonin on invasive and metastatic properties of MCF-7 human breast cancer cells. Can Res. 1998;58(19):4383–90. Cos S, Fernández R, Güézmes A, Sánchez-Barceló EJ. Influence of melatonin on invasive and metastatic properties of MCF-7 human breast cancer cells. Can Res. 1998;58(19):4383–90.
38.
Zurück zum Zitat Bergers G, Benjamin LE. Tumorigenesis and the angiogenic switch. Nat Rev Cancer. 2003;3(6):401–10.PubMedCrossRef Bergers G, Benjamin LE. Tumorigenesis and the angiogenic switch. Nat Rev Cancer. 2003;3(6):401–10.PubMedCrossRef
39.
Zurück zum Zitat Vaupel P. The role of hypoxia-induced factors in tumor progression. Oncologist. 2004;9(Suppl 5):10–7.PubMedCrossRef Vaupel P. The role of hypoxia-induced factors in tumor progression. Oncologist. 2004;9(Suppl 5):10–7.PubMedCrossRef
40.
Zurück zum Zitat Hurwitz H, Fehrenbacher L, Novotny W, Cartwright T, Hainsworth J, Heim W, Berlin J, Baron A, Griffing S, Holmgren E, et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med. 2004;350(23):2335–42.PubMedCrossRef Hurwitz H, Fehrenbacher L, Novotny W, Cartwright T, Hainsworth J, Heim W, Berlin J, Baron A, Griffing S, Holmgren E, et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med. 2004;350(23):2335–42.PubMedCrossRef
41.
Zurück zum Zitat Jeong JW, Bae MK, Ahn MY, Kim SH, Sohn TK, Bae MH, Yoo MA, Song EJ, Lee KJ, Kim KW. Regulation and destabilization of HIF-1alpha by ARD1-mediated acetylation. Cell. 2002;111(5):709–20.PubMedCrossRef Jeong JW, Bae MK, Ahn MY, Kim SH, Sohn TK, Bae MH, Yoo MA, Song EJ, Lee KJ, Kim KW. Regulation and destabilization of HIF-1alpha by ARD1-mediated acetylation. Cell. 2002;111(5):709–20.PubMedCrossRef
42.
Zurück zum Zitat Mirza-Aghazadeh-Attari M, Reiter RJ, Rikhtegar R, Jalili J, Hajalioghli P, Mihanfar A, Majidinia M, Yousefi B. Melatonin: an atypical hormone with major functions in the regulation of angiogenesis. IUBMB Life. 2020;72(8):1560–84.PubMedCrossRef Mirza-Aghazadeh-Attari M, Reiter RJ, Rikhtegar R, Jalili J, Hajalioghli P, Mihanfar A, Majidinia M, Yousefi B. Melatonin: an atypical hormone with major functions in the regulation of angiogenesis. IUBMB Life. 2020;72(8):1560–84.PubMedCrossRef
43.
Zurück zum Zitat Ma Q, Reiter RJ, Chen Y. Role of melatonin in controlling angiogenesis under physiological and pathological conditions. Angiogenesis. 2020;23(2):91–104.PubMedCrossRef Ma Q, Reiter RJ, Chen Y. Role of melatonin in controlling angiogenesis under physiological and pathological conditions. Angiogenesis. 2020;23(2):91–104.PubMedCrossRef
44.
Zurück zum Zitat Park SY, Jang WJ, Yi EY, Jang JY, Jung Y, Jeong JW, Kim YJ. Melatonin suppresses tumor angiogenesis by inhibiting HIF-1alpha stabilization under hypoxia. J Pineal Res. 2010;48(2):178–84.PubMedCrossRef Park SY, Jang WJ, Yi EY, Jang JY, Jung Y, Jeong JW, Kim YJ. Melatonin suppresses tumor angiogenesis by inhibiting HIF-1alpha stabilization under hypoxia. J Pineal Res. 2010;48(2):178–84.PubMedCrossRef
45.
Zurück zum Zitat Cho SY, Lee HJ, Jeong SJ, Lee HJ, Kim HS, Chen CY, Lee EO, Kim SH. Sphingosine kinase 1 pathway is involved in melatonin-induced HIF-1α inactivation in hypoxic PC-3 prostate cancer cells. J Pineal Res. 2011;51(1):87–93.PubMedCrossRef Cho SY, Lee HJ, Jeong SJ, Lee HJ, Kim HS, Chen CY, Lee EO, Kim SH. Sphingosine kinase 1 pathway is involved in melatonin-induced HIF-1α inactivation in hypoxic PC-3 prostate cancer cells. J Pineal Res. 2011;51(1):87–93.PubMedCrossRef
46.
Zurück zum Zitat Alvarez-García V, González A, Alonso-González C, Martínez-Campa C, Cos S. Regulation of vascular endothelial growth factor by melatonin in human breast cancer cells. J Pineal Res. 2013;54(4):373–80.PubMedCrossRef Alvarez-García V, González A, Alonso-González C, Martínez-Campa C, Cos S. Regulation of vascular endothelial growth factor by melatonin in human breast cancer cells. J Pineal Res. 2013;54(4):373–80.PubMedCrossRef
47.
Zurück zum Zitat Siu SW, Lau KW, Tam PC, Shiu SY. Melatonin and prostate cancer cell proliferation: interplay with castration, epidermal growth factor, and androgen sensitivity. Prostate. 2002;52(2):106–22.PubMedCrossRef Siu SW, Lau KW, Tam PC, Shiu SY. Melatonin and prostate cancer cell proliferation: interplay with castration, epidermal growth factor, and androgen sensitivity. Prostate. 2002;52(2):106–22.PubMedCrossRef
48.
Zurück zum Zitat Liu R, Wang HL, Deng MJ, Wen XJ, Mo YY, Chen FM, Zou CL, Duan WF, Li L, Nie X. Melatonin inhibits reactive oxygen species-driven proliferation, epithelial-mesenchymal transition, and vasculogenic mimicry in oral cancer. Oxid Med Cell Longev. 2018;2018:3510970.PubMedPubMedCentralCrossRef Liu R, Wang HL, Deng MJ, Wen XJ, Mo YY, Chen FM, Zou CL, Duan WF, Li L, Nie X. Melatonin inhibits reactive oxygen species-driven proliferation, epithelial-mesenchymal transition, and vasculogenic mimicry in oral cancer. Oxid Med Cell Longev. 2018;2018:3510970.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Liu H, Zhu Y, Zhu H, Cai R, Wang KF, Song J, Wang RX, Zhou RX. Role of transforming growth factor β1 in the inhibition of gastric cancer cell proliferation by melatonin in vitro and in vivo. Oncol Rep. 2019;42(2):753–62.PubMed Liu H, Zhu Y, Zhu H, Cai R, Wang KF, Song J, Wang RX, Zhou RX. Role of transforming growth factor β1 in the inhibition of gastric cancer cell proliferation by melatonin in vitro and in vivo. Oncol Rep. 2019;42(2):753–62.PubMed
50.
Zurück zum Zitat León J, Casado J, Jiménez Ruiz SM, Zurita MS, González-Puga C, Rejón JD, Gila A, Muñoz de Rueda P, Pavón EJ, Reiter RJ, et al. Melatonin reduces endothelin-1 expression and secretion in colon cancer cells through the inactivation of FoxO-1 and NF-κβ. J Pineal Res. 2014;56(4):415–26.PubMedCrossRef León J, Casado J, Jiménez Ruiz SM, Zurita MS, González-Puga C, Rejón JD, Gila A, Muñoz de Rueda P, Pavón EJ, Reiter RJ, et al. Melatonin reduces endothelin-1 expression and secretion in colon cancer cells through the inactivation of FoxO-1 and NF-κβ. J Pineal Res. 2014;56(4):415–26.PubMedCrossRef
51.
Zurück zum Zitat Goncalves Ndo N, Rodrigues RV, Jardim-Perassi BV, Moschetta MG, Lopes JR, Colombo J, Zuccari DA. Molecular markers of angiogenesis and metastasis in lines of oral carcinoma after treatment with melatonin. Anticancer Agents Med Chem. 2014;14(9):1302–11.PubMedCrossRef Goncalves Ndo N, Rodrigues RV, Jardim-Perassi BV, Moschetta MG, Lopes JR, Colombo J, Zuccari DA. Molecular markers of angiogenesis and metastasis in lines of oral carcinoma after treatment with melatonin. Anticancer Agents Med Chem. 2014;14(9):1302–11.PubMedCrossRef
52.
Zurück zum Zitat Maschio-Signorini LB, Gelaleti GB, Moschetta MG, Borin TF, Jardim-Perassi BV, Lopes JR, Lacerda JZ, Roela RA, Bordin NA, Corrêa LA, et al. Melatonin regulates angiogenic and inflammatory proteins in MDA-MB-231 cell line and in co-culture with cancer-associated fibroblasts. Anticancer Agents Med Chem. 2016;16(11):1474–84.PubMedCrossRef Maschio-Signorini LB, Gelaleti GB, Moschetta MG, Borin TF, Jardim-Perassi BV, Lopes JR, Lacerda JZ, Roela RA, Bordin NA, Corrêa LA, et al. Melatonin regulates angiogenic and inflammatory proteins in MDA-MB-231 cell line and in co-culture with cancer-associated fibroblasts. Anticancer Agents Med Chem. 2016;16(11):1474–84.PubMedCrossRef
53.
Zurück zum Zitat Wang RX, Liu H, Xu L, Zhang H, Zhou RX. Melatonin downregulates nuclear receptor RZR/RORγ expression causing growth-inhibitory and anti-angiogenesis activity in human gastric cancer cells in vitro and in vivo. Oncol Lett. 2016;12(2):897–903.PubMedPubMedCentralCrossRef Wang RX, Liu H, Xu L, Zhang H, Zhou RX. Melatonin downregulates nuclear receptor RZR/RORγ expression causing growth-inhibitory and anti-angiogenesis activity in human gastric cancer cells in vitro and in vivo. Oncol Lett. 2016;12(2):897–903.PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Lissoni P, Rovelli F, Malugani F, Bucovec R, Conti A, Maestroni GJ. Anti-angiogenic activity of melatonin in advanced cancer patients. Neuro Endocrinol Lett. 2001;22(1):45–7.PubMed Lissoni P, Rovelli F, Malugani F, Bucovec R, Conti A, Maestroni GJ. Anti-angiogenic activity of melatonin in advanced cancer patients. Neuro Endocrinol Lett. 2001;22(1):45–7.PubMed
55.
Zurück zum Zitat Vimalraj S, Saravanan S, Raghunandhakumar S, Anuradha D. Melatonin regulates tumor angiogenesis via miR-424-5p/VEGFA signaling pathway in osteosarcoma. Life Sci. 2020;256:118011.PubMedCrossRef Vimalraj S, Saravanan S, Raghunandhakumar S, Anuradha D. Melatonin regulates tumor angiogenesis via miR-424-5p/VEGFA signaling pathway in osteosarcoma. Life Sci. 2020;256:118011.PubMedCrossRef
56.
57.
Zurück zum Zitat Bagherifard A, Kadijani AA, Yahyazadeh H, Rezazadeh J, Azizi M, Akbari A, Mirzaei A. The value of serum total oxidant to the antioxidant ratio as a biomarker of knee osteoarthritis. Clin Nutr ESPEN. 2020;38:118–23.PubMedCrossRef Bagherifard A, Kadijani AA, Yahyazadeh H, Rezazadeh J, Azizi M, Akbari A, Mirzaei A. The value of serum total oxidant to the antioxidant ratio as a biomarker of knee osteoarthritis. Clin Nutr ESPEN. 2020;38:118–23.PubMedCrossRef
58.
Zurück zum Zitat Saha SK, Lee SB, Won J, Choi HY, Kim K, Yang G-M, Dayem AA, Cho S-G. Correlation between oxidative stress, nutrition, and cancer initiation. Int J Mol Sci. 2017;18(7):1544.PubMedCentralCrossRef Saha SK, Lee SB, Won J, Choi HY, Kim K, Yang G-M, Dayem AA, Cho S-G. Correlation between oxidative stress, nutrition, and cancer initiation. Int J Mol Sci. 2017;18(7):1544.PubMedCentralCrossRef
59.
Zurück zum Zitat Reuter S, Gupta SC, Chaturvedi MM, Aggarwal BB. Oxidative stress, inflammation, and cancer: how are they linked? Free Radic Biol Med. 2010;49(11):1603–16.PubMedPubMedCentralCrossRef Reuter S, Gupta SC, Chaturvedi MM, Aggarwal BB. Oxidative stress, inflammation, and cancer: how are they linked? Free Radic Biol Med. 2010;49(11):1603–16.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Zhang HM, Zhang Y. Melatonin: a well-documented antioxidant with conditional pro-oxidant actions. J Pineal Res. 2014;57(2):131–46.PubMedCrossRef Zhang HM, Zhang Y. Melatonin: a well-documented antioxidant with conditional pro-oxidant actions. J Pineal Res. 2014;57(2):131–46.PubMedCrossRef
61.
Zurück zum Zitat Galano A, Tan DX, Reiter RJ. Melatonin as a natural ally against oxidative stress: a physicochemical examination. J Pineal Res. 2011;51(1):1–16.PubMedCrossRef Galano A, Tan DX, Reiter RJ. Melatonin as a natural ally against oxidative stress: a physicochemical examination. J Pineal Res. 2011;51(1):1–16.PubMedCrossRef
62.
63.
Zurück zum Zitat Sun TC, Liu XC, Yang SH, Song LL, Zhou SJ, Deng SL, Tian L, Cheng LY. Melatonin inhibits oxidative stress and apoptosis in cryopreserved ovarian tissues via Nrf2/HO-1 signaling pathway. Front Mol Biosci. 2020;7:163.PubMedPubMedCentralCrossRef Sun TC, Liu XC, Yang SH, Song LL, Zhou SJ, Deng SL, Tian L, Cheng LY. Melatonin inhibits oxidative stress and apoptosis in cryopreserved ovarian tissues via Nrf2/HO-1 signaling pathway. Front Mol Biosci. 2020;7:163.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat He L, He T, Farrar S, Ji L, Liu T, Ma X. Antioxidants maintain cellular redox homeostasis by elimination of reactive oxygen species. Cell Physiol Biochem. 2017;44(2):532–53.PubMedCrossRef He L, He T, Farrar S, Ji L, Liu T, Ma X. Antioxidants maintain cellular redox homeostasis by elimination of reactive oxygen species. Cell Physiol Biochem. 2017;44(2):532–53.PubMedCrossRef
65.
Zurück zum Zitat Yoo YS, Han HG, Jeon YJ. Unfolded protein response of the endoplasmic reticulum in tumor progression and immunogenicity. Oxid Med Cell Longev. 2017;2017:2969271.PubMedPubMedCentralCrossRef Yoo YS, Han HG, Jeon YJ. Unfolded protein response of the endoplasmic reticulum in tumor progression and immunogenicity. Oxid Med Cell Longev. 2017;2017:2969271.PubMedPubMedCentralCrossRef
66.
69.
Zurück zum Zitat Mehrzadi S, Hemati K, Reiter RJ, Hosseinzadeh A. Mitochondrial dysfunction in age-related macular degeneration: melatonin as a potential treatment. Expert Opin Ther Targets. 2020;24(4):359–78.PubMedCrossRef Mehrzadi S, Hemati K, Reiter RJ, Hosseinzadeh A. Mitochondrial dysfunction in age-related macular degeneration: melatonin as a potential treatment. Expert Opin Ther Targets. 2020;24(4):359–78.PubMedCrossRef
70.
Zurück zum Zitat Lin Y, Jiang M, Chen W, Zhao T, Wei Y. Cancer and ER stress: Mutual crosstalk between autophagy, oxidative stress and inflammatory response. Biomed Pharmacother. 2019;118:109249.PubMedCrossRef Lin Y, Jiang M, Chen W, Zhao T, Wei Y. Cancer and ER stress: Mutual crosstalk between autophagy, oxidative stress and inflammatory response. Biomed Pharmacother. 2019;118:109249.PubMedCrossRef
71.
Zurück zum Zitat Yun CW, Kim S, Lee JH, Lee SH. Melatonin promotes apoptosis of colorectal cancer cells via superoxide-mediated ER stress by inhibiting cellular prion protein expression. Anticancer Res. 2018;38(7):3951–60.PubMedCrossRef Yun CW, Kim S, Lee JH, Lee SH. Melatonin promotes apoptosis of colorectal cancer cells via superoxide-mediated ER stress by inhibiting cellular prion protein expression. Anticancer Res. 2018;38(7):3951–60.PubMedCrossRef
72.
Zurück zum Zitat Kim HS, Han TY, Yoo YM. Melatonin-mediated intracellular insulin during 2-deoxy-d-glucose treatment is reduced through autophagy and EDC3 protein in insulinoma INS-1E cells. Oxid Med Cell Longev. 2016;2016:2594703.PubMedPubMedCentralCrossRef Kim HS, Han TY, Yoo YM. Melatonin-mediated intracellular insulin during 2-deoxy-d-glucose treatment is reduced through autophagy and EDC3 protein in insulinoma INS-1E cells. Oxid Med Cell Longev. 2016;2016:2594703.PubMedPubMedCentralCrossRef
73.
Zurück zum Zitat Zha L, Fan L, Sun G, Wang H, Ma T, Zhong F, Wei W. Melatonin sensitizes human hepatoma cells to endoplasmic reticulum stress-induced apoptosis. J Pineal Res. 2012;52(3):322–31.PubMedCrossRef Zha L, Fan L, Sun G, Wang H, Ma T, Zhong F, Wei W. Melatonin sensitizes human hepatoma cells to endoplasmic reticulum stress-induced apoptosis. J Pineal Res. 2012;52(3):322–31.PubMedCrossRef
74.
Zurück zum Zitat Bu LJ, Yu HQ, Fan LL, Li XQ, Wang F, Liu JT, Zhong F, Zhang CJ, Wei W, Wang H, et al. Melatonin, a novel selective ATF-6 inhibitor, induces human hepatoma cell apoptosis through COX-2 downregulation. World J Gastroenterol. 2017;23(6):986–98.PubMedPubMedCentralCrossRef Bu LJ, Yu HQ, Fan LL, Li XQ, Wang F, Liu JT, Zhong F, Zhang CJ, Wei W, Wang H, et al. Melatonin, a novel selective ATF-6 inhibitor, induces human hepatoma cell apoptosis through COX-2 downregulation. World J Gastroenterol. 2017;23(6):986–98.PubMedPubMedCentralCrossRef
75.
Zurück zum Zitat Moreira AJ, Ordoñez R, Cerski CT, Picada JN, García-Palomo A, Marroni NP, Mauriz JL, González-Gallego J. Melatonin activates endoplasmic reticulum stress and apoptosis in rats with diethylnitrosamine-induced hepatocarcinogenesis. PLoS ONE. 2015;10(12):e0144517.PubMedPubMedCentralCrossRef Moreira AJ, Ordoñez R, Cerski CT, Picada JN, García-Palomo A, Marroni NP, Mauriz JL, González-Gallego J. Melatonin activates endoplasmic reticulum stress and apoptosis in rats with diethylnitrosamine-induced hepatocarcinogenesis. PLoS ONE. 2015;10(12):e0144517.PubMedPubMedCentralCrossRef
76.
Zurück zum Zitat Zhou B, Lu Q, Liu J, Fan L, Wang Y, Wei W, Wang H, Sun G. Melatonin increases the sensitivity of hepatocellular carcinoma to sorafenib through the PERK-ATF4-beclin1 pathway. Int J Biol Sci. 2019;15(9):1905–20.PubMedPubMedCentralCrossRef Zhou B, Lu Q, Liu J, Fan L, Wang Y, Wei W, Wang H, Sun G. Melatonin increases the sensitivity of hepatocellular carcinoma to sorafenib through the PERK-ATF4-beclin1 pathway. Int J Biol Sci. 2019;15(9):1905–20.PubMedPubMedCentralCrossRef
77.
Zurück zum Zitat Zheng Y, Tu J, Wang X, Yu Y, Li J, Jin Y, Wu J. The therapeutic effect of melatonin on GC by inducing cell apoptosis and autophagy induced by endoplasmic reticulum stress. Onco Targets Ther. 2019;12:10187–98.PubMedPubMedCentralCrossRef Zheng Y, Tu J, Wang X, Yu Y, Li J, Jin Y, Wu J. The therapeutic effect of melatonin on GC by inducing cell apoptosis and autophagy induced by endoplasmic reticulum stress. Onco Targets Ther. 2019;12:10187–98.PubMedPubMedCentralCrossRef
78.
Zurück zum Zitat Yoo YM. Melatonin-mediated insulin synthesis during endoplasmic reticulum stress involves HuD expression in rat insulinoma INS-1E cells. J Pineal Res. 2013;55(2):207–20.PubMedCrossRef Yoo YM. Melatonin-mediated insulin synthesis during endoplasmic reticulum stress involves HuD expression in rat insulinoma INS-1E cells. J Pineal Res. 2013;55(2):207–20.PubMedCrossRef
79.
Zurück zum Zitat Fan L, Sun G, Ma T, Zhong F, Lei Y, Li X, Wei W. Melatonin reverses tunicamycin-induced endoplasmic reticulum stress in human hepatocellular carcinoma cells and improves cytotoxic response to doxorubicin by increasing CHOP and decreasing survivin. J Pineal Res. 2013;55(2):184–94.PubMedCrossRef Fan L, Sun G, Ma T, Zhong F, Lei Y, Li X, Wei W. Melatonin reverses tunicamycin-induced endoplasmic reticulum stress in human hepatocellular carcinoma cells and improves cytotoxic response to doxorubicin by increasing CHOP and decreasing survivin. J Pineal Res. 2013;55(2):184–94.PubMedCrossRef
80.
Zurück zum Zitat Zhu B, Ma AQ, Yang L, Dang XM. Atorvastatin attenuates bleomycin-induced pulmonary fibrosis via suppressing iNOS expression and the CTGF (CCN2)/ERK signaling pathway. Int J Mol Sci. 2013;14(12):24476–91.PubMedPubMedCentralCrossRef Zhu B, Ma AQ, Yang L, Dang XM. Atorvastatin attenuates bleomycin-induced pulmonary fibrosis via suppressing iNOS expression and the CTGF (CCN2)/ERK signaling pathway. Int J Mol Sci. 2013;14(12):24476–91.PubMedPubMedCentralCrossRef
81.
Zurück zum Zitat Bento CF, Renna M, Ghislat G, Puri C, Ashkenazi A, Vicinanza M, Menzies FM, Rubinsztein DC. Mammalian autophagy: how does it work? Annu Rev Biochem. 2016;85:685–713.PubMedCrossRef Bento CF, Renna M, Ghislat G, Puri C, Ashkenazi A, Vicinanza M, Menzies FM, Rubinsztein DC. Mammalian autophagy: how does it work? Annu Rev Biochem. 2016;85:685–713.PubMedCrossRef
84.
Zurück zum Zitat Stolz A, Ernst A, Dikic I. Cargo recognition and trafficking in selective autophagy. Nat Cell Biol. 2014;16(6):495–501.PubMedCrossRef Stolz A, Ernst A, Dikic I. Cargo recognition and trafficking in selective autophagy. Nat Cell Biol. 2014;16(6):495–501.PubMedCrossRef
85.
Zurück zum Zitat Song S, Tan J, Miao Y, Li M, Zhang Q. Crosstalk of autophagy and apoptosis: Involvement of the dual role of autophagy under ER stress. J Cell Physiol. 2017;232:2977–84.PubMedCrossRef Song S, Tan J, Miao Y, Li M, Zhang Q. Crosstalk of autophagy and apoptosis: Involvement of the dual role of autophagy under ER stress. J Cell Physiol. 2017;232:2977–84.PubMedCrossRef
86.
Zurück zum Zitat B’Chir W, Maurin AC, Carraro V, Averous J, Jousse C, Muranishi Y, Parry L, Stepien G, Fafournoux P, Bruhat A. The eIF2alpha/ATF4 pathway is essential for stress-induced autophagy gene expression. Nucleic Acids Res. 2013;41(16):7683–99.PubMedPubMedCentralCrossRef B’Chir W, Maurin AC, Carraro V, Averous J, Jousse C, Muranishi Y, Parry L, Stepien G, Fafournoux P, Bruhat A. The eIF2alpha/ATF4 pathway is essential for stress-induced autophagy gene expression. Nucleic Acids Res. 2013;41(16):7683–99.PubMedPubMedCentralCrossRef
87.
Zurück zum Zitat Kouroku Y, Fujita E, Tanida I, Ueno T, Isoai A, Kumagai H, Ogawa S, Kaufman RJ, Kominami E, Momoi T. ER stress (PERK/eIF2alpha phosphorylation) mediates the polyglutamine-induced LC3 conversion, an essential step for autophagy formation. Cell Death Differ. 2007;14(2):230–9.PubMedCrossRef Kouroku Y, Fujita E, Tanida I, Ueno T, Isoai A, Kumagai H, Ogawa S, Kaufman RJ, Kominami E, Momoi T. ER stress (PERK/eIF2alpha phosphorylation) mediates the polyglutamine-induced LC3 conversion, an essential step for autophagy formation. Cell Death Differ. 2007;14(2):230–9.PubMedCrossRef
88.
Zurück zum Zitat Margariti A, Li H, Chen T, Martin D, Vizcay-Barrena G, Alam S, Karamariti E, Xiao Q, Zampetaki A, Zhang Z, et al. XBP1 mRNA splicing triggers an autophagic response in endothelial cells through BECLIN-1 transcriptional activation. J Biol Chem. 2013;288(2):859–72.PubMedCrossRef Margariti A, Li H, Chen T, Martin D, Vizcay-Barrena G, Alam S, Karamariti E, Xiao Q, Zampetaki A, Zhang Z, et al. XBP1 mRNA splicing triggers an autophagic response in endothelial cells through BECLIN-1 transcriptional activation. J Biol Chem. 2013;288(2):859–72.PubMedCrossRef
89.
Zurück zum Zitat Pattingre S, Tassa A, Qu X, Garuti R, Liang XH, Mizushima N, Packer M, Schneider MD, Levine B. Bcl-2 antiapoptotic proteins inhibit Beclin 1-dependent autophagy. Cell. 2005;122(6):927–39.PubMedCrossRef Pattingre S, Tassa A, Qu X, Garuti R, Liang XH, Mizushima N, Packer M, Schneider MD, Levine B. Bcl-2 antiapoptotic proteins inhibit Beclin 1-dependent autophagy. Cell. 2005;122(6):927–39.PubMedCrossRef
90.
Zurück zum Zitat Yamamoto K, Ichijo H, Korsmeyer SJ. BCL-2 is phosphorylated and inactivated by an ASK1/Jun N-terminal protein kinase pathway normally activated at G(2)/M. Mol Cell Biol. 1999;19(12):8469–78.PubMedPubMedCentralCrossRef Yamamoto K, Ichijo H, Korsmeyer SJ. BCL-2 is phosphorylated and inactivated by an ASK1/Jun N-terminal protein kinase pathway normally activated at G(2)/M. Mol Cell Biol. 1999;19(12):8469–78.PubMedPubMedCentralCrossRef
91.
Zurück zum Zitat Hoyer-Hansen M, Bastholm L, Szyniarowski P, Campanella M, Szabadkai G, Farkas T, Bianchi K, Fehrenbacher N, Elling F, Rizzuto R, et al. Control of macroautophagy by calcium, calmodulin-dependent kinase kinase-beta, and Bcl-2. Mol Cell. 2007;25(2):193–205.PubMedCrossRef Hoyer-Hansen M, Bastholm L, Szyniarowski P, Campanella M, Szabadkai G, Farkas T, Bianchi K, Fehrenbacher N, Elling F, Rizzuto R, et al. Control of macroautophagy by calcium, calmodulin-dependent kinase kinase-beta, and Bcl-2. Mol Cell. 2007;25(2):193–205.PubMedCrossRef
92.
Zurück zum Zitat Zalckvar E, Berissi H, Eisenstein M, Kimchi A. Phosphorylation of Beclin 1 by DAP-kinase promotes autophagy by weakening its interactions with Bcl-2 and Bcl-XL. Autophagy. 2009;5(5):720–2.PubMedCrossRef Zalckvar E, Berissi H, Eisenstein M, Kimchi A. Phosphorylation of Beclin 1 by DAP-kinase promotes autophagy by weakening its interactions with Bcl-2 and Bcl-XL. Autophagy. 2009;5(5):720–2.PubMedCrossRef
93.
Zurück zum Zitat Fernandez-Gil BI, Guerra-Librero A, Shen YQ, Florido J, Martínez-Ruiz L, García-López S, Adan C, Rodríguez-Santana C, Acuña-Castroviejo D, Quiñones-Hinojosa A, et al. Melatonin enhances cisplatin and radiation cytotoxicity in head and neck squamous cell carcinoma by stimulating mitochondrial ROS generation, apoptosis, and autophagy. Oxid Med Cell Longev. 2019;2019:7187128.PubMedPubMedCentralCrossRef Fernandez-Gil BI, Guerra-Librero A, Shen YQ, Florido J, Martínez-Ruiz L, García-López S, Adan C, Rodríguez-Santana C, Acuña-Castroviejo D, Quiñones-Hinojosa A, et al. Melatonin enhances cisplatin and radiation cytotoxicity in head and neck squamous cell carcinoma by stimulating mitochondrial ROS generation, apoptosis, and autophagy. Oxid Med Cell Longev. 2019;2019:7187128.PubMedPubMedCentralCrossRef
94.
Zurück zum Zitat Shen YQ, Guerra-Librero A, Fernandez-Gil BI, Florido J, García-López S, Martinez-Ruiz L, Mendivil-Perez M, Soto-Mercado V, Acuña-Castroviejo D, Ortega-Arellano H, et al. Combination of melatonin and rapamycin for head and neck cancer therapy: Suppression of AKT/mTOR pathway activation, and activation of mitophagy and apoptosis via mitochondrial function regulation. J Pineal Res. 2018;64(3):e12461.CrossRef Shen YQ, Guerra-Librero A, Fernandez-Gil BI, Florido J, García-López S, Martinez-Ruiz L, Mendivil-Perez M, Soto-Mercado V, Acuña-Castroviejo D, Ortega-Arellano H, et al. Combination of melatonin and rapamycin for head and neck cancer therapy: Suppression of AKT/mTOR pathway activation, and activation of mitophagy and apoptosis via mitochondrial function regulation. J Pineal Res. 2018;64(3):e12461.CrossRef
95.
Zurück zum Zitat Xiao W, Xiong Z, Xiong W, Yuan C, Xiao H, Ruan H, Song Z, Wang C, Bao L, Cao Q, et al. Melatonin/PGC1A/UCP1 promotes tumor slimming and represses tumor progression by initiating autophagy and lipid browning. J Pineal Res. 2019;67(4):e12607.PubMedCrossRef Xiao W, Xiong Z, Xiong W, Yuan C, Xiao H, Ruan H, Song Z, Wang C, Bao L, Cao Q, et al. Melatonin/PGC1A/UCP1 promotes tumor slimming and represses tumor progression by initiating autophagy and lipid browning. J Pineal Res. 2019;67(4):e12607.PubMedCrossRef
96.
Zurück zum Zitat Ordonez R, Fernandez A, Prieto-Dominguez N, Martinez L, Garcia-Ruiz C, Fernandez-Checa JC, Mauriz JL, Gonzalez-Gallego J. Ceramide metabolism regulates autophagy and apoptotic cell death induced by melatonin in liver cancer cells. J Pineal Res. 2015;59(2):178–89.PubMedPubMedCentralCrossRef Ordonez R, Fernandez A, Prieto-Dominguez N, Martinez L, Garcia-Ruiz C, Fernandez-Checa JC, Mauriz JL, Gonzalez-Gallego J. Ceramide metabolism regulates autophagy and apoptotic cell death induced by melatonin in liver cancer cells. J Pineal Res. 2015;59(2):178–89.PubMedPubMedCentralCrossRef
97.
98.
Zurück zum Zitat Fan T, Pi H, Li M, Ren Z, He Z, Zhu F, Tian L, Tu M, Xie J, Liu M, et al. Inhibiting MT2-TFE3-dependent autophagy enhances melatonin-induced apoptosis in tongue squamous cell carcinoma. J Pineal Res. 2018;64(2):e12457.CrossRef Fan T, Pi H, Li M, Ren Z, He Z, Zhu F, Tian L, Tu M, Xie J, Liu M, et al. Inhibiting MT2-TFE3-dependent autophagy enhances melatonin-induced apoptosis in tongue squamous cell carcinoma. J Pineal Res. 2018;64(2):e12457.CrossRef
99.
Zurück zum Zitat Trivedi PP, Jena GB, Tikoo KB, Kumar V. Melatonin modulated autophagy and Nrf2 signaling pathways in mice with colitis-associated colon carcinogenesis. Mol Carcinog. 2016;55(3):255–67.PubMedCrossRef Trivedi PP, Jena GB, Tikoo KB, Kumar V. Melatonin modulated autophagy and Nrf2 signaling pathways in mice with colitis-associated colon carcinogenesis. Mol Carcinog. 2016;55(3):255–67.PubMedCrossRef
100.
Zurück zum Zitat Chen L, Liu L, Li Y, Gao J. Melatonin increases human cervical cancer HeLa cells apoptosis induced by cisplatin via inhibition of JNK/Parkin/mitophagy axis. In Vitro Cell Dev Biol Anim. 2018;54(1):1–10.PubMedCrossRef Chen L, Liu L, Li Y, Gao J. Melatonin increases human cervical cancer HeLa cells apoptosis induced by cisplatin via inhibition of JNK/Parkin/mitophagy axis. In Vitro Cell Dev Biol Anim. 2018;54(1):1–10.PubMedCrossRef
101.
Zurück zum Zitat Bennukul K, Numkliang S, Leardkamolkarn V. Melatonin attenuates cisplatin-induced HepG2 cell death via the regulation of mTOR and ERCC1 expressions. World J Hepatol. 2014;6(4):230–42.PubMedPubMedCentralCrossRef Bennukul K, Numkliang S, Leardkamolkarn V. Melatonin attenuates cisplatin-induced HepG2 cell death via the regulation of mTOR and ERCC1 expressions. World J Hepatol. 2014;6(4):230–42.PubMedPubMedCentralCrossRef
102.
Zurück zum Zitat Fattman CL. Apoptosis in pulmonary fibrosis: too much or not enough? Antioxid Redox Signal. 2008;10(2):379–86.PubMedCrossRef Fattman CL. Apoptosis in pulmonary fibrosis: too much or not enough? Antioxid Redox Signal. 2008;10(2):379–86.PubMedCrossRef
103.
Zurück zum Zitat Huang K, Zhang J, O’Neill KL, Gurumurthy CB, Quadros RM, Tu Y, Luo X. Cleavage by Caspase 8 and mitochondrial membrane association activate the BH3-only protein bid during TRAIL-induced apoptosis. J Biol Chem. 2016;291(22):11843–51.PubMedCrossRef Huang K, Zhang J, O’Neill KL, Gurumurthy CB, Quadros RM, Tu Y, Luo X. Cleavage by Caspase 8 and mitochondrial membrane association activate the BH3-only protein bid during TRAIL-induced apoptosis. J Biol Chem. 2016;291(22):11843–51.PubMedCrossRef
105.
Zurück zum Zitat Hattori T, Ookawa N, Fujita R, Fukuchi K. Heterodimerization of Bcl-2 and Bcl-X(L) with Bax and Bad in colorectal cancer. Acta oncologica (Stockholm, Sweden). 2000;39(4):495–500.CrossRef Hattori T, Ookawa N, Fujita R, Fukuchi K. Heterodimerization of Bcl-2 and Bcl-X(L) with Bax and Bad in colorectal cancer. Acta oncologica (Stockholm, Sweden). 2000;39(4):495–500.CrossRef
106.
Zurück zum Zitat Thomas S, Quinn BA, Das SK, Dash R, Emdad L, Dasgupta S, Wang X-Y, Dent P, Reed JC, Pellecchia M. Targeting the Bcl-2 family for cancer therapy. Expert Opin Ther Targets. 2013;17(1):61–75.PubMedCrossRef Thomas S, Quinn BA, Das SK, Dash R, Emdad L, Dasgupta S, Wang X-Y, Dent P, Reed JC, Pellecchia M. Targeting the Bcl-2 family for cancer therapy. Expert Opin Ther Targets. 2013;17(1):61–75.PubMedCrossRef
107.
108.
Zurück zum Zitat Suzuki Y, Takahashi-Niki K, Akagi T, Hashikawa T, Takahashi R. Mitochondrial protease Omi/HtrA2 enhances caspase activation through multiple pathways. Cell Death Differ. 2004;11(2):208–16.PubMedCrossRef Suzuki Y, Takahashi-Niki K, Akagi T, Hashikawa T, Takahashi R. Mitochondrial protease Omi/HtrA2 enhances caspase activation through multiple pathways. Cell Death Differ. 2004;11(2):208–16.PubMedCrossRef
109.
110.
Zurück zum Zitat Kim C, Kim B. Anti-cancer natural products and their bioactive compounds inducing ER stress-mediated apoptosis: a review. Nutrients. 2018;10(8):1021.PubMedCentralCrossRef Kim C, Kim B. Anti-cancer natural products and their bioactive compounds inducing ER stress-mediated apoptosis: a review. Nutrients. 2018;10(8):1021.PubMedCentralCrossRef
111.
Zurück zum Zitat Liao Y, Fung TS, Huang M, Fang SG, Zhong Y, Liu DX. Upregulation of CHOP/GADD153 during coronavirus infectious bronchitis virus infection modulates apoptosis by restricting activation of the extracellular signal-regulated kinase pathway. J Virol. 2013;87(14):8124–34.PubMedPubMedCentralCrossRef Liao Y, Fung TS, Huang M, Fang SG, Zhong Y, Liu DX. Upregulation of CHOP/GADD153 during coronavirus infectious bronchitis virus infection modulates apoptosis by restricting activation of the extracellular signal-regulated kinase pathway. J Virol. 2013;87(14):8124–34.PubMedPubMedCentralCrossRef
112.
Zurück zum Zitat Song J, Ma SJ, Luo JH, Liu H, Li L, Zhang ZG, Chen LS, Zhou RX. Downregulation of AKT and MDM2, melatonin induces apoptosis in AGS and MGC803 Cells. Anatomical Rec. 2019;302(9):1544–51.CrossRef Song J, Ma SJ, Luo JH, Liu H, Li L, Zhang ZG, Chen LS, Zhou RX. Downregulation of AKT and MDM2, melatonin induces apoptosis in AGS and MGC803 Cells. Anatomical Rec. 2019;302(9):1544–51.CrossRef
113.
Zurück zum Zitat Proietti S, Cucina A, Dobrowolny G, D’Anselmi F, Dinicola S, Masiello MG, Pasqualato A, Palombo A, Morini V, Reiter RJ, et al. Melatonin down-regulates MDM2 gene expression and enhances p53 acetylation in MCF-7 cells. J Pineal Res. 2014;57(1):120–9.PubMedCrossRef Proietti S, Cucina A, Dobrowolny G, D’Anselmi F, Dinicola S, Masiello MG, Pasqualato A, Palombo A, Morini V, Reiter RJ, et al. Melatonin down-regulates MDM2 gene expression and enhances p53 acetylation in MCF-7 cells. J Pineal Res. 2014;57(1):120–9.PubMedCrossRef
114.
Zurück zum Zitat Lee YJ, Lee JH, Moon JH, Park SY. Overcoming hypoxic-resistance of tumor cells to TRAIL-induced apoptosis through melatonin. Int J Mol Sci. 2014;15(7):11941–56.PubMedPubMedCentralCrossRef Lee YJ, Lee JH, Moon JH, Park SY. Overcoming hypoxic-resistance of tumor cells to TRAIL-induced apoptosis through melatonin. Int J Mol Sci. 2014;15(7):11941–56.PubMedPubMedCentralCrossRef
115.
Zurück zum Zitat Martín-Renedo J, Mauriz JL, Jorquera F, Ruiz-Andrés O, González P, González-Gallego J. Melatonin induces cell cycle arrest and apoptosis in hepatocarcinoma HepG2 cell line. J Pineal Res. 2008;45(4):532–40.PubMedCrossRef Martín-Renedo J, Mauriz JL, Jorquera F, Ruiz-Andrés O, González P, González-Gallego J. Melatonin induces cell cycle arrest and apoptosis in hepatocarcinoma HepG2 cell line. J Pineal Res. 2008;45(4):532–40.PubMedCrossRef
116.
Zurück zum Zitat Cucina A, Proietti S, D’Anselmi F, Coluccia P, Dinicola S, Frati L, Bizzarri M. Evidence for a biphasic apoptotic pathway induced by melatonin in MCF-7 breast cancer cells. J Pineal Res. 2009;46(2):172–80.PubMedCrossRef Cucina A, Proietti S, D’Anselmi F, Coluccia P, Dinicola S, Frati L, Bizzarri M. Evidence for a biphasic apoptotic pathway induced by melatonin in MCF-7 breast cancer cells. J Pineal Res. 2009;46(2):172–80.PubMedCrossRef
117.
Zurück zum Zitat Wei JY, Li WM, Zhou LL, Lu QN, He W. Melatonin induces apoptosis of colorectal cancer cells through HDAC4 nuclear import mediated by CaMKII inactivation. J Pineal Res. 2015;58(4):429–38.PubMedCrossRef Wei JY, Li WM, Zhou LL, Lu QN, He W. Melatonin induces apoptosis of colorectal cancer cells through HDAC4 nuclear import mediated by CaMKII inactivation. J Pineal Res. 2015;58(4):429–38.PubMedCrossRef
118.
Zurück zum Zitat Ma Z, Liu D, Di S, Zhang Z, Li W, Zhang J, Xu L, Guo K, Zhu Y, Li X, et al. Histone deacetylase 9 downregulation decreases tumor growth and promotes apoptosis in non-small cell lung cancer after melatonin treatment. J Pineal Res. 2019;67(2):e12587.PubMedCrossRef Ma Z, Liu D, Di S, Zhang Z, Li W, Zhang J, Xu L, Guo K, Zhu Y, Li X, et al. Histone deacetylase 9 downregulation decreases tumor growth and promotes apoptosis in non-small cell lung cancer after melatonin treatment. J Pineal Res. 2019;67(2):e12587.PubMedCrossRef
119.
Zurück zum Zitat Zhao Q, Wang W, Cui J. Melatonin enhances TNF-alpha-mediated cervical cancer HeLa cells death via suppressing CaMKII/Parkin/mitophagy axis. Cancer Cell Int. 2019;19:58.PubMedPubMedCentralCrossRef Zhao Q, Wang W, Cui J. Melatonin enhances TNF-alpha-mediated cervical cancer HeLa cells death via suppressing CaMKII/Parkin/mitophagy axis. Cancer Cell Int. 2019;19:58.PubMedPubMedCentralCrossRef
120.
Zurück zum Zitat El-Magd MA, Mohamed Y, El-Shetry ES, Elsayed SA, Abo Gazia M, Abdel-Aleem GA, Shafik NM, Abdo WS, El-Desouki NI, Basyony MA. Melatonin maximizes the therapeutic potential of non-preconditioned MSCs in a DEN-induced rat model of HCC. Biomed Pharmacother. 2019;114:108732.PubMedCrossRef El-Magd MA, Mohamed Y, El-Shetry ES, Elsayed SA, Abo Gazia M, Abdel-Aleem GA, Shafik NM, Abdo WS, El-Desouki NI, Basyony MA. Melatonin maximizes the therapeutic potential of non-preconditioned MSCs in a DEN-induced rat model of HCC. Biomed Pharmacother. 2019;114:108732.PubMedCrossRef
121.
Zurück zum Zitat Jung JH, Shin EA, Kim JH, Sim DY, Lee H, Park JE, Lee HJ, Kim SH. NEDD9 inhibition by miR-25–5p activation is critically involved in co-treatment of melatonin- and pterostilbene-induced apoptosis in colorectal cancer cells. Cancers. 2019;11(11):1684.PubMedCentralCrossRef Jung JH, Shin EA, Kim JH, Sim DY, Lee H, Park JE, Lee HJ, Kim SH. NEDD9 inhibition by miR-25–5p activation is critically involved in co-treatment of melatonin- and pterostilbene-induced apoptosis in colorectal cancer cells. Cancers. 2019;11(11):1684.PubMedCentralCrossRef
122.
Zurück zum Zitat Kim JH, Jeong SJ, Kim B, Yun SM, Choi DY, Kim SH. Melatonin synergistically enhances cisplatin-induced apoptosis via the dephosphorylation of ERK/p90 ribosomal S6 kinase/heat shock protein 27 in SK-OV-3 cells. J Pineal Res. 2012;52(2):244–52.PubMedCrossRef Kim JH, Jeong SJ, Kim B, Yun SM, Choi DY, Kim SH. Melatonin synergistically enhances cisplatin-induced apoptosis via the dephosphorylation of ERK/p90 ribosomal S6 kinase/heat shock protein 27 in SK-OV-3 cells. J Pineal Res. 2012;52(2):244–52.PubMedCrossRef
123.
Zurück zum Zitat Woo SM, Min KJ, Kwon TK. Melatonin-mediated Bim up-regulation and cyclooxygenase-2 (COX-2) down-regulation enhances tunicamycin-induced apoptosis in MDA-MB-231 cells. J Pineal Res. 2015;58(3):310–20.PubMedCrossRef Woo SM, Min KJ, Kwon TK. Melatonin-mediated Bim up-regulation and cyclooxygenase-2 (COX-2) down-regulation enhances tunicamycin-induced apoptosis in MDA-MB-231 cells. J Pineal Res. 2015;58(3):310–20.PubMedCrossRef
124.
Zurück zum Zitat Zakki SA, Muhammad JS, Li JL, Sun L, Li ML, Feng QW, Li YL, Cui ZG, Inadera H. Melatonin triggers the anticancer potential of phenylarsine oxide via induction of apoptosis through ROS generation and JNK activation. Metallomics Integr Biometal Sci. 2020;12:396–407.CrossRef Zakki SA, Muhammad JS, Li JL, Sun L, Li ML, Feng QW, Li YL, Cui ZG, Inadera H. Melatonin triggers the anticancer potential of phenylarsine oxide via induction of apoptosis through ROS generation and JNK activation. Metallomics Integr Biometal Sci. 2020;12:396–407.CrossRef
125.
Zurück zum Zitat Gonzalez-Gonzalez A, Gonzalez A, Alonso-Gonzalez C, Menendez-Menendez J, Martinez-Campa C, Cos S. Complementary actions of melatonin on angiogenic factors, the angiopoietin/Tie2 axis and VEGF, in cocultures of human endothelial and breast cancer cells. Oncol Rep. 2018;39(1):433–41.PubMed Gonzalez-Gonzalez A, Gonzalez A, Alonso-Gonzalez C, Menendez-Menendez J, Martinez-Campa C, Cos S. Complementary actions of melatonin on angiogenic factors, the angiopoietin/Tie2 axis and VEGF, in cocultures of human endothelial and breast cancer cells. Oncol Rep. 2018;39(1):433–41.PubMed
126.
Zurück zum Zitat Kumari R, Rawat K, Kumari A, Shrivastava A. Amelioration of Dalton’s lymphoma-induced angiogenesis by melatonin. Tumour Biol. 2017;39(6):1010428317705758.PubMedCrossRef Kumari R, Rawat K, Kumari A, Shrivastava A. Amelioration of Dalton’s lymphoma-induced angiogenesis by melatonin. Tumour Biol. 2017;39(6):1010428317705758.PubMedCrossRef
127.
Zurück zum Zitat Zonta YR, Martinez M, Camargo IC, Domeniconi RF, Lupi Junior LA, Pinheiro PF, Reiter RJ, Martinez FE, Chuffa LG. Melatonin reduces angiogenesis in serous papillary ovarian carcinoma of ethanol-preferring rats. Int J Mol Sci. 2017;18(4):763.PubMedCentralCrossRef Zonta YR, Martinez M, Camargo IC, Domeniconi RF, Lupi Junior LA, Pinheiro PF, Reiter RJ, Martinez FE, Chuffa LG. Melatonin reduces angiogenesis in serous papillary ovarian carcinoma of ethanol-preferring rats. Int J Mol Sci. 2017;18(4):763.PubMedCentralCrossRef
128.
Zurück zum Zitat Gelaleti GB, Borin TF, Maschio-Signorini LB, Moschetta MG, Hellmen E, Viloria-Petit AM, Zuccari D. Melatonin and IL-25 modulate apoptosis and angiogenesis mediators in metastatic (CF-41) and non-metastatic (CMT-U229) canine mammary tumour cells. Vet Comp Oncol. 2017;15(4):1572–84.PubMedCrossRef Gelaleti GB, Borin TF, Maschio-Signorini LB, Moschetta MG, Hellmen E, Viloria-Petit AM, Zuccari D. Melatonin and IL-25 modulate apoptosis and angiogenesis mediators in metastatic (CF-41) and non-metastatic (CMT-U229) canine mammary tumour cells. Vet Comp Oncol. 2017;15(4):1572–84.PubMedCrossRef
129.
Zurück zum Zitat González A, González-González A, Alonso-González C, Menéndez-Menéndez J, Martínez-Campa C, Cos S. Melatonin inhibits angiogenesis in SH-SY5Y human neuroblastoma cells by downregulation of VEGF. Oncol Rep. 2017;37(4):2433–40.PubMedCrossRef González A, González-González A, Alonso-González C, Menéndez-Menéndez J, Martínez-Campa C, Cos S. Melatonin inhibits angiogenesis in SH-SY5Y human neuroblastoma cells by downregulation of VEGF. Oncol Rep. 2017;37(4):2433–40.PubMedCrossRef
130.
Zurück zum Zitat Wang RX, Liu H, Xu L, Zhang H, Zhou RX. Melatonin downregulates nuclear receptor RZR/RORgamma expression causing growth-inhibitory and anti-angiogenesis activity in human gastric cancer cells in vitro and in vivo. Oncol Lett. 2016;12(2):897–903.PubMedPubMedCentralCrossRef Wang RX, Liu H, Xu L, Zhang H, Zhou RX. Melatonin downregulates nuclear receptor RZR/RORgamma expression causing growth-inhibitory and anti-angiogenesis activity in human gastric cancer cells in vitro and in vivo. Oncol Lett. 2016;12(2):897–903.PubMedPubMedCentralCrossRef
131.
Zurück zum Zitat Jardim-Perassi BV, Lourenco MR, Doho GM, Grigolo IH, Gelaleti GB, Ferreira LC, Borin TF, Moschetta MG, Pires de Campos Zuccari DA. Melatonin regulates angiogenic factors under hypoxia in breast cancer cell lines. Anticancer Agents Med Chem. 2016;16(3):347–58.PubMedCrossRef Jardim-Perassi BV, Lourenco MR, Doho GM, Grigolo IH, Gelaleti GB, Ferreira LC, Borin TF, Moschetta MG, Pires de Campos Zuccari DA. Melatonin regulates angiogenic factors under hypoxia in breast cancer cell lines. Anticancer Agents Med Chem. 2016;16(3):347–58.PubMedCrossRef
132.
Zurück zum Zitat Talib WH, Saleh S. Propionibacterium acnes augments antitumor, anti-angiogenesis and immunomodulatory effects of melatonin on breast cancer implanted in mice. PLoS ONE. 2015;10(4):e0124384.PubMedPubMedCentralCrossRef Talib WH, Saleh S. Propionibacterium acnes augments antitumor, anti-angiogenesis and immunomodulatory effects of melatonin on breast cancer implanted in mice. PLoS ONE. 2015;10(4):e0124384.PubMedPubMedCentralCrossRef
133.
Zurück zum Zitat Sohn EJ, Won G, Lee J, Lee S, Kim SH. Upregulation of miRNA3195 and miRNA374b mediates the anti-angiogenic properties of melatonin in hypoxic PC-3 prostate cancer cells. J Cancer. 2015;6(1):19–28.PubMedPubMedCentralCrossRef Sohn EJ, Won G, Lee J, Lee S, Kim SH. Upregulation of miRNA3195 and miRNA374b mediates the anti-angiogenic properties of melatonin in hypoxic PC-3 prostate cancer cells. J Cancer. 2015;6(1):19–28.PubMedPubMedCentralCrossRef
134.
Zurück zum Zitat Carbajo-Pescador S, Ordonez R, Benet M, Jover R, Garcia-Palomo A, Mauriz JL, Gonzalez-Gallego J. Inhibition of VEGF expression through blockade of Hif1alpha and STAT3 signalling mediates the anti-angiogenic effect of melatonin in HepG2 liver cancer cells. Br J Cancer. 2013;109(1):83–91.PubMedPubMedCentralCrossRef Carbajo-Pescador S, Ordonez R, Benet M, Jover R, Garcia-Palomo A, Mauriz JL, Gonzalez-Gallego J. Inhibition of VEGF expression through blockade of Hif1alpha and STAT3 signalling mediates the anti-angiogenic effect of melatonin in HepG2 liver cancer cells. Br J Cancer. 2013;109(1):83–91.PubMedPubMedCentralCrossRef
135.
Zurück zum Zitat Kim KJ, Choi JS, Kang I, Kim KW, Jeong CH, Jeong JW. Melatonin suppresses tumor progression by reducing angiogenesis stimulated by HIF-1 in a mouse tumor model. J Pineal Res. 2013;54(3):264–70.PubMedCrossRef Kim KJ, Choi JS, Kang I, Kim KW, Jeong CH, Jeong JW. Melatonin suppresses tumor progression by reducing angiogenesis stimulated by HIF-1 in a mouse tumor model. J Pineal Res. 2013;54(3):264–70.PubMedCrossRef
136.
Zurück zum Zitat Jardim-Perassi BV, Arbab AS, Ferreira LC, Borin TF, Varma NR, Iskander AS, Shankar A, Ali MM, de Campos Zuccari DA. Effect of melatonin on tumor growth and angiogenesis in xenograft model of breast cancer. PLoS ONE. 2014;9(1):e85311.PubMedPubMedCentralCrossRef Jardim-Perassi BV, Arbab AS, Ferreira LC, Borin TF, Varma NR, Iskander AS, Shankar A, Ali MM, de Campos Zuccari DA. Effect of melatonin on tumor growth and angiogenesis in xenograft model of breast cancer. PLoS ONE. 2014;9(1):e85311.PubMedPubMedCentralCrossRef
137.
Zurück zum Zitat Marques JHM, Mota AL, Oliveira JG, Lacerda JZ, Stefani JP, Ferreira LC, Castro TB, Aristizabal-Pachon AF, Zuccari D. Melatonin restrains angiogenic factors in triple-negative breast cancer by targeting miR-152-3p: In vivo and in vitro studies. Life Sci. 2018;208:131–8.PubMedCrossRef Marques JHM, Mota AL, Oliveira JG, Lacerda JZ, Stefani JP, Ferreira LC, Castro TB, Aristizabal-Pachon AF, Zuccari D. Melatonin restrains angiogenic factors in triple-negative breast cancer by targeting miR-152-3p: In vivo and in vitro studies. Life Sci. 2018;208:131–8.PubMedCrossRef
138.
Zurück zum Zitat Lacerda JZ, Ferreira LC, Lopes BC, Aristizabal-Pachon AF, Bajgelman MC, Borin TF, Zuccari D. Therapeutic potential of melatonin in the regulation of MiR-148a-3p and angiogenic factors in breast cancer. MicroRNA (Shariqah, United Arab Emirates). 2019;8(3):237–47. Lacerda JZ, Ferreira LC, Lopes BC, Aristizabal-Pachon AF, Bajgelman MC, Borin TF, Zuccari D. Therapeutic potential of melatonin in the regulation of MiR-148a-3p and angiogenic factors in breast cancer. MicroRNA (Shariqah, United Arab Emirates). 2019;8(3):237–47.
139.
Zurück zum Zitat Hsieh MJ, Lin CW, Su SC, Reiter RJ, Chen AW, Chen MK, Yang SF. Effects of miR-34b/miR-892a upregulation and inhibition of ABCB1/ABCB4 on melatonin-induced apoptosis in VCR-resistant oral cancer cells. Mol Ther Nucleic Acids. 2020;19:877–89.PubMedPubMedCentralCrossRef Hsieh MJ, Lin CW, Su SC, Reiter RJ, Chen AW, Chen MK, Yang SF. Effects of miR-34b/miR-892a upregulation and inhibition of ABCB1/ABCB4 on melatonin-induced apoptosis in VCR-resistant oral cancer cells. Mol Ther Nucleic Acids. 2020;19:877–89.PubMedPubMedCentralCrossRef
140.
Zurück zum Zitat Talib WH. A ketogenic diet combined with melatonin overcomes cisplatin and vincristine drug resistance in breast carcinoma syngraft. Nutrition (Burbank, Los Angeles County, Calif). 2019;72:110659.CrossRef Talib WH. A ketogenic diet combined with melatonin overcomes cisplatin and vincristine drug resistance in breast carcinoma syngraft. Nutrition (Burbank, Los Angeles County, Calif). 2019;72:110659.CrossRef
141.
Zurück zum Zitat Amin AH, El-Missiry MA, Othman AI, Ali DA, Gouida MS, Ismail AH. Ameliorative effects of melatonin against solid Ehrlich carcinoma progression in female mice. J Pineal Res. 2019;67(2):e12585.PubMedCrossRef Amin AH, El-Missiry MA, Othman AI, Ali DA, Gouida MS, Ismail AH. Ameliorative effects of melatonin against solid Ehrlich carcinoma progression in female mice. J Pineal Res. 2019;67(2):e12585.PubMedCrossRef
142.
Zurück zum Zitat Hao J, Fan W, Li Y, Tang R, Tian C, Yang Q, Zhu T, Diao C, Hu S, Chen M, et al. Melatonin synergizes BRAF-targeting agent vemurafenib in melanoma treatment by inhibiting iNOS/hTERT signaling and cancer-stem cell traits. J Exp Clin Cancer Res. 2019;38(1):48.PubMedPubMedCentralCrossRef Hao J, Fan W, Li Y, Tang R, Tian C, Yang Q, Zhu T, Diao C, Hu S, Chen M, et al. Melatonin synergizes BRAF-targeting agent vemurafenib in melanoma treatment by inhibiting iNOS/hTERT signaling and cancer-stem cell traits. J Exp Clin Cancer Res. 2019;38(1):48.PubMedPubMedCentralCrossRef
143.
Zurück zum Zitat Sonehara NM, Lacerda JZ, Jardim-Perassi BV, de Paula R, Moschetta-Pinheiro MG, Souza YST, de Andrade JCJ, De Campos Zuccari DAP. Melatonin regulates tumor aggressiveness under acidosis condition in breast cancer cell lines. Oncol Lett. 2019;17(2):1635–45.PubMed Sonehara NM, Lacerda JZ, Jardim-Perassi BV, de Paula R, Moschetta-Pinheiro MG, Souza YST, de Andrade JCJ, De Campos Zuccari DAP. Melatonin regulates tumor aggressiveness under acidosis condition in breast cancer cell lines. Oncol Lett. 2019;17(2):1635–45.PubMed
144.
Zurück zum Zitat Leja-Szpak A, Nawrot-Porabka K, Goralska M, Jastrzebska M, Link-Lenczowski P, Bonior J, Pierzchalski P, Jaworek J. Melatonin and its metabolite N1-acetyl-N2-formyl-5-methoxykynuramine (afmk) enhance chemosensitivity to gemcitabine in pancreatic carcinoma cells (PANC-1). Pharmacol Rep. 2018;70(6):1079–88.PubMedCrossRef Leja-Szpak A, Nawrot-Porabka K, Goralska M, Jastrzebska M, Link-Lenczowski P, Bonior J, Pierzchalski P, Jaworek J. Melatonin and its metabolite N1-acetyl-N2-formyl-5-methoxykynuramine (afmk) enhance chemosensitivity to gemcitabine in pancreatic carcinoma cells (PANC-1). Pharmacol Rep. 2018;70(6):1079–88.PubMedCrossRef
145.
Zurück zum Zitat Kocak N, Donmez H, Yildirim IH. Effects of melatonin on apoptosis and cell differentiation in MCF-7 derived cancer stem cells. Cell Mol Biol. 2018;64(12):56–61.PubMedCrossRef Kocak N, Donmez H, Yildirim IH. Effects of melatonin on apoptosis and cell differentiation in MCF-7 derived cancer stem cells. Cell Mol Biol. 2018;64(12):56–61.PubMedCrossRef
146.
Zurück zum Zitat Krestinina O, Fadeev R, Lomovsky A, Baburina Y, Kobyakova M, Akatov V. Melatonin can strengthen the effect of retinoic acid in HL-60 cells. Int J Mol Sci. 2018;19(10):2873.PubMedCentralCrossRef Krestinina O, Fadeev R, Lomovsky A, Baburina Y, Kobyakova M, Akatov V. Melatonin can strengthen the effect of retinoic acid in HL-60 cells. Int J Mol Sci. 2018;19(10):2873.PubMedCentralCrossRef
147.
Zurück zum Zitat Odeh LH, Talib WH, Basheti IA. Synergistic effect of thymoquinone and melatonin against breast cancer implanted in mice. J Cancer Res Ther. 2018;14(Supplement):S324-s330.PubMed Odeh LH, Talib WH, Basheti IA. Synergistic effect of thymoquinone and melatonin against breast cancer implanted in mice. J Cancer Res Ther. 2018;14(Supplement):S324-s330.PubMed
148.
Zurück zum Zitat Safaroghli-Azar A, Pourbagheri-Sigaroodi A, Bashash D, Nooshinfar E, Anjam-Najmedini A, Sadeghi S, Rezaie-Tavirani M, Akbari ME. Stimulatory effect of indolic hormone on As2O3 cytotoxicity in breast cancer cells: NF-kappaB-dependent mechanism of action of melatonin. Int J Mol Cell Med. 2018;7(3):158–68.PubMedPubMedCentral Safaroghli-Azar A, Pourbagheri-Sigaroodi A, Bashash D, Nooshinfar E, Anjam-Najmedini A, Sadeghi S, Rezaie-Tavirani M, Akbari ME. Stimulatory effect of indolic hormone on As2O3 cytotoxicity in breast cancer cells: NF-kappaB-dependent mechanism of action of melatonin. Int J Mol Cell Med. 2018;7(3):158–68.PubMedPubMedCentral
149.
Zurück zum Zitat Fang Z, Jung KH, Yan HH, Kim SJ, Rumman M, Park JH, Han B, Lee JE, Kang YW, Lim JH, et al. Melatonin synergizes with sorafenib to suppress pancreatic cancer via melatonin receptor and PDGFR-beta/STAT3 Pathway. Cell Physiol Biochem. 2018;47(5):1751–68.PubMedCrossRef Fang Z, Jung KH, Yan HH, Kim SJ, Rumman M, Park JH, Han B, Lee JE, Kang YW, Lim JH, et al. Melatonin synergizes with sorafenib to suppress pancreatic cancer via melatonin receptor and PDGFR-beta/STAT3 Pathway. Cell Physiol Biochem. 2018;47(5):1751–68.PubMedCrossRef
150.
Zurück zum Zitat Franco DG, Moretti IF, Marie SKN. Mitochondria transcription factor A: a putative target for the effect of melatonin on u87mg malignant glioma cell line. Molecules. 2018;23(5):1129.PubMedCentralCrossRef Franco DG, Moretti IF, Marie SKN. Mitochondria transcription factor A: a putative target for the effect of melatonin on u87mg malignant glioma cell line. Molecules. 2018;23(5):1129.PubMedCentralCrossRef
151.
Zurück zum Zitat Wei X, Qi Y, Jia N, Zhou Q, Zhang S, Wang Y. Hyperbaric oxygen treatment sensitizes gastric cancer cells to melatonin-induced apoptosis through multiple pathways. J Cell Biochem. 2018;119(8):6723–31.PubMedCrossRef Wei X, Qi Y, Jia N, Zhou Q, Zhang S, Wang Y. Hyperbaric oxygen treatment sensitizes gastric cancer cells to melatonin-induced apoptosis through multiple pathways. J Cell Biochem. 2018;119(8):6723–31.PubMedCrossRef
152.
Zurück zum Zitat Zou ZW, Liu T, Li Y, Chen P, Peng X, Ma C, Zhang WJ, Li PD. Melatonin suppresses thyroid cancer growth and overcomes radioresistance via inhibition of p65 phosphorylation and induction of ROS. Redox Biol. 2018;16:226–36.PubMedPubMedCentralCrossRef Zou ZW, Liu T, Li Y, Chen P, Peng X, Ma C, Zhang WJ, Li PD. Melatonin suppresses thyroid cancer growth and overcomes radioresistance via inhibition of p65 phosphorylation and induction of ROS. Redox Biol. 2018;16:226–36.PubMedPubMedCentralCrossRef
153.
Zurück zum Zitat Song J, Ma SJ, Luo JH, Zhang H, Wang RX, Liu H, Li L, Zhang ZG, Zhou RX. Melatonin induces the apoptosis and inhibits the proliferation of human gastric cancer cells via blockade of the AKT/MDM2 pathway. Oncol Rep. 2018;39(4):1975–83.PubMed Song J, Ma SJ, Luo JH, Zhang H, Wang RX, Liu H, Li L, Zhang ZG, Zhou RX. Melatonin induces the apoptosis and inhibits the proliferation of human gastric cancer cells via blockade of the AKT/MDM2 pathway. Oncol Rep. 2018;39(4):1975–83.PubMed
154.
Zurück zum Zitat Yu Z, Tian X, Peng Y, Sun Z, Wang C, Tang N, Li B, Jian Y, Wang W, Huo X, et al. Mitochondrial cytochrome P450 (CYP) 1B1 is responsible for melatonin-induced apoptosis in neural cancer cells. J Pineal Res. 2018;65(1):e12478.PubMedCrossRef Yu Z, Tian X, Peng Y, Sun Z, Wang C, Tang N, Li B, Jian Y, Wang W, Huo X, et al. Mitochondrial cytochrome P450 (CYP) 1B1 is responsible for melatonin-induced apoptosis in neural cancer cells. J Pineal Res. 2018;65(1):e12478.PubMedCrossRef
155.
Zurück zum Zitat Zhu C, Huang Q, Zhu H. Melatonin inhibits the proliferation of gastric cancer cells through regulating the miR-16-5p-Smad3 pathway. DNA Cell Biol. 2018;37(3):244–52.PubMedCrossRef Zhu C, Huang Q, Zhu H. Melatonin inhibits the proliferation of gastric cancer cells through regulating the miR-16-5p-Smad3 pathway. DNA Cell Biol. 2018;37(3):244–52.PubMedCrossRef
156.
Zurück zum Zitat Chovancova B, Hudecova S, Lencesova L, Babula P, Rezuchova I, Penesova A, Grman M, Moravcik R, Zeman M, Krizanova O. Melatonin-induced changes in cytosolic calcium might be responsible for apoptosis induction in tumour cells. Cell Physiol Biochem. 2017;44(2):763–77.PubMedCrossRef Chovancova B, Hudecova S, Lencesova L, Babula P, Rezuchova I, Penesova A, Grman M, Moravcik R, Zeman M, Krizanova O. Melatonin-induced changes in cytosolic calcium might be responsible for apoptosis induction in tumour cells. Cell Physiol Biochem. 2017;44(2):763–77.PubMedCrossRef
157.
Zurück zum Zitat Gatti G, Lucini V, Dugnani S, Calastretti A, Spadoni G, Bedini A, Rivara S, Mor M, Canti G, Scaglione F, et al. Antiproliferative and pro-apoptotic activity of melatonin analogues on melanoma and breast cancer cells. Oncotarget. 2017;8(40):68338–53.PubMedPubMedCentralCrossRef Gatti G, Lucini V, Dugnani S, Calastretti A, Spadoni G, Bedini A, Rivara S, Mor M, Canti G, Scaglione F, et al. Antiproliferative and pro-apoptotic activity of melatonin analogues on melanoma and breast cancer cells. Oncotarget. 2017;8(40):68338–53.PubMedPubMedCentralCrossRef
158.
Zurück zum Zitat Shrestha S, Zhu J, Wang Q, Du X, Liu F, Jiang J, Song J, Xing J, Sun D, Hou Q, et al. Melatonin potentiates the antitumor effect of curcumin by inhibiting IKKbeta/NF-kappaB/COX-2 signaling pathway. Int J Oncol. 2017;51(4):1249–60.PubMedPubMedCentralCrossRef Shrestha S, Zhu J, Wang Q, Du X, Liu F, Jiang J, Song J, Xing J, Sun D, Hou Q, et al. Melatonin potentiates the antitumor effect of curcumin by inhibiting IKKbeta/NF-kappaB/COX-2 signaling pathway. Int J Oncol. 2017;51(4):1249–60.PubMedPubMedCentralCrossRef
159.
Zurück zum Zitat Pariente R, Bejarano I, Rodriguez AB, Pariente JA, Espino J. Melatonin increases the effect of 5-fluorouracil-based chemotherapy in human colorectal adenocarcinoma cells in vitro. Mol Cell Biochem. 2018;440(1–2):43–51.PubMedCrossRef Pariente R, Bejarano I, Rodriguez AB, Pariente JA, Espino J. Melatonin increases the effect of 5-fluorouracil-based chemotherapy in human colorectal adenocarcinoma cells in vitro. Mol Cell Biochem. 2018;440(1–2):43–51.PubMedCrossRef
160.
Zurück zum Zitat Lu JJ, Fu L, Tang Z, Zhang C, Qin L, Wang J, Yu Z, Shi D, Xiao X, Xie F, et al. Melatonin inhibits AP-2beta/hTERT, NF-kappaB/COX-2 and Akt/ERK and activates caspase/Cyto C signaling to enhance the antitumor activity of berberine in lung cancer cells. Oncotarget. 2016;7(3):2985–3001.PubMedCrossRef Lu JJ, Fu L, Tang Z, Zhang C, Qin L, Wang J, Yu Z, Shi D, Xiao X, Xie F, et al. Melatonin inhibits AP-2beta/hTERT, NF-kappaB/COX-2 and Akt/ERK and activates caspase/Cyto C signaling to enhance the antitumor activity of berberine in lung cancer cells. Oncotarget. 2016;7(3):2985–3001.PubMedCrossRef
161.
Zurück zum Zitat Li W, Fan M, Chen Y, Zhao Q, Song C, Yan Y, Jin Y, Huang Z, Lin C, Wu J. Melatonin induces cell apoptosis in AGS Cells through the activation of JNK and P38 MAPK and the suppression of nuclear Factor-Kappa B: a novel therapeutic implication for gastric cancer. Cell Physiol Biochem. 2015;37(6):2323–38.PubMedCrossRef Li W, Fan M, Chen Y, Zhao Q, Song C, Yan Y, Jin Y, Huang Z, Lin C, Wu J. Melatonin induces cell apoptosis in AGS Cells through the activation of JNK and P38 MAPK and the suppression of nuclear Factor-Kappa B: a novel therapeutic implication for gastric cancer. Cell Physiol Biochem. 2015;37(6):2323–38.PubMedCrossRef
162.
Zurück zum Zitat Chuffa LG, Alves MS, Martinez M, Camargo IC, Pinheiro PF, Domeniconi RF, Junior LA, Martinez FE. Apoptosis is triggered by melatonin in an in vivo model of ovarian carcinoma. Endocr Relat Cancer. 2016;23(2):65–76.PubMedCrossRef Chuffa LG, Alves MS, Martinez M, Camargo IC, Pinheiro PF, Domeniconi RF, Junior LA, Martinez FE. Apoptosis is triggered by melatonin in an in vivo model of ovarian carcinoma. Endocr Relat Cancer. 2016;23(2):65–76.PubMedCrossRef
163.
Zurück zum Zitat Pariente R, Pariente JA, Rodríguez AB, Espino J. Melatonin sensitizes human cervical cancer HeLa cells to cisplatin-induced cytotoxicity and apoptosis: effects on oxidative stress and DNA fragmentation. J Pineal Res. 2016;60(1):55–64.PubMedCrossRef Pariente R, Pariente JA, Rodríguez AB, Espino J. Melatonin sensitizes human cervical cancer HeLa cells to cisplatin-induced cytotoxicity and apoptosis: effects on oxidative stress and DNA fragmentation. J Pineal Res. 2016;60(1):55–64.PubMedCrossRef
164.
Zurück zum Zitat Codenotti S, Battistelli M, Burattini S, Salucci S, Falcieri E, Rezzani R, Faggi F, Colombi M, Monti E, Fanzani A. Melatonin decreases cell proliferation, impairs myogenic differentiation and triggers apoptotic cell death in rhabdomyosarcoma cell lines. Oncol Rep. 2015;34(1):279–87.PubMedCrossRef Codenotti S, Battistelli M, Burattini S, Salucci S, Falcieri E, Rezzani R, Faggi F, Colombi M, Monti E, Fanzani A. Melatonin decreases cell proliferation, impairs myogenic differentiation and triggers apoptotic cell death in rhabdomyosarcoma cell lines. Oncol Rep. 2015;34(1):279–87.PubMedCrossRef
165.
Zurück zum Zitat Tosun M, Soysal Y, Mas NG, Karabekir HS. Comparison of the effects of 13-cis retinoic acid and melatonin on the viabilities of SH-SY5Y neuroblastoma cell line. J Korean Neurosurg Soc. 2015;57(3):147–51.PubMedPubMedCentralCrossRef Tosun M, Soysal Y, Mas NG, Karabekir HS. Comparison of the effects of 13-cis retinoic acid and melatonin on the viabilities of SH-SY5Y neuroblastoma cell line. J Korean Neurosurg Soc. 2015;57(3):147–51.PubMedPubMedCentralCrossRef
166.
Zurück zum Zitat 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):1443–9.PubMedCrossRef 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):1443–9.PubMedCrossRef
167.
Zurück zum Zitat Plaimee P, Weerapreeyakul N, Barusrux S, Johns NP. Melatonin potentiates cisplatin-induced apoptosis and cell cycle arrest in human lung adenocarcinoma cells. Cell Prolif. 2015;48(1):67–77.PubMedPubMedCentralCrossRef Plaimee P, Weerapreeyakul N, Barusrux S, Johns NP. Melatonin potentiates cisplatin-induced apoptosis and cell cycle arrest in human lung adenocarcinoma cells. Cell Prolif. 2015;48(1):67–77.PubMedPubMedCentralCrossRef
168.
Zurück zum Zitat Xu L, Jin QD, Gong X, Liu H, Zhou RX. Anti-gastric cancer effect of melatonin and Bcl-2, Bax, p21 and p53 expression changes. Sheng li xue bao : [Acta physiologica Sinica]. 2014;66(6):723–9. Xu L, Jin QD, Gong X, Liu H, Zhou RX. Anti-gastric cancer effect of melatonin and Bcl-2, Bax, p21 and p53 expression changes. Sheng li xue bao : [Acta physiologica Sinica]. 2014;66(6):723–9.
169.
Zurück zum Zitat Plaimee P, Weerapreeyakul N, Thumanu K, Tanthanuch W, Barusrux S. Melatonin induces apoptosis through biomolecular changes, in SK-LU-1 human lung adenocarcinoma cells. Cell Prolif. 2014;47(6):564–77.PubMedPubMedCentralCrossRef Plaimee P, Weerapreeyakul N, Thumanu K, Tanthanuch W, Barusrux S. Melatonin induces apoptosis through biomolecular changes, in SK-LU-1 human lung adenocarcinoma cells. Cell Prolif. 2014;47(6):564–77.PubMedPubMedCentralCrossRef
170.
Zurück zum Zitat Kim W, Jeong JW, Kim JE. CCAR2 deficiency augments genotoxic stress-induced apoptosis in the presence of melatonin in non-small cell lung cancer cells. Tumour Biol. 2014;35(11):10919–29.PubMedCrossRef Kim W, Jeong JW, Kim JE. CCAR2 deficiency augments genotoxic stress-induced apoptosis in the presence of melatonin in non-small cell lung cancer cells. Tumour Biol. 2014;35(11):10919–29.PubMedCrossRef
171.
Zurück zum Zitat Hong Y, Won J, Lee Y, Lee S, Park K, Chang KT, Hong Y. Melatonin treatment induces interplay of apoptosis, autophagy, and senescence in human colorectal cancer cells. J Pineal Res. 2014;56(3):264–74.PubMedCrossRef Hong Y, Won J, Lee Y, Lee S, Park K, Chang KT, Hong Y. Melatonin treatment induces interplay of apoptosis, autophagy, and senescence in human colorectal cancer cells. J Pineal Res. 2014;56(3):264–74.PubMedCrossRef
172.
Zurück zum Zitat Park EJ, Woo SM, Min KJ, Kwon TK. Transcriptional and post-translational regulation of Bim controls apoptosis in melatonin-treated human renal cancer Caki cells. J Pineal Res. 2014;56(1):97–106.PubMedCrossRef Park EJ, Woo SM, Min KJ, Kwon TK. Transcriptional and post-translational regulation of Bim controls apoptosis in melatonin-treated human renal cancer Caki cells. J Pineal Res. 2014;56(1):97–106.PubMedCrossRef
173.
Zurück zum Zitat Perdomo J, Cabrera J, Estevez F, Loro J, Reiter RJ, Quintana J. Melatonin induces apoptosis through a caspase-dependent but reactive oxygen species-independent mechanism in human leukemia Molt-3 cells. J Pineal Res. 2013;55(2):195–206.PubMedCrossRef Perdomo J, Cabrera J, Estevez F, Loro J, Reiter RJ, Quintana J. Melatonin induces apoptosis through a caspase-dependent but reactive oxygen species-independent mechanism in human leukemia Molt-3 cells. J Pineal Res. 2013;55(2):195–206.PubMedCrossRef
174.
Zurück zum Zitat Zhang S, Qi Y, Zhang H, He W, Zhou Q, Gui S, Wang Y. Melatonin inhibits cell growth and migration, but promotes apoptosis in gastric cancer cell line, SGC7901. Biotechn Histochem. 2013;88(6):281–9.CrossRef Zhang S, Qi Y, Zhang H, He W, Zhou Q, Gui S, Wang Y. Melatonin inhibits cell growth and migration, but promotes apoptosis in gastric cancer cell line, SGC7901. Biotechn Histochem. 2013;88(6):281–9.CrossRef
175.
Zurück zum Zitat Wang J, Guo W, Chen W, Yu W, Tian Y, Fu L, Shi D, Tong B, Xiao X, Huang W, et al. Melatonin potentiates the antiproliferative and pro-apoptotic effects of ursolic acid in colon cancer cells by modulating multiple signaling pathways. J Pineal Res. 2013;54(4):406–16.PubMedCrossRef Wang J, Guo W, Chen W, Yu W, Tian Y, Fu L, Shi D, Tong B, Xiao X, Huang W, et al. Melatonin potentiates the antiproliferative and pro-apoptotic effects of ursolic acid in colon cancer cells by modulating multiple signaling pathways. J Pineal Res. 2013;54(4):406–16.PubMedCrossRef
176.
Zurück zum Zitat Xu C, Wu A, Zhu H, Fang H, Xu L, Ye J, Shen J. Melatonin is involved in the apoptosis and necrosis of pancreatic cancer cell line SW-1990 via modulating of Bcl-2/Bax balance. Biomed Pharmacother. 2013;67(2):133–9.PubMedCrossRef Xu C, Wu A, Zhu H, Fang H, Xu L, Ye J, Shen J. Melatonin is involved in the apoptosis and necrosis of pancreatic cancer cell line SW-1990 via modulating of Bcl-2/Bax balance. Biomed Pharmacother. 2013;67(2):133–9.PubMedCrossRef
177.
Zurück zum Zitat Li W, Wang Z, Chen Y, Wang K, Lu T, Ying F, Fan M, Li Z, Wu J. Melatonin treatment induces apoptosis through regulating the nuclear factor-kappaB and mitogen-activated protein kinase signaling pathways in human gastric cancer SGC7901 cells. Oncol Lett. 2017;13(4):2737–44.PubMedPubMedCentralCrossRef Li W, Wang Z, Chen Y, Wang K, Lu T, Ying F, Fan M, Li Z, Wu J. Melatonin treatment induces apoptosis through regulating the nuclear factor-kappaB and mitogen-activated protein kinase signaling pathways in human gastric cancer SGC7901 cells. Oncol Lett. 2017;13(4):2737–44.PubMedPubMedCentralCrossRef
178.
Zurück zum Zitat Gu J, Lu Z, Ji C, Chen Y, Liu Y, Lei Z, Wang L, Zhang HT, Li X. Melatonin inhibits proliferation and invasion via repression of miRNA-155 in glioma cells. Biomed Pharmacother. 2017;93:969–75.PubMedCrossRef Gu J, Lu Z, Ji C, Chen Y, Liu Y, Lei Z, Wang L, Zhang HT, Li X. Melatonin inhibits proliferation and invasion via repression of miRNA-155 in glioma cells. Biomed Pharmacother. 2017;93:969–75.PubMedCrossRef
179.
Zurück zum Zitat Gelaleti GB, Borin TF, Maschio-Signorini LB, Moschetta MG, Jardim-Perassi BV, Calvinho GB, Facchini MC, Viloria-Petit AM, de Campos Zuccari DAP. Efficacy of melatonin, IL-25 and siIL-17B in tumorigenesis-associated properties of breast cancer cell lines. Life Sci. 2017;183:98–109.PubMedCrossRef Gelaleti GB, Borin TF, Maschio-Signorini LB, Moschetta MG, Jardim-Perassi BV, Calvinho GB, Facchini MC, Viloria-Petit AM, de Campos Zuccari DAP. Efficacy of melatonin, IL-25 and siIL-17B in tumorigenesis-associated properties of breast cancer cell lines. Life Sci. 2017;183:98–109.PubMedCrossRef
180.
Zurück zum Zitat Bejarano I, Espino J, Marchena AM, Barriga C, Paredes SD, Rodríguez AB, Pariente JA. Melatonin enhances hydrogen peroxide-induced apoptosis in human promyelocytic leukaemia HL-60 cells. Mol Cell Biochem. 2011;353(1–2):167–76.PubMedCrossRef Bejarano I, Espino J, Marchena AM, Barriga C, Paredes SD, Rodríguez AB, Pariente JA. Melatonin enhances hydrogen peroxide-induced apoptosis in human promyelocytic leukaemia HL-60 cells. Mol Cell Biochem. 2011;353(1–2):167–76.PubMedCrossRef
181.
Zurück zum Zitat Um HJ, Park JW, Kwon TK. Melatonin sensitizes Caki renal cancer cells to kahweol-induced apoptosis through CHOP-mediated up-regulation of PUMA. J Pineal Res. 2011;50(4):359–66.PubMedCrossRef Um HJ, Park JW, Kwon TK. Melatonin sensitizes Caki renal cancer cells to kahweol-induced apoptosis through CHOP-mediated up-regulation of PUMA. J Pineal Res. 2011;50(4):359–66.PubMedCrossRef
182.
Zurück zum Zitat Leja-Szpak A, Jaworek J, Pierzchalski P, Reiter RJ. Melatonin induces pro-apoptotic signaling pathway in human pancreatic carcinoma cells (PANC-1). J Pineal Res. 2010;49(3):248–55.PubMedCrossRef Leja-Szpak A, Jaworek J, Pierzchalski P, Reiter RJ. Melatonin induces pro-apoptotic signaling pathway in human pancreatic carcinoma cells (PANC-1). J Pineal Res. 2010;49(3):248–55.PubMedCrossRef
183.
Zurück zum Zitat Casado-Zapico S, Rodriguez-Blanco J, García-Santos G, Martín V, Sánchez-Sánchez AM, Antolín I, Rodriguez C. Synergistic antitumor effect of melatonin with several chemotherapeutic drugs on human Ewing sarcoma cancer cells: potentiation of the extrinsic apoptotic pathway. J Pineal Res. 2010;48(1):72–80.PubMedCrossRef Casado-Zapico S, Rodriguez-Blanco J, García-Santos G, Martín V, Sánchez-Sánchez AM, Antolín I, Rodriguez C. Synergistic antitumor effect of melatonin with several chemotherapeutic drugs on human Ewing sarcoma cancer cells: potentiation of the extrinsic apoptotic pathway. J Pineal Res. 2010;48(1):72–80.PubMedCrossRef
184.
Zurück zum Zitat Martín V, García-Santos G, Rodriguez-Blanco J, Casado-Zapico S, Sanchez-Sanchez A, Antolín I, Medina M, Rodriguez C. Melatonin sensitizes human malignant glioma cells against TRAIL-induced cell death. Cancer Lett. 2010;287(2):216–23.PubMedCrossRef Martín V, García-Santos G, Rodriguez-Blanco J, Casado-Zapico S, Sanchez-Sanchez A, Antolín I, Medina M, Rodriguez C. Melatonin sensitizes human malignant glioma cells against TRAIL-induced cell death. Cancer Lett. 2010;287(2):216–23.PubMedCrossRef
185.
Zurück zum Zitat Jang SS, Kim WD, Park WY. Melatonin exerts differential actions on X-ray radiation-induced apoptosis in normal mice splenocytes and Jurkat leukemia cells. J Pineal Res. 2009;47(2):147–55.PubMedCrossRef Jang SS, Kim WD, Park WY. Melatonin exerts differential actions on X-ray radiation-induced apoptosis in normal mice splenocytes and Jurkat leukemia cells. J Pineal Res. 2009;47(2):147–55.PubMedCrossRef
186.
Zurück zum Zitat Raza H, John A, Brown EM, Benedict S, Kambal A. Alterations in mitochondrial respiratory functions, redox metabolism and apoptosis by oxidant 4-hydroxynonenal and antioxidants curcumin and melatonin in PC12 cells. Toxicol Appl Pharmacol. 2008;226(2):161–8.PubMedCrossRef Raza H, John A, Brown EM, Benedict S, Kambal A. Alterations in mitochondrial respiratory functions, redox metabolism and apoptosis by oxidant 4-hydroxynonenal and antioxidants curcumin and melatonin in PC12 cells. Toxicol Appl Pharmacol. 2008;226(2):161–8.PubMedCrossRef
187.
Zurück zum Zitat García-Santos G, Antolín I, Herrera F, Martín V, Rodriguez-Blanco J, Carrera MdP, Rodriguez C. Melatonin induces apoptosis in human neuroblastoma cancer cells. J Pineal Res. 2006;41(2):130–5.PubMedCrossRef García-Santos G, Antolín I, Herrera F, Martín V, Rodriguez-Blanco J, Carrera MdP, Rodriguez C. Melatonin induces apoptosis in human neuroblastoma cancer cells. J Pineal Res. 2006;41(2):130–5.PubMedCrossRef
188.
Zurück zum Zitat Majsterek I, Gloc E, Blasiak J, Reiter RJ. A comparison of the action of amifostine and melatonin on DNA-damaging effects and apoptosis induced by idarubicin in normal and cancer cells. J Pineal Res. 2005;38(4):254–63.PubMedCrossRef Majsterek I, Gloc E, Blasiak J, Reiter RJ. A comparison of the action of amifostine and melatonin on DNA-damaging effects and apoptosis induced by idarubicin in normal and cancer cells. J Pineal Res. 2005;38(4):254–63.PubMedCrossRef
189.
Zurück zum Zitat Wenzel U, Nickel A, Daniel H. Melatonin potentiates flavone-induced apoptosis in human colon cancer cells by increasing the level of glycolytic end products. Int J Cancer. 2005;116(2):236–42.PubMedCrossRef Wenzel U, Nickel A, Daniel H. Melatonin potentiates flavone-induced apoptosis in human colon cancer cells by increasing the level of glycolytic end products. Int J Cancer. 2005;116(2):236–42.PubMedCrossRef
190.
Zurück zum Zitat Cos S, Mediavilla MD, Fernández R, González-Lamuño D, Sánchez-Barceló EJ. Does melatonin induce apoptosis in MCF-7 human breast cancer cells in vitro? J Pineal Res. 2002;32(2):90–6.PubMedCrossRef Cos S, Mediavilla MD, Fernández R, González-Lamuño D, Sánchez-Barceló EJ. Does melatonin induce apoptosis in MCF-7 human breast cancer cells in vitro? J Pineal Res. 2002;32(2):90–6.PubMedCrossRef
191.
Zurück zum Zitat Lu Y-X, Chen D-L, Wang D-S, Chen L-Z, Mo H-Y, Sheng H, Bai L, Wu Q-N, Yu H-E, Xie D. Melatonin enhances sensitivity to fluorouracil in oesophageal squamous cell carcinoma through inhibition of Erk and Akt pathway. Cell Death Dis. 2016;7(10):e2432–e2432.PubMedPubMedCentralCrossRef Lu Y-X, Chen D-L, Wang D-S, Chen L-Z, Mo H-Y, Sheng H, Bai L, Wu Q-N, Yu H-E, Xie D. Melatonin enhances sensitivity to fluorouracil in oesophageal squamous cell carcinoma through inhibition of Erk and Akt pathway. Cell Death Dis. 2016;7(10):e2432–e2432.PubMedPubMedCentralCrossRef
192.
Zurück zum Zitat Lin PH, Tung YT, Chen HY, Chiang YF, Hong HC, Huang KC, Hsu SP, Huang TC, Hsia SM. Melatonin activates cell death programs for the suppression of uterine leiomyoma cell proliferation. J Pineal Res. 2020;68(1):e12620.PubMedCrossRef Lin PH, Tung YT, Chen HY, Chiang YF, Hong HC, Huang KC, Hsu SP, Huang TC, Hsia SM. Melatonin activates cell death programs for the suppression of uterine leiomyoma cell proliferation. J Pineal Res. 2020;68(1):e12620.PubMedCrossRef
193.
Zurück zum Zitat Lee WJ, Chen LC, Lin JH, Cheng TC, Kuo CC, Wu CH, Chang HW, Tu SH, Ho YS. Melatonin promotes neuroblastoma cell differentiation by activating hyaluronan synthase 3-induced mitophagy. Cancer Med. 2019;8(10):4821–35.PubMedPubMedCentralCrossRef Lee WJ, Chen LC, Lin JH, Cheng TC, Kuo CC, Wu CH, Chang HW, Tu SH, Ho YS. Melatonin promotes neuroblastoma cell differentiation by activating hyaluronan synthase 3-induced mitophagy. Cancer Med. 2019;8(10):4821–35.PubMedPubMedCentralCrossRef
194.
Zurück zum Zitat Nopparat C, Sinjanakhom P, Govitrapong P. Melatonin reverses H(2) O(2) -induced senescence in SH-SY5Y cells by enhancing autophagy via sirtuin 1 deacetylation of the RelA/p65 subunit of NF-κB. Journal of pineal research 2017;63(1). Nopparat C, Sinjanakhom P, Govitrapong P. Melatonin reverses H(2) O(2) -induced senescence in SH-SY5Y cells by enhancing autophagy via sirtuin 1 deacetylation of the RelA/p65 subunit of NF-κB. Journal of pineal research 2017;63(1).
195.
Zurück zum Zitat Martín V, Sanchez-Sanchez AM, Puente-Moncada N, Gomez-Lobo M, Alvarez-Vega MA, Antolín I, Rodriguez C. Involvement of autophagy in melatonin-induced cytotoxicity in glioma-initiating cells. J Pineal Res. 2014;57(3):308–16.PubMedCrossRef Martín V, Sanchez-Sanchez AM, Puente-Moncada N, Gomez-Lobo M, Alvarez-Vega MA, Antolín I, Rodriguez C. Involvement of autophagy in melatonin-induced cytotoxicity in glioma-initiating cells. J Pineal Res. 2014;57(3):308–16.PubMedCrossRef
196.
Zurück zum Zitat Liu C, Jia Z, Zhang X, Hou J, Wang L, Hao S, Ruan X, Yu Z, Zheng Y. Involvement of melatonin in autophagy-mediated mouse hepatoma H22 cell survival. Int Immunopharmacol. 2012;12(2):394–401.PubMedCrossRef Liu C, Jia Z, Zhang X, Hou J, Wang L, Hao S, Ruan X, Yu Z, Zheng Y. Involvement of melatonin in autophagy-mediated mouse hepatoma H22 cell survival. Int Immunopharmacol. 2012;12(2):394–401.PubMedCrossRef
197.
Zurück zum Zitat Sagrillo-Fagundes L, Bienvenue-Pariseault J, Vaillancourt C. Melatonin: The smart molecule that differentially modulates autophagy in tumor and normal placental cells. PLoS ONE. 2019;14(1):e0202458.PubMedPubMedCentralCrossRef Sagrillo-Fagundes L, Bienvenue-Pariseault J, Vaillancourt C. Melatonin: The smart molecule that differentially modulates autophagy in tumor and normal placental cells. PLoS ONE. 2019;14(1):e0202458.PubMedPubMedCentralCrossRef
198.
Zurück zum Zitat Galley HF, McCormick B, Wilson KL, Lowes DA, Colvin L, Torsney C. Melatonin limits paclitaxel-induced mitochondrial dysfunction in vitro and protects against paclitaxel-induced neuropathic pain in the rat. J Pineal Res. 2017;63(4):e12444.PubMedCentralCrossRef Galley HF, McCormick B, Wilson KL, Lowes DA, Colvin L, Torsney C. Melatonin limits paclitaxel-induced mitochondrial dysfunction in vitro and protects against paclitaxel-induced neuropathic pain in the rat. J Pineal Res. 2017;63(4):e12444.PubMedCentralCrossRef
199.
Zurück zum Zitat Waseem M, Sahu U, Salman M, Choudhury A, Kar S, Tabassum H, Parvez S. Melatonin pre-treatment mitigates SHSY-5Y cells against oxaliplatin induced mitochondrial stress and apoptotic cell death. PLoS ONE. 2017;12(7):e0180953.PubMedPubMedCentralCrossRef Waseem M, Sahu U, Salman M, Choudhury A, Kar S, Tabassum H, Parvez S. Melatonin pre-treatment mitigates SHSY-5Y cells against oxaliplatin induced mitochondrial stress and apoptotic cell death. PLoS ONE. 2017;12(7):e0180953.PubMedPubMedCentralCrossRef
200.
Zurück zum Zitat Koşar PA, Nazıroğlu M, Övey İS, Çiğ B. Synergic effects of doxorubicin and melatonin on apoptosis and mitochondrial oxidative stress in MCF-7 breast cancer cells: involvement of TRPV1 channels. J Membr Biol. 2016;249(1–2):129–40.PubMedCrossRef Koşar PA, Nazıroğlu M, Övey İS, Çiğ B. Synergic effects of doxorubicin and melatonin on apoptosis and mitochondrial oxidative stress in MCF-7 breast cancer cells: involvement of TRPV1 channels. J Membr Biol. 2016;249(1–2):129–40.PubMedCrossRef
201.
Zurück zum Zitat Plaimee P, Khamphio M, Weerapreeyakul N, Barusrux S, Johns NP. Immunomodulatory effect of melatonin in SK-LU-1 human lung adenocarcinoma cells co-cultured with peripheral blood mononuclear cells. Cell Prolif. 2014;47(5):406–15.PubMedPubMedCentralCrossRef Plaimee P, Khamphio M, Weerapreeyakul N, Barusrux S, Johns NP. Immunomodulatory effect of melatonin in SK-LU-1 human lung adenocarcinoma cells co-cultured with peripheral blood mononuclear cells. Cell Prolif. 2014;47(5):406–15.PubMedPubMedCentralCrossRef
202.
Zurück zum Zitat Ruiz-Rabelo J, Vázquez R, Arjona A, Perea D, Montilla P, Túnez I, Muntané J, Padillo J. Improvement of capecitabine antitumoral activity by melatonin in pancreatic cancer. Pancreas. 2011;40(3):410–4.PubMedCrossRef Ruiz-Rabelo J, Vázquez R, Arjona A, Perea D, Montilla P, Túnez I, Muntané J, Padillo J. Improvement of capecitabine antitumoral activity by melatonin in pancreatic cancer. Pancreas. 2011;40(3):410–4.PubMedCrossRef
203.
Zurück zum Zitat Lin ZY, Chuang WL. Pharmacologic concentrations of melatonin have diverse influence on differential expressions of angiogenic chemokine genes in different hepatocellular carcinoma cell lines. Biomed Pharmacother. 2010;64(10):659–62.PubMedCrossRef Lin ZY, Chuang WL. Pharmacologic concentrations of melatonin have diverse influence on differential expressions of angiogenic chemokine genes in different hepatocellular carcinoma cell lines. Biomed Pharmacother. 2010;64(10):659–62.PubMedCrossRef
204.
Zurück zum Zitat Padillo FJ, Ruiz-Rabelo JF, Cruz A, Perea MD, Tasset I, Montilla P, Túnez I, Muntané J. Melatonin and celecoxib improve the outcomes in hamsters with experimental pancreatic cancer. J Pineal Res. 2010;49(3):264–70.PubMedCrossRef Padillo FJ, Ruiz-Rabelo JF, Cruz A, Perea MD, Tasset I, Montilla P, Túnez I, Muntané J. Melatonin and celecoxib improve the outcomes in hamsters with experimental pancreatic cancer. J Pineal Res. 2010;49(3):264–70.PubMedCrossRef
205.
Zurück zum Zitat Moselhy SS, Al mslmani MA. Chemopreventive effect of lycopene alone or with melatonin against the genesis of oxidative stress and mammary tumors induced by 7,12 dimethyl(a)benzanthracene in sprague dawely female rats. Mol Cell Biochem. 2008;319(1–2):175–80.PubMedCrossRef Moselhy SS, Al mslmani MA. Chemopreventive effect of lycopene alone or with melatonin against the genesis of oxidative stress and mammary tumors induced by 7,12 dimethyl(a)benzanthracene in sprague dawely female rats. Mol Cell Biochem. 2008;319(1–2):175–80.PubMedCrossRef
206.
Zurück zum Zitat Ruiz-Rabelo JF, Vázquez R, Perea MD, Cruz A, González R, Romero A, Muñoz-Villanueva MC, Túnez I, Montilla P, Muntané J, et al. Beneficial properties of melatonin in an experimental model of pancreatic cancer. J Pineal Res. 2007;43(3):270–5.PubMedCrossRef Ruiz-Rabelo JF, Vázquez R, Perea MD, Cruz A, González R, Romero A, Muñoz-Villanueva MC, Túnez I, Montilla P, Muntané J, et al. Beneficial properties of melatonin in an experimental model of pancreatic cancer. J Pineal Res. 2007;43(3):270–5.PubMedCrossRef
207.
Zurück zum Zitat Zhu H, Chen Y, Bai LC, Cao XR, Xu R. Different effects of melatonin on X-rays-irradiated cancer cells in a dose-dependent manner. Dose-Response. 2019;17(3):1559325819877271.PubMedPubMedCentralCrossRef Zhu H, Chen Y, Bai LC, Cao XR, Xu R. Different effects of melatonin on X-rays-irradiated cancer cells in a dose-dependent manner. Dose-Response. 2019;17(3):1559325819877271.PubMedPubMedCentralCrossRef
208.
Zurück zum Zitat Chen YT, Yang CC, Shao PL, Huang CR, Yip HK. Melatonin-mediated downregulation of ZNF746 suppresses bladder tumorigenesis mainly through inhibiting the AKT-MMP-9 signaling pathway. J Pineal Res. 2019;66(1):e12536.PubMedCrossRef Chen YT, Yang CC, Shao PL, Huang CR, Yip HK. Melatonin-mediated downregulation of ZNF746 suppresses bladder tumorigenesis mainly through inhibiting the AKT-MMP-9 signaling pathway. J Pineal Res. 2019;66(1):e12536.PubMedCrossRef
209.
Zurück zum Zitat Gurunathan S, Jeyaraj M, Kang MH, Kim JH. Melatonin Enhances Palladium-Nanoparticle-Induced Cytotoxicity and Apoptosis in Human Lung Epithelial Adenocarcinoma Cells A549 and H1229. Antioxidants (Basel, Switzerland). 2020;9(4):357. Gurunathan S, Jeyaraj M, Kang MH, Kim JH. Melatonin Enhances Palladium-Nanoparticle-Induced Cytotoxicity and Apoptosis in Human Lung Epithelial Adenocarcinoma Cells A549 and H1229. Antioxidants (Basel, Switzerland). 2020;9(4):357.
210.
Zurück zum Zitat Li M, Wu C, Muhammad JS, Yan D, Tsuneyama K, Hatta H, Cui ZG, Inadera H. Melatonin sensitises shikonin-induced cancer cell death mediated by oxidative stress via inhibition of the SIRT3/SOD2-AKT pathway. Redox Biol. 2020;36:101632.PubMedPubMedCentralCrossRef Li M, Wu C, Muhammad JS, Yan D, Tsuneyama K, Hatta H, Cui ZG, Inadera H. Melatonin sensitises shikonin-induced cancer cell death mediated by oxidative stress via inhibition of the SIRT3/SOD2-AKT pathway. Redox Biol. 2020;36:101632.PubMedPubMedCentralCrossRef
211.
Zurück zum Zitat Zakki SA, Muhammad JS, Li JL, Sun L, Li ML, Feng QW, Li YL, Cui ZG, Inadera H. Melatonin triggers the anticancer potential of phenylarsine oxide via induction of apoptosis through ROS generation and JNK activation. Metallomics Integr Biometal Sci. 2020;12(3):396–407.CrossRef Zakki SA, Muhammad JS, Li JL, Sun L, Li ML, Feng QW, Li YL, Cui ZG, Inadera H. Melatonin triggers the anticancer potential of phenylarsine oxide via induction of apoptosis through ROS generation and JNK activation. Metallomics Integr Biometal Sci. 2020;12(3):396–407.CrossRef
Metadaten
Titel
An updated review of mechanistic potentials of melatonin against cancer: pivotal roles in angiogenesis, apoptosis, autophagy, endoplasmic reticulum stress and oxidative stress
verfasst von
Saeed Mehrzadi
Mohammad Hossein Pourhanifeh
Alireza Mirzaei
Farid Moradian
Azam Hosseinzadeh
Publikationsdatum
01.12.2021
Verlag
BioMed Central
Erschienen in
Cancer Cell International / Ausgabe 1/2021
Elektronische ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-021-01892-1

Weitere Artikel der Ausgabe 1/2021

Cancer Cell International 1/2021 Zur Ausgabe

Alphablocker schützt vor Miktionsproblemen nach der Biopsie

16.05.2024 alpha-1-Rezeptorantagonisten Nachrichten

Nach einer Prostatabiopsie treten häufig Probleme beim Wasserlassen auf. Ob sich das durch den periinterventionellen Einsatz von Alphablockern verhindern lässt, haben australische Mediziner im Zuge einer Metaanalyse untersucht.

Antikörper-Wirkstoff-Konjugat hält solide Tumoren in Schach

16.05.2024 Zielgerichtete Therapie Nachrichten

Trastuzumab deruxtecan scheint auch jenseits von Lungenkrebs gut gegen solide Tumoren mit HER2-Mutationen zu wirken. Dafür sprechen die Daten einer offenen Pan-Tumor-Studie.

Mammakarzinom: Senken Statine das krebsbedingte Sterberisiko?

15.05.2024 Mammakarzinom Nachrichten

Frauen mit lokalem oder metastasiertem Brustkrebs, die Statine einnehmen, haben eine niedrigere krebsspezifische Mortalität als Patientinnen, die dies nicht tun, legen neue Daten aus den USA nahe.

Labor, CT-Anthropometrie zeigen Risiko für Pankreaskrebs

13.05.2024 Pankreaskarzinom Nachrichten

Gerade bei aggressiven Malignomen wie dem duktalen Adenokarzinom des Pankreas könnte Früherkennung die Therapiechancen verbessern. Noch jedoch klafft hier eine Lücke. Ein Studienteam hat einen Weg gesucht, sie zu schließen.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.