Next Article in Journal
What Is the Evolutionary Fingerprint in Neutrophil Granulocytes?
Previous Article in Journal
Innovative Three-Dimensional Microscopic Analysis of Uremic Growth Plate Discloses Alterations in the Process of Chondrocyte Hypertrophy: Effects of Growth Hormone Treatment
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Melatonin and Mesenchymal Stem Cells as a Key for Functional Integrity for Liver Cancer Treatment

1
Department for Management of Science and Technology Development, Ton Duc Thang University, Ho Chi Minh, Vietnam
2
Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, Vietnam
3
Laboratory of Kafr El Sheikh Fever Hospital, Kafr El Sheikh Fever Hospital, 33511 Kafr El-Sheikh, Egypt
4
Department of Zoology, Faculty of Science, Tanta University, Tanta 31527, Egypt
5
Department of Pathology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El Sheikh Governorate 33516, Egypt
6
Laboratory of Wildlife and Forensic Pathology/Biomedical Science Examination and Research Center, Department of Veterinary Medicine, Faculty of Veterinary Medicine, Okayama University of Science, Okayama 700-8530, Japan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2020, 21(12), 4521; https://doi.org/10.3390/ijms21124521
Submission received: 25 May 2020 / Revised: 19 June 2020 / Accepted: 21 June 2020 / Published: 25 June 2020
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)

Abstract

:
Hepatocellular carcinoma (HCC) is the most common hepatobiliary malignancy with limited therapeutic options. On the other hand, melatonin is an indoleamine that modulates a variety of potential therapeutic effects. In addition to its important role in the regulation of sleep–wake rhythms, several previous studies linked the biologic effects of melatonin to various substantial endocrine, neural, immune and antioxidant functions, among others. Furthermore, the effects of melatonin could be influenced through receptor dependent and receptor independent manner. Among the other numerous physiological and therapeutic effects of melatonin, controlling the survival and differentiation of mesenchymal stem cells (MSCs) has been recently discussed. Given its controversial interaction, several previous reports revealed the therapeutic potential of MSCs in controlling the hepatocellular carcinoma (HCC). Taken together, the intention of the present review is to highlight the effects of melatonin and mesenchymal stem cells as a key for functional integrity for liver cancer treatment. We hope to provide solid piece of information that may be helpful in designing novel drug targets to control HCC.

1. Introduction

The last few years have witnessed extraordinary increase in the reports of liver cancers worldwide [1,2]. In 2018, around 841,000 cases and 782,000 deaths were recorded due to these types of cancer [1,2]. Furthermore, this type of cancer has been considered the 5th cancer type between the male, the 7th between female and the 4th fatal between other cancers [1,2]. Therefore, it is not surprising to state that liver cancer has been considered one of the tumors with the fastest rising incidence and highest mortality in recent years [3,4]. Among others, hepatocellular carcinoma (HCC) has been considered the most common type of liver cancers.
HCC is mostly linked to miscellaneous predisposing etiologies including viral hepatitis or exposure to toxins such as aflatoxin [5,6,7]. Given its global distribution, most HCC cases are estimated to occur in Asia and sub-Saharan Africa [6,8]. Several factors are considered inclining agents for developing HCC such as hemochromatosis and alpha 1-antitrypsin deficiency and metabolic syndrome [2]. Taken into account, the prognosis of HCC mainly depends on several factors including the degree of tumor spread, size of tumor and the general healthy status, among others [6]. Despite the great progress achieved in understanding of HCC, resistance of apoptosis treatment is still challenged by apoptosis resistance [9]. Some recent studies showed that inhibitor of apoptosis proteins (IAPs) have been involved in resistance to apoptosis in HCC through inhibition of caspases activation [10,11]. Indeed, it seems mandatory searching effective novel therapeutic agents that can improve the treatment courses of HCC and future prognosis of such cases.
To our knowledge, melatonin has been identified as a natural antioxidant with numerous immunoenhancing properties, while mesenchymal stem cells has shown a potential promising strategy either in preventing or arresting neoplastic growth [12,13]. The intention of the following sections is to give an overview about mesenchymal stem cells (MSCs) and highlight several physiological and biologic effects of melatonin followed by discussing the potential promising effects of the combined use of melatonin and MSCs in treatment of HCC.

2. Mesenchymal Stem Cells (MSCs)

2.1. An Overview of Mesenchymal Stem Cells (MSCs)

Mesenchymal stem cells (MSCs) are multipotent cells capable for differentiation into cartilage, bone, muscle, tendon, ligament, fat and hepatocyte. MSCs population is one of the major stem cell populations in the adult bone marrow [14]. They represent only 0.01% to 0.001% of all mononuclear cells in the bone marrow [14], making the identification of a native MSC niche is difficult [15]. Several locations for in vivo MSC niche within the bone marrow have been proposed including the periosteal niche, the pericytic niche and the perivascular niche [16]. Surface marker expression studies referred to the perivascular niche as the true “home” for MSCs, allowing the easier access of MScs progeny to the circulation [17]. The recent years gave more attention towards pluripotent mesenchymal stem cells, which are found in bone marrow stem cells (BMSCs) and adipose tissue (AD-MSC) [18]. In this regard, MSCs have been proposed as promising sources for restoring tissue and organ function [19]. However, several potential health hazards for their clinical application were reported, including shortage in their availability, their sensitivity to toxic environments, senescence and tumorigenicity [19]. In addition, MSCs- based treatment has shown regeneration of organ function via the production of cytokines and other several anti-inflammatory mechanisms [20].

2.2. Isolation and Characterization of MSCs

To authors’ knowledge, MSCs are mostly isolated from bone barrow, fat and cord tissues [21,22]. However, pluripotent stem cells could be isolated from other numerous tissues and organs, including adipose tissue, skin, dental tissues, placenta, umbilical cord blood, liver, menstrual blood, dental tissue, perinatal tissues and ear [23]. Among others, isolation of MSCs from adipose connective tissue has been considered as an ubiquitous techniques in stem cell-based therapy with minimal invasive protocol [24]. In addition, the outer surface of the ear has plenty of MSCs expressing multiple stromal markers besides their ability to differentiate into different lineage including fat, cartilaginous and osseous tissues [25,26]. It is noteworthy to mention that some modifications have been carried out for isolation of mulitpotent MSCs, including alterations to culture media supplements and serum percentage, growth on various substrates such as collagen and fibronectin, and depletion of hematopoietic cell contaminants by surface marker-based negative selection [27]. These methods allow enrichment of a fibroblastic spindle-cell population [27]. Although a heterogeneous mixture of spindle cells, star-shaped cells and large flattened cells is frequently observed, a characteristic pattern of surface marker co-expression indicates the self-renewal and multipotence capabilities was noticed [28]. Additional surface markers reveal subpopulations of MSC that are differentially committed to various stromal cell types were also recorded [29]. Taking into account that human and murine MSC generally are not able to express hematopoietic markers cluster of differentiations (CD) as CD34 and CD45, however, subpopulations of cells express a low level of these markers [30]. Human MSC were reported to be positive for the following surface markers: CD44, CD73, CD90, CD105, CD106 and STRO-1, while murine MSC express stem cell antigen-1 (Sca-1 or Ly6A) and all these previously mentioned markers except STRO-1 [31]. Likewise, these markers have been used in combination to get pure MSCs from bone marrow isolates [32]. Additionally, it was reported that certain multipotent bone marrow stromal cells did not display these markers in vivo. Consequently, sorted MSCs populations may not contain all multipotent marrow stromal subtypes [33,34]. The absence of MSCs specific genes and markers is another hurdle faced in the characterization of these cells. The trypsin-resistant antigen denoted STRO-1 remains the best choice, since it is absent in peripheral tissues, the hematopoietic compartment or in mature mesenchymal cells, however, it is expressed on endothelial progenitors [35]. Unfortunately, the structural and functional characteristics of STRO-1 have yet to be determined combined with STRO-1 negativity in mice before its use in preclinical studies [36]. It is noteworthy to state that the pluripotent stem cell-derived MSCs overcome many disadvantages of adult MSCs such as reduced batch-to-batch variations and stem cell senescence and produced some unique cytokines different from bone marrow-or cord-derived MSCs [37,38,39]. Interestingly, GMP-grade MSCs are currently being used in clinical trials for various viable diseases, including industrial sectors. In this concern, Mayo Clinic initiated a phase I/II trial to find the side effects and best dose of MSCs infected with oncolytic measles virus encoding NIS (MV-NIS) and to observe its effect on patients with ovarian cancer. These studies suggested further future in-depth research about MSCs timing in patients [40,41].

2.3. Recruitment of MSCs

Tissue repair, inflammation and neoplasia represent few of the processes that encourage engraftment of circulating MSCs [42]. Tumor tropism of MSC can be examined by cell trafficking assays in vitro and in vivo. A number of modalities, such as intravenous injection, utilizing fluorescence, magnetic resonance and bioluminescence, can be used to track ex vivo expanded MSC [43]. No previous studies have deeply explored the migration of endogenous MSC into tumors, but some previous reports have shown that recruitment of labeled MSC home to tumor stroma following bone marrow engraftment in sublethally irradiated mice [44].
The process of MSCs recruitment into tumors follows a similar pattern of recruitment of the activated inflammatory cells during tissue repair [45]. MSCs show graded responses to leukocyte and endothelial activating Transforming growth factor-beta (TGFβ-1), interleukin-6 (IL-6), IL-8, IL-37 and neurotrophin 3 (NT-3) [46]. However, under hypoxic conditions, breast cancer cells produce high amounts of IL-6 which activates and attracts MSCs. It should be stressed that IL-6 also acts in a paracrine fashion on MSC, resulting in activation of STAT3 and MAPK signaling pathways that together trigger the survival of the cell and their migratory potential [47]. Furthermore, LL-37 (Leucine leucine-37), which presents in many tumors, stimulates the migratory activity of MSCs and facilitates the progression of ovarian tumor via recruitment of MSCs to act as pro-angiogenic factor-expressing tumor stromal cells [48].
In addition, tumor cells secrete many chemoattractants that promote the migratory activity of MSCs and MSCs express receptors of the four chemokine subfamilies: CC, CXC, CX(3)C and C [49]. It should be born in mind that several chemotaxis assays in vitro have shown that Dose-dependent migration of MSCs could be induced by chemokines like CCL2/MCP1 (monocyte chemoattractant protein-1), CCL25 (thymus expressed chemokine), CXCL8 (IL8), CXCL12/SDF1α and CXCL13 (BCA1) [50]. In addition, sphingosine 1 phosphate (S1P) exerted a strong chemoattraction on MSCs through matrix metalloproteinase (MMP)-mediated signaling events and the RhoA/ROCK and MEK1/ERK intracellular pathways [51].

2.4. Dual Roles of MSCs in Liver Cancer

It is noteworthy to state that the considerable difference of intrahepatic microenvironment from other organs seems to influence the development of cancer [52]. MSCs constitute an important components within the microenvironment of both normal liver and liver with tumors, suggesting their pleiotropic functionality which is shown in Figure 1. Clearly, MSCs may exert a tumor-promoting or a tumor-limiting effects depending on the experimental circumstances [13]. Several hypotheses have been proposed to explore this dualistic behavior of MSCs in cancer [53,54]. One of these theories L is related to the role played by TLRs in immuno-polarization of MSCs. MSCs express several TLRs combined with their capabilities to migrate, invade and secrete immune modulating factors [13]. Interestingly, TLR4-primed MSCs and TLR3-primed MSCs are polarized into two phenotypes; a proinflammatory MSC1 and the classical immunosuppressive MSC2 phenotype, respectively [55]. In cancer models, MSC1-based treatment of established tumors in an immune competent animal models impaired the tumor growth and metastasis [56]. On the contrary, MSC2-treated animals displayed an increase tumor growth and metastasis [56]. The other theory proposes a developmental phase-dependent MSC functionality [57]. In this hypothesis, MSCs may promote tumor growth in case of co-injection with tumor cells, while their administration in established tumors inhibit progression of tumors. This means that the presence of MSCs during the early stage of tumorigenesis may contribute to angiogenesis [58]. Clearly, the tumor cells and their microenvironment may have an influence on the action of recruited MSCs [59]. Taken together, both postulations seem concomitantly true and therefore, it is very hard to make a prediction to the effects of MSCs on the cancerous process [60,61].

2.5. Mechanisms of MSC-Dependent Tumor Suppression in Liver Cancer

To author knowledge, MSCs have shown tumor suppressive effects in induced murine HCC that were linked to down regulation of Wnt signaling target genes [60,62]. There are several suggested mechanisms lying behind this action. In this concern, TLR signals can stimulate downstream effectors that may interfere with LPS–TLR4 pathway and the active secretion of Wnt inhibitors, including Dickkopf-1, combined with MSC-dependent inhibition of NF-kB signaling in cancer cells [13,63]. Additionally, MSCs release microvesicles that exert several actions include inhibition of both cell cycling and the growth of different established tumors in vivo, besides induction of the apoptosis of HCC cell lines in vitro, which in turn provides another antioncogenic pathway [62]. However, it should be borne in mind that the direct effectors are still unclear, but it seems that the secretome of MSCs play an crucial role in suppression of tumors [64]. It should be stressed that various molecular mechanisms seem to be involved in therapeutic MSC actions in vivo and increase their regenerative potential [65]. Among others, direct MSC differentiation into cardiac and endothelial cells and the paracrine activity mediated by MSC-derived soluble molecules and vesicles are currently considered as major factors mediating beneficial effects of MSC-based therapies in various viable diseases [65,66,67,68,69]. However, recent studies showed that paracrine function of MSCs is the main mechanism by which these cells participate in tissue repair [68]. Importantly, MSCs can promote normal tissue regeneration through enhancement of angiogenesis, tissue remodeling and activation of endogenous stem cells [70], which endorsed the paracrine actions rather than cell differentiation. MSCs paracrine functions seem tightly regulated by Rap1/nuclear factor-kappaB (NF-κB) signaling pathway. It is noteworthy to mention that the absence of Rap1 of MSCs markedly enhance the secretion profile of these cells and their resistance to any stressful challenge combined with reduction the production of proinflammatory cytokines [71]. Clearly, NF-kb signaling pathway play a critical role in HCC and therefore, the anti-inflammatory properties of MSCs were found remarkable in the earlier stages [72]. Another interesting mechanism of MSCs is through transfer of mitochondria as reported in several previous studies [73,74]. Mitochondria have been considered a key player involved in many biologic processes in health and disease, including in HCC [75,76,77,78,79]. Some proinflammatory cytokines such as Il-6 and TNF-α can induce MSCs skeletal rearrangement and form tunneling nanotubes (TNT) through which mitochondria mobility occurs from MSCs to neighbor cells [73,80]. Inflammation-driven mitochondrial transfer of MSC to neighbor cells including retinal cells and cancer cells were also reported recently [73]. It should be stressed that the therapeutic effects of MSCs and direction of mitochondrial transfer highly depend on the niche where MSCs is located, including in case of HCC [73]. It seems that the proinflammatory environment can enhance MSCs—mitochondrial transfer and MSC—mitochondrial transfer to T cells, which in turn trigger various immune cells, including CD4+T cells [81].

2.6. Therapeutic Application of MSCs in Liver Cancer

To our knowledge, several preclinical models showed that MSCs can migrate into different types of tumors and therefore, this notion has inspired many experts in the field about the potential use of MSCs in anticancer drug/gene delivery [82,83]. In this regards, the genetically modified MSCs demonstrated a clear inhibition the proliferation of HCC in vitro and in vivo [84]. Delivering oncolytic viruses such as Measles virus into the tumor cells via MSCs represent another approach to avoid pre-existing immunity against the virus [85]. These interesting findings seems very promising for designing novel trials for treating HCC using MSCs as a vector. In accordance with the clinical trials, MSCs have been extensively investigated in treatment of various types of cancer such as ovarian cancer, head and neck cancer and prostate cancer [86,87,88].
Moreover, adoptive immunotherapy which relies on transfer of naturally occurring or genetically engineered T cells represents another novel shape of cancer therapy [89,90]. This previously mentioned technique could be carried out using induced pluripotent stem cells (iPSCs) that may provide unlimited source of highly reactive antigen-specific cytotoxic T-lymphocytes, which in turn target, infiltrate and eradicate tumors upon their transfer into the patient [91]. Interestingly, bone marrow-derived MSCs transduced with a lentiviral vector stTRAIL have shown promising results in to treatment of heat-shocked residual cancer cells that target tumor growth inhibition [92].

3. Melatonin

3.1. Synthesis and Precursors of Melatonin

Melatonin (MLT), N-acetyl-5-methoxytryptamine, is a natural substance that has been recognized in all major living species including plants, animals, bacteria, other unicellular microorganisms and human being [44,93]. This natural substance is normally secreted during the dark phase of the daily light–dark cycle [94]. Given its lipophilic nature, MLT is mainly produced by the pineal gland, then released into the circulation and gains access to various fluids, tissues and cellular compartments [95,96]. Other peripheral organs and tissues rather than pineal gland are also get involved in secretion of melatonin including retina, Harderian gland, gastrointestinal tract, leukocytes, thymus and bone marrow cells, however, the chronobiotic properties is retained to the pineal secretions [97]. As result of its amphiphilic nature, melatonin gets infiltrated inside subcellular compartment, enabling it to cross all biologic barriers and gets free access to all cellular compartments.
Regarding its synthesis, MLT is synthesized from the amino acid tryptophan, taken up from blood and converted to serotonin [98]. Serotonin is then acetylated to N-acetylserotonin by arylalkylamine N-acetyltransferase enzyme. N-acetylserotonin is subsequently converted into MLT by hydroxyindole-O-methyltransferase (HIOMT) enzyme. Interestingly, the enzymes of MLT biosynthesis have recently been identified in human lymphocytes and therefore, locally synthesized MLT is probably modulate the immune system [99]. Among other extra-pineal major production sites of MLT, the gastrointestinal (GI) tract is of particular interest since it contains several hundred-fold of MLT exceeding those amounts of the pineal gland [100]. Taketo into consideration, GI MLT may release into circulation under certain circumstances such as under the influence of high dietary tryptophan levels [101]. The following section highlights some facts about the physiological and therapeutic implications of melatonin, particularly against cancer (Figure 2).

3.2. Signaling Mechanism of Melatonin

To best of author’s knowledge, two mammalian subtypes of G protein-coupled receptor (GPCR) binds to melatonin receptors; MT1 (Mel1a) and MT2 (Mel1b) [102]. These two mammalian subtypes play an important role in exerting some of MLT actions [102]. Moreover, MT3, has been identified initially as third binding site, then it was subsequently characterized as quinone reductase 2 enzyme [103]. It was reported that the decrease in Cyclic adenosine monophosphate (cAMP) production, caused by MLT via MT1 and MT2 receptor interaction, results in reduction the uptake of linoleic acid by affecting special fatty acid transporter [104,105]. Linoleic acid can be oxidized to 13- hydroxyoctadecadienoic acid by 15-lipoxygenase that serves as an energy source for tumor growth and tumor growth-signaling molecules. Moreover, inhibition of linoleic acid uptake by MLT has been considered a mechanism of the antiproliferative effects in case of cancer [106]. Taken into account, MLT also acts through binding to cytoplasmic proteins like the calcium binding proteins such as calmodulin or tubulin and to nuclear receptors like RZR/ROR [105,107,108]. Some studies also suggested that modulation of the expression and function of nuclear receptors (RZR/ROR) could influence the biologic effects of MLT [109,110]. By binding to nuclear receptors, MLT alters the process of transcription of several genes that play a role in cellular proliferation (i.e., 5-lipoxygenase, p21 or bone sialoprotein) [110,111].
Another suggested mechanism of action of melatonin is modulation of intracellular calcium and calmodulin activity [112,113]. Calcium-activated calmodulin is linked to the initiation of the S and M phases of the cell cycle during the cell cycle-related gene expression regulation and in the reentry of quiescent cells from G0 back into the cell cycle [114]. Melatonin has shown to increase calmodulin degradation through a direct binding and redistributing it, thereby inhibiting cell cycle progression [115,116]. It also serves as a potent modulator of gene transcriptional activity and targets a considerable number of genes, in central or in peripheral tissues [117]. It was hypothesized that melatonin mediates the seasonal photoperiodic control via phasing clock genes expression in the pars tuberalis [118]. In addition, MLT downregulates the expression of integrin and integrin-associated protein encoding genes in rat retina, while upregulates the cAMP response element binding protein cAMP response element-binding (CREB) gene in retinal pigmentary cells [119]. Notably, melatonin has also shown a striking effects on the expression of certain genes related to oncogenesis (e.g., Mybl1, Mllt3, Rasa1 and Enigma homolog 2) and calcium metabolism (Kcnn4 and Dcakl1) [12,120]. It should be stressed that the exact mechanisms of melatonin underlying the suppression of these oncogenes are still unclear, however, some reports linked it to the direct interaction of with Bridging Integrator 1 (BIN1) that considers as HCC suppressant gene with c-Myc, leading to downregulation of c-Myc associated with HCC [121]. Interestingly, linked bridging integrator 1 (BIN1) or Myc box-dependent-interacting protein 1 is also highly expressed in pineal gland [122]. In addition, MLT has a significant effects on mitochondrial genes expression, like genes encoding cytochrome C oxidase subunits I and II (mt-Co1, mt-Co3), 16S ribosomal RNA (mt-RNr2), NADH dehydrogenase 1 (mt-Nd1),) and ATP synthase subunit 6 (mt-ATP6; downregulated) [123].

3.3. Antioxidant Effect of Melatonin

Interestingly, it seems that the function of melatonin in phylogeny is related to its antioxidant activity [124]. Several herbs have been used by Chinese in ancient ages due to its high levels of melatonin to retard aging and to treat diseases associated with the production of free radicals [114]. Besides its actions as free radical scavenger and its role in membrane stabilization, melatonin acts on enzymes that generate or metabolize reactive oxygen intermediates, there by further increasing its protective activity toward free radicals [125,126]. Furthermore, melatonin influences the antioxidant enzymes gene expression as it increases mRNA levels for both Cu–Zn–SOD and Mn–SOD in the Harderian gland and brain cortex of rodents [127]. Moreover, melatonin enhances the activity of glutathione peroxidase (GPx) to remove hydrogen peroxide (H2O2) from cells [128]. Therefore, several important antioxidative enzymes seem to be stimulated by MLT, protecting cells from oxidative damage [129]. Meanwhile, recent report indicated that the engineered MSC with overexpression of GPx can enhance the protection of hepatocytes [130].

3.4. Anticancer Effect of Melatonin

To our knowledge, many natural mechanisms are widely known to protect against carcinogenesis, and they fall into two main categories, immune and non-immune [131]. Importantly, immunosurveillance has been proposed as one of the major processes by which cancerous cells are detected and eliminated [132]. Studies of knockout mice have shown the important role played by the immune system in controlling the spontaneous generation of tumors [133]. Understanding the immune changes in the elderly can provide new insights into the complex relationship between immunity and cancer [134]. The age-related impairment of the immune system appears around the sixth decade of age coinciding with a normal decline in plasma MLT concentration [135]. Aging has been associated with a decrease in immune function and an increased incidence of cancer [136]. In this respect, the decline in the production of MLT with aging was suggested to play a crucial role in triggering immunosenescence, especially age-associated neoplastic diseases [137]. antitumor defense assumes a primary role among the various functions attributed to melatonin in modulation of the immune system. The nighttime physiological surge of MLT in blood or extracellular fluid has been proposed to serve as a “natural restraint” for tumor initiation, promotion and/or progressions [138]. The activation of lymphocytes and monocytes/macrophages by MLT can be one of the major mechanisms in preventing tumor development besides its crucial immunomodulatory role in the immunocompromised state [139]. Some previous reports have investigated the potential beneficial effects of melatonin in induction of HCC and HepG-2 cell death) and ovarian cancer in animal models through enhancement the apoptosis of cancerous cells [140,141,142].
Moreover, administration of melatonin increases the production of both NK cells and monocytes (which contain MLT receptors) in bone marrow and spleen within 7–14 days of treatment [143]. Since both cell types are components of the non-specific immune system, melatonin can be effective in prevent the growth of cancer [144]. Indeed, melatonin was able to rescue hematopoiesis from the toxic effect of cancer chemotherapy in several experimental models [145]. This evidence actually poses the basis for the therapeutic use of MLT as an adjuvant in combination with myelotoxic anticancer therapeutic protocols [146].

3.5. Regulation Effects of Melatonin on the Immune System

It seems that the level of melatonin secretion in human beings could be influenced along the different season of the year that reflects the significant role played by MLT on immune system modulation [147]. In addition, the synthesis of MLT by human lymphocytes support the hypothesis proposes that MLT has a role in the regulation of immune function [148,149]. Furthermore, melatonin can enhance the immune response that may be helpful in correction of the immunodeficiencies secondary to viral diseases or acute stress [150,151]. It should be stressed that melatonin plays an important role in modulation of hematopoiesis, immune cell production and function [149]. MLT also stimulates cytokine production, enhanced phagocytosis, increased NK cell activity and skewing of the immune response toward a helper T cell type 1 profile (Th1) [152]. Likewise, up regulation of cytokine production and immune function occur as a result of binding of melatonin to its receptors [153]. Both membrane and nuclear receptors have been identified on leukocytes. Importantly, membrane receptors were found mainly on CD4 T lymphocytes, but also on CD8 T and B cells and it was reported that melatonin modulates the proliferative response of stimulated lymphocytes via these receptors [154]. On the other hand, MLT induces cytokine production by mononuclear cells through its influence on the nuclear MLT receptors [155]. Indeed, treatment with MLT enhanced antigen presentation by splenic macrophages to T cells together with a concurrent increase in MHC class II expression and synthesis of the proinflammatory cytokines IL-1, IL-2 and Tumor Necrosis factor (TNF [156].
Additionally, treatment of mice with melatonin resulted in enhancing the expression of Macrophage colony-stimulating factor (M-CSF), TNF-α, TGF-β and stem cell factor (SCF) in peritoneal macrophages, while IL-1β, IFN-γ, M-CSF, TNF-α and SCF was increased in spleen cells of mice [157]. The presence of high levels of melatonin in cultured rat thymocytes and expression of mRNAs encoding for Arylalkylamine N-acetyltransferase (AANAT) and HIOMT in the rat and human thymus cells support the hypothesis that MLT is also synthesized by thymocytes [158]. Likewise, the pineal neurohormone MLT has been widely shown to exert an immunostimulatory and potent inhibitor of apoptosis in immune cells through its action on Th cells and on T- and B-cell precursors, respectively [156,159].

4. Potential Beneficial Effect of the Combination between Melatonin and MSCs on Triggering HCC

Given the above information, several previous works have suggested the potential contribution of melatonin to overcome HCC through various mechanisms including targeting the expression survivin and X-linked inhibitor of apoptosis (XIAP) via the cyclooxygenase-2 (COX-2)/phosphatidylinositol 3-kinase pathway (PI3K/Akt) pathway [160]. Furthermore, melatonin has shown to exerts numerous anticancer effects through enhancement the expression of various pro-apoptotic markers (mainly Bax and caspase 3) besides induction of apoptosis and inhibition of oxidative stress, inflammation and angiogenesis [143,161,162,163]. Given the fact that Mel receptors are expressed in bone marrow-derived MSCs, melatonin also exerted various receptor-mediated effects on MSCs including enhancement their survival, motility, engraftment and cell differentiation which seems linked to receptors/matrix enzymes interaction, and therefore higher homing effects of MSCs followed pre-administration of a combination of melatonin with MSCs [163,164,165]. On the other hand, various processes have been postulated in relation to MSC-dependent tumor suppression. In this regard, MSCs pulsed with tumor-derived microvesicles showed an enhanced antitumor activity in HCC [166]. Some previous report revealed the use of melatonin enhanced the potential therapeutic role MSCs in treatment various diseases such as acute kidney injury, metabolic syndromes including diabetes through various mechanisms that include through the activation of antioxidative pathways, inhibition of the inflammatory response and reduction of apoptosis and fibrosis [19,167,168,169,170]. More interestingly, few recent reports revealed the beneficial effects of the combination between melatonin and MSCs on targeting inflammation in HCC [171,172,173,174,175]. However, it should be stressed that there is a clear shortage in the available data about the combined use of melatonin and MSCs and their possible synergistic effects in treatment of HCC. In this concern, some of these reports favor the administration of melatonin before MSCs transplantation and it seems this method offer many advantages over the single use of either factor [172]. Interestingly, pre-administration of melatonin prior to MSCs transplantation in HCC resulted in series of actions include, promoted the homing potential of bone marrow-derived MSCs (BMMSCs) and decrease the carcinogenic effect induced by diethylnitrosamine (DEN) which is known as a potent liver carcinogen in rats [176]. Moreover, a significant decrease in the following parameters has been reported, proliferating cell nuclear antigen (PCNA) index, glutathione S-transferase placental positive foci (GST-P), tumor biomarkers in serum besides reduction of inflammation, angiogenesis and metastasis in HCC [171,172,176]. On the other hand, this combination resulted in induction of apoptosis and antioxidant enzymes, tissue matrix and liver repairs in HCC [171]. This was evidenced by lower level of apoptosis in liver tissues which indicated by marked increase in levels of PCNA immunoreactivity and decrease in levels of fragmented DNA and expression of p53, caspase 9 and caspase 3 genes [171,172,176]. Similarly, another study was carried out by Basyony et al. (2019) revealed that treatment using a combination of melatonin and MSCs were reported to decrease malondialdehyde (MDA) which is a known marker of oxidative stress and the antioxidant status in cancer [176]. In addition, in the same study, this combination increases the superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPX) combined with attenuation of PCNA, Bcl2 and programmed death ligand 1 (PD-L1) immunostain markers and down regulate the expression of inflammation and cell proliferation genes [171,176].
Taken together, the combined use of melatonin and MSCs may provide promising beneficial effects via triggering the apoptosis resistance and as consequences target HCC Figure 3. It should be stressed that this action was confirmed in rat models where the combined treatment restored the liver function and decreased the HCC versus the treatment with either factor alone or in combination with preconditioning in HCC rats [171,172,176]. Given the lack of available data in this topic, further research seems mandatory to explore more about the effects of this combination together with investigation the main mechanisms underlying the reported actions for better understanding the potential use of targeted stems cell therapy in treatment of HCC.

5. Conclusions

In conclusion, the present review highlights the main biologic, physiological and therapeutic effects of melatonin that could be very beneficial for controlling of HCC. Furthermore, an overview about MSCs was given with explaining their potential roles in controlling of HCC and their recruitments and suggested actions. It should be stressed that there is a shortage in the available data that explore the effects of the combined use of melatonin and MSCs-based treatments. The present review suggests further future research for exploring the possible beneficial effects of the combination of MSCs and melatonin in treatment of HCC, together with exploring the role of receptors in this possible underlying activity. We hope that this information may contribute to develop novel drug targets with anticancer activity.

Author Contributions

Y.M. and E.K.E. designed the idea of the review, drafted the manuscript and submitted the manuscript to the journal. W.A. and T.Y. involved in the conception of the idea of the manuscript and contributed his scientific advice and revision of the manuscript. The manuscript was read and approved by all the authors. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

The authors would like to thank Mahmoud Abbas for his technical support and help.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

AKTprotein kinase B mybl1 MYB proto-oncogene like 1
cAMPcyclic adenosine monophosphate NF-kB nuclear factor kappa B
COX-2cyclooxygenase-2 NK natural killer cells
IFN-γinterferon gamma PI3 K phosphoinositide 3-kinase
IL-1βinterleukin 1 beta rasa1 ras GTPase activating protein
Kcnn4potassium calcium-activated channel subfamily N member 4 ROR related orphan receptor
MAPK Amitogen-activated protein kinase member 4 TLR Toll-like receptors
Mllt3protein AF-9 STAT3 Signal transducer and activator of transcription 3
mt-Co1mitochondrially encoded cytochrome c oxidase I TGF-β Transforming growth factor beta
mt-Co3mitochondrially encoded cytochrome c oxidase 3 TNF-α tumor necrosis factor-alpha
mt-Nd1mitochondrially NADH-ubiquinone oxidoreductase chain 1
mt-RNr2mitochondrially encoded 16S RNA TRAIL TNF-related apoptosis-inducing ligand
XIAPX-linked inhibitor of apoptosis protein

References

  1. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Howlader, M.; Noone, A.; Krapcho, M.; Miller, K.D.; Brest, A.; Yu, M.C.; Ruhl, J.; Tatalovich, Z.; Mariotto, A.; Lewis, D.R.; et al. SEER Cancer Statistics Review, 1975–2016; National Cancer Institute: Bethesda, MD, USA, 2019. Available online: https://seer.cancer.gov/csr/1975_2016/ (accessed on 15 June 2020).
  3. Rawla, P.; Sunkara, T.; Muralidharan, P.; Raj, J.P. Update in global trends and aetiology of hepatocellular carcinoma. Contemp. Oncol. (Pozn.) 2018, 22, 141–150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. European Association for the Study of the Liver. EASL–EORTC clinical practice guidelines: Management of hepatocellular carcinoma. J. Hepatol. 2012, 56, 908–943. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Forner, A.; Llovet, J.M.; Bruix, J. Hepatocellular carcinoma. Lancet 2012, 379, 1245–1255. [Google Scholar] [CrossRef]
  6. Yang, J.D.; Hainaut, P.; Gores, G.J.; Amadou, A.; Plymoth, A.; Roberts, L.R. A global view of hepatocellular carcinoma: Trends, risk, prevention and management. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 589–604. [Google Scholar] [CrossRef]
  7. Chen, C.; Lou, T. Hypoxia inducible factors in hepatocellular carcinoma. Oncotarget 2017, 8, 46691–46703. [Google Scholar] [CrossRef] [Green Version]
  8. Kumar, V.; Abbas, A.K.; Aster, J.C. Robbins and Cotran Pathologic Basis of Disease, 9th ed.; Elsevier: Amsterdam, The Netherlands, 2014; p. 1391. [Google Scholar]
  9. Fan, L.; Song, B.; Sun, G.; Ma, T.; Zhong, F.; Wei, W. Endoplasmic reticulum stress-induced resistance to doxorubicin is reversed by paeonol treatment in human hepatocellular carcinoma cells. PLoS ONE 2013, 8, e62627. [Google Scholar] [CrossRef]
  10. Augello, C.; Caruso, L.; Maggioni, M.; Donadon, M.; Montorsi, M.; Santambrogio, R.; Torzilli, G.; Vaira, V.; Pellegrini, C.; Roncalli, M.; et al. Inhibitors of apoptosis proteins (IAPs) expression and their prognostic significance in hepatocellular carcinoma. BMC Cancer 2009, 9, 125. [Google Scholar] [CrossRef]
  11. Straub, C.S. Targeting IAPs as an approach to anti-cancer therapy. Curr. Top. Med. Chem. 2011, 11, 291–316. [Google Scholar] [CrossRef] [PubMed]
  12. Finati, E. Melatonin: A Pleiotropic Molecule of Natural Origin. Evaluation of the Different Therapeutic Activities in Animal Models and/or Human Patients and a Study of the Metabolic-Biochemical Pathways Related to Them. Ph.D.Thesis, Università Degli Studi Di, Milano, Italy, 2013. [Google Scholar]
  13. Hernanda, P.Y.; Pedroza-Gonzalez, A.; Sprengers, D.; Peppelenbosch, M.P.; Pan, Q. Multipotent mesenchymal stromal cells in liver cancer: Implications for tumor biology and therapy. Biochim. Biophys. Acta 2014, 1846, 439–445. [Google Scholar] [CrossRef]
  14. Montemurro, T.; Vigano, M.; Parazzi, V.; Baluce, B.; Lavazza, C.; Budelli, S.; Montelatici, E. Not all the stem cells meet all the clinical needs: Mesenchymal stem cells in regenerative medicine. Cytotherapy 2014, 16, S90. [Google Scholar] [CrossRef]
  15. Hsiao, S.T.; Asgari, A.; Lokmic, Z.; Sinclair, R.; Dusting, G.J.; Lim, S.Y.; Dilley, R.J. Comparative analysis of paracrine factor expression in human adult mesenchymal stem cells derived from bone marrow, adipose, and dermal tissue. Stem Cells Dev. 2012, 21, 2189–2203. [Google Scholar] [CrossRef] [Green Version]
  16. Da Silva Meirelles, L.; Caplan, A.I.; Nardi, N.B. In search of the in vivo identity of mesenchymal stem cells. Stem Cells 2008, 26, 2287–2299. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Hu, X.; Garcia, M.; Weng, L.; Jung, X.; Murakami, J.L.; Kumar, B.; Warden, C.D.; Todorov, I.; Chen, C.C. Identification of a common mesenchymal stromal progenitor for the adult haematopoietic niche. Nat. Commun. 2016, 7, 13095. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Webb, T.L.; Quimby, J.M.; Dow, S.W. In vitro comparison of feline bone marrow-derived and adipose tissue-derived mesenchymal stem cells. J. Feline Med. Surg. 2012, 14, 165–168. [Google Scholar] [CrossRef]
  19. Hu, C.; Li, L. Melatonin plays critical role in mesenchymal stem cell-based regenerative medicine in vitro and in vivo. Stem Cell Res. Ther. 2019, 10, 13. [Google Scholar] [CrossRef] [Green Version]
  20. Rodriguez-Lozano, F.J.; Garcia-Bernal, D.; Ros-Roca Mde, L.; Alguero Mdel, C.; Onate-Sanchez, R.E.; Camacho-Alonso, F.; Moraleda, J.M. Cytoprotective effects of melatonin on zoledronic acid-treated human mesenchymal stem cells in vitro. J. Cranio-Maxillofac. Surg. 2015, 43, 855–862. [Google Scholar] [CrossRef] [PubMed]
  21. Orbay, H.; Tobita, M.; Mizuno, H. Mesenchymal stem cells isolated from adipose and other tissues: Basic biological properties and clinical applications. Stem Cells Int. 2012, 2012, 461718. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Han, Y.; Li, X.; Zhang, Y.; Chang, F.; Ding, J. Mesenchymal Stem Cells for Regenerative Medicine. Cells 2019, 8, 886. [Google Scholar] [CrossRef] [Green Version]
  23. Ayala-Cuellar, A.P.; Kang, J.H.; Jeung, E.B.; Choi, K.C. Roles of Mesenchymal Stem Cells in Tissue Regeneration and Immunomodulation. Biomol. Ther. (Seoul) 2019, 27, 25–33. [Google Scholar] [CrossRef]
  24. Mushahary, D.; Spittler, A.; Kasper, C.; Weber, V.; Charwat, V. Isolation, cultivation, and characterization of human mesenchymal stem cells. Cytom. Part A 2018, 93, 19–31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Gawronska-Kozak, B.; Manuel, J.A.; Prpic, V. Ear mesenchymal stem cells (EMSC) can differentiate into spontaneously contracting muscle cells. J. Cell Biochem. 2007, 102, 122–135. [Google Scholar] [CrossRef] [PubMed]
  26. Rim, J.S.; Mynatt, R.L.; Gawronska-Kozak, B. Mesenchymal stem cells from the outer ear: A novel adult stem cell model system for the study of adipogenesis. FASEB J. 2005, 19, 1205–1207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Al-Nbaheen, M.; Vishnubalaji, R.; Ali, D.; Bouslimi, A.; Al-Jassir, F.; Megges, M.; Prigione, A.; Adjaye, J.; Kassem, M.; Aldahmash, A. Human stromal (mesenchymal) stem cells from bone marrow, adipose tissue and skin exhibit differences in molecular phenotype and differentiation potential. Stem Cell Rev. Rep. 2013, 9, 32–43. [Google Scholar] [CrossRef] [Green Version]
  28. Gurung, S.; Deane, J.A.; Masuda, H.; Maruyama, T.; Gargett, C.E. Stem Cells in Endometrial Physiology. Semin. Reprod. Med. 2015, 33, 326–332. [Google Scholar] [CrossRef]
  29. Lv, F.J.; Tuan, R.S.; Cheung, K.M.; Leung, V.Y. Concise review: The surface markers and identity of human mesenchymal stem cells. Stem Cells 2014, 32, 1408–1419. [Google Scholar] [CrossRef]
  30. Liu, S.; Hou, K.D.; Yuan, M.; Peng, J.; Zhang, L.; Sui, X.; Zhao, B.; Xu, W.; Wang, A.; Lu, S.; et al. Characteristics of mesenchymal stem cells derived from Wharton’s jelly of human umbilical cord and for fabrication of non-scaffold tissue-engineered cartilage. J. Biosci. Bioeng. 2014, 117, 229–235. [Google Scholar] [CrossRef]
  31. Ramos, T.L.; Sanchez-Abarca, L.I.; Muntion, S.; Preciado, S.; Puig, N.; Lopez-Ruano, G.; Hernandez-Hernandez, A.; Redondo, A.; Ortega, R.; Rodriguez, C.; et al. MSC surface markers (CD44, CD73, and CD90) can identify human MSC-derived extracellular vesicles by conventional flow cytometry. Cell Commun. Signal. 2016, 14, 2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Mabuchi, Y.; Houlihan, D.D.; Akazawa, C.; Okano, H.; Matsuzaki, Y. Prospective isolation of murine and human bone marrow mesenchymal stem cells based on surface markers. Stem Cells Int. 2013, 2013, 507301. [Google Scholar] [CrossRef] [Green Version]
  33. Fitter, S.; Gronthos, S.; Ooi, S.S.; Zannettino, A.C. The Mesenchymal Precursor Cell Marker Antibody STRO-1 Binds to Cell Surface Heat Shock Cognate 70. Stem Cells 2017, 35, 940–951. [Google Scholar] [CrossRef] [Green Version]
  34. Lechanteur, C.; Briquet, A.; Giet, O.; Delloye, O.; Baudoux, E.; Beguin, Y. Clinical-scale expansion of mesenchymal stromal cells: A large banking experience. J. Transl. Med. 2016, 14, 145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Karp, J.M.; Leng Teo, G.S. Mesenchymal stem cell homing: The devil is in the details. Cell Stem Cell 2009, 4, 206–216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Isobe, Y.; Koyama, N.; Nakao, K.; Osawa, K.; Ikeno, M.; Yamanaka, S.; Okubo, Y.; Fujimura, K.; Bessho, K. Comparison of human mesenchymal stem cells derived from bone marrow, synovial fluid, adult dental pulp, and exfoliated deciduous tooth pulp. Int. J. Oral. Maxillofac. Surg. 2016, 45, 124–131. [Google Scholar] [CrossRef] [PubMed]
  37. Mastrolia, I.; Foppiani, E.M.; Murgia, A.; Candini, O.; Samarelli, A.V.; Grisendi, G.; Veronesi, E.; Horwitz, E.M.; Dominici, M. Challenges in Clinical Development of Mesenchymal Stromal/Stem Cells: Concise Review. Stem Cells Transl. Med. 2019, 8, 1135–1148. [Google Scholar] [CrossRef] [Green Version]
  38. Neri, S. Genetic Stability of Mesenchymal Stromal Cells for Regenerative Medicine Applications: A Fundamental Biosafety Aspect. Int. J. Mol. Sci. 2019, 20, 2406. [Google Scholar] [CrossRef] [Green Version]
  39. Giuliani, M.; Oudrhiri, N.; Noman, M.Z.; Vernochet, A.; Chouaib, S.; Azzarone, B.; Durrbach, A.; Bennaceur-Griscelli, A. Human mesenchymal stem cells derived from induced pluripotent stem cells down-regulate NK-cell cytolytic machinery. Blood 2011, 118, 3254–3262. [Google Scholar] [CrossRef]
  40. Schweizer, M.T.; Wang, H.; Bivalacqua, T.J.; Partin, A.W.; Lim, S.J.; Chapman, C.; Abdallah, R.; Levy, O.; Bhowmick, N.A.; Karp, J.M.; et al. A Phase I Study to Assess the Safety and Cancer-Homing Ability of Allogeneic Bone Marrow-Derived Mesenchymal Stem Cells in Men with Localized Prostate Cancer. Stem Cells Transl. Med. 2019, 8, 441–449. [Google Scholar] [CrossRef] [Green Version]
  41. Galipeau, J.; Sensebe, L. Mesenchymal Stromal Cells: Clinical Challenges and Therapeutic Opportunities. Cell Stem Cell 2018, 22, 824–833. [Google Scholar] [CrossRef] [Green Version]
  42. Heissig, B.; Dhahri, D.; Eiamboonsert, S.; Salama, Y.; Shimazu, H.; Munakata, S.; Hattori, K. Role of mesenchymal stem cell-derived fibrinolytic factor in tissue regeneration and cancer progression. Cell Mol. Life Sci. 2015, 72, 4759–4770. [Google Scholar] [CrossRef]
  43. Lee, J.W.; Krasnodembskaya, A.; McKenna, D.H.; Song, Y.; Abbott, J.; Matthay, M.A. Therapeutic effects of human mesenchymal stem cells in ex vivo human lungs injured with live bacteria. Am. J. Respir. Crit. Care Med. 2013, 187, 751–760. [Google Scholar] [CrossRef] [Green Version]
  44. Wen, S.; Dooner, M.; Cheng, Y.; Papa, E.; Del Tatto, M.; Pereira, M.; Deng, Y.; Goldberg, L.; Aliotta, J.; Chatterjee, D.; et al. Mesenchymal stromal cell-derived extracellular vesicles rescue radiation damage to murine marrow hematopoietic cells. Leukemia 2016, 30, 2221–2231. [Google Scholar] [CrossRef] [PubMed]
  45. Mantovani, A.; Biswas, S.K.; Galdiero, M.R.; Sica, A.; Locati, M. Macrophage plasticity and polarization in tissue repair and remodelling. J. Pathol. 2013, 229, 176–185. [Google Scholar] [CrossRef]
  46. Lichtman, M.K.; Otero-Vinas, M.; Falanga, V. Transforming growth factor beta (TGF-beta) isoforms in wound healing and fibrosis. Wound Repair Regen 2016, 24, 215–222. [Google Scholar] [CrossRef] [PubMed]
  47. Bharti, R.; Dey, G.; Mandal, M. Cancer development, chemoresistance, epithelial to mesenchymal transition and stem cells: A snapshot of IL-6 mediated involvement. Cancer Lett. 2016, 375, 51–61. [Google Scholar] [CrossRef] [PubMed]
  48. Piktel, E.; Niemirowicz, K.; Wnorowska, U.; Watek, M.; Wollny, T.; Gluszek, K.; Gozdz, S.; Levental, I.; Bucki, R. The Role of Cathelicidin LL-37 in Cancer Development. Arch Immunol. Exp. (Warsz.) 2016, 64, 33–46. [Google Scholar] [CrossRef] [Green Version]
  49. Roccaro, A.M.; Sacco, A.; Purschke, W.G.; Moschetta, M.; Buchner, K.; Maasch, C.; Zboralski, D.; Zollner, S.; Vonhoff, S.; Mishima, Y.; et al. SDF-1 inhibition targets the bone marrow niche for cancer therapy. Cell Rep. 2014, 9, 118–128. [Google Scholar] [CrossRef] [Green Version]
  50. Xu, Y.; Chu, N.; Qiu, X.; Gober, H.J.; Li, D.; Wang, L. The interconnected role of chemokines and estrogen in bone metabolism. Biosci. Trends 2017, 10, 433–444. [Google Scholar] [CrossRef] [Green Version]
  51. Geranmayeh, M.H.; Nourazarian, A.; Avci, C.B.; Rahbarghazi, R.; Farhoudi, M. Stem Cells as a Promising Tool for the Restoration of Brain Neurovascular Unit and Angiogenic Orientation. Mol. Neurobiol. 2017, 54, 7689–7705. [Google Scholar] [CrossRef]
  52. Montano-Loza, A.J.; Bhanji, R.A.; Wasilenko, S.; Mason, A.L. Systematic review: Recurrent autoimmune liver diseases after liver transplantation. Aliment Pharm. Ther. 2017, 45, 485–500. [Google Scholar] [CrossRef]
  53. Frese, L.; Dijkman, P.E.; Hoerstrup, S.P. Adipose Tissue-Derived Stem Cells in Regenerative Medicine. Transfus. Med. Hemother. 2016, 43, 268–274. [Google Scholar] [CrossRef]
  54. Holly, J.M.P.; Biernacka, K.; Perks, C.M. The Neglected Insulin: IGF-II, a Metabolic Regulator with Implications for Diabetes, Obesity, and Cancer. Cells 2019, 8, 1207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Deng, S.Z.; Sun, K.; Wang, R.; Wang, J. and Lin, Y. Fundamental Concepts and Features of Mesenchymal Stem Cells: Proliferation, Differentiation, Migration and Immunomodulatory Characteristics. Mesenchymal Stem Cells Craniofacial Regen. 2016, 3, 2896–2902. [Google Scholar]
  56. Osiecki, M.J. Isolation and Expansion of Placental Derived Mesenchymal Stromal Cells in a Packed Bed Bioreactor. Ph.D. Thesis, Queensland University of Technology, Brisbane City, Australia, 2016. [Google Scholar]
  57. Zang, W.; Xie, L.H.; Zhu, B.H.; Cui, D.W. Construction of human HepG-2 cells infected by lentivirus carrying green fluorescent protein gene. Int. J. Clin. Exp. Med. 2016, 9, 8161–8168. [Google Scholar]
  58. Quail, D.F.; Joyce, J.A. Microenvironmental regulation of tumor progression and metastasis. Nat. Med. 2013, 19, 1423–1437. [Google Scholar] [CrossRef] [PubMed]
  59. Hill, B.S.; Sarnella, A.; D’Avino, G.; Zannetti, A. Recruitment of stromal cells into tumour microenvironment promote the metastatic spread of breast cancer. Semin. Cancer Biol. 2019. [Google Scholar] [CrossRef]
  60. Ai, J.; Ketabchi, N.; Verdi, J.; Gheibi, N.; Khadem Haghighian, H.; Kavianpour, M. Mesenchymal stromal cells induce inhibitory effects on hepatocellular carcinoma through various signaling pathways. Cancer Cell Int. 2019, 19, 329. [Google Scholar] [CrossRef] [Green Version]
  61. Chowdhury, R. Prostate Cancer Exosomes Differentiate BM-MSCs into Pro-Angiogenic and Pro-Invasive Myofibroblasts. Ph.D. Thesis, Cardiff University, Cardiff, UK, 2015. [Google Scholar]
  62. Yuan, Y.; Zhou, C.; Chen, X.; Tao, C.; Cheng, H.; Lu, X. Suppression of tumor cell proliferation and migration by human umbilical cord mesenchymal stem cells: A possible role for apoptosis and Wnt signaling. Oncol. Lett. 2018, 15, 8536–8544. [Google Scholar] [CrossRef]
  63. Parys, M.; Nelson, N.; Koehl, K.; Miller, R.; Kaneene, J.B.; Kruger, J.M.; Yuzbasiyan-Gurkan, V. Safety of Intraperitoneal Injection of Adipose Tissue-Derived Autologous Mesenchymal Stem Cells in Cats. J. Vet. Intern. Med. 2016, 30, 157–163. [Google Scholar] [CrossRef] [Green Version]
  64. Marks, D.L.; Olson, R.L.; Fernandez-Zapico, M.E. Epigenetic control of the tumor microenvironment. Epigenomics 2016, 8, 1671–1687. [Google Scholar] [CrossRef] [Green Version]
  65. Jimenez-Puerta, G.J.; Marchal, J.A.; Lopez-Ruiz, E.; Galvez-Martin, P. Role of Mesenchymal Stromal Cells as Therapeutic Agents: Potential Mechanisms of Action and Implications in Their Clinical Use. J. Clin. Med. 2020, 9, 445. [Google Scholar] [CrossRef] [Green Version]
  66. Park, K.S.; Bandeira, E.; Shelke, G.V.; Lasser, C.; Lotvall, J. Enhancement of therapeutic potential of mesenchymal stem cell-derived extracellular vesicles. Stem Cell Res. Ther. 2019, 10, 288. [Google Scholar] [CrossRef]
  67. Park, W.S.; Ahn, S.Y.; Sung, S.I.; Ahn, J.Y.; Chang, Y.S. Strategies to enhance paracrine potency of transplanted mesenchymal stem cells in intractable neonatal disorders. Pediatric Res. 2018, 83, 214–222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Maacha, S.; Sidahmed, H.; Jacob, S.; Gentilcore, G.; Calzone, R.; Grivel, J.C.; Cugno, C. Paracrine Mechanisms of Mesenchymal Stromal Cells in Angiogenesis. Stem Cells Int. 2020, 2020, 4356359. [Google Scholar] [CrossRef] [PubMed]
  69. Fan, X.L.; Zhang, Y.; Li, X.; Fu, Q.L. Mechanisms underlying the protective effects of mesenchymal stem cell-based therapy. Cell Mol. Life Sci. 2020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Siu, C.W.; Liao, S.Y.; Liu, Y.; Lian, Q.; Tse, H.F. Stem cells for myocardial repair. Thromb. Haemost. 2010, 104, 6–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Zhang, Y.; Chiu, S.; Liang, X.; Gao, F.; Zhang, Z.; Liao, S.; Liang, Y.; Chai, Y.H.; Low, D.J.; Tse, H.F.; et al. Rap1-mediated nuclear factor-kappaB (NF-kappaB) activity regulates the paracrine capacity of mesenchymal stem cells in heart repair following infarction. Cell Death Discov. 2015, 1, 15007. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Zong, C.; Zhang, H.; Yang, X.; Gao, L.; Hou, J.; Ye, F.; Jiang, J.; Yang, Y.; Li, R.; Han, Z.; et al. The distinct roles of mesenchymal stem cells in the initial and progressive stage of hepatocarcinoma. Cell Death Dis. 2018, 9, 345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Li, C.; Cheung, M.K.H.; Han, S.; Zhang, Z.; Chen, L.; Chen, J.; Zeng, H.; Qiu, J. Mesenchymal stem cells and their mitochondrial transfer: A double-edged sword. Biosci. Rep. 2019, 39. [Google Scholar] [CrossRef] [Green Version]
  74. Kholodenko, I.V.; Kurbatov, L.K.; Kholodenko, R.V.; Manukyan, G.V.; Yarygin, K.N. Mesenchymal Stem Cells in the Adult Human Liver: Hype or Hope? Cells 2019, 8, 1127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Ogunwobi, O.O.; Harricharran, T.; Huaman, J.; Galuza, A.; Odumuwagun, O.; Tan, Y.; Ma, G.X.; Nguyen, M.T. Mechanisms of hepatocellular carcinoma progression. World J. Gastroenterol. 2019, 25, 2279–2293. [Google Scholar] [CrossRef]
  76. Auger, C.; Alhasawi, A.; Contavadoo, M.; Appanna, V.D. Dysfunctional mitochondrial bioenergetics and the pathogenesis of hepatic disorders. Front. Cell Dev. Biol. 2015, 3, 40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Herst, P.M.; Rowe, M.R.; Carson, G.M.; Berridge, M.V. Functional Mitochondria in Health and Disease. Front. Endocrinol. (Lausanne) 2017, 8, 296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Zimmermann, A. Mitochondrial Biology in Hepatobiliary Tumors: Changes of the Cellular Energy Factory. In Tumors and Tumor-Like Lesions of the Hepatobiliary Tract: General and Surgical Pathology; Springer International Publishing: Cham, Switzerland, 2017; pp. 3091–3124. [Google Scholar]
  79. Léveillé, M.; Estall, J. Mitochondrial Dysfunction in the Transition from NASH to HCC. Metabolites 2019, 9, 233. [Google Scholar] [CrossRef] [Green Version]
  80. Vignais, M.L.; Caicedo, A.; Brondello, J.M.; Jorgensen, C. Cell Connections by Tunneling Nanotubes: Effects of Mitochondrial Trafficking on Target Cell Metabolism, Homeostasis, and Response to Therapy. Stem Cells Int. 2017, 2017, 6917941. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Court, A.C.; Le-Gatt, A.; Luz-Crawford, P.; Parra, E.; Aliaga-Tobar, V.; Batiz, L.F.; Contreras, R.A.; Ortuzar, M.I.; Kurte, M.; Elizondo-Vega, R.; et al. Mitochondrial transfer from MSCs to T cells induces Treg differentiation and restricts inflammatory response. EMBO Rep. 2020, 21, e48052. [Google Scholar] [CrossRef] [PubMed]
  82. Lin, W.; Huang, L.; Li, Y.; Fang, B.; Li, G.; Chen, L.; Xu, L. Mesenchymal Stem Cells and Cancer: Clinical Challenges and Opportunities. BioMed Res. Int. 2019, 2019, 2820853. [Google Scholar] [CrossRef] [Green Version]
  83. Powell, A.B.; Williams, K.; Cruz, C.R.Y. Gene-modified, cell-based therapies-an overview. Cytotherapy 2016, 18, 1351–1359. [Google Scholar] [CrossRef]
  84. Woodsworth, D.J.; Holt, R.A. Cell-Based Therapeutics: Making a Faustian Pact with Biology. Trends Mol. Med. 2017, 23, 104–115. [Google Scholar] [CrossRef]
  85. Aurelian, L. Oncolytic viruses as immunotherapy: Progress and remaining challenges. Onco. Targets Ther. 2016, 9, 2627–2637. [Google Scholar] [CrossRef] [Green Version]
  86. Kim, N.; Cho, S.G. New strategies for overcoming limitations of mesenchymal stem cell-based immune modulation. Int. J. Stem Cells 2015, 8, 54–68. [Google Scholar] [CrossRef] [Green Version]
  87. Nowakowski, A.; Drela, K.; Rozycka, J.; Janowski, M.; Lukomska, B. Engineered Mesenchymal Stem Cells as an Anti-Cancer Trojan Horse. Stem Cells Dev. 2016, 25, 1513–1531. [Google Scholar] [CrossRef] [PubMed]
  88. Hmadcha, A.; Martin-Montalvo, A.; Gauthier, B.R.; Soria, B.; Capilla-Gonzalez, V. Therapeutic Potential of Mesenchymal Stem Cells for Cancer Therapy. Front. Bioeng. Biotechnol. 2020, 8, 43. [Google Scholar] [CrossRef] [PubMed]
  89. Zhao, L.; Cao, Y.J. Engineered T Cell Therapy for Cancer in the Clinic. Front. Immunol. 2019, 10, 2250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Melief, C.J.; van Hall, T.; Arens, R.; Ossendorp, F.; van der Burg, S.H. Therapeutic cancer vaccines. J. Clin. Investig. 2015, 125, 3401–3412. [Google Scholar] [CrossRef]
  91. Brauer, P.M.; Singh, J.; Xhiku, S.; Zuniga-Pflucker, J.C. T Cell Genesis: In Vitro Veritas Est? Trends Immunol. 2016, 37, 889–901. [Google Scholar] [CrossRef] [Green Version]
  92. Niess, H.; Thomas, M.N.; Schiergens, T.S.; Kleespies, A.; Jauch, K.W.; Bruns, C.; Werner, J.; Nelson, P.J.; Angele, M.K. Genetic engineering of mesenchymal stromal cells for cancer therapy: Turning partners in crime into Trojan horses. Innov. Surg. Sci. 2016, 1, 19–32. [Google Scholar] [CrossRef]
  93. Hardeland, R. Melatonin in Plants-Diversity of Levels and Multiplicity of Functions. Front. Plant Sci. 2016, 7, 198. [Google Scholar] [CrossRef]
  94. Tenorio, F.; Simoes Mde, J.; Teixeira, V.W.; Teixeira, A.A. Effects of melatonin and prolactin in reproduction: Review of literature. Rev. Assoc. Med. Bras. 2015, 61, 269–274. [Google Scholar] [CrossRef] [Green Version]
  95. Malpaux, B.; Migaud, M.; Tricoire, H.; Chemineau, P. Biology of mammalian photoperiodism and the critical role of the pineal gland and melatonin. J. Biol. Rhythm. 2001, 16, 336–347. [Google Scholar] [CrossRef]
  96. Sanchez, A.; Calpena, A.C.; Clares, B. Evaluating the Oxidative Stress in Inflammation: Role of Melatonin. Int. J. Mol. Sci. 2015, 16, 16981–17004. [Google Scholar] [CrossRef] [Green Version]
  97. Hardeland, R.; Cardinali, D.P.; Srinivasan, V.; Spence, D.W.; Brown, G.M.; Pandi-Perumal, S.R. Melatonin—A pleiotropic, orchestrating regulator molecule. Prog. Neurobiol. 2011, 93, 350–384. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Brzęczek, M.S.K.; Hyla-Klekot, L. Melatonina–hormon plejotropowym działaniu. Pediatr. Med. Rodz. 2016, 12, 127–133. [Google Scholar] [CrossRef]
  99. Xin, Z.; Jiang, S.; Jiang, P.; Yan, X.; Fan, C.; Di, S.; Wu, G.; Yang, Y.; Reiter, R.J.; Ji, G. Melatonin as a treatment for gastrointestinal cancer: A review. J. Pineal Res. 2015, 58, 375–387. [Google Scholar] [CrossRef] [Green Version]
  100. Galano, A. The role of indoleamines in reducing free radical damage and oxidative stress: A physicochemical perspective. In Indoleamines: Sources, Role in Biological Processes and Health Effects; Nova Science Publishers, Inc.: Hauppauge, NY, USA, 2015; pp. 1–41. [Google Scholar]
  101. Wongprayoon, P.; Govitrapong, P. Melatonin Attenuates Methamphetamine-Induced Neurotoxicity. Curr. Pharm. Des. 2016, 22, 1022–1032. [Google Scholar] [CrossRef]
  102. Reppert, S.M.; Weaver, D.R.; Godson, C. Melatonin receptors step into the light: Cloning and classification of subtypes. Trends Pharm. Sci. 1996, 17, 100–102. [Google Scholar] [CrossRef]
  103. Zee, P.C.; Reid, K.J. 35 Melatonin in Sleep-Wake Regulation. In Diagnosis and Treatment; CRC Press: Boca Raton, FL, USA, 2016; p. 410. [Google Scholar]
  104. Mektepbayeva, D.; Alibek, K.; Atinbayeva, N.; Irving, S.; Zhaisanbayeva, B.; Mussurova, S.; Mussakhan, S. Anticancer Effects and uses of Melatonin: A Review. Austin J. Cancer Clin. Res. 2015, 2, 1052. [Google Scholar]
  105. Boutin, J.A. Melatonin binding site MT3 is QR2: State of the art. J. Soc. Biol. 2007, 201, 97–103. [Google Scholar] [CrossRef]
  106. Goncalves Ndo, N.; Colombo, J.; Lopes, J.R.; Gelaleti, G.B.; Moschetta, M.G.; Sonehara, N.M.; Hellmen, E.; Zanon Cde, F.; Oliani, S.M.; Zuccari, D.A. Effect of Melatonin in Epithelial Mesenchymal Transition Markers and Invasive Properties of Breast Cancer Stem Cells of Canine and Human Cell Lines. PLoS ONE 2016, 11, e0150407. [Google Scholar] [CrossRef] [Green Version]
  107. Panzer, A.; Viljoen, M. The validity of melatonin as an oncostatic agent. J. Pineal Res. 1997, 22, 184–202. [Google Scholar] [CrossRef] [Green Version]
  108. Pandi-Perumal, S.R.; Trakht, I.; Srinivasan, V.; Spence, D.W.; Maestroni, G.J.; Zisapel, N.; Cardinali, D.P. Physiological effects of melatonin: Role of melatonin receptors and signal transduction pathways. Prog. Neurobiol. 2008, 85, 335–353. [Google Scholar] [CrossRef]
  109. Cutando, A.; Aneiros-Fernandez, J.; Lopez-Valverde, A.; Arias-Santiago, S.; Aneiros-Cachaza, J.; Reiter, R.J. A new perspective in Oral health: Potential importance and actions of melatonin receptors MT1, MT2, MT3, and RZR/ROR in the oral cavity. Arch Oral. Biol. 2011, 56, 944–950. [Google Scholar] [CrossRef] [PubMed]
  110. Garcia-Maurino, S.; Pozo, D.; Calvo, J.R.; Guerrero, J.M. Correlation between nuclear melatonin receptor expression and enhanced cytokine production in human lymphocytic and monocytic cell lines. J. Pineal Res. 2000, 29, 129–137. [Google Scholar] [CrossRef] [PubMed]
  111. Tuli, H.S.; Kashyap, D.; Sharma, A.K.; Sandhu, S.S. Molecular aspects of melatonin (MLT)-mediated therapeutic effects. Life Sci. 2015, 135, 147–157. [Google Scholar] [CrossRef] [PubMed]
  112. Benitez-King, G.; Anton-Tay, F. Calmodulin mediates melatonin cytoskeletal effects. Experientia 1993, 49, 635–641. [Google Scholar] [CrossRef]
  113. Hardeland, R. Melatonin: Signaling mechanisms of a pleiotropic agent. Biofactors 2009, 35, 183–192. [Google Scholar] [CrossRef]
  114. Srinivasan, V.; RPandi-Perumal, S.; Brzezinski, A.; PBhatnagar, K.; PCardinali, D. Melatonin, immune function and cancer. Recent Pat. Endocr. Metab. Immune Drug Discov. 2011, 5, 109–123. [Google Scholar] [CrossRef] [Green Version]
  115. Gupta, B.B.; Spessert, R.; Vollrath, L. Molecular components and mechanism of adrenergic signal transduction in mammalian pineal gland: Regulation of melatonin synthesis. Indian J. Exp. Biol. 2005, 43, 115–149. [Google Scholar]
  116. Dubocovich, M.L.; Yun, K.; Al-Ghoul, W.M.; Benloucif, S.; Masana, M.I. Selective MT2 melatonin receptor antagonists block melatonin-mediated phase advances of circadian rhythms. FASEB J. 1998, 12, 1211–1220. [Google Scholar] [CrossRef] [Green Version]
  117. Fischer, T.W.; Slominski, A.; Zmijewski, M.A.; Reiter, R.J.; Paus, R. Melatonin as a major skin protectant: From free radical scavenging to DNA damage repair. Exp. Derm. 2008, 17, 713–730. [Google Scholar] [CrossRef]
  118. Loureiro, R.; Magalhaes-Novais, S.; Mesquita, K.A.; Baldeiras, I.; Sousa, I.S.; Tavares, L.C.; Barbosa, I.A.; Oliveira, P.J.; Vega-Naredo, I. Melatonin antiproliferative effects require active mitochondrial function in embryonal carcinoma cells. Oncotarget 2015, 6, 17081–17096. [Google Scholar] [CrossRef] [Green Version]
  119. Lopez-Munoz, F.; Molina, J.D.; Rubio, G.; Alamo, C. An historical view of the pineal gland and mental disorders. J. Clin. Neurosci. 2011, 18, 1028–1037. [Google Scholar] [CrossRef] [PubMed]
  120. Lee, H.; Lee, H.J.; Jung, J.H.; Shin, E.A.; Kim, S.H. Melatonin disturbs SUMOylation-mediated crosstalk between c-Myc and nestin via MT1 activation and promotes the sensitivity of paclitaxel in brain cancer stem cells. J. Pineal Res. 2018, 65, e12496. [Google Scholar] [CrossRef] [PubMed]
  121. Pan, K.; Liang, X.-T.; Zhang, H.-k.; Zhao, J.-J.; Wang, D.-D.; Li, J.-J.; Lian, Q.; Chang, A.E.; Li, Q.; Xia, J.-C. Characterization of Bridging Integrator 1 (BIN1) as a Potential Tumor Suppressor and Prognostic Marker in Hepatocellular Carcinoma. Mol. Med. 2012, 18, 507–518. [Google Scholar] [CrossRef] [PubMed]
  122. Holler, C.J.; Davis, P.R.; Beckett, T.L.; Platt, T.L.; Webb, R.L.; Head, E.; Murphy, M.P. Bridging integrator 1 (BIN1) protein expression increases in the Alzheimer’s disease brain and correlates with neurofibrillary tangle pathology. J. Alzheimers Dis. 2014, 42, 1221–1227. [Google Scholar] [CrossRef] [Green Version]
  123. Paradies, G.; Paradies, V.; Ruggiero, F.M.; Petrosillo, G. Protective role of melatonin in mitochondrial dysfunction and related disorders. Arch. Toxicol. 2015, 89, 923–939. [Google Scholar] [CrossRef]
  124. Reiter, R.J.; Tan, D.X.; Osuna, C.; Gitto, E. Actions of melatonin in the reduction of oxidative stress. A review. J. BioMed. Sci. 2000, 7, 444–458. [Google Scholar] [CrossRef]
  125. Reiter, R.J.; Tan, D.X.; Cabrera, J.; D’Arpa, D. Melatonin and tryptophan derivatives as free radical scavengers and antioxidants. Adv. Exp. Med. Biol. 1999, 467, 379–387. [Google Scholar] [CrossRef]
  126. Shyma, M.S.; Ansar, E.B.; Gayathri, V.; Varma, H.K.; Mohanan, P.V. Attenuation of Cisplatin Induced Toxicity by Melatonin, Loaded on a Dextran Modified Iron Oxide Nanoparticles: An In Vitro Study. J. Forensic. Toxicol. Pharm. 2015, 4. [Google Scholar]
  127. Anisimov, S.V.; Popovic, N. Genetic aspects of melatonin biology. Rev. Neurosci. 2004, 15, 209–230. [Google Scholar] [CrossRef] [PubMed]
  128. Maharaj, D.S.; Glass, B.D.; Daya, S. Melatonin: New places in therapy. Biosci. Rep. 2007, 27, 299–320. [Google Scholar] [CrossRef] [PubMed]
  129. Letra-Vilela, R.; Sanchez-Sanchez, A.M.; Rocha, A.M.; Martin, V.; Branco-Santos, J.; Puente-Moncada, N.; Santa-Marta, M.; Outeiro, T.F.; Antolin, I.; Rodriguez, C.; et al. Distinct roles of N-acetyl and 5-methoxy groups in the antiproliferative and neuroprotective effects of melatonin. Mol. Cell Endocrinol. 2016, 434, 238–249. [Google Scholar] [CrossRef] [PubMed]
  130. Qi, X.; Ng, K.T.; Lian, Q.; Li, C.X.; Geng, W.; Ling, C.C.; Yeung, W.H.; Ma, Y.Y.; Liu, X.B.; Liu, H.; et al. Glutathione Peroxidase 3 Delivered by hiPSC-MSCs Ameliorated Hepatic IR Injury via Inhibition of Hepatic Senescence. Theranostics 2018, 8, 212–222. [Google Scholar] [CrossRef] [PubMed]
  131. Jakobisiak, M.; Lasek, W.; Golab, J. Natural mechanisms protecting against cancer. Immunol. Lett. 2003, 90, 103–122. [Google Scholar] [CrossRef] [PubMed]
  132. Swann, J.B.; Smyth, M.J. Immune surveillance of tumors. J. Clin. Investig. 2007, 117, 1137–1146. [Google Scholar] [CrossRef] [Green Version]
  133. Hakim, F.T.; Flomerfelt, F.A.; Boyiadzis, M.; Gress, R.E. Aging, immunity and cancer. Curr. Opin. Immunol. 2004, 16, 151–156. [Google Scholar] [CrossRef]
  134. Deleidi, M.; Jaggle, M.; Rubino, G. Immune aging, dysmetabolism, and inflammation in neurological diseases. Front. Neurosci. 2015, 9, 172. [Google Scholar] [CrossRef] [Green Version]
  135. Carrillo-Vico, A.; Lardone, P.J.; Alvarez-Sanchez, N.; Rodriguez-Rodriguez, A.; Guerrero, J.M. Melatonin: Buffering the immune system. Int. J. Mol. Sci. 2013, 14, 8638–8683. [Google Scholar] [CrossRef] [Green Version]
  136. Plackett, T.P.; Boehmer, E.D.; Faunce, D.E.; Kovacs, E.J. Aging and innate immune cells. J. Leukoc. Biol. 2004, 76, 291–299. [Google Scholar] [CrossRef]
  137. Su, S.C.; Hsieh, M.J.; Yang, W.E.; Chung, W.H.; Reiter, R.J.; Yang, S.F. Cancer metastasis: Mechanisms of inhibition by melatonin. J. Pineal Res. 2017, 62. [Google Scholar] [CrossRef]
  138. Guerrero, J.M.; Reiter, R.J. Melatonin-immune system relationships. Curr. Top. Med. Chem. 2002, 2, 167–179. [Google Scholar] [CrossRef]
  139. Medrano-Campillo, P.; Sarmiento-Soto, H.; Alvarez-Sanchez, N.; Alvarez-Rios, A.I.; Guerrero, J.M.; Rodriguez-Prieto, I.; Castillo-Palma, M.J.; Lardone, P.J.; Carrillo-Vico, A. Evaluation of the immunomodulatory effect of melatonin on the T-cell response in peripheral blood from systemic lupus erythematosus patients. J. Pineal Res. 2015, 58, 219–226. [Google Scholar] [CrossRef] [PubMed]
  140. Chuffa, L.G.; Lupi Junior, L.A.; Seiva, F.R.; Martinez, M.; Domeniconi, R.F.; Pinheiro, P.F.; Dos Santos, L.D.; Martinez, F.E. Quantitative Proteomic Profiling Reveals That Diverse Metabolic Pathways Are Influenced by Melatonin in an in Vivo Model of Ovarian Carcinoma. J. Proteome Res. 2016, 15, 3872–3882. [Google Scholar] [CrossRef] [PubMed]
  141. Martin-Renedo, J.; Mauriz, J.L.; Jorquera, F.; Ruiz-Andres, O.; Gonzalez, P.; Gonzalez-Gallego, J. Melatonin induces cell cycle arrest and apoptosis in hepatocarcinoma HepG2 cell line. J. Pineal Res. 2008, 45, 532–540. [Google Scholar] [CrossRef] [PubMed]
  142. Moreira, A.J.; Ordonez, R.; Cerski, C.T.; Picada, J.N.; Garcia-Palomo, A.; Marroni, N.P.; Mauriz, J.L.; González-Gallego, J. Melatonin activates endoplasmic reticulum stress and apoptosis in rats with diethylnitrosamine-induced hepatocarcinogenesis. PLoS ONE 2015, 10, e0144517. [Google Scholar] [CrossRef]
  143. Currier, N.L.; Sun, L.Z.; Miller, S.C. Exogenous melatonin: Quantitative enhancement in vivo of cells mediating non-specific immunity. J. Neuroimmunol. 2000, 104, 101–108. [Google Scholar] [CrossRef]
  144. Srinivasan, V.; Maestroni, G.J.; Cardinali, D.P.; Esquifino, A.I.; Perumal, S.R.; Miller, S.C. Melatonin, immune function and aging. Immun. Ageing 2005, 2, 17. [Google Scholar] [CrossRef] [Green Version]
  145. Maestroni, G.J. T-helper-2 lymphocytes as a peripheral target of melatonin. J. Pineal Res. 1995, 18, 84–89. [Google Scholar] [CrossRef]
  146. Cardinali, D.E.; Acuña-Castroviejo, D.; Ortiz, F. and Fernández-Gil, B. Melatonin-induced oncostasis, mechanisms and clinical relevance. J. Integ. Oncol. 2016. [Google Scholar] [CrossRef] [Green Version]
  147. Fourtillan, J.B.; Brisson, A.M.; Fourtillan, M.; Ingrand, I.; Decourt, J.P.; Girault, J. Melatonin secretion occurs at a constant rate in both young and older men and women. Am. J. Physiol. Endocrinol. Metab. 2001, 280, E11–E22. [Google Scholar] [CrossRef] [Green Version]
  148. Hardeland, R.; Cardinali, D.P.; Brown, G.M.; Pandi-Perumal, S.R. Melatonin and brain inflammaging. Prog. Neurobiol. 2015, 127–128, 46–63. [Google Scholar] [CrossRef] [Green Version]
  149. Cutolo, M.; Maestroni, G.J. The melatonin-cytokine connection in rheumatoid arthritis. Ann. Rheum. Dis. 2005, 64, 1109–1111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Maestroni, G.J. Therapeutic potential of melatonin in immunodeficiency states, viral diseases, and cancer. In Tryptophan, Serotonin, and Melatonin; Springer: Boston, MA, USA, 1999; pp. 217–226. [Google Scholar]
  151. Elmahallawy, E.K.; Luque, J.O.; Aloweidi, A.S.; Gutierrez-Fernandez, J.; Sampedro-Martinez, A.; Rodriguez-Granger, J.; Kaki, A.; Agil, A. Potential Relevance of Melatonin Against Some Infectious Agents: A Review and Assessment of Recent Research. Curr. Med. Chem. 2015, 22, 3848–3861. [Google Scholar] [CrossRef]
  152. Reiter, R.J.; Mayo, J.C.; Tan, D.X.; Sainz, R.M.; Alatorre-Jimenez, M.; Qin, L. Melatonin as an antioxidant: Under promises but over delivers. J. Pineal Res. 2016, 61, 253–278. [Google Scholar] [CrossRef] [PubMed]
  153. Garcia-Maurino, S.; Gonzalez-Haba, M.G.; Calvo, J.R.; Rafii-El-Idrissi, M.; Sanchez-Margalet, V.; Goberna, R.; Guerrero, J.M. Melatonin enhances IL-2, IL-6, and IFN-gamma production by human circulating CD4+ cells: A possible nuclear receptor-mediated mechanism involving T helper type 1 lymphocytes and monocytes. J. Immunol. 1997, 159, 574–581. [Google Scholar] [PubMed]
  154. Szczepanik, M. Melatonin and its influence on immune system. J Physiol Pharm. 2007, 58 (Suppl. 6), 115–124. [Google Scholar]
  155. Lardone, P.J.; Carrillo-Vico, A.; Molinero, P.; Rubio, A.; Guerrero, J.M. A novel interplay between membrane and nuclear melatonin receptors in human lymphocytes: Significance in IL-2 production. Cell Mol. Life Sci. 2009, 66, 516–525. [Google Scholar] [CrossRef]
  156. Paterniti, I.; Cordaro, M.; Esposito, E.; Cuzzocrea, S. The antioxidative property of melatonin against brain ischemia. Expert Rev. Neurother. 2016, 16, 841–848. [Google Scholar] [CrossRef]
  157. Liu, F.; Ng, T.B.; Fung, M.C. Pineal indoles stimulate the gene expression of immunomodulating cytokines. J. Neural. Transm. (Vienna) 2001, 108, 397–405. [Google Scholar] [CrossRef]
  158. Conti, A.; Conconi, S.; Hertens, E.; Skwarlo-Sonta, K.; Markowska, M.; Maestroni, J.M. Evidence for melatonin synthesis in mouse and human bone marrow cells. J. Pineal Res. 2000, 28, 193–202. [Google Scholar] [CrossRef] [PubMed]
  159. Singh, M.; Jadhav, H.R. Melatonin: Functions and ligands. Drug Discov. Today 2014, 19, 1410–1418. [Google Scholar] [CrossRef]
  160. Fan, L.; Sun, G.; Ma, T.; Zhong, F.; Wei, W. Melatonin overcomes apoptosis resistance in human hepatocellular carcinoma by targeting survivin and XIAP. J. Pineal Res. 2013, 55, 174–183. [Google Scholar] [CrossRef] [PubMed]
  161. Wang, Y.; Wang, P.; Zheng, X.; Du, X. Therapeutic strategies of melatonin in cancer patients: A systematic review and meta-analysis. Oncotargets Ther. 2018, 11, 7895. [Google Scholar] [CrossRef] [Green Version]
  162. Rodriguez, C.; Martín, V.; Herrera, F.; García-Santos, G.; Rodriguez-Blanco, J.; Casado-Zapico, S.; Sánchez-Sánchez, A.M.; Suárez, S.; Puente-Moncada, N.; Anítua, M.J. Mechanisms involved in the pro-apoptotic effect of melatonin in cancer cells. Int. J. Mol. Sci. 2013, 14, 6597–6613. [Google Scholar] [CrossRef] [PubMed]
  163. Lee, S.J.; Jung, Y.H.; Oh, S.Y.; Yun, S.P.; Han, H.J. Melatonin enhances the human mesenchymal stem cells motility via melatonin receptor 2 coupling with Galphaq in skin wound healing. J. Pineal Res. 2014, 57, 393–407. [Google Scholar] [CrossRef] [PubMed]
  164. Mias, C.; Lairez, O.; Trouche, E.; Roncalli, J.; Calise, D.; Seguelas, M.H.; Ordener, C.; Piercecchi-Marti, M.D.; Auge, N.; Salvayre, A.N.; et al. Mesenchymal stem cells promote matrix metalloproteinase secretion by cardiac fibroblasts and reduce cardiac ventricular fibrosis after myocardial infarction. Stem Cells 2009, 27, 2734–2743. [Google Scholar] [CrossRef] [PubMed]
  165. Zaminy, A.; Ragerdi Kashani, I.; Barbarestani, M.; Hedayatpour, A.; Mahmoudi, R.; Farzaneh Nejad, A. Osteogenic differentiation of rat mesenchymal stem cells from adipose tissue in comparison with bone marrow mesenchymal stem cells: Melatonin as a differentiation factor. Iran BioMed J. 2008, 12, 133–141. [Google Scholar] [PubMed]
  166. Zheng, W.; Yang, Y.; Sequeira, R.C.; Bishop, C.E.; Atala, A.; Gu, Z.; Zhao, W. Effects of Extracellular Vesicles Derived from Mesenchymal Stem/Stromal Cells on Liver Diseases. Curr. Stem Cell Res. Ther. 2019, 14, 442–452. [Google Scholar] [CrossRef]
  167. Cho, Y.A.; Noh, K.; Jue, S.S.; Lee, S.Y.; Kim, E.C. Melatonin promotes hepatic differentiation of human dental pulp stem cells: Clinical implications for the prevention of liver fibrosis. J. Pineal Res. 2015, 58, 127–135. [Google Scholar] [CrossRef]
  168. Kadry, S.M.; El-Dakdoky, M.H.; Haggag, N.Z.; Rashed, L.A.; Hassen, M.T. Melatonin improves the therapeutic role of mesenchymal stem cells in diabetic rats. Toxicol. Mech. Methods 2018, 28, 529–538. [Google Scholar] [CrossRef]
  169. Wang, X.; Liang, T.; Qiu, J.; Qiu, X.; Gao, B.; Gao, W.; Lian, C.; Chen, T.; Zhu, Y.; Liang, A.; et al. Melatonin Reverses the Loss of Stemness Induced by TNF- α in Human Bone Marrow Mesenchymal Stem Cells through Upregulation of YAP Expression. Stem Cells Int. 2019, 2019, 1–16. [Google Scholar] [CrossRef]
  170. Jiang, T.; Xia, C.; Chen, X.; Hu, Y.; Wang, Y.; Wu, J.; Chen, S.; Gao, Y. Melatonin promotes the BMP9-induced osteogenic differentiation of mesenchymal stem cells by activating the AMPK/β-catenin signalling pathway. Stem Cell Res. Ther. 2019, 10, 408. [Google Scholar] [CrossRef]
  171. Mohamed, Y.; Basyony, M.A.; El-Desouki, N.I.; Abdo, W.S.; El-Magd, M.A. The potential therapeutic effect for melatonin and mesenchymal stem cells on hepatocellular carcinoma. BioMed (Taipei) 2019, 9, 24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. El-Magd, M.A.; Mohamed, Y.; El-Shetry, E.S.; Elsayed, S.A.; Abo Gazia, M.; Abdel-Aleem, G.A.; Shafik, N.M.; Abdo, W.S.; El-Desouki, N.I.; Basyony, M.A. Melatonin maximizes the therapeutic potential of non-preconditioned MSCs in a DEN-induced rat model of HCC. BioMed Pharm. 2019, 114, 108732. [Google Scholar] [CrossRef] [PubMed]
  173. Mortezaee, K.; Khanlarkhani, N.; Sabbaghziarani, F.; Nekoonam, S.; Majidpoor, J.; Hosseini, A.; Pasbakhsh, P.; Kashani, I.R.; Zendedel, A. Preconditioning with melatonin improves therapeutic outcomes of bone marrow-derived mesenchymal stem cells in targeting liver fibrosis induced by CCl4. Cell Tissue Res. 2017, 369, 303–312. [Google Scholar] [CrossRef] [PubMed]
  174. Mortezaee, K.; Pasbakhsh, P.; Ragerdi Kashani, I.; Sabbaghziarani, F.; Omidi, A.; Zendedel, A.; Ghasemi, S.; Dehpour, A.R. Melatonin Pretreatment Enhances the Homing of Bone Marrow-derived Mesenchymal Stem Cells Following Transplantation in a Rat Model of Liver Fibrosis. Iran BioMed J. 2016, 20, 207–216. [Google Scholar] [CrossRef] [PubMed]
  175. Chen, H.H.; Lin, K.C.; Wallace, C.G.; Chen, Y.T.; Yang, C.C.; Leu, S.; Chen, Y.C.; Sun, C.K.; Tsai, T.H.; Chen, Y.L.; et al. Additional benefit of combined therapy with melatonin and apoptotic adipose-derived mesenchymal stem cell against sepsis-induced kidney injury. J. Pineal Res. 2014, 57, 16–32. [Google Scholar] [CrossRef] [PubMed]
  176. Basyony, M.; Desouki, N.; Sobhy, W.; Hegazy, R.; Mohamed, Y. Melatonin improves the anticancer effects of mesenchymal stem cell against HCC in rat. Egypt. J. Exp. Biol. (Zool.) 2019, 1. [Google Scholar] [CrossRef]
Figure 1. The pleiotropic functionality of mesenchymal stem cells (MSCs) which tumor-limiting and tumor-promoting effects.
Figure 1. The pleiotropic functionality of mesenchymal stem cells (MSCs) which tumor-limiting and tumor-promoting effects.
Ijms 21 04521 g001
Figure 2. Tumor-limiting effects of melatonin and their suggested mechanisms.
Figure 2. Tumor-limiting effects of melatonin and their suggested mechanisms.
Ijms 21 04521 g002
Figure 3. Effects of the combined use of melatonin and MSCs on hepatocellular carcinoma (HCC); ((GSTPS: glutathione S-transferases), (PCNA: proliferating cell nuclear antigen)) [171,172,173,174,175,176].
Figure 3. Effects of the combined use of melatonin and MSCs on hepatocellular carcinoma (HCC); ((GSTPS: glutathione S-transferases), (PCNA: proliferating cell nuclear antigen)) [171,172,173,174,175,176].
Ijms 21 04521 g003

Share and Cite

MDPI and ACS Style

Elmahallawy, E.K.; Mohamed, Y.; Abdo, W.; Yanai, T. Melatonin and Mesenchymal Stem Cells as a Key for Functional Integrity for Liver Cancer Treatment. Int. J. Mol. Sci. 2020, 21, 4521. https://doi.org/10.3390/ijms21124521

AMA Style

Elmahallawy EK, Mohamed Y, Abdo W, Yanai T. Melatonin and Mesenchymal Stem Cells as a Key for Functional Integrity for Liver Cancer Treatment. International Journal of Molecular Sciences. 2020; 21(12):4521. https://doi.org/10.3390/ijms21124521

Chicago/Turabian Style

Elmahallawy, Ehab Kotb, Yasser Mohamed, Walied Abdo, and Tokuma Yanai. 2020. "Melatonin and Mesenchymal Stem Cells as a Key for Functional Integrity for Liver Cancer Treatment" International Journal of Molecular Sciences 21, no. 12: 4521. https://doi.org/10.3390/ijms21124521

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop