Effects of melatonin and its analogues on neural stem cells

Effects of melatonin and its analogues on neural stem cells

Accepted Manuscript Effects of melatonin and its analogues on neural stem cells Jiaqi Chu, Yalin Tu, Jingkao Chen, Dunxian Tan, Xingguo Liu, Rongbiao ...

2MB Sizes 3 Downloads 77 Views

Accepted Manuscript Effects of melatonin and its analogues on neural stem cells Jiaqi Chu, Yalin Tu, Jingkao Chen, Dunxian Tan, Xingguo Liu, Rongbiao Pi PII:

S0303-7207(15)30113-1

DOI:

10.1016/j.mce.2015.10.012

Reference:

MCE 9314

To appear in:

Molecular and Cellular Endocrinology

Received Date: 3 May 2015 Revised Date:

27 September 2015

Accepted Date: 18 October 2015

Please cite this article as: Chu, J., Tu, Y., Chen, J., Tan, D., Liu, X., Pi, R., Effects of melatonin and its analogues on neural stem cells, Molecular and Cellular Endocrinology (2015), doi: 10.1016/ j.mce.2015.10.012. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT

Effects of melatonin and its analogues on neural stem cells

Department of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510080, China.

2

International Joint Laboratory (SYSU-PolyU HK) of Novel Anti-Dementia Drugs of

3

M AN U

Guangdong, Guangzhou 510006, China.

SC

1

RI PT

Jiaqi Chu1,2,3, Yalin Tu1,2,3, Jingkao Chen1,2,3, Dunxian Tan4, Xingguo Liu5, Rongbiao Pi1,2,3*

National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Sun Yat-Sen University, Guangzhou 510080, China.

4

Department of Cellular and Structural Biology, The University of Texas, Health Science

5

TE D

Center at San Antonio, 7703 Floyd Curl, San Antonio, TX 78229, USA.

School of Chemical Engineering and Light Industry, Guangdong University of Technology,

EP

Guangzhou, P.R.China

AC C

*To whom correspondence should be addressed; Telephone: +86-20-3994-3122, E-mail: [email protected] .

ACCEPTED MANUSCRIPT

Abstract

Neural stem cells (NSCs) are multipotent cells which are capable of self-replication and

RI PT

differentiation into neurons, astrocytes or oligodendrocytes in the central nervous system (CNS). NSCs are found in two main regions in the adult brain: the subgranular zone (SGZ) in the hippocampal dentate gyrus (DG) and the subventricular zone (SVZ). The recent discovery

SC

of NSCs in the adult mammalian brain has fostered a plethora of translational and preclinical studies to investigate novel approaches for the therapy of neurodegenerative diseases.

M AN U

Melatonin is the major secretory product synthesized and secreted by the pineal gland and shows both a wide distribution within phylogenetically distant organisms from bacteria to humans and a great functional versatility. Recently, accumulated experimental evidence showed that melatonin plays an important role in NSCs, including its proliferation,

TE D

differentiation and survival, which are modulated by many factors including MAPK/ERK signaling pathway, histone acetylation, neurotrophic factors, transcription factors, and apoptotic genes. The purpose of this review is to summarize the beneficial effects of

EP

melatonin on NSCs and further to discuss the potential usage of melatonin and its derivatives

AC C

or analogues in the treatment of CNS neurodegenerative diseases.

Keywords

melatonin; neural stem cells; proliferation; differentiation; neurogenesis

ACCEPTED MANUSCRIPT

Abbreviations:

Parkinson's disease; AD, Alzheimer's disease; TH, tyroxine hydroxylase ;CVS,cardiovascular system; PVN,

RI PT

paraventricular nucleus; Aβ, β-amyloid protein; MSCs, mesenchymal stem cells; GFAP, glial fibrillary acidic protein; MAP2, microtubule associated protein 2; bHLH, helix–loop–helix; MT, melatonin receptors; MAPK, mitogen-activated protein kinase; ERK, Extracellular signal-regulated kinase; BDNF,

SC

Brain-derived neurotrophic factor; PI3K, phosphoinositide 3-kinase; HD, Huntington's disease; GDNF, Glial cell line-derived neurotrophic factor; Bcl-2, B-cell lymphoma 2; Nrf2, nuclear factor erythroid

M AN U

2-related factor 2; ARE, antioxidant response element; Keap1, kelch-like ECH associating protein 1; AChE, acetylcholinesterase; MTLs, multi-targeted ligands.

TE D

1. Introduction

Neural stem cells (NSCs) are multipotent cells present in the developing and adult brain.

EP

They have the potential to generate both neurons and glia of the developing brain and they also account for the limited regenerative potential in the adult brain. In the adult brain, NSCs

AC C

reside in defined regions (“neurogenic niches”) that sustain their multipotency and regulate the balance between symmetrical self-renewal and fate-committed asymmetric divisions (Sommer and Rao, 2002).

NSCs have become one of the most intensively studied cell types in neurobiology. NSCs offer a unique and powerful tool for basic research and regenerative medicine in the field of Central Nervous System (CNS). The discovery of NSCs and neurogenesis in the adult

ACCEPTED MANUSCRIPT

mammalian CNS has tremendously changed our view of the plasticity and function of the brain. This has prompted excitement for the possible exploitement of intrinsic neurogenic

RI PT

activity to cure CNS diseases and rescue brain function after injury or dysfunction. Mobilization of endogenous NSCs has thus emerged as a potential therapeutic approach for neural repair (Honmou, 2015,Tong, Fong and Huang, 2015).

SC

Melatonin is an endogenous indoleamine present in different tissues, cellular compartments and organelles including mitochondria. In the past decades, among the

proliferation,

migration

and

M AN U

numerous functions of melatonin, the effects of the indoleamine on NSCs, including survival, differentiation,

had

been

extensively

researched.

Receptor-dependent and -independent responses to melatonin are suggested to occur in NSCs.

TE D

1.1 NSCs and neurogenesis

NSCs develop into new neurons, astrocytes and oligodendrocytes in successive process of proliferation, migration and differentiation (Wade, McKinney and Phillips, 2014) and

EP

neurogenesis occurs throughout whole life in the mammalian brain (Wang, Liu, Liu et al.,

AC C

2011). In the early embryo, embryonic stem cells (ESCs) receive the differentiation signal and then the formation of mesoderm, endoderm and ectoderm is guided (Thomson, Itskovitz-Eldor, Shapiro et al., 1998). There exists a type of neurally specified ectoderm cells known as the earliest NSCs within the ectoderm, which are called neuroepithelial cells and the neuroepithelial cells in the telencephalon later develop into radial glial cells in the ventricular zone (VZ) (Fishell and Kriegstein, 2003). At early stages, the cells undergo rapid expansion through symmetrical divisions to generate a stem cell pool (Temple, 2001) and

ACCEPTED MANUSCRIPT

then they initiate self-renewal through asymmetrical divisions, generating a committed neuronal precursor cell and a daughter cell identical to the parent. Resembling the

RI PT

transit-amplifying cells in many adult lineages, the committed neuronal cells might undergo further symmetrical divisions before differentiation, and are thus called neural precursor cells (NPCs) (Gotz and Huttner, 2005). Throughout the stem cell divisions, radial glial cells remain

SC

in the region of the VZ while the committed precursors created migrate from the VZ into an overlying secondary proliferative region called the subventricular zone (SVZ), where they can

M AN U

undergo a small number of symmetrical divisions before differentiating into neuronal or glial cells (Miyata, Kawaguchi, Saito et al., 2004,Noctor, Martinez-Cerdeno, Ivic et al., 2004,Tabata and Nakajima, 2003). Along with the development, the VZ shrinks, while the SVZ increases in size till adulthood. The SVZ and the subgranular zone (SGZ) in the

TE D

hippocampal dentate gyrus (DG) of the hippocampus are considered as the two neurogenic niches in the adult brain (Zhao, Deng and Gage, 2008).

NSCs or neural precursor cells (NPCs) in SGZ and SVZ continuously participating in

EP

neurogenesis and gliogenesis under both normal and pathological conditions (Kriegstein and

AC C

Alvarez-Buylla, 2009,Zhao et al., 2008). NPCs-based therapy has been considered as a promising therapeutic approach to protect and to restore the damaged CNS, which were reinforced by discovery of potential benefits of NSCs in several animal models of various neural diseases, such as stroke, Parkinson's disease (PD) and Alzheimer's disease (AD) (Martino, Pluchino, Bonfanti et al., 2011). However, a large percentage of neurons generated by NSCs die during the first two weeks, and only a few of them survive for a relatively long period of time (Kempermann, Gast, Kronenberg et al., 2003), though the continuous

ACCEPTED MANUSCRIPT

neurogenesis and gliogenesis. Thus, further investigation of the mechanisms of NSCs proliferation and differentiation are needed.

RI PT

1.2 Functions of melatonin

Melatonin is an ancient and highly conserved indoleamine derived from tryptophan and it

SC

is present in all phyla of muticellular animals. It is recognized as a neurohormone that possesses numerous functions and has a wide range of distribution (Hardeland and Poeggeler,

M AN U

2003). In mammals, melatonin is mainly synthesized by the pineal gland in a circadian manner and released into blood and cerebrospinal fluid to exert regulatory roles on seasonal and circadian rhythms (Luchetti, Canonico, Betti et al., 2010), which is critical for the physiological functions of the neuroimmuno-endocrine system such as sleep-wake cycle,

TE D

pubertal development and seasonal adaptation (Reiter, Tan, Manchester et al., 2009,Salti, Galluzzi, Bindi et al., 2000,Shochat, Luboshitzky and Lavie, 1997).

During the daytime, the melatonin secretion level is very low (below 10 pg/mL), yet the

EP

concentration exhibit higher values (10–15-fold increase, up to 120 pg/mL) during the night

AC C

(Karasek and Winczyk, 2006,Konturek, Konturek, Brzozowska et al., 2007,Sanders, Chaturvedi and Hordinsky, 1999). This circadian rhythm is present in all living organisms. For example, in human beings, the rhythm is developed during the first months of life and reaches the greatest magnitude between the 4th and 7th year of age and then decreases (Escames, Lopez, Garcia et al., 2010). Accordingly, the melatonin level varies during the lifespan. In the fetal period, the fetus uses maternal melatonin that crosses the placenta and the level of melatonin increases from birth to a peak around puberty, and then declines in

ACCEPTED MANUSCRIPT

middle-aged and elderly individuals (Sharma, Palacios-Bois, Schwartz et al., 1989,Waddell, Wharfe, Crew et al., 2012). The association between the decline in melatonin’s levels with

RI PT

aging and insomnia as well as the development of neurodegenerative diseases such as AD has been reported (Bubenik and Konturek, 2011).

In addition to its multiple physiological functions, much attention has been attracted to the

SC

antioxidant action of melatonin (Reiter, Paredes, Manchester et al., 2009). Studies indicated that melatonin prevented neuronal cell death induced by toxins, such as 6-hydroxydopamine

M AN U

(Sharma, McMillan, Tenn et al., 2006), 1-methyl-4-phenylpyridinium ion (MPP+) (Chetsawang, Govitrapong and Chetsawang, 2007), amphetamine (Klongpanichapak, Phansuwan-Pujito, Ebadi et al., 2007), β-amyloid protein (Aβ) (Gunasingh, Philip, Ashok et al., 2008) and kainic acid (Lee, Chun, Kong et al., 2006) via its free radical-scavenging

TE D

mechanisms. Thus, melatonin has been proposed to be a hormone that could play a role in the development of diseases associated with oxidative damages.

EP

Amounting evidence shows that melatonin has numerous functions in mesenchymal stem cells (MSCs), a heterogeneous population of multipotent elements resident in tissues such as

AC C

bone marrow, muscle, and adipose tissue, which are primarily involved in developmental and regeneration processes, tissue repair and restoration. The roles of melatonin in MSC lineage commitment and adipogenic differentiation as well as their mechanism are well discussed in a wonderful review published recently (Luchetti, Canonico, Bartolini et al., 2014). Very recently, several reports indicated the potential clinical applications of melatonin on MSCs to treat the kidney injury (Chen, Lin, Wallace et al., 2014), acute interstitial cystitis (Chen, Tan,

ACCEPTED MANUSCRIPT

Jing et al., 2014), skin wound (Lee, Jung, Oh et al., 2014), and also cerebral ischemia (Tang, Cai, Yuan et al., 2014). Interestingly, emerging evidence suggests that melatonin receptors are

RI PT

expressed in NSCs/NPCs (Niles, Armstrong, Rincon Castro et al., 2004,Ramirez-Rodriguez, Klempin, Babu et al., 2009) and raises the obvious question of whether melatonin also play a key role in the development of NSCs/NPCs. Moreover, it has been documented that

SC

neurogenesis in the DG occurs under a circadian rhythmic pattern. BrdU-labeled proliferating cells show a dark/light cycle–dependent pattern (Guzman-Marin, Suntsova, Bashir et al.,

M AN U

2007). M-phase cells increase during the night (Tamai, Sanada and Fukada, 2008), and constant light exposure decreases neurogenesis in the DG (Fujioka, Fujioka, Tsuruta et al., 2011). Clock genes (Per1, Per2, Cry1, Cry2, Bmal1, and Clock), which are expressed in NPCs in the DG of the hippocampus (Jilg, Lesny, Peruzki et al., 2010), can regulate the differentiation

TE D

transcription factors including NeuroD1, Id2, Hey1, Olig1 (Kimiwada, Sakurai, Ohashi et al., 2009). These data suggest melatonin, which secretes following light/dark rhythms (Zawilska, Skene and Arendt, 2009) and acts as a regulator of circadian rhythms in mammals (Stehle,

EP

von Gall and Korf, 2003), can play an important role in the development of NSCs. Therefore, a better understanding of the potential interaction between melatonin and NSCs is required to

AC C

shed novel lights on the effects and their mechanisms of melatonin on NSCs and to facilitate the development of new therapies for neurodegenerative diseases.

2. Effects of Melatonin on NSCs

The survival, proliferation and differentiation and also migration of NSCs are very important for their functions in the developing or adult brain as well as in the pathological

ACCEPTED MANUSCRIPT

conditions. Amounting evidence demonstrated that melatonin plays important roles in these aspects (summarized in Table 1).

RI PT

2.1 Melatonin modulates cell survival of NSCs

The fate of NSCs is very important. Consistent with previous studies that melatonin

SC

protects damaged neurons, it was reported that melatonin regulates the cell viability of NSCs from different regions of brain. Kong X, et al. found that melatonin (0.05-1 nM) increased the

M AN U

viability of cultured NSCs obtained from rat midbrain in a dose-dependent manner by MTT assay (Kong, Li, Cai et al., 2008). Ramirez-Rodriguez, G., et al. suggested that melatonin promoted survival of precursor cells in vitro and increased survival of newborn neurons in vivo. By quantifying cells that underwent differentiation after 5 days, melatonin (0.1 µM) was

TE D

found to significantly increase the survival of differentiated cells, which is confirmed with the WST-1 assay. In addition, melatonin increased the survival of newborn neurons in the DG of adult mice after 14 days of treatment by BrdU-labelling. The number of surviving cells in the

EP

DG in melatonin-treated mice (8 mg/kg) was increased by 63%, comparing to that in vehicle-treated mice (Ramirez-Rodriguez et al., 2009). Studies also showed that melatonin

AC C

protected NSCs against hypoxia and increased the cell viability in vitro. Melatonin (0.001-100 µM) obviously improved the cell viability of NSCs in a dose-dependent manner. Meanwhile, melatonin (100 nM) significantly decreased the death of cells subjected to hypoxia when detected by Hoechst staining (Fu, Zhao, Liu et al., 2011). Most recently, melatonin was reported to protect NSCs against LPS-induced inflammation. Melatonin (100 nM) decreased approximately 20% and 35% cytotoxicity level of the NSCs treated with 100ng/mL and

ACCEPTED MANUSCRIPT

1µg/mL LPS respectively by lactate dehydrogenase (LDH) assay, which might be related to attenuating NO production in LPS treated NSCs (Song, Kang, Lee et al., 2015). In addition,

RI PT

the protection effects were also found in the human bone marrow derived MSCs (BM-MSCs). Pretreatment of melatonin (0.01-100 µM) successfully reversed the H2O2-induced senescent phenotypes of BM-MSCs (Zhou, Chen, Liu et al., 2015).

SC

2.2 Melatonin promotes NSCs proliferation

M AN U

The ability of proliferation of NSCs affects its function. Melatonin was found to induce cell proliferation in various NSCs. Melatonin (1 nM) significantly increased the total number of neural spheres and the number of neural spheres over 60 µm in diameter in NSCs derived from rat ventral midbrain (Kong et al., 2008). Similarly, melatonin (0.001-10 µM) induced

TE D

SVZ precursor cell proliferation in a concentration-dependent manner. The number of neurosphere generated in melatonin-treated cells in vitro was significantly increased. Furthermore, an increase in neurosphere size was observed in melatonin-treated cells

EP

(Sotthibundhu, Phansuwan-Pujito and Govitrapong, 2010). Moreover, melatonin (0.1-10 µM) treatment for 5 days increased the number of neurosphere of adult hippocampal NSCs

AC C

(Tocharus, Puriboriboon, Junmanee et al., 2014). By using bromodeoxyuridine labeling and immunostaining, melatonin (100 nM) was shown to promote the proliferation of hypoxic NSCs. The ratio of BrdU/nestin positive cells against total nestin-positive NSCs (>95% of all cells) was significantly increased comparing with the untreated cells at 3 days after hypoxia (Fu et al., 2011). Melatonin (5 or 10 mg/kg) treatment dramatically enhanced cell proliferation of ischemic-stroke NSCs in a time-dependent manner, evaluated by ki67 (a protein strictly

ACCEPTED MANUSCRIPT

associated with cell proliferation) staining (Chern, Liao, Wang et al., 2012). In addition, synergistic effect of melatonin (10 mg/kg) and exercising on the proliferation of endogenous

RI PT

NSCs/NPCs after Spinal Cord Injury were reported (Lee, Lee, Lee et al., 2014). However, contrary to the above, melatonin (0.01–10 nM, physiological concentrations) failed to affect the proliferation of NSCs derived from the mouse embryo striatum by WST-8 assay.

SC

Pharmacological concentrations (1–100 µM) of melatonin also did not affect cell proliferation after 1 day incubation, but suppressed cell proliferation on days 4 and 7 in a

M AN U

concentration-dependent manner, examined by WST-8 assay and BrdU incorporation analysis (Moriya, Horie, Mitome et al., 2007). These results seem to be contrary to other results demonstrating that melatonin promote the proliferation of NSCs and the reason for the difference in the effects of melatonin is unclear at present. The difference in the source of

TE D

NSCs or the experiment system might be the explanation. Recently, melatonin was found to minimize the inhibitory effects of dexamethasone on adult hippocampal progenitor cell proliferation.

Addition

of

dexamethasone

to

hippocampal

progenitor

cells

from

EP

eight-week-old rats resulted in a decrease in the number of neurospheres, which could be precluded by pretreatment of melatonin (1 µM) (Ekthuwapranee, Sotthibundhu, Tocharus et

AC C

al., 2014).

2.3 Melatonin promotes NSCs differentiation

Some studies demonstrated that melatonin facilitates NSCs differentiation into neurons. The cultured ventral midbrain NSCs with melatonin treatment (1 nM) were found to differentiate into tyroxine hydroxylase (TH), which is a key enzyme of dopamine synthesis

ACCEPTED MANUSCRIPT

and is usually regarded as a marker of dopaminergic neurons, positive neurons. By immunostaining, increased percentage of BrdU-positive cells detected were β-III-tubulin (a

RI PT

neuronal marker) positive when treated with melatonin (1 nM), while the glial fibrillary acidic protein (GFAP) positive cells decreased (Kong et al., 2008). The nestin (653 bp) and β-III-tubulin (442 bp) mRNA transcripts were readily detected in all samples of C17.2 cells (a

SC

neural stem cell line derived from the external germinal layer of mouse cerebellum) treated with melatonin (0.05-100 nM) for 24 hr by RT-PCR, in contrast, the mRNA transcript for

M AN U

GFAP (592 bp) was not detected in any samples (Sharma, Ottenhof, Rzeczkowska et al., 2008). Fluorescence-based immunocytochemistry showed that 1µM melatonin-treated cells, isolated from the SVZ of adult mice, exhibited significantly increased differentiation into the β-III-tubulin positive cells, but differentiation into GFAP remained unchanged. These cells

TE D

also showed characteristic neuronal morphologies and established contacts between each other (Sotthibundhu et al., 2010). Adult hippocampal NPCs cultured in the presence of melatonin (0.01-10,000 nM) caused an increase in the density of β-III-tubulin positive cells

EP

and measuring β-III-tubulin levels by western blot confirmed the increase in neuronal differentiation in response to melatonin (Ramirez-Rodriguez et al., 2009). The percentage of

AC C

neurons in relation to the total cell number was decreased when NSCs were exposed to hypoxia, yet melatonin (100 nM) reversed this and increased the percentage of microtubule associated protein 2 (MAP2, a neuronal marker) positive cells in all melatonin treated groups, especially in the 7 days differentiation group, by immunocytochemistry, as corroborated by Western blot analysis. When compared with the control group, hypoxia did not appear to affect the differentiation of NSCs into astrocytes (Fu et al., 2011). Takahiro Moriya, et al.

ACCEPTED MANUSCRIPT

reported that treating with physiological concentration of melatonin (0.01–10 nM) during the proliferation period and differentiation period failed to affect the differentiation of the NSCs either

β-III-tubulin+

neurons

or

GFAP+

astrocytes

when

examined

by

RI PT

into

immunocytochemistry and ELISA. On the other hand, NSCs which had been treated with higher concentrations of melatonin (1–25 µM) during the proliferation period differentiated more

abundant

neurons

than

those

in

control

medium,

evaluated

by

SC

into

immunocytochemistry and ELISA. In contrast, melatonin (1–100 µM) decreased the neural

M AN U

differentiation of the NSCs into β-III-tubulin+ neurons when cell were treated during the differentiation period in a concentration-dependent manner. However, melatonin failed to affect the differentiation into GFAP+ astrocytes and the number of total viable cells under differentiation-preferring conditions (Moriya et al., 2007). Melatonin might modulate adult

TE D

neurogenesis by different mechanisms under different concentrations. Thus, melatonin exerts its effects at both physiological (0.01-10 nM) and pharmacological concentrations (1-100 µM).

AC C

of NSCs

EP

3. The mechanisms of melatonin regulating proliferation and differentiation activities

Studies from several sources indicate that melatonin exerts NSCs regulation via different

mechanisms involving melatonin receptors, MAPK/ERK signaling, histone acetylation, neurotrophic factors, basic helix–loop–helix (bHLH) factors, etc. A brief summary of this evidence was described as follows (summarized in Fig. 1).

3.1 Melatonin receptors

ACCEPTED MANUSCRIPT

Till now, four types of melatonin receptors have been identified, membrane receptors (MT1, MT2), cytosolic binding sites (MT3 and calmodulin), and nuclear receptors of the RZR/ROR

RI PT

family (Cutando, Aneiros-Fernandez, Lopez-Valverde et al., 2011). Numerous biological functions of melatonin in mammals are mediated via activation of the G-protein-coupled high-affinity melatonin receptors 1 and 2 (MT1 and MT2). Both MT1 and MT2 receptors are

SC

expressed in various organs, the MT1 subtype is found in retina, ovary, testis, mammary gland, coronary arteries, gall bladder, aorta, liver, kidney, skin and brain, high density of melatonin

M AN U

receptors has been shown to be in the SCNs, particularly in the adenohypophysis, suprachiasmatic nucleus (SCN), paraventricular nucleus (PVN) and area postremaand thus should be related to the function of melatonin in SCNs. The MT2 subtype is found in the retina, SCN, hippocampus, substantia nigra, ventral tegmental area (Luchetti et al.,

TE D

2010,Singh and Jadhav, 2014).

MT1 is a 350- amino protein, presenting a sequence homology of 60% with the 362-amino acid MT2 and has higher homology than MT2 with rhodopsin (Reppert, Godson, Mahle et al.,

EP

1995). MT1 and MT2 are typical the G-protein-coupled receptor (GPCR) with

AC C

7-transmembrane (TM) α-helices and 4 intracellular and 4 extracellular domains. The receptors couple to G proteins (Gai2/3, Gaq, and Gbg) during activation by binding sites of the MT isoforms, which lie in the same part of the receptor: in the immediate vicinity of TM5 and in the extracellular half of the transmembrane domain (Mazna, Grycova, Balik et al., 2008,Rivara, Lorenzi, Mor et al., 2005).

The effects of melatonin have been found to be linked with the activation of functional

ACCEPTED MANUSCRIPT

melatonin receptors. The transcripts of melatonin receptors are detected in precursor cells and the increased proportion of neurons induced by melatonin was almost entirely blocked (84%)

RI PT

by luzindole, a melatonin receptor antagonist. The antagonist alone, however, did not induce changes in neuronal differentiation (Ramirez-Rodriguez et al., 2009). Luzindole partially suppressed the melatonin-induced proliferation of precursor cells isolated from SVZ of adult

SC

mouse in a concentration-dependent manner. Double labeling showed that MT1 was expressed in cells during both proliferation and differentiation periods (Sotthibundhu et al.,

M AN U

2010). Chern CM, et al. found that MT2 melatonin receptor is involved in ameliorating neural function in ischemic-stroke mice (Chern et al., 2012). The protective effect of melatonin was largely reversed by pretreating with either luzindole or 4P-PDOT, two MT antagonists with different chemical structures. The latter is a much more selective MT2 antagonist than

TE D

luzindole (Dubocovich, Masana, Iacob et al., 1997).

3.2 MAPK/ERK signaling

EP

The mitogen-activated protein kinase (MAPK) signal pathway is evolutionarily conserved among eukaryotes and plays important roles in numerous biological functions, including cell

AC C

proliferation and cell death. Extracellular signal-regulated kinase (ERK) is one of the main subgroups of the MAPKs. ERK1 and ERK2 are key transducers of proliferation, differentiation and survival signals. It was originally found to be phosphorylated on Tyr and Thr residues, and almost expressed in all tissues, mainly affecting the growth of cells by G1-to S-phase progression. Generally, growth factors are the most well-known activators, which then cause the activation of upstream kinases MEK1/2 MAPK kinases and Raf MAPK

ACCEPTED MANUSCRIPT

kinases (Lei, Wang, Mei et al., 2014). ERK1/2 is reported to be activated by phosphorylation in response to melatonin in neuronal cell cultures (Roy and Belsham, 2002). Studies found

RI PT

that the MAPK/ERK pathway, which has been linked to histone acetylation (Cohen-Armon, Visochek, Rozensal et al., 2007,Lee, McCool, Murdoch et al., 2006), is involved in melatonin-induced proliferation and differentiation of NSCs. Data showed that melatonin

SC

significantly increased the phosphorylation of the main MAPK (ERK1/2) and c-Raf , which are protein kinases involved in the MAPK signaling pathway, and the phosphorylation was

M AN U

significantly reduced when melatonin receptor antagonist was pretreated (Fu et al., 2011,Niles, Pan, Kang et al., 2013,Tocharus et al., 2014). However, whether melatonin will exert its effect when ERK is blocked was not discussed.

TE D

3.3 Neurotrophic factors

Neurotrophic factors are secreted proteins that play important roles in the development and maintenance of the nervous system, influencing the development of distinct neuronal

EP

populations, and maintaining the survival and neuritic arbors of mature neurons into adulthood (Hegarty, O'Keeffe and Sullivan, 2014). Brain-derived neurotrophic factor (BDNF)

AC C

can maintain neuronal survival and regulate synaptic plasticity through its downstream signals including phosphoinositide 3-kinase (PI3K)/Akt, ERK and phospholipase Cγ pathways (Kuczewski, Porcher and Gaiarsa, 2010,Numakawa, Adachi, Richards et al., 2013). Glial cell line-derived neurotrophic factor (GDNF) promotes survival of Dopamine and other neurons, and are proposed to protect neurons in PD (Hong, Mukhida and Mendez, 2008), regulates neuronal survival through activation of cellular signaling, mRNA translation and new protein

ACCEPTED MANUSCRIPT

synthesis. GDNF also protects neurons by suppressing death receptor–caspase pathway, translocation of apoptogenic Bax, and by upregulating of anti-apoptotic Bcl-2 and Bcl-XL

RI PT

(Maruyama and Naoi, 2013).

Studies have found that neurotrophic factors, especially BDNF and GDNF, significantly increased in the cultured NSCs treated with melatonin. It suggests that melatonin could

SC

promote the viability and facilitate neuronal differentiation of NSCs through increasing the

M AN U

level of BDNF and GDNF (Kong et al., 2008,Niles et al., 2004,Sharma et al., 2008).

3.4 Histone acetylation

There is evidence that gene alteration by melatonin is associated with the effects on NSCs (Kim and Rosenfeld, 2010,Sarlak, Jenwitheesuk, Chetsawang et al., 2013). The epigenetic

TE D

mechanisms that modulate DNA without altering genomic sequences involve nucleosomes, which consist of DNA and an octamer of histones. The control of transcription is crucial in cell differentiation and development of the nervous system. In the last decade, histone

EP

modifications have been shown to control many aspects of transcription including higher

AC C

order chromatin structure and gene expression (Lilja, Heldring and Hermanson, 2013). Histones can be post-translationally modified in many ways, for example by acetylation, methylation, phosphorylation, ubiquitination, and sumoylation (Kouzarides, 2007).

Histone acetylation is found to associate with neuronal differentiation (Hsieh, Nakashima, Kuwabara et al., 2004,Marin-Husstege, Muggironi, Liu et al., 2002,Stockhausen, Sjolund, Manetopoulos et al., 2005). Histone hyperacetylation in progenitor cells has been shown to

ACCEPTED MANUSCRIPT

promote neuronal differentiation, while blocking the development of a glial phenotype (Balasubramaniyan, Boddeke, Bakels et al., 2006,Hsieh et al., 2004). Sharma R et al. found

RI PT

that melatonin induced the mRNA expression of HDAC3, HDAC5, and HDAC7. It is likely that enhanced HDAC mRNA levels represent a compensatory feedback mechanism following melatonin-induced histone hyperacetylation (Sharma et al., 2008). Melatonin induced

SC

significant increases in histone H3 and H4 acetylation in the hippocampus and striatum but not in the midbrain and cerebellum were detected by Niles LP, et al. (Niles et al., 2013),

M AN U

suggesting that hippocampus and striatum may be the targets for the epigenetic effects of melatonin, and it also shows the regional differences of melatonin signaling.

In addition to acetylation, the methylation of histone is also closely related to NSCs development. The methylation of histones especially at residues H3K4, H3K9 and H3K27 is

TE D

emerging as some of the key epigenetic marks that control transcription in stem cells and progenitors (Lilja et al., 2013). Nevertheless, the crosstalk between melatonin and histone methylation has not been reported yet, thus the modification of histone and the enzymes

EP

regulating these modifications can be a focus of attention when dissecting the epigenetic

AC C

mechanisms of melatonin influencing NSCs state and fate.

3.5 bHLH transcription factors

In neurodevelopment, determination of neuron and glia cell fates from NSCs during fetal

and adult brain development depends on some bHLH factors (Kageyama, Ohtsuka, Hatakeyama et al., 2005,Ross, Greenberg and Stiles, 2003,Zhou and Anderson, 2002). There are two types of bHLH genes, the repressor type and the activator type. The repressor-type

ACCEPTED MANUSCRIPT

bHLH genes include Hes genes, homologs of Drosophila hairy and Enhancer of split [E(spl)], while the activator-type bHLH genes include Mash1, Math and Neurogenin (Ngn), homologs

RI PT

of Drosophila proneural genes achaete-scute complex and atonal (Kageyama et al., 2005). The activator-type bHLH genes Mash1, Math and Ngn are expressed by differentiating neurons. These bHLH factors form a heterodimer with a ubiquitously expressed bHLH factor,

SC

E47, and activate gene expression by binding to the E box (CANNTG), they not only induce the neuronal-specific gene expression but also inhibit the glia-specific gene expression. While

M AN U

the Hes genes can represses activator-type bHLH genes expression by directly binding to the promoter E47 and antagonizes the activity (Kageyama et al., 2005).

Fu J et al. found that melatonin treatment induce up-regulation of Mash1, NeuroG1, and NeuroD2, which are involved in vertebrate neurogenesis, in the differentiated NSCs compared

TE D

with untreated cells in hypoxia. But other bHLH factors such as Hes1, Hes5, and Id which can regulate the specification of gliogenesis (Jen, Manova and Benezra, 1997,Ohtsuka, Ishibashi, Gradwohl et al., 1999,Ohtsuka, Sakamoto, Guillemot et al., 2001) are not affected by

EP

melatonin (Fu et al., 2011). It is possible that increased neurogenesis caused by melatonin

AC C

supplementation is the consequence of enhanced expression of these transcription factors.

3.6 Anti-apoptotic Bcl-2 family proteins

The anti-apoptotic B-cell lymphoma 2 (Bcl-2) protein family act as key regulators in the

intrinsic or “mitochondrial” apoptosis pathway, they are classified into three different classes depending on their structural and functional properties: (a) anti-apoptotic Bcl-2 proteins including Bcl-2 itself, Bcl-XL (Bcl-extra long), Mcl-1 and Bcl-W; (b) pro-apoptotic proteins

ACCEPTED MANUSCRIPT

Bax (Bcl-2-associated × protein), Bak (Bcl-2-antagonist/killer-1) and potentially Bok (Bcl-2 related ovarian killer); and (c) BH3-only proteins including Bim (Bcl-2 interacting mediator),

RI PT

PUMA (p53 upregulated modulator of apoptosis), Bid (BH3 interacting domain death agonist), Bik (Bcl-2 interacting killer), Bad (Bcl-2 associated death promoter), Bmf (Bcl-2 modifying factor), Hrk (Hara-kiri) and Noxa (Latin for “damage”) (Anilkumar and Prehn, 2014).

SC

Anti-apoptotic Bcl-2 family proteins as well as Bax and Bak are often constitutively expressed in cells and activation of the mitochondrial apoptosis pathway through

M AN U

pro-apoptotic Bcl-2 proteins is able to activate different cell death pathways including apoptosis (Kilbride and Prehn, 2013). Numerous literatures show that Bcl-2 plays an important role in initiating or inhibiting apoptosis during neuronal development and injury. For example, overexpression of Bcl-2 inhibited Bax-mediated cytochrome-c release, caspase

TE D

activation and cell death (Putcha, Deshmukh and Johnson, 1999); Bcl-2 overexpression protected hippocampal neurons against glutamate mediated excitotoxicity (Wong, Ralph, Walmsley et al., 2005). Interestingly, melatonin was also found to induced a significant

EP

increase in the ratio of Bcl-2/Bax and inhibition of caspase-3 activation (Fu et al., 2011) which indicate that the overexpression of the Bcl-2 family proteins may contributes to the

AC C

protection effects of melatonin on injured NSCs.

3.7 Nrf2 signaling

The nuclear factor erythroid 2-related factor 2 (Nrf2)-antioxidant response element (ARE) pathway is a primary sensor and a master regulator of oxidative stress via its ability to modulate the expression of hundreds of antioxidant and detoxifying genes (Johnson, Johnson,

ACCEPTED MANUSCRIPT

Kraft et al., 2008). Nrf2 is one of the cap'n'collar (CNC) transcription factors and ubiquitously expressed in all human tissues including the brain (Moi, Chan, Asunis et al., 1994). Nrf2

RI PT

binds to the ARE, which is an enhancer element located in the 5 flanking region of many phase II detoxifying and antioxidant genes, to regulate this pathway. Under basal conditions, Nrf2 is negatively regulated in the cytoplasm by kelch-like ECH associating protein 1

SC

(Keap1). Keap1 prevents the nuclear translocation of Nrf2 and functions as an adaptor protein to E3 ligase to promote the degradation of Nrf2 via the ubiquitin proteasome system (UPS)

M AN U

(Gan and Johnson, 2014). Upon disruption of Keap1-Nrf2 binding, Nrf2 translocates to the nucleus and binds with small Maf proteins. The formed heterodimer binds to the ARE and coordinates the transcription of genes involved in phase II detoxification and antioxidant defense as so to maintain redox balance (Itoh, Chiba, Takahashi et al., 1997).

TE D

A number of studies have shown the dynamic changes of Nrf2/ARE pathway in disease demonstrating the accumulation of oxidative damage and exert neuroprotection effect against oxidative stress and protect against protein oxidation and misfolding, such as Aβ and tau

EP

neurofilament tangles (NFTs) in AD (Gan and Johnson, 2014). Recently, some studies also

AC C

suggest the protection effect against oxidative stress of Nrf2 pathway on NSCs/NPCs (Abdanipour, Tiraihi, Noori-Zadeh et al., 2014,Li, Johnson, Calkins et al., 2005,Ni, Liu, Ren et al., 2014). Kärkkäinen V., et al. found that Nrf2 regulates neurogenesis and protects NPCs against Aβ toxicity. Nrf2 prevents the ischemia-induced decrease in newborn neurons in the SVZ of the DG; lentivirus-mediated overexpression of Nrf2 gene or by treatment with pyrrolidine dithiocarbamate (PDTC), an Nrf2 activating compound, increased the growth of NPC neurospheres as well as the neuronal differentiation of NPCs. Aβ-induced toxicity and

ACCEPTED MANUSCRIPT

reduction in neurosphere proliferation were prevented by Nrf2 overexpression, conversely, Nrf2 deficiency enhanced the Aβ-induced reduction of neuronal differentiation (Karkkainen,

RI PT

Pomeshchik, Savchenko et al., 2014).

Accordingly, some literatures demonstrated that melatonin protect cells against oxidative stress, brain injury and neurotoxicity through the activation of Nrf2/ARE signaling pathway

SC

(Deng, Zhu, Mi et al., 2014,Ding, Wang, Xu et al., 2014,Wang, Ma, Meng et al., 2012), yet few literature indicate the involvement of Nrf2 in the mechanism of melatonin in NSCs/NPCs,

M AN U

raising the question whether the pathway play a role in the mechanism of melatonin regulating the NSCs survival and development.

3.8 Others

TE D

Interestingly, studies showed that melatonin ameliorates dexamethasone-induced inhibitory effects on the proliferation of cultured progenitor cells obtained from adult rat hippocampus. Dexamethasone (the glucocorticoid receptor agonist) increased the glucocorticoid receptor

EP

protein but decreased the level of MT1 melatonin receptor, whereas melatonin increased the

AC C

level of MT1 melatonin receptor but decreased the glucocorticoid receptor protein, suggesting the crosstalk and cross regulation between the melatonin receptor and the glucocorticoid receptor on hippocampal progenitor cell proliferation (Ekthuwapranee et al., 2014).

4. Melatonin derivative and analogues

Since melatonin plays an important role in NSC growth and differentiation, melatonin-based compounds could show a neurogenic profile of paramount importance in the

ACCEPTED MANUSCRIPT

search of new therapies for AD or other neurodegenerative diseases and neurodevelopmental diseases. However, pharmacokinetic issues such as limited oral bioavailability and short

RI PT

half-life time limit its usage. Thus, to design and synthesis of melatonin analogues, especially, the multi-targeted derivatives with improved PD/PK characters is evoking the interest of the scientific community.

SC

Recently, patents and patent applications from 2012 to September 2014 of which melatonin or synthetic analogues were claimed for the prevention or treatment of pathological conditions (Rivara, Pala, Bedini et al., 2015). For example, a series of novel

M AN U

had been well reviewed

hybrid molecules obtained by linking melatonin, or its oxidation products, to a tetrahydroacridine unit via a carbamate bond, which are described as potent cholinesterase enzyme inhibitors, antioxidants and able to prevent the aggregation of β-amyloid (Rivara et

TE D

al., 2015). Here the melatonin derivatives that are supposed to play a role in stem cells were summarized below (also summarized in Table 2).

EP

Agomelatine (S20098), a synthetic melatonin analogue that binds to MT1 and MT2 receptors (Audinot, Mailliet, Lahaye-Brasseur et al., 2003), have been reported to increases

AC C

progenitor cell proliferation in the DG, which requires an intact diurnal corticosterone rhythm (AlAhmed and Herbert, 2010). SSH-BM-I was synthesized from tryptamine by using a newly developed synthetic method, and it has structural similarity to melatonin. It had been reported that SSH-BM-I increases osteoblasts in scales of gold fish (Suzuki, Somei, Kitamura et al., 2008) and stimulates terminal osteoblast differentiation in a cell differentiation stage–dependent manner (Mikami, Somei and Tsuda, 2011). Data showed that SSH-BM-I

ACCEPTED MANUSCRIPT

increased the formation of mineralized nodules in mature osteoblast ROS17/2.8 cells yet suppressed bone morphogenetic protein 2 (BMP-2, which is one of the most powerful

RI PT

cytokines to promote mesenchymal progenitors differentiate into particular cell types) induced osteoblast differentiation in mesenchymal progenitor ROB-C26 cells. Several 2-vinyl-8-hydroxyquinoline derivatives were found as potential antioxidants and regulators of

SC

H2O2-induced oxidative stress in rat bone marrow mesenchymal stem cells, and one of these compounds, Compound 4, which has similar structure with melatonin, shows better

M AN U

proliferative activities and exhibited strong protection effects against oxidative stress in MSCs (Wang, Zeng, Li et al., 2010). Pradoldej Sompol et al. reported that N-acetylserotonin (NAS), the immediate precursor of melatonin, regulates an early event of neurogenesis by increasing NPCs proliferation in both active and sleeping phases of the mice and it significantly

TE D

enhances NPCs proliferation in sleep-deprived mice (Sompol, Liu, Baba et al., 2011).

Recently, a new family of melatonin−N,N-dibenzyl (N-methyl) amine hybrids with a balanced multifunctional profile including neurogenic, antioxidant, cholinergic, and

EP

neuroprotective properties at low micromolar concentrations were synthesized by linking the

AC C

melatonin framework and the protonable N,N-dibenzyl (N-methyl) amine, which is present in the well-known acetylcholinesterase (AChE) inhibitor AP2238 and could interact with the AChE catalytic active site (Piazzi, Cavalli, Belluti et al., 2007,Piazzi, Rampa, Bisi et al., 2003,Rizzo, Bartolini, Ceccarini et al., 2010). They promoted the maturation of NSCs into a neuronal phenotype and protected neural cells against mitochondrial oxidative stress. They also have antioxidant properties, and could inhibit human AChE (Lopez-Iglesias, Perez, Morales-Garcia et al., 2014). A series of melatonin analogues resulting from the hybridization

ACCEPTED MANUSCRIPT

of both pinoline and melatonin structures was synthesized and pharmacological evaluation indicated that pinoline at trace concentration and compound 2 were able to stimulate early

RI PT

neurogenesis and neuronal maturation in an in vitro model of neural stem cells isolated from the adult rat SVZ (de la Fuente Revenga, Perez, Morales-Garcia et al., 2015). In addition, several melatonin based compounds were synthesized by replacing the acetamido group with

SC

a series of reversed amides and azoles, and were found to promote differentiation of rat NSCs to a neuronal phenotype in vitro (de la Fuente Revenga, Fernandez-Saez, Herrera-Arozamena

M AN U

et al., 2015).

As the complexity of AD pathology, the therapeutic paradigm one-compound-one-target has failed till now. Thus, the design of molecules that are able to interact with two or more complementary targets is catching much more people’s attention, with the expectation that

TE D

these multi-targeted ligands (MTLs) may represent important advantages in the treatment of AD and other neurodegenerative pathologies (Cavalli, Bolognesi, Minarini et al., 2008,Leon,

EP

Garcia and Marco-Contelles, 2013,Morphy and Rankovic, 2005).

AC C

5. Perspective and conclusions

NSCs are self-renewing progenitors that are capable of acquiring a neuronal or glial

phenotype. As such, NSC transplantation may serve as a potential therapeutic approach for several neurodegenerative diseases. Yet engrafted stem cells in neurodegenerative diseases’ models have poor survival rates and may not acquire the desired phenotype (Arenas, 2002). Therefore, stem cell transplantation could benefit from combined treatment with compounds that improve graft integration and differentiation. Varies data in the literature have shown that

ACCEPTED MANUSCRIPT

melatonin modulates NSCs proliferation, survival and differentiation. Because melatonin can cross the blood brain barrier and is soluble in both lipid and water (Reiter, Tan, Manchester et

RI PT

al., 2007), this endogenous modulator might be beneficially used for modulating NSCs, pro-proliferation and pro-survival of endogenous NSCs as well as engrafted NSCs for CNS diseases.

SC

Signaling and transcriptional responses elicited by the exposure of NSCs to melatonin are now partially disclosed (see Fig. 1), but obviously further work is needed to provide a more

M AN U

precise picture of melatonin-mediated regulatory and differentiation mechanisms. For instance, the correlation between melatonin and microRNAs (miRNAs) on influencing the cell fate of NSCs, since an emerging hypothesis is that miRNAs play a central role in controlling stem cell-fate determination (Stevanato and Sinden, 2014). miRNAs are integrated

TE D

within networks that form both positive and negative feedback loops to promote and stabilize cell fate choice at multiple levels (Shenoy and Blelloch, 2014) and channelize the cellular physiology toward neuronal differentiation or indirectly influence neurogenesis by

EP

regulating the proliferation and self-renewal of NSCs and are dysregulated in several

AC C

neurodegenerative diseases (Iyengar, Choudhary, Sarangdhar et al., 2014). Meza-Sosa KF et al. found that, by regulating different target genes, microRNAs let-7, microRNA-9 and microRNA-124 have been shown to promote the differentiation of NSCs/NPCs into specific neural cell types while microRNA-25, microRNA-134 and microRNA-137 have been characterized as microRNAs that induce the proliferation of NSCs/NPCs (Meza-Sosa, Pedraza-Alva and Perez-Martinez, 2014). Interestingly, recent studies found that melatonin significantly modulates the expression of miRNAs (Fu, Zhao, Zheng et al., 2014,Lee, Kim,

ACCEPTED MANUSCRIPT

Youn et al., 2011), including let-7a and miR-124, which are involved with neurogenesis, indicating that regulating the expression of miRNAs may be involved in the mechanism of

RI PT

melatonin to modulate the development of NSCs, which is so far poorly investigated. Further studies should be carried out to uncover the roles of miRNAs in the effects of melatonin on NSCs.

SC

Given small molecules are more druggable, it evokes a great interest to develop potential orally administrated small molecules to modulate NSCs/NPCs in vivo (Rishton,

M AN U

2008,Swaminathan, Kumar, Halder Sinha et al., 2014). As we know, CNS diseases, including AD and PD, are multifactorial pathologies. MTLs should be more efficiency to combat these diseases. To design of MTLs-based melatonin derivatives or analogues could be a new frontier of melatonin in near future. Further investigations into the targets and functions of

TE D

melatonin-based MTLs will be beneficial to develop new strategies for the therapy of neurodegenerative diseases.

study

was

AC C

This

EP

Acknowledgments

supported

by

Guangdong

Provincial

International

Cooperation Project of Science & Technology (No. 2013B051000038) and National Science

Foundation

of

China

(No.31371070) to Pi R.,

and

Natural

Scientific

and

Technological Cooperation between the Italian Republic and the People’s Republic of China for Year 2013-2015 (No. MAE-M00705\CN13MO9) to Pi R. and Macchia M..

Conflict of interests

ACCEPTED MANUSCRIPT

None.

References

RI PT

Abdanipour, A., Tiraihi, T., Noori-Zadeh, A., Majdi, A. and Gosaili, R., 2014. Evaluation of lovastatin effects on expression of anti-apoptotic Nrf2 and PGC-1alpha genes in neural stem cells treated with hydrogen peroxide, Mol Neurobiol. 49, 1364-72.

AlAhmed, S. and Herbert, J., 2010. Effect of agomelatine and its interaction with the daily corticosterone rhythm on progenitor cell proliferation in the dentate gyrus of the adult rat, Neuropharmacology. 59, 375-9.

SC

Anilkumar, U. and Prehn, J.H., 2014. Anti-apoptotic BCL-2 family proteins in acute neural injury, Front Cell Neurosci. 8, 281.

Arenas, E., 2002. Stem cells in the treatment of Parkinson's disease, Brain Res Bull. 57, 795-808.

M AN U

Audinot, V., Mailliet, F., Lahaye-Brasseur, C., Bonnaud, A., Le Gall, A., Amosse, C., Dromaint, S., Rodriguez, M., Nagel, N., Galizzi, J.P., Malpaux, B., Guillaumet, G., Lesieur, D., Lefoulon, F., Renard, P., Delagrange, P. and Boutin, J.A., 2003. New selective ligands of human cloned melatonin MT1 and MT2 receptors, Naunyn Schmiedebergs Arch Pharmacol. 367, 553-61.

Balasubramaniyan, V., Boddeke, E., Bakels, R., Kust, B., Kooistra, S., Veneman, A. and Copray, S., 2006. Effects of histone deacetylation inhibition on neuronal differentiation of embryonic mouse neural stem cells, Neuroscience. 143, 939-51.

Bubenik, G.A. and Konturek, S.J., 2011. Melatonin and aging: prospects for human treatment, J Physiol Pharmacol. 62, 13-9.

TE D

Cavalli, A., Bolognesi, M.L., Minarini, A., Rosini, M., Tumiatti, V., Recanatini, M. and Melchiorre, C., 2008. Multi-target-directed ligands to combat neurodegenerative diseases, J Med Chem. 51, 347-72. 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., Chung, S.Y., Chang, C.L. and Yip, H.K., 2014. Additional benefit of combined therapy with melatonin and apoptotic adipose-derived mesenchymal stem cell against sepsis-induced kidney injury, J Pineal Res. 57, 16-32.

EP

Chen, M., Tan, M., Jing, M., Liu, A., Liu, Q., Wen, S., Chen, Z., Chao, X., He, X., Ramassamy, C., Gao, Y. and Pi, R., 2014. Berberine protects homocysteic acid-induced HT-22 cell death: involvement of Akt pathway, Metab Brain Dis.

AC C

Chern, C.M., Liao, J.F., Wang, Y.H. and Shen, Y.C., 2012. Melatonin ameliorates neural function by promoting endogenous neurogenesis through the MT2 melatonin receptor in ischemic-stroke mice, Free Radic Biol Med. 52, 1634-47.

Chetsawang, J., Govitrapong, P. and Chetsawang, B., 2007. Melatonin inhibits MPP+-induced caspase-mediated death pathway and DNA fragmentation factor-45 cleavage in SK-N-SH cultured cells, J Pineal Res. 43, 115-20.

Cohen-Armon, M., Visochek, L., Rozensal, D., Kalal, A., Geistrikh, I., Klein, R., Bendetz-Nezer, S., Yao, Z. and Seger, R., 2007. DNA-independent PARP-1 activation by phosphorylated ERK2 increases Elk1 activity: a link to histone acetylation, Mol Cell. 25, 297-308. Cutando, A., Aneiros-Fernandez, J., Lopez-Valverde, A., Arias-Santiago, S., Aneiros-Cachaza, J. and Reiter, R.J., 2011. 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. 56, 944-50. de la Fuente Revenga, M., Fernandez-Saez, N., Herrera-Arozamena, C., Morales-Garcia, J.A., Alonso-Gil, S.,

ACCEPTED MANUSCRIPT

Perez-Castillo, A., Caignard, D.H., Rivara, S. and Rodriguez-Franco, M.I., 2015. Novel N-Acetyl Bioisosteres of Melatonin: Melatonergic Receptor Pharmacology, Physicochemical Studies, and Phenotypic Assessment of Their Neurogenic Potential, J Med Chem. 58, 4998-5014. de la Fuente Revenga, M., Perez, C., Morales-Garcia, J.A., Alonso-Gil, S., Perez-Castillo, A., Caignard, D.H., Yanez, M., Gamo, A.M. and Rodriguez-Franco, M.I., 2015. Neurogenic Potential Assessment and Pharmacological Characterization of 6-Methoxy-1,2,3,4-tetrahydro-beta-carboline (Pinoline) and Melatonin-Pinoline

RI PT

Hybrids, ACS Chem Neurosci.

Deng, Y., Zhu, J., Mi, C., Xu, B., Jiao, C., Li, Y., Xu, D., Liu, W. and Xu, Z., 2014. Melatonin Antagonizes Mn-Induced Oxidative Injury Through the Activation of Keap1-Nrf2-ARE Signaling Pathway in the Striatum of Mice, Neurotox Res.

Ding, K., Wang, H., Xu, J., Li, T., Zhang, L., Ding, Y., Zhu, L., He, J. and Zhou, M., 2014. Melatonin stimulates antioxidant enzymes and reduces oxidative stress in experimental traumatic brain injury: the Nrf2-ARE

SC

signaling pathway as a potential mechanism, Free Radic Biol Med. 73, 1-11.

Dubocovich, M.L., Masana, M.I., Iacob, S. and Sauri, D.M., 1997. Melatonin receptor antagonists that differentiate between the human Mel1a and Mel1b recombinant subtypes are used to assess the Pharmacol. 355, 365-75.

M AN U

pharmacological profile of the rabbit retina ML1 presynaptic heteroreceptor, Naunyn Schmiedebergs Arch Ekthuwapranee, K., Sotthibundhu, A., Tocharus, C. and Govitrapong, P., 2014. Melatonin ameliorates dexamethasone-induced inhibitory effects on the proliferation of cultured progenitor cells obtained from adult rat hippocampus, J Steroid Biochem Mol Biol.

Escames, G., Lopez, A., Garcia, J.A., Garcia, L., Acuna-Castroviejo, D., Garcia, J.J. and Lopez, L.C., 2010. The role of mitochondria in brain aging and the effects of melatonin, Curr Neuropharmacol. 8, 182-93. Fishell, G. and Kriegstein, A.R., 2003. Neurons from radial glia: the consequences of asymmetric inheritance,

TE D

Curr Opin Neurobiol. 13, 34-41.

Fu, J., Zhao, S.D., Liu, H.J., Yuan, Q.H., Liu, S.M., Zhang, Y.M., Ling, E.A. and Hao, A.J., 2011. Melatonin promotes proliferation and differentiation of neural stem cells subjected to hypoxia in vitro, J Pineal Res. 51, 104-12. Fu, S., Zhao, H., Zheng, Z., Li, J. and Zhang, W., 2014. Melatonin regulating the expression of miRNAs involved in hair folli-cle cycle of cashmere goats skin, Yi Chuan. 36, 1235-42. Fujioka, A., Fujioka, T., Tsuruta, R., Izumi, T., Kasaoka, S. and Maekawa, T., 2011. Effects of a constant light

EP

environment on hippocampal neurogenesis and memory in mice, Neurosci Lett. 488, 41-4. Gan, L. and Johnson, J.A., 2014. Oxidative damage and the Nrf2-ARE pathway in neurodegenerative diseases, Biochim Biophys Acta. 1842, 1208-18.

AC C

Gotz, M. and Huttner, W.B., 2005. The cell biology of neurogenesis, Nat Rev Mol Cell Biol. 6, 777-88. Gunasingh, M.J., Philip, J.E., Ashok, B.S., Kirubagaran, R., Jebaraj, W.C., Davis, G.D., Vignesh, S., Dhandayuthapani, S. and Jayakumar, R., 2008. Melatonin prevents amyloid protofibrillar induced oxidative imbalance and biogenic amine catabolism, Life Sci. 83, 96-102.

Guzman-Marin, R., Suntsova, N., Bashir, T., Szymusiak, R. and McGinty, D., 2007. Cell proliferation in the dentate gyrus of the adult rat fluctuates with the light-dark cycle, Neurosci Lett. 422, 198-201.

Hardeland, R. and Poeggeler, B., 2003. Non-vertebrate melatonin, J Pineal Res. 34, 233-41. Hegarty, S.V., O'Keeffe, G.W. and Sullivan, A.M., 2014. Neurotrophic factors: from neurodevelopmental regulators to novel therapies for Parkinson's disease, Neural Regen Res. 9, 1708-1711. Hong, M., Mukhida, K. and Mendez, I., 2008. GDNF therapy for Parkinson's disease, Expert Rev Neurother. 8, 1125-39. Honmou, O., 2015. [Stem cell therapy for dementia], Brain Nerve. 67, 93-8.

ACCEPTED MANUSCRIPT

Hsieh, J., Nakashima, K., Kuwabara, T., Mejia, E. and Gage, F.H., 2004. Histone deacetylase inhibition-mediated neuronal differentiation of multipotent adult neural progenitor cells, Proc Natl Acad Sci U S A. 101, 16659-64. Itoh, K., Chiba, T., Takahashi, S., Ishii, T., Igarashi, K., Katoh, Y., Oyake, T., Hayashi, N., Satoh, K., Hatayama, I., Yamamoto, M. and Nabeshima, Y., 1997. An Nrf2/small Maf heterodimer mediates the induction of phase 313-22.

RI PT

II detoxifying enzyme genes through antioxidant response elements, Biochem Biophys Res Commun. 236, Iyengar, B.R., Choudhary, A., Sarangdhar, M.A., Venkatesh, K.V., Gadgil, C.J. and Pillai, B., 2014. Non-coding RNA interact to regulate neuronal development and function, Front Cell Neurosci. 8, 47.

Jen, Y., Manova, K. and Benezra, R., 1997. Each member of the Id gene family exhibits a unique expression pattern in mouse gastrulation and neurogenesis, Dev Dyn. 208, 92-106.

Jilg, A., Lesny, S., Peruzki, N., Schwegler, H., Selbach, O., Dehghani, F. and Stehle, J.H., 2010. Temporal dynamics

SC

of mouse hippocampal clock gene expression support memory processing, Hippocampus. 20, 377-88.

Johnson, J.A., Johnson, D.A., Kraft, A.D., Calkins, M.J., Jakel, R.J., Vargas, M.R. and Chen, P.C., 2008. The Nrf2-ARE pathway: an indicator and modulator of oxidative stress in neurodegeneration, Ann N Y Acad Sci.

M AN U

1147, 61-9.

Kageyama, R., Ohtsuka, T., Hatakeyama, J. and Ohsawa, R., 2005. Roles of bHLH genes in neural stem cell differentiation, Exp Cell Res. 306, 343-8.

Karasek, M. and Winczyk, K., 2006. Melatonin in humans, J Physiol Pharmacol. 57 Suppl 5, 19-39. Karkkainen, V., Pomeshchik, Y., Savchenko, E., Dhungana, H., Kurronen, A., Lehtonen, S., Naumenko, N., Tavi, P., Levonen, A.L., Yamamoto, M., Malm, T., Magga, J., Kanninen, K.M. and Koistinaho, J., 2014. Nrf2 regulates neurogenesis and protects neural progenitor cells against Abeta toxicity, Stem Cells. 32, 1904-16. Kempermann, G., Gast, D., Kronenberg, G., Yamaguchi, M. and Gage, F.H., 2003. Early determination and

TE D

long-term persistence of adult-generated new neurons in the hippocampus of mice, Development. 130, 391-9.

Kilbride, S.M. and Prehn, J.H., 2013. Central roles of apoptotic proteins in mitochondrial function, Oncogene. 32, 2703-11.

Kim, H.J. and Rosenfeld, M.G., 2010. Epigenetic control of stem cell fate to neurons and glia, Arch Pharm Res. 33, 1467-73.

EP

Kimiwada, T., Sakurai, M., Ohashi, H., Aoki, S., Tominaga, T. and Wada, K., 2009. Clock genes regulate neurogenic transcription factors, including NeuroD1, and the neuronal differentiation of adult neural stem/progenitor cells, Neurochem Int. 54, 277-85.

AC C

Klongpanichapak, S., Phansuwan-Pujito, P., Ebadi, M. and Govitrapong, P., 2007. Melatonin protects SK-N-SH neuroblastoma cells from amphetamine-induced neurotoxicity, J Pineal Res. 43, 65-73.

Kong, X., Li, X., Cai, Z., Yang, N., Liu, Y., Shu, J., Pan, L. and Zuo, P., 2008. Melatonin regulates the viability and differentiation of rat midbrain neural stem cells, Cell Mol Neurobiol. 28, 569-79.

Konturek, S.J., Konturek, P.C., Brzozowska, I., Pawlik, M., Sliwowski, Z., Czesnikiewicz-Guzik, M., Kwiecien, S., Brzozowski, T., Bubenik, G.A. and Pawlik, W.W., 2007. Localization and biological activities of melatonin in intact and diseased gastrointestinal tract (GIT), J Physiol Pharmacol. 58, 381-405.

Kouzarides, T., 2007. Chromatin modifications and their function, Cell. 128, 693-705. Kriegstein, A. and Alvarez-Buylla, A., 2009. The glial nature of embryonic and adult neural stem cells, Annu Rev Neurosci. 32, 149-84. Kuczewski, N., Porcher, C. and Gaiarsa, J.L., 2010. Activity-dependent dendritic secretion of brain-derived neurotrophic factor modulates synaptic plasticity, Eur J Neurosci. 32, 1239-44.

ACCEPTED MANUSCRIPT

Lee, E.R., McCool, K.W., Murdoch, F.E. and Fritsch, M.K., 2006. Dynamic changes in histone H3 phosphoacetylation during early embryonic stem cell differentiation are directly mediated by mitogenand stress-activated protein kinase 1 via activation of MAPK pathways, J Biol Chem. 281, 21162-72. Lee, S.E., Kim, S.J., Youn, J.P., Hwang, S.Y., Park, C.S. and Park, Y.S., 2011. MicroRNA and gene expression analysis of melatonin-exposed human breast cancer cell lines indicating involvement of the anticancer effect, J Pineal Res. 51, 345-52.

RI PT

Lee, S.H., Chun, W., Kong, P.J., Han, J.A., Cho, B.P., Kwon, O.Y., Lee, H.J. and Kim, S.S., 2006. Sustained activation of Akt by melatonin contributes to the protection against kainic acid-induced neuronal death in hippocampus, J Pineal Res. 40, 79-85.

Lee, S.J., Jung, Y.H., Oh, S.Y., Yun, S.P. and Han, H.J., 2014. Melatonin enhances the human mesenchymal stem cells motility via melatonin receptor 2 coupling with Galphaq in skin wound healing, J Pineal Res. 57, 393-407.

SC

Lee, Y., Lee, S., Lee, S.R., Park, K., Hong, Y., Lee, M., Park, S., Jin, Y., Chang, K.T. and Hong, Y., 2014. Beneficial effects of melatonin combined with exercise on endogenous neural stem/progenitor cells proliferation after spinal cord injury, Int J Mol Sci. 15, 2207-22.

M AN U

Lei, Y.Y., Wang, W.J., Mei, J.H. and Wang, C.L., 2014. Mitogen-Activated Protein Kinase Signal Transduction in Solid Tumors, Asian Pac J Cancer Prev. 15, 8539-8548.

Leon, R., Garcia, A.G. and Marco-Contelles, J., 2013. Recent advances in the multitarget-directed ligands approach for the treatment of Alzheimer's disease, Med Res Rev. 33, 139-89. Li, J., Johnson, D., Calkins, M., Wright, L., Svendsen, C. and Johnson, J., 2005. Stabilization of Nrf2 by tBHQ confers protection against oxidative stress-induced cell death in human neural stem cells, Toxicol Sci. 83, 313-28.

Lilja, T., Heldring, N. and Hermanson, O., 2013. Like a rolling histone: epigenetic regulation of neural stem cells 1830, 2354-60.

TE D

and brain development by factors controlling histone acetylation and methylation, Biochim Biophys Acta. Lopez-Iglesias, B., Perez, C., Morales-Garcia, J.A., Alonso-Gil, S., Perez-Castillo, A., Romero, A., Lopez, M.G., Villarroya, M., Conde, S. and Rodriguez-Franco, M.I., 2014. New melatonin-N,N-dibenzyl(N-methyl)amine hybrids: potent neurogenic agents with antioxidant, cholinergic, and neuroprotective properties as innovative drugs for Alzheimer's disease, J Med Chem. 57, 3773-85.

EP

Luchetti, F., Canonico, B., Bartolini, D., Arcangeletti, M., Ciffolilli, S., Murdolo, G., Piroddi, M., Papa, S., Reiter, R.J. and Galli, F., 2014. Melatonin regulates mesenchymal stem cell differentiation: a review, J Pineal Res. 56, 382-97.

AC C

Luchetti, F., Canonico, B., Betti, M., Arcangeletti, M., Pilolli, F., Piroddi, M., Canesi, L., Papa, S. and Galli, F., 2010. Melatonin signaling and cell protection function, FASEB J. 24, 3603-24.

Marin-Husstege, M., Muggironi, M., Liu, A. and Casaccia-Bonnefil, P., 2002. Histone deacetylase activity is necessary for oligodendrocyte lineage progression, J Neurosci. 22, 10333-45.

Martino, G., Pluchino, S., Bonfanti, L. and Schwartz, M., 2011. Brain regeneration in physiology and pathology: the immune signature driving therapeutic plasticity of neural stem cells, Physiol Rev. 91, 1281-304.

Maruyama, W. and Naoi, M., 2013. ''70th Birthday Professor Riederer'' induction of glial cell line-derived and brain-derived neurotrophic factors by rasagiline and (-)deprenyl: a way to a disease-modifying therapy?, J Neural Transm. 120, 83-9. Mazna, P., Grycova, L., Balik, A., Zemkova, H., Friedlova, E., Obsilova, V., Obsil, T. and Teisinger, J., 2008. The role of proline residues in the structure and function of human MT2 melatonin receptor, J Pineal Res. 45, 361-72.

ACCEPTED MANUSCRIPT

Meza-Sosa, K.F., Pedraza-Alva, G. and Perez-Martinez, L., 2014. microRNAs: key triggers of neuronal cell fate, Front Cell Neurosci. 8, 175. Mikami, Y., Somei, M. and Tsuda, H., 2011. SSH-BM-I, a tryptamine derivative, stimulates mineralization in terminal osteoblast differentiation but inhibits osteogenesis of pre-committed progenitor cells, J Pharmacol Sci. 116, 63-72. Miyata, T., Kawaguchi, A., Saito, K., Kawano, M., Muto, T. and Ogawa, M., 2004. Asymmetric production of

RI PT

surface-dividing and non-surface-dividing cortical progenitor cells, Development. 131, 3133-45.

Moi, P., Chan, K., Asunis, I., Cao, A. and Kan, Y.W., 1994. Isolation of NF-E2-related factor 2 (Nrf2), a NF-E2-like basic leucine zipper transcriptional activator that binds to the tandem NF-E2/AP1 repeat of the beta-globin locus control region, Proc Natl Acad Sci U S A. 91, 9926-30.

Moriya, T., Horie, N., Mitome, M. and Shinohara, K., 2007. Melatonin influences the proliferative and differentiative activity of neural stem cells, J Pineal Res. 42, 411-8. Chem. 48, 6523-43.

SC

Morphy, R. and Rankovic, Z., 2005. Designed multiple ligands. An emerging drug discovery paradigm, J Med Ni, N., Liu, Q., Ren, H., Wu, D., Luo, C., Li, P., Wan, J.B. and Su, H., 2014. Ginsenoside Rb1 protects rat neural

M AN U

progenitor cells against oxidative injury, Molecules. 19, 3012-24.

Niles, L.P., Armstrong, K.J., Rincon Castro, L.M., Dao, C.V., Sharma, R., McMillan, C.R., Doering, L.C. and Kirkham, D.L., 2004. Neural stem cells express melatonin receptors and neurotrophic factors: colocalization of the MT1 receptor with neuronal and glial markers, BMC Neurosci. 5, 41.

Niles, L.P., Pan, Y., Kang, S. and Lacoul, A., 2013. Melatonin induces histone hyperacetylation in the rat brain, Neurosci Lett. 541, 49-53.

Noctor, S.C., Martinez-Cerdeno, V., Ivic, L. and Kriegstein, A.R., 2004. Cortical neurons arise in symmetric and asymmetric division zones and migrate through specific phases, Nat Neurosci. 7, 136-44.

TE D

Numakawa, T., Adachi, N., Richards, M., Chiba, S. and Kunugi, H., 2013. Brain-derived neurotrophic factor and glucocorticoids: reciprocal influence on the central nervous system, Neuroscience. 239, 157-72. Ohtsuka, T., Ishibashi, M., Gradwohl, G., Nakanishi, S., Guillemot, F. and Kageyama, R., 1999. Hes1 and Hes5 as notch effectors in mammalian neuronal differentiation, EMBO J. 18, 2196-207. Ohtsuka, T., Sakamoto, M., Guillemot, F. and Kageyama, R., 2001. Roles of the basic helix-loop-helix genes Hes1 and Hes5 in expansion of neural stem cells of the developing brain, J Biol Chem. 276, 30467-74. A.,

EP

Piazzi, L., Cavalli, A., Belluti, F., Bisi, A., Gobbi, S., Rizzo, S., Bartolini, M., Andrisano, V., Recanatini, M. and Rampa, 2007.

Extensive

SAR

and

computational

studies

of

3-{4-[(benzylmethylamino)methyl]phenyl}-6,7-dimethoxy-2H-2-chromenone (AP2238) derivatives, J Med

AC C

Chem. 50, 4250-4.

Piazzi, L., Rampa, A., Bisi, A., Gobbi, S., Belluti, F., Cavalli, A., Bartolini, M., Andrisano, V., Valenti, P. and Recanatini,

M.,

2003.

3-(4-[[Benzyl(methyl)amino]methyl]phenyl)-6,7-dimethoxy-2H-2-chromenone

(AP2238) inhibits both acetylcholinesterase and acetylcholinesterase-induced beta-amyloid aggregation: a

dual function lead for Alzheimer's disease therapy, J Med Chem. 46, 2279-82.

Putcha, G.V., Deshmukh, M. and Johnson, E.M., Jr., 1999. BAX translocation is a critical event in neuronal apoptosis: regulation by neuroprotectants, BCL-2, and caspases, J Neurosci. 19, 7476-85.

Ramirez-Rodriguez, G., Klempin, F., Babu, H., Benitez-King, G. and Kempermann, G., 2009. Melatonin modulates cell survival of new neurons in the hippocampus of adult mice, Neuropsychopharmacology. 34, 2180-91. Reiter, R.J., Paredes, S.D., Manchester, L.C. and Tan, D.X., 2009. Reducing oxidative/nitrosative stress: a newly-discovered genre for melatonin, Crit Rev Biochem Mol Biol. 44, 175-200. Reiter, R.J., Tan, D.X., Manchester, L.C., Paredes, S.D., Mayo, J.C. and Sainz, R.M., 2009. Melatonin and

ACCEPTED MANUSCRIPT

reproduction revisited, Biol Reprod. 81, 445-56. Reiter, R.J., Tan, D.X., Manchester, L.C., Pilar Terron, M., Flores, L.J. and Koppisepi, S., 2007. Medical implications of melatonin: receptor-mediated and receptor-independent actions, Adv Med Sci. 52, 11-28. Reppert, S.M., Godson, C., Mahle, C.D., Weaver, D.R., Slaugenhaupt, S.A. and Gusella, J.F., 1995. Molecular characterization of a second melatonin receptor expressed in human retina and brain: the Mel1b melatonin receptor, Proc Natl Acad Sci U S A. 92, 8734-8.

RI PT

Rishton, G.M., 2008. Small molecules that promote neurogenesis in vitro, Recent Pat CNS Drug Discov. 3, 200-8. Rivara, S., Lorenzi, S., Mor, M., Plazzi, P.V., Spadoni, G., Bedini, A. and Tarzia, G., 2005. Analysis of structure-activity relationships for MT2 selective antagonists by melatonin MT1 and MT2 receptor models, J Med Chem. 48, 4049-60.

Rivara, S., Pala, D., Bedini, A. and Spadoni, G., 2015. Therapeutic uses of melatonin and melatonin derivatives: a patent review (2012 - 2014), Expert Opin Ther Pat. 1-17.

SC

Rizzo, S., Bartolini, M., Ceccarini, L., Piazzi, L., Gobbi, S., Cavalli, A., Recanatini, M., Andrisano, V. and Rampa, A., 2010. Targeting Alzheimer's disease: Novel indanone hybrids bearing a pharmacophoric fragment of AP2238, Bioorg Med Chem. 18, 1749-60.

M AN U

Ross, S.E., Greenberg, M.E. and Stiles, C.D., 2003. Basic helix-loop-helix factors in cortical development, Neuron. 39, 13-25.

Roy, D. and Belsham, D.D., 2002. Melatonin receptor activation regulates GnRH gene expression and secretion in GT1-7 GnRH neurons. Signal transduction mechanisms, J Biol Chem. 277, 251-8. Salti, R., Galluzzi, F., Bindi, G., Perfetto, F., Tarquini, R., Halberg, F. and Cornelissen, G., 2000. Nocturnal melatonin patterns in children, J Clin Endocrinol Metab. 85, 2137-44.

Sanders, D.C., Chaturvedi, A.K. and Hordinsky, J.R., 1999. Melatonin: aeromedical, toxicopharmacological, and analytical aspects, J Anal Toxicol. 23, 159-67.

TE D

Sarlak, G., Jenwitheesuk, A., Chetsawang, B. and Govitrapong, P., 2013. Effects of melatonin on nervous system aging: neurogenesis and neurodegeneration, J Pharmacol Sci. 123, 9-24. Sharma, M., Palacios-Bois, J., Schwartz, G., Iskandar, H., Thakur, M., Quirion, R. and Nair, N.P., 1989. Circadian rhythms of melatonin and cortisol in aging, Biol Psychiatry. 25, 305-19. Sharma, R., McMillan, C.R., Tenn, C.C. and Niles, L.P., 2006. Physiological neuroprotection by melatonin in a 6-hydroxydopamine model of Parkinson's disease, Brain Res. 1068, 230-6.

EP

Sharma, R., Ottenhof, T., Rzeczkowska, P.A. and Niles, L.P., 2008. Epigenetic targets for melatonin: induction of histone H3 hyperacetylation and gene expression in C17.2 neural stem cells, J Pineal Res. 45, 277-84. Shenoy, A. and Blelloch, R.H., 2014. Regulation of microRNA function in somatic stem cell proliferation and

AC C

differentiation, Nat Rev Mol Cell Biol. 15, 565-76. Shochat, T., Luboshitzky, R. and Lavie, P., 1997. Nocturnal melatonin onset is phase locked to the primary sleep gate, Am J Physiol. 273, R364-70.

Singh, M. and Jadhav, H.R., 2014. Melatonin: functions and ligands, Drug Discov Today. 19, 1410-8. Sommer, L. and Rao, M., 2002. Neural stem cells and regulation of cell number, Prog Neurobiol. 66, 1-18. Sompol, P., Liu, X., Baba, K., Paul, K.N., Tosini, G., Iuvone, P.M. and Ye, K., 2011. N-acetylserotonin promotes hippocampal neuroprogenitor cell proliferation in sleep-deprived mice, Proc Natl Acad Sci U S A. 108, 8844-9. Song, J., Kang, S.M., Lee, K.M. and Lee, J.E., 2015. The protective effect of melatonin on neural stem cell against LPS-induced inflammation, Biomed Res Int. 2015, 854359. Sotthibundhu, A., Phansuwan-Pujito, P. and Govitrapong, P., 2010. Melatonin increases proliferation of cultured neural stem cells obtained from adult mouse subventricular zone, J Pineal Res. 49, 291-300.

ACCEPTED MANUSCRIPT

Stehle, J.H., von Gall, C. and Korf, H.W., 2003. Melatonin: a clock-output, a clock-input, J Neuroendocrinol. 15, 383-9. Stevanato, L. and Sinden, J.D., 2014. The effects of microRNAs on human neural stem cell differentiation in twoand three-dimensional cultures, Stem Cell Res Ther. 5, 49. Stockhausen, M.T., Sjolund, J., Manetopoulos, C. and Axelson, H., 2005. Effects of the histone deacetylase inhibitor valproic acid on Notch signalling in human neuroblastoma cells, Br J Cancer. 92, 751-9.

RI PT

Suzuki, N., Somei, M., Kitamura, K., Reiter, R.J. and Hattori, A., 2008. Novel bromomelatonin derivatives suppress osteoclastic activity and increase osteoblastic activity: implications for the treatment of bone diseases, J Pineal Res. 44, 326-34.

Swaminathan, A., Kumar, M., Halder Sinha, S., Schneider-Anthony, A., Boutillier, A.L. and Kundu, T.K., 2014. Modulation of neurogenesis by targeting epigenetic enzymes using small molecules: an overview, ACS Chem Neurosci. 5, 1164-77. developing cerebral cortex, J Neurosci. 23, 9996-10001.

SC

Tabata, H. and Nakajima, K., 2003. Multipolar migration: the third mode of radial neuronal migration in the Tamai, S., Sanada, K. and Fukada, Y., 2008. Time-of-day-dependent enhancement of adult neurogenesis in the

M AN U

hippocampus, PLoS One. 3, e3835.

Tang, Y., Cai, B., Yuan, F., He, X., Lin, X., Wang, J., Wang, Y. and Yang, G.Y., 2014. Melatonin pretreatment improves the survival and function of transplanted mesenchymal stem cells after focal cerebral ischemia, Cell Transplant. 23, 1279-91.

Temple, S., 2001. The development of neural stem cells, Nature. 414, 112-7.

Thomson, J.A., Itskovitz-Eldor, J., Shapiro, S.S., Waknitz, M.A., Swiergiel, J.J., Marshall, V.S. and Jones, J.M., 1998. Embryonic stem cell lines derived from human blastocysts, Science. 282, 1145-7. Tocharus, C., Puriboriboon, Y., Junmanee, T., Tocharus, J., Ekthuwapranee, K. and Govitrapong, P., 2014.

TE D

Melatonin enhances adult rat hippocampal progenitor cell proliferation via ERK signaling pathway through melatonin receptor, Neuroscience. 275, 314-21.

Tong, L.M., Fong, H. and Huang, Y., 2015. Stem cell therapy for Alzheimer's disease and related disorders: current status and future perspectives, Exp Mol Med. 47, e151. Waddell, B.J., Wharfe, M.D., Crew, R.C. and Mark, P.J., 2012. A rhythmic placenta? Circadian variation, clock genes and placental function, Placenta. 33, 533-9.

EP

Wade, A., McKinney, A. and Phillips, J.J., 2014. Matrix regulators in neural stem cell functions, Biochim Biophys Acta. 1840, 2520-5.

Wang, C., Liu, F., Liu, Y.Y., Zhao, C.H., You, Y., Wang, L., Zhang, J., Wei, B., Ma, T., Zhang, Q., Zhang, Y., Chen, R.,

AC C

Song, H. and Yang, Z., 2011. Identification and characterization of neuroblasts in the subventricular zone and rostral migratory stream of the adult human brain, Cell Res. 21, 1534-50.

Wang, T.T., Zeng, G.C., Li, X.C. and Zeng, H.P., 2010. In vitro studies on the antioxidant and protective effect of 2-substituted -8-hydroxyquinoline derivatives against H(2)O(2)-induced oxidative stress in BMSCs, Chem Biol Drug Des. 75, 214-22.

Wang, Z., Ma, C., Meng, C.J., Zhu, G.Q., Sun, X.B., Huo, L., Zhang, J., Liu, H.X., He, W.C., Shen, X.M., Shu, Z. and Chen, G., 2012. Melatonin activates the Nrf2-ARE pathway when it protects against early brain injury in a subarachnoid hemorrhage model, J Pineal Res. 53, 129-37. Wong, L.F., Ralph, G.S., Walmsley, L.E., Bienemann, A.S., Parham, S., Kingsman, S.M., Uney, J.B. and Mazarakis, N.D., 2005. Lentiviral-mediated delivery of Bcl-2 or GDNF protects against excitotoxicity in the rat hippocampus, Mol Ther. 11, 89-95. Zawilska, J.B., Skene, D.J. and Arendt, J., 2009. Physiology and pharmacology of melatonin in relation to

ACCEPTED MANUSCRIPT

biological rhythms, Pharmacol Rep. 61, 383-410. Zhao, C., Deng, W. and Gage, F.H., 2008. Mechanisms and functional implications of adult neurogenesis, Cell. 132, 645-60. Zhou, L., Chen, X., Liu, T., Gong, Y., Chen, S., Pan, G., Cui, W., Luo, Z.P., Pei, M., Yang, H. and He, F., 2015. Melatonin reverses H2 O2 -induced premature senescence in mesenchymal stem cells via the SIRT1-dependent pathway, J Pineal Res. 59, 190-205.

RI PT

Zhou, Q. and Anderson, D.J., 2002. The bHLH transcription factors OLIG2 and OLIG1 couple neuronal and glial

SC

subtype specification, Cell. 109, 61-73.

Legend:

M AN U

Fig.1. Involved signaling pathways in melatonin mechanism to promote NSCs survival and modulate NSCs proliferation and differentiation. PARP, poly ADP-ribose polymerases EIK-1, ets like gene-1

c-fos, EIK-1 target gene

histone acetyl transferase

MSK, mitogen

AC C

EP

Nuclear factor erythroid2 related factor-2

ribosomal s6 kinase

and stress activated kinase

HAT, CREB,

Mash, Basic Helix-Loop-Helix Factors

TE D

cAMP-response element binding protein

RSK, p90

Keap1, Kelch-like ECH-associated protein 1.

Nrf2,

ACCEPTED MANUSCRIPT

Table 1. The effects and most effective concentrations of melatonin in different neural stem cells.

Effects

C17.2 Cortical NSCs

Differentiation survival

Most effective concentration 1 nM 100 nM

NSCs obtained from pregnant rat midbrain

survival

1 nM

Proliferation

1 µM

M AN U

Differentiation

25 µM

Proliferation

1 µM

Sotthibundhu, A., et al. 2010

Survival

100 nM

Fu, J., et al. 2011

Proliferation

1 µM

Tocharus, C., et al. 2014

Survival

100 nM

EP

AC C

Ischemic-stroke mice

MSCs exposed to H2O2

1 µM

TE D

Differentiation

New neurons in the hippocampus of adult mice

Kong, X., et al. 2008

Moriya, T., et al. 2007

NSCs obtained from adult mouse SVZ NSCs subjected to hypoxia Adult rat hippocampal progenitor cell

Sharma, R., et al. 2008 Song, J., et al. 2015

SC

NSCs derived from the mouse embryo striatum

Reference

RI PT

Cells

Ramirez-Rodriguez, G., et al. 2009

Differentiation

1 nM

Neurogenesis

10 mg/kg

Proliferation protection

10 mg/kg 100 µM

Chern, C., et al. 2012 Zhou, L., et al. 2015

ACCEPTED MANUSCRIPT

Name

Structure

Reference

Agomelatine (S20098)

Increase NPCs proliferation

AlAhmed, S., et al. 2010

SSH-BM-I

Stimulate terminal osteoblast differentiation

Mikami, Y., et al. 2011

2-vinyl-8-hydroxyquinoline derivatives (Compound 4)

Increase proliferation; Protection effects against oxidative stress in MSCs

Wang, T., et al. 2010

Increase NPCs proliferation

Sompol, P., et al. 2011

Differentiation Protection Inhibitary of AChE

Lopez-Iglesias B., et al. 2014

Stimulate early neurogenesis and neuronal maturation

de la Fuente Revenga, M., et al. 2015

Stimulate early neurogenesis and neuronal maturation

de la Fuente Revenga, M., et al. 2015

promote differentiation of rat NSCs to a neuronal phenotype

de la Fuente Revenga, M., et al. 2015

M AN U

RI PT

Activity

SC

Table 2. The effects of Melatonin derivatives/analogues on neural stem cells.

Melatonin−N,Ndibenzyl (Nmethyl) amine hybrids

Pinoline

EP

Melatonin-pinoline hybrid (compound 2)

TE D

N-acetylserotonin (NAS)

AC C

N‑Acetyl Bioisosteres of Melatonin (compound 16)

Note: MSCs, mesenchymal stem cells; NPCs, neural progenitor cells.

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIPT

Highlights: : Melatonin modulates cell fate of neural stem cells (NSCs).



Melatonin modulates cell fate of NSCs via multiple mechanisms.



Melatonin analogues or derivatives show good effects on NSCs.



The multi-targeted compounds show therapeutic potential in Alzheimer’s disease.

AC C

EP

TE D

M AN U

SC

RI PT