+ All Categories
Home > Documents > Comparing the Effects of Melatonin with Caloric ... · Isao Shimokawa and Piyarat Govitrapong have...

Comparing the Effects of Melatonin with Caloric ... · Isao Shimokawa and Piyarat Govitrapong have...

Date post: 01-Feb-2021
Category:
Upload: others
View: 0 times
Download: 0 times
Share this document with a friend
9
Vol.:(0123456789) 1 3 Neurochem Res (2018) 43:153–161 DOI 10.1007/s11064-017-2369-7 ORIGINAL PAPER Comparing the Effects of Melatonin with Caloric Restriction in the Hippocampus of Aging Mice: Involvement of Sirtuin1 and the FOXOs Pathway Anorut Jenwitheesuk 1  · Seongjoon Park 2  · Prapimpun Wongchitrat 3  · Jiraporn Tocharus 4  · Sujira Mukda 1  · Isao Shimokawa 2  · Piyarat Govitrapong 1,5,6  Received: 23 April 2017 / Revised: 25 July 2017 / Accepted: 26 July 2017 / Published online: 2 August 2017 © Springer Science+Business Media, LLC 2017 control, CR, Mel and CR+Mel treated groups. The Mel and CR+Mel were treated with melatonin 10 mg/kg, daily, sub- cutaneously for 7 consecutive days. Mel treatment upregu- lated the mRNA expression of Sirt1, FOXOs (FoxO1 and FoxO3a) target genes that regulated the cell cycle [e.g., cyclin-dependent kinase inhibitor 1B (p27)], Wingless and INT-1 (Wnt1) and inducible signaling pathway protein 1 (Wisp1) in the aged mouse hippocampus. CR treatment also showed the similar actions. However, the mRNA expres- sion of Sirt1, FoxO1, FoxO3a, p27 or Wisp1 did not alter in the CR+Mel group when compared with CR or Mel group. Melatonin could not produce any additive effect on the CR treatment group, suggesting that both treatments mim- icked the effect, possibly via the same pathway. NPY which mediates physiological adaptations to energy deficits is an essential link between CR and longevity in mice. In order to focus on the role of Npy in mediating the effects of mela- tonin, the gene expression between NpyKO and WT male mice were compared. Our data showed that, in the absence of Npy, melatonin could not mediate effects on those gene expressions, suggesting that Npy was required for mela- tonin to mediate the effect, possibly, on life extension. Keywords Caloric restriction · Melatonin · Sirtuin1 · FoxO1 · Neuropeptide Y · Aging Abbreviations ADAM10 A disintegrin and metalloprotease domain 10 AgRP Agouti-related peptide AMPK Adenosine monophosphate-activated protein kinase CART Cocaine and amphetamine regulated transcript CBP CREB-binding protein CR Caloric restriction Abstract It has been suggested that age-related neu- rodegeneration might be associated with neuropeptide Y (NPY); sirtuin1 (SIRT1) and forkhead box transcrip- tion factors O subfamily (FOXOs) pathways. Melatonin, a hormone mainly secreted by the pineal gland, is another anti-aging agent associated with the SIRT1-FOXOs path- way. This study aimed to compare the effects of melatonin (Mel) and caloric restriction (CR) on the expression of Sirt1, FoxO1, FoxO3a and FOXOs target genes in the aging mouse hippocampus. Neuropeptide Y-knockout (NpyKO) and wild-type (WT) male mice aged 19 months were pre- viously treated either with food ad libitum or CR for 16 months. WT old animals were divided into four groups: Isao Shimokawa and Piyarat Govitrapong have contributed equally to this work. * Isao Shimokawa [email protected] * Piyarat Govitrapong [email protected]; [email protected] 1 Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Thailand 2 Department of Pathology, Nagasaki University School of Medicine and Graduate School of Biomedical Sciences, 1-12-4, Sakamoto, Nagasaki 852-8523, Japan 3 Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Salaya, Nakon Pathom 73170, Thailand 4 Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand 5 Center for Neuroscience and Department of Pharmacology, Faculty of Science, Mahidol University, Salaya, Thailand 6 Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Kamphaeng Phet 6 Road, Lak Si, Bangkok 10210, Thailand
Transcript
  • Vol.:(0123456789)1 3

    Neurochem Res (2018) 43:153–161 DOI 10.1007/s11064-017-2369-7

    ORIGINAL PAPER

    Comparing the Effects of Melatonin with Caloric Restriction in the Hippocampus of Aging Mice: Involvement of Sirtuin1 and the FOXOs Pathway

    Anorut Jenwitheesuk1 · Seongjoon Park2 · Prapimpun Wongchitrat3 · Jiraporn Tocharus4 · Sujira Mukda1 · Isao Shimokawa2 · Piyarat Govitrapong1,5,6 

    Received: 23 April 2017 / Revised: 25 July 2017 / Accepted: 26 July 2017 / Published online: 2 August 2017 © Springer Science+Business Media, LLC 2017

    control, CR, Mel and CR+Mel treated groups. The Mel and CR+Mel were treated with melatonin 10 mg/kg, daily, sub-cutaneously for 7 consecutive days. Mel treatment upregu-lated the mRNA expression of Sirt1, FOXOs (FoxO1 and FoxO3a) target genes that regulated the cell cycle [e.g., cyclin-dependent kinase inhibitor 1B (p27)], Wingless and INT-1 (Wnt1) and inducible signaling pathway protein 1 (Wisp1) in the aged mouse hippocampus. CR treatment also showed the similar actions. However, the mRNA expres-sion of Sirt1, FoxO1, FoxO3a, p27 or Wisp1 did not alter in the CR+Mel group when compared with CR or Mel group. Melatonin could not produce any additive effect on the CR treatment group, suggesting that both treatments mim-icked the effect, possibly via the same pathway. NPY which mediates physiological adaptations to energy deficits is an essential link between CR and longevity in mice. In order to focus on the role of Npy in mediating the effects of mela-tonin, the gene expression between NpyKO and WT male mice were compared. Our data showed that, in the absence of Npy, melatonin could not mediate effects on those gene expressions, suggesting that Npy was required for mela-tonin to mediate the effect, possibly, on life extension.

    Keywords Caloric restriction · Melatonin · Sirtuin1 · FoxO1 · Neuropeptide Y · Aging

    AbbreviationsADAM10 A disintegrin and metalloprotease domain 10AgRP Agouti-related peptideAMPK Adenosine monophosphate-activated protein

    kinaseCART Cocaine and amphetamine regulated transcriptCBP CREB-binding proteinCR Caloric restriction

    Abstract It has been suggested that age-related neu-rodegeneration might be associated with neuropeptide Y (NPY); sirtuin1 (SIRT1) and forkhead box transcrip-tion factors O subfamily (FOXOs) pathways. Melatonin, a hormone mainly secreted by the pineal gland, is another anti-aging agent associated with the SIRT1-FOXOs path-way. This study aimed to compare the effects of melatonin (Mel) and caloric restriction (CR) on the expression of Sirt1, FoxO1, FoxO3a and FOXOs target genes in the aging mouse hippocampus. Neuropeptide Y-knockout (NpyKO) and wild-type (WT) male mice aged 19 months were pre-viously treated either with food ad  libitum or CR for 16 months. WT  old animals were divided into four groups:

    Isao Shimokawa and Piyarat Govitrapong have contributed equally to this work.

    * Isao Shimokawa [email protected]

    * Piyarat Govitrapong [email protected]; [email protected]

    1 Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Thailand

    2 Department of Pathology, Nagasaki University School of Medicine and Graduate School of Biomedical Sciences, 1-12-4, Sakamoto, Nagasaki 852-8523, Japan

    3 Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Salaya, Nakon Pathom 73170, Thailand

    4 Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand

    5 Center for Neuroscience and Department of Pharmacology, Faculty of Science, Mahidol University, Salaya, Thailand

    6 Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Kamphaeng Phet 6 Road, Lak Si, Bangkok 10210, Thailand

    http://crossmark.crossref.org/dialog/?doi=10.1007/s11064-017-2369-7&domain=pdf

  • 154 Neurochem Res (2018) 43:153–161

    1 3

    DKK1 Wnt inhibitor Dickkopf WNT signaling path-way inhibitor 1

    FOXOs Forkhead box transcription factors and the O subfamily

    GAPDH Glyceraldehyde 3-phosphate dehydrogenaseIGF-1 Insulin-like growth factor-1Mel MelatoninNAD Nicotinamide adenine dinucleotideNAMPT Nicotinamide phosphoribosyltransferaseNF-κB Nuclear factor-κBNMNAT Nicotinamide/nicotinic acid mononucleotide

    adenylyltransferaseNPY Neuropeptide YNrf2 Nuclear factor (erythroid-derived 2)-like 2POMC Anorexigenic proopiomelanocortinp27 Cyclin-dependent kinase inhibitor 1Bp300 E1A binding protein p300p53 Tumor suppressor protein 53PGC-1α Peroxisome proliferator-activated receptor

    gamma coactivator 1-alphaPI3K Phosphoinositide-3-kinasePOMC ProopiomelanocortinPPAR-γ Peroxisome proliferator-activated receptor

    gammaSAMP8 Senescence-accelerated mouse prone 8SAMR1 Senescence-accelerated-resistant mouseSIRT SirtuinWisp1 Wn1 inducible pathway protein1WNT Wingless-type

    Introduction

    Caloric restriction (CR) has been shown to delay the onset and severity of brain diseases associated with aging and to extend the functional health span of important functions, such as cognition. Reduced caloric intake has enhanced longevity and prevented age-related diseases in rodents and monkeys. High calorie diets have been associated with the risk of Alzheimer’s disease; therefore, it has been proposed that a low calorie diet might afford protection against neu-rodegenerative diseases [1–5]. However, the underlying mechanisms of how CR could protect the brain from aging have not been clearly elucidated.

    The neuroprotective effects of CR might result from pathways that respond to energy status, including the sir-tuin1 (SIRT1) [6–8] and forkhead box transcription factors and the O subfamily (FOXOs) [9, 10]. The FOXOs tran-scription family is one of the targets of SIRT1. FOXO1 and FOXO3a are expressed in the hippocampus and play an important role in modulating hippocampal neuronal home-ostasis [11, 12]. CR inhibits insulin-like growth factor-1

    (IGF-1) signaling, which negatively regulates the FOXO transcription factor [13, 14].

    Neuropeptide Y (NPY), a neuropeptide from the hypo-thalamic arcuate nucleus, is elevated by CR. Aging reduces levels of NPY in the brain and in response to fasting. NPY is present in the hippocampus and plays a role in learning and memory; thus, the possible involvement of this peptide in the pathogenesis of neurodegenerative diseases, such as Alzheimer’s disease [15] and Parkinson’s disease, has been suggested [16]. The beneficial effects of CR might result from two pathways that respond to low energy status: NPY [17] and SIRT1 [7].

    Melatonin, a hormone mainly secreted by the pineal gland, is another anti-aging agent that could potentially be applied in the treatment of neurodegenerative diseases [18, 19]. This hormone was recently found to be associated with the SIRT1-FOXOs pathway. Melatonin preserved the rela-tive protein levels of SIRT1 in senescence-accelerated mice (SAMP8) [20–22] and in the hippocampus of total sleep-deprived rats [23]. In addition, melatonin plays a potent protective role against brain injury via the actions of pro-tein kinase B (AKT) [24] and FOXOs [25].

    The present study aimed to compare the effects of CR and melatonin on the expressions of the Sirt1, FoxO1, FoxO3a and the FOXOs target genes, including cyclin-dependent kinase inhibitor 1B (p27) in the hippocampus of aging mice. In addition, Npy knockout (NpyKO) mice were used to determine whether NPY was required to regulate the melatonin SIRT1/FOXOs pathway.

    Experimental Procedure

    Animals and Experimental Design

    Npy-knockout (NpyKO) and wild type (WT) mice were breed and raised in animal facilities of Center for Frontier Life Sciences at Nagasaki University as described in our previous study [26]. Animals were handled according to the guidelines approved by the Ethics Review Committee for Animal Experimentation at Nagasaki University. All mice were housed in 12:12 h light/dark cycle in an ambi-ent temperature of 22–25 °C under specific pathogen-free conditions. Animals were fed ad  libitum with a Charles River-LPF diet (Oriental Yeast Co. Ltd., Tsukuba, Japan). NpyKO and WT mice with 3  months of age were used for experimental procedures. Animals were fed with the dietary regimens either normal receive food ad  libitum or caloric restriction (CR) throughout the experiments for 16 months. The CR-treated mice were received a food allot-ment according to previous study [27], consisting of 70% of the mean daily food intake of the normal group every day, 30 min before the lights were turned off.

  • 155Neurochem Res (2018) 43:153–161

    1 3

    At 19-month-old, mice were groups to test whether melatonin could increase Sirt1 and FoxOs expression in the hippocampus. WT mice were divided to four groups: a con-trol group was continued to receive food ad  libitum, CR-treated group, melatonin-treated group (Mel) and CR and melatonin-treated group (CR+Mel). NpyKo mice which normally show a phenotype to reduce in a food intake [28] was continued to receive food ad libitum and were divided to 2 groups: a control group and Mel group. In the Mel and CR+Mel group, 10 mg/kg of melatonin (Sigma-Aldrich, St Louis, MO, USA) were administered daily via subcutane-ous injection for 7 consecutive days. Control and CR group animals received vehicle injections at the same time. All mice were sacrificed, and hippocampal brain tissues were dissected, collected and stored at −80  °C until assayed. mRNAs of Sirt1, FoxO1, FoxO3a and selected genes involved in cell cycle arrest (p27) and FOXO3a modulation (Wisp1) were determined.

    RNA Isolation and Reverse Transcription

    Total RNAs were extracted from the hippocampus of dif-ferent groups of mice by using RNeasy tissue kit (Qia-gen, Valencia, CA, USA) according to the manufacturer’s instructions. RNA concentrations were determined by UV spectrophotometry at a wavelength of 260  nm. The qual-ity of RNA was evaluated as the densitometric ratio of 28S and 18S ribosomal RNA by electrophoresis. The extracted RNA was reverse-transcribed using a Bio-Rad reverse transcriptase reagent kit (Bio-Rad, Hercules, CA, USA), according to the manufacturer’s instructions. The cDNA solution was then stored at −20 °C until use.

    Quantitative Real‑time PCR (qPCR)

    The relative gene expression was performed by using SYBR-based real-time PCR. Commercially available, prevalidated primer sets for FoxO1, FoxO3a, p27 and Gapdh were purchased from the TAKARA Perfect Real Time support system (TAKARA BIO Inc., Shiga, Japan)

    as described in our previous report [29]. The other primer pairs for Sirt1 and Wisp1 were purchased from Takara Bio Inc. See Table 1 for all primers used details. ThunderBird SYBR qPCR Mix (TOYOBO Co. Ltd., Osaka, Japan) was used for PCR amplification, and the reactions were per-formed according to the manufacturer’s protocol. Gapdh was used as an endogenous control and non-template con-trols were systematically included to check for contamina-tion. The amplification, data quantification, and analysis were performed on an ABI PRISM 7900HT Sequence Detector (Applied Biosystems, Foster City, CA, USA).

    Statistical Analysis

    The data are expressed as the means ± SEM. Signifi-cance was assessed using one-way analysis of variance (ANOVA), followed by the Tukey–Kramer tests using GraphPad Prism version 5. P values less than 0.05 were considered significant.

    Results

    Melatonin, CR and NpyKO in Sirt1 Expression

    We compared the Sirt1 mRNA between young and old mice and found that Sirt1 mRNA was downregulated by aging (P < 0.05) (Fig.  1a). Therefore, we explored the effects of melatonin and CR on Sirt1 mRNA expression in the hippocampus obtained from old mice. The Sirt1 mRNA expression in the hippocampus was compared with that among old mice. Both the CR group and the Mel group significantly (P < 0.05) increased Sirt1 mRNA in old mice, compared with the old control group (Fig.  1b). Sirt1 mRNA expression in all groups of the NpyKO mice were significantly (P < 0.05) lower than that in the old WT control group. Ten milligrams per kilogram of mela-tonin did not alter Sirt1 level in the CR+Mel group com-pared with CR group and Mel group of WT mice. The Sirt1 mRNA level in the NpyKO Mel-treated group showed no

    Table 1 Primers used details Gene name GenBank accession no. Takara primer set ID

    FoxO1 NM_019739.3 MA072856FoxO3a NM_019740.2 MA075309p27 NM_001293757.1 MA099459Gapdh NM_001289726.1 MA050371

    Primer sequenceSirt1 NM_001159589.2 Forward CAG ACC CTC AAG CCA TGT TTGAT

    Reverse TTG GAT TCC TGC AAC CTG CTCWisp1 NM_018865.2 Forward GTC CTG AGG GTG GGC AAC AT

    Reverse GGG CGT GTA GTC GTT TCC TCT

  • 156 Neurochem Res (2018) 43:153–161

    1 3

    significant difference, compared with old NpyKO control mice. NpyKO mice treated with melatonin had lower Sirt1 mRNA than WT mice treated with melatonin (P < 0.001) (Fig. 1b).

    Melatonin, CR and NpyKO in FoxOs Expression

    Because FoxO1 is expressed in the hippocampus, and several lines of evidence have shown that FoxO1 was

    downregulated by aging. In WT mice, both the CR group and the Mel group significantly increased FoxO1 mRNA in the hippocampus, compared with the old control group (P < 0.01) (Fig. 2A). FoxO1 mRNA levels in all groups of the NpyKO mice were significantly lower than in the old WT mice (P < 0.05). In CR+Mel group, FoxO1 mRNA expression did not alter compared with CR group and Mel group of WT mice. Melatonin did not affect FoxO mRNA

    Fig. 1 Sirt1 mRNA expression in young and old mouse hippocam-pus. a Sirt1 mRNA was compared between young and old wild-type (WT) mice. b The effect of melatonin on Sirt1 mRNA expression in the hippocampus of old WT mice on a normal diet and caloric restric-tion (CR). Npy-knockout (NpyKO) and WT mice were treated with melatonin (Mel) dosages at 10 mg/kg daily for 7 consecutive days and saline via a subcutaneous route. The bars represent means ± standard errors of 4–5 mice. Graphs represent percentages of the young (a) and old mice (b), & denotes significant difference with P < 0.05 com-pared with the WT young control group, * denotes significant dif-ference with P < 0.05 compared with the WT old control group, ### denotes significant difference with P < 0.001 compared with the WT melatonin-treated group

    Fig. 2 The effects of melatonin on FoxO1 (a) and FoxO3a (b) expressions in the hippocampus of old mice on a normal diet and caloric restriction (CR). Wild-type (WT) and Npy-knockout (NpyKO) were treated with melatonin dosages at 10 mg/kg daily for 7 consecu-tive days and saline subcutaneously. The WT mice were divided into ad  libitum as control group, CR, Mel and CR+Mel group. NpyKo mice were divided into ad  libitum as control group and Mel group. The bars represent means ± standard errors of 4–5 mice. Graphs rep-resent the percentage of the old mice. (*, ** denote significant dif-ference with P < 0.05 and P < 0.01, respectively, compared with the WT old control group, and ### denotes significant difference with P < 0.001 compared with the WT melatonin-treated group)

  • 157Neurochem Res (2018) 43:153–161

    1 3

    expression in NpyKO mice and had lower mRNA levels than WT mice treated with melatonin (P < 0.001) (Fig. 2a).

    We determined the FoxO3a mRNA expression in the animals treated with CR or melatonin because FoxO3a is associated with brain apoptotic pathways and age-related neurodegeneration. In WT mice, both the CR group and the Mel group significantly increased FoxO3a mRNA expression, compared with the old control group (P < 0.01) (Fig.  2b). FoxO3a mRNA levels in all NpyKO groups were significantly lower than that in the old WT group (P < 0.05). Ten milligrams per kilogram of melatonin treatment did not significantly alter FoxO3a expression in the CR animals, compared with CR or Mel group of WT mice. FoxO3a mRNA levels in the NpyKO Mel-treated group showed no significant difference, compared with old NpyKO controls. NpyKO mice treated with melatonin had lower FoxO3a mRNA than WT mice treated with mela-tonin (P < 0.001) (Fig. 2b).

    Melatonin, CR and NpyKO in Cell Cycle Arrest Signals

    The cell cycle arrest of p27 in FOXOs target genes was examined for FOXOs activity. In WT mice, both the CR group and the Mel group significantly increased p27 mRNA expression in old mice, compared with old control mice (P < 0.05, P < 0.01, respectively) (Fig. 3). In CR+Mel group, p27 mRNA expression did not alter compared with CR group and Mel group of WT mice. p27 mRNA levels in the NpyKO Mel-treated group showed no significant differ-ence, compared with the old NpyKO controls (Fig. 3).

    Melatonin, CR and NpyKO in Wisp1 Expression

    Because melatonin and CR increased FoxO3a mRNA expression, which was modulated by the Wnt/β-catenin pathway, the CR or 10  mg/kg melatonin significantly (P < 0.01) increased Wisp1 mRNA expression in old WT mice. Wisp1 mRNA levels in the Mel-treated NpyKO mice showed no significant difference, compared with the old NpyKO controls. However, Wisp1 mRNA expression in both group the NpyKO mice was significantly higher than that in the old WT controls (P < 0.05) but was signifi-cantly lower than all of the CR, Mel and CR+Mel group of WT mice (P < 0.001). NpyKO Mel-treated mice had lower Wisp1 mRNA than WT mice treated with melatonin (P < 0.001) (Fig. 4).

    Discussion

    The present study found that 10  mg/kg melatonin treat-ment for 7 days attenuated the reduction in Sirt1 expres-sion in the hippocampus of aged mice. Many studies have

    demonstrated that melatonin increased SIRT1 expression in a variety of models [21, 23]. The supportive effect of melatonin on SIRT1 acts via the nicotinamideadenine dinu-cleotide (NAD) system. Melatonin preserves NAD lev-els under oxidative stress. Oxidative stress plays a crucial role in cognitive impairment in aging model such as klotho mutant mice. Klotho mutant mice have an extremely short lifespan and develop cognitive impairment [30]. Melatonin decreases glutathione/oxidized glutathione (GSH/GSSG) ratio, phospho-ERK expression, Nuclear factor (eryth-roid-derived 2)-like 2 (Nrf2) nuclear translocation, Nrf2 DNA-binding activity, and GCL mRNA expression in the hippocampi of klotho mutant mice via melatonin receptor type 2 (MT2 receptor) [31]. When melatonin was incu-bated with PC12 cell line in an oxidative environment, the oxidation was reduced, and melatonin donated an electron to NAD radical, resulting in the recreation of NAD [32]. CR induced the expression of the lifespan-regulating pro-tein SIRT1 in lower organisms such as yeast and worms to higher mammalian animals [8, 33, 34]. Like melatonin, CR induced an increase in SIRT1 by maintaining the nor-mal energetic nicotinamide adenine dinucleotide (NAD) system [35–37] and cAMP responsive-element binding protein (CREB) [14]. In yeast, CR decreased the levels of

    Fig. 3 The effects of melatonin on p27 expression in the hippocam-pus of old mice on a normal diet and caloric restriction (CR). Wild-type (WT) and Npy-knockout (NpyKO) were treated with melatonin dosages at 10 mg/kg daily for 7 consecutive days and saline subcu-taneously. The WT mice were divided into ad  libitum as control group, CR, Mel and CR+Mel group. NpyKo mice were divided into ad  libitum as control group and Mel group. The bars represent means ± standard errors of 4–5 mice. Graphs represent percentages of old mice. *, ** denote significant difference P < 0.05 and P < 0.01, respectively, compared with the WT old control group, ## denotes significant difference with P < 0.01 compared with the WT mela-tonin-treated group

  • 158 Neurochem Res (2018) 43:153–161

    1 3

    NADH or nicotinamide, leading to the activation of Sir2, a homolog of mammalian SIRT1 [38, 39]. In addition, nico-tinamide phosphoribosyltransferase (NAMPT) was recently found to be required for CR-induced-SIRT1 [40–42].

    Recent studies have found that CR or melatonin admin-istration could restore the reduction in FOXO1 expression and activity in senescence models and during aging [21, 27, 43]. The anti-aging properties of CR are expected to restore FOXO1 levels during senescent periods. CR upreg-ulated FoxO1 mRNA in skeletal muscle and many tissues, including the brain [9]. FOXO1-overexpressing transgenic mice increased GADD45α and glutamine synthase proteins in skeletal muscle. Gadd45α, glutamine synthase, and cata-lase are longevity-related genes [44].

    Melatonin might promote FOXOs activities via SIRT1 deacetylation action and also act by suppressing CREB-binding protein (CBP) and p300. The acetylation of FOXOs via CBP and p300 decreased DNA binding also decreased FOXOs activity [45, 46]. Melatonin suppressed p300 histone acetyltransferase activity and p300-mediated NF-κB acetylation in the human vascular smooth muscle cell line CRL1999 [47]. In addition, melatonin was found to increase the deacetylation of several substrates of SIRT1, such as p53, peroxisome proliferator-activated receptor γ

    coactivator 1α (PGC-1α), FOXO1, a disintegrin and metal-loprotease domain 10 (ADAM10) and NF-κB in a murine senescence model [21].

    The present study showed that cell cycle arrest p27 mRNA was induced by CR and melatonin, suggesting that both factors were beneficial for protecting against neuro-degenerative diseases during aging. The cyclin-dependent kinase inhibitor p27, which is a critical determinant of cell cycle progression, is also an important regulation target of mitogenic signals. In young animals, the upregulation of these cell cycle arrest molecules has adverse effects by reducing neurogenesis in the hippocampus [48, 49]. In con-trast, p27 protects the brain during chronic stress to arrest the G1 stage, leading to additional time for DNA repair and fewer cycling cells left to engage in apoptosis [50]. In old animals, the attempts of neurons to re-enter mitosis in response to external stimuli can lead to abnormal cell cycle processes and neuronal degeneration.

    The action of FOXO3a could be regulated through the WNT-β-catenin pathway [51]. Wisp1 is a target gene of the WNT/β-catenin pathway [52, 53]. WISP1 induced the inhibition of apoptosis, mediated AKT activation, and enhanced survivin expression and the inhibition of pro-apoptotic BAX, accompanied by blocked cytochrome c [53]. WISP1 phosphorylated FOXO3a and prevented cas-pase degradation [52]. CR and melatonin increase Wisp1 by enhancing the WNT pathway. CR and melatonin decrease glycogen synthase kinase (GSK)-3β activity. Increased GSK-3β activity during aging, results in inactiva-tion of β-catenin by a phosphorylation process. CR induced increase in insulin-stimulated GSK-3β (Ser9) phosphoryla-tion in rat muscles [54]. Melatonin attenuated the increase in phosphorylated GSK-3β in primary cultures of cerebellar granule neurons [55], in a SAMP8 mice model [56], and in an Alzheimer’s model [57, 58].

    NPY, which mediates physiological adaptation in energy deficiency, is an essential link between CR and longevity in mice. Several studies have suggested a key role for Npy in mediating the effects of CR. The results of the present study showed that melatonin could not alter Sirt1, FoxO1, FoxO3a, p27 or Wisp1 expressions in Npy-knockout hip-pocampus. NPY might directly activate Sirt1 expression or might indirectly affect Sirt1 expression via melatonin. SIRT1 regulated food intake and body weight by acting on NPY/AGRP and an anorexigenic proopiomelanocortin (POMC) neurons [59]. Melatonin and NPY have agonistic effects on each other. NPY is present in high concentrations in the mammalian pineal gland, mainly localized in the neu-ronal innervations. The rat pineal gland expresses the gene coding for Y1- receptor NPY increased melatonin secre-tion due to the induced Ca2+ increase in rat pinealocytes, and this effect was inhibited by the Y1-receptor antago-nist BIBP3226 [60]. In contrast, melatonin increases NPY

    Fig. 4 The effects of melatonin on Wisp1 mRNA expression in the hippocampus of old mice on a normal diet and caloric restriction (CR). Wild-type (WT) and Npy-knockout (NpyKO) were treated with melatonin dosages at 10  mg/kg daily for 7 consecutive days and saline subcutaneously. The WT mice were divided into ad  libi-tum as control group, CR, Mel and CR+Mel group. NpyKo mice were divided into ad  libitum as control group and Mel group. The bars represent means ± standard errors of 4–5 mice. Graphs represent percentages of the old mice *, **, *** denote significant difference with P < 0.05, P < 0.01, and P < 0.001, respectively, compared with the WT old control group, and ### denotes significant difference with P < 0.001 compared with the WT melatonin-treated group

  • 159Neurochem Res (2018) 43:153–161

    1 3

    levels. Prenatal exposure to melatonin increased NPY syn-thesis in the hypothalamus [61]. Pinealectomy decreased NPY synthesis, and exogenous melatonin restored NPY levels in the hypothalamic arcuate nucleus of the rat [62]. Our data showed that melatonin induced Sirt1 expression in the wild-type, but not in the Npy-knockout mouse hip-pocampus, suggesting that Npy is essential for melatonin to upregulate Sirt1 expression. In addition, it also requires several downstream factors, i.e., FoxO1, FoxO3a, p27 and Wisp1 upregulation.

    Conclusion

    The present study compared two anti-aging treatments, melatonin administration and caloric restriction, which are involved with the SIRT1 and FOXOs pathways (Fig.  5). Both CR and melatonin significantly upregulated Sirt1, FoxO1 and FoxO3a mRNAs in aged mouse hippocam-pus. Both upregulated FOXOs target genes including p27, which plays a role in controlling the cell cycle. Melatonin could not produce additive actions on CR treatment. The results suggested that melatonin administration and CR might share the same regulatory pathways in controlling the SIRT1-FOXOs cascade for lifespan extension. In addi-tion, our results also indicated that melatonin regulated the SIRT1-FOXOs pathway required the presence of NPY. Fur-ther studies need to be fully elucidated to clarify the effects of melatonin- and CR-mediated life span extension, par-ticularly the connections between SIRT1 and other longev-ity factors.

    Acknowledgements This study was supported in part by the Thai-land Research Fund (TRF) (DPG5780001), a Mahidol University Research Grant to PG and Goho Life Sciences International Funding to AJ.

    References

    1. Morris JK, Bomhoff GL, Stanford JA, Geiger PC (2010) Neu-rodegeneration in an animal model of Parkinson’s disease is exacerbated by a high-fat diet. Am J Physiol Regul Integr Comp Physiol 299:R1082–R1090

    2. Rotermund C, Truckenmuller FM, Schell H, Kahle PJ (2014) Diet-induced obesity accelerates the onset of terminal phe-notypes in alpha-synuclein transgenic mice. J Neurochem 131:848-858

    3. le Coutre J, Mattson MP, Dillin A, Friedman J, Bistrian B (2013) Nutrition and the biology of human ageing: cognitive decline/food intake and caloric restriction. J Nutr Health Aging 17:717–720

    4. Wu P, Shen Q, Dong S, Xu Z, Tsien JZ, Hu Y (2008) Calorie restriction ameliorates neurodegenerative phenotypes in fore-brain-specific presenilin-1 and presenilin-2 double knockout mice. Neurobiol Aging 29:1502–1511

    5. Van Cauwenberghe C, Vandendriessche C, Libert C, Vandenb-roucke RE (2016) Caloric restriction: beneficial effects on brain aging and Alzheimer’s disease. Mamm Genome 27:300–319

    6. Maiese K, Chong ZZ, Shang YC, Wang S (2011) Translating cell survival and cell longevity into treatment strategies with SIRT1. Rom J Morphol Embryol 52:1173–1185

    7. Narasimhan SD, Yen K, Tissenbaum HA (2009) Converging pathways in lifespan regulation. Curr Biol 19:R657-R666

    8. Quintas A, de Solis AJ, Diez-Guerra FJ, Carrascosa JM, Bogonez E (2012) Age-associated decrease of SIRT1 expression in rat hippocampus: prevention by late onset caloric restriction. Exp Gerontol 47:198–201

    Fig. 5 Summary of the possible mechanisms that explain the ben-eficial effects of CR and melatonin on SIRT1 and the FOXOs path-way that protect the aging brain. Both CR and 10 mg/kg melatonin significantly upregulated Sirt1, FoxO1, and FoxO3a mRNA. In addi-tion, CR and melatonin also upregulated FOXOs target genes, includ-ing p27, which plays a role in cell cycle control. Although melatonin

    and CR upregulated FoxO3a expression, melatonin and CR might decrease FOXO3a apoptotic activity by upregulating WISP1. Neu-ropeptide Y (NPY) is essential for melatonin to upregulate Sirt1 expression, and it is needed for downstream factors, including FoxO1, FoxO3a, p27 and Wisp1 upregulation

  • 160 Neurochem Res (2018) 43:153–161

    1 3

    9. Furuyama T, Yamashita H, Kitayama K, Higami Y, Shimokawa I, Mori N (2002) Effects of aging and caloric restriction on the gene expression of Foxo1, 3, and 4 (FKHR, FKHRL1, and AFX) in the rat skeletal muscles. Microsc Res Tech 59:331–334

    10. Yamaza H, Chiba T, Higami Y, Shimokawa I (2002) Lifespan extension by caloric restriction: an aspect of energy metabolism. Microsc Res Tech 59:325–330

    11. Fukunaga K, Shioda N (2009) Pathophysiological relevance of forkhead transcription factors in brain ischemia. Adv Exp Med Biol 665:130–142

    12. Zemva J, Schilbach K, Stohr O, Moll L, Franko A, Krone W, Wiesner RJ, Schubert M (2012) Central FoxO3a and FoxO6 expression is down-regulated in obesity induced diabetes but not in aging. Exp Clin Endocrinol Diabetes 120:340–350

    13. Shimokawa I, Trindade LS (2010) Dietary restriction and aging in rodents: a current view on its molecular mechanisms. Aging Dis 1:89–107

    14. Xu BL, Wang R, Ma LN, Dong W, Zhao ZW, Zhang JS, Wang YL, Zhang X (2015) Effects of caloric intake on learning and memory function in juvenile C57BL/6J mice. Biomed Res Int 2015:759803

    15. Smialowska M, Domin H, Zieba B, Kozniewska E, Michalik R, Piotrowski P, Kajta M (2009) Neuroprotective effects of neuro-peptide Y-Y2 and Y5 receptor agonists in vitro and in vivo. Neu-ropeptides 43:235–249

    16. Decressac M, Pain S, Chabeauti PY, Frangeul L, Thiriet N, Her-zog H, Vergote J, Chalon S, Jaber M, Gaillard A (2012) Neuro-protection by neuropeptide Y in cell and animal models of Par-kinson’s disease. Neurobiol Aging 33:2125–2137

    17. Minor RK, Chang JW, de Cabo R (2009) Hungry for life: how the arcuate nucleus and neuropeptide Y may play a critical role in mediating the benefits of calorie restriction. Mol Cell Endo-crinol 299:79–88

    18. Karasek M (2004) Melatonin, human aging, and age-related dis-eases. Exp Gerontol 39:1723–1729

    19. Karasek M (2007) Does melatonin play a role in aging pro-cesses? J Physiol Pharmacol 58:105–113

    20. Gutierrez-Cuesta J, Tajes M, Jimenez A, Coto-Montes A, Camins A, Pallas M (2008) Evaluation of potential pro-survival pathways regulated by melatonin in a murine senescence model. J Pineal Res 45:497–505

    21. Tajes M, Gutierrez-Cuesta J, Ortuno-Sahagun D, Camins A, Pal-las M (2009) Anti-aging properties of melatonin in an in  vitro murine senescence model: involvement of the sirtuin 1 pathway. J Pineal Res 47:228–237

    22. Cristofol R, Porquet D, Corpas R, Coto-Montes A, Serret J, Camins A, Pallas M, Sanfeliu C (2012) Neurons from senes-cence-accelerated SAMP8 mice are protected against frailty by the sirtuin 1 promoting agents melatonin and resveratrol. J Pineal Res 52:271–281

    23. Chang HM, Wu UI, Lan CT (2009) Melatonin preserves lon-gevity protein (sirtuin 1) expression in the hippocampus of total sleep-deprived rats. J Pineal Res 47:211–220

    24. Kong PJ, Byun JS, Lim SY, Lee JJ, Hong SJ, Kwon KJ, Kim SS (2008) Melatonin induces Akt phosphorylation through mela-tonin receptor- and PI3K-dependent pathways in primary astro-cytes. Korean J Physiol Pharmacol 12:37–41

    25. Koh PO (2008) Melatonin prevents the injury-induced decline of Akt/forkhead transcription factors phosphorylation. J Pineal Res 45:199–203

    26. Park S, Fujishita C, Komatsu T, Kim SE, Chiba T, Mori R, Shimokawa I (2014) NPY antagonism reduces adiposity and attenuates age-related imbalance of adipose tissue metabolism. FASEB J 28:5337–5348

    27. Hayashi H, Yamaza H, Komatsu T, Park S, Chiba T, Higami Y, Nagayasu T (2008) Calorie restriction minimizes activation of

    insulin signaling in response to glucose: potential involvement of the growth hormone-insulin-like growth factor 1 axis. Exp Ger-ontol 43:827–832

    28. Bannon AW, Seda J, Carmouche M, Francis JM, Norman MH, Karbon B, McCaleb ML (2000) Behavioral characterization of neuropeptide Y knockout mice. Brain Res 868:79–87

    29. Chiba T, Tamashiro Y, Park D, Kusudo T, Fujie R, Komatsu T, Kim SE, Park S, Hayashi H, Mori R, Yamashita H, Chung HY, Shimokawa I (2014) A key role for neuropeptide Y in lifespan extension and cancer suppression via dietary restriction. Sci Rep 4:4517

    30. Park SJ, Shin EJ, Min SS, An J, Li Z, Hee Chung Y, Hoon Jeong J, Bach JH, Nah SY, Kim WK, Jang CG, Kim YS, Nabeshima Y, Nabeshima T, Kim HC (2013) Inactivation of JAK2/STAT3 signaling axis and downregulation of M1 mAChR cause cogni-tive impairment in klotho mutant mice, a genetic model of aging. Neuropsychopharmacology 38:1426–1437

    31. Shin EJ, Chung YH, Le HL, Jeong JH, Dang DK, Nam Y, Wie MB, Nah SY, Nabeshima Y, Nabeshima T, Kim HC (2014) Mel-atonin attenuates memory impairment induced by Klotho gene deficiency via interactive signaling between MT2 receptor, ERK, and Nrf2-related antioxidant potential. Int J Neuropsychophar-macol. doi:10.1093/ijnp/pyu105

    32. Tan DX, Manchester LC, Sainz RM, Mayo JC, Leon J, Harde-land R, Poeggeler B, Reiter RJ (2005) Interactions between mel-atonin and nicotinamide nucleotide: NADH preservation in cells and in cell-free systems by melatonin. J Pineal Res 39:185–194

    33. Graff J, Kahn M, Samiei A, Gao J, Ota KT, Rei D, Tsai LH (2013) A dietary regimen of caloric restriction or pharmacologi-cal activation of SIRT1 to delay the onset of neurodegeneration. J Neurosci 33:8951–8960

    34. Turan B, Tuncay E, Vassort G (2012) Resveratrol and diabetic cardiac function: focus on recent in vitro and in vivo studies. J Bioenerg Biomembr 44:281–296

    35. Liu D, Pitta M, Mattson MP (2008) Preventing NAD(+) deple-tion protects neurons against excitotoxicity: bioenergetic effects of mild mitochondrial uncoupling and caloric restriction. Ann NY Acad Sci 1147:275–282

    36. Lu SP, Lin SJ (2010) Regulation of yeast sirtuins by NAD(+) metabolism and calorie restriction. Biochim Biophys Acta 1804:1567–1575

    37. Ramis MR, Esteban S, Miralles A, Tan DX, Reiter RJ (2015) Caloric restriction, resveratrol and melatonin: role of SIRT1 and implications for aging and related-diseases. Mech Ageing Dev 146–148:28–41

    38. Rahat O, Maoz N, Cohen HY (2011) Multiple pathways regu-lating the calorie restriction response in yeast. J Gerontol A 66:163–169

    39. Anderson RM, Bitterman KJ, Wood JG, Medvedik O, Sinclair DA (2003) Nicotinamide and PNC1 govern lifespan exten-sion by calorie restriction in Saccharomyces cerevisiae. Nature 423:181–185

    40. Song J, Ke SF, Zhou CC, Zhang SL, Guan YF, Xu TY, Sheng CQ, Wang P, Miao CY (2014) Nicotinamide phosphoribo-syltransferase is required for the calorie restriction-mediated improvements in oxidative stress, mitochondrial biogenesis, and metabolic adaptation. J Gerontol A Biol Sci Med Sci 69:44–57

    41. Yang H, Lavu S, Sinclair DA (2006) Nampt/PBEF/Visfatin: a regulator of mammalian health and longevity? Exp Gerontol 41:718–726

    42. Yamamoto T, Byun J, Zhai P, Ikeda Y, Oka S, Sadoshima J (2014) Nicotinamide mononucleotide, an intermediate of NAD+ synthesis, protects the heart from ischemia and reperfusion. PLoS ONE 9:e98972

    43. Caricasole A, Copani A, Caraci F, Aronica E, Rozemuller AJ, Caruso A, Storto M, Gaviraghi G, Terstappen GC, Nicoletti F

    https://doi.org/10.1093/ijnp/pyu105

  • 161Neurochem Res (2018) 43:153–161

    1 3

    (2004) Induction of Dickkopf-1, a negative modulator of the Wnt pathway, is associated with neuronal degeneration in Alzhei-mer’s brain. J Neurosci 24:6021–6027

    44. Chiba T, Kamei Y, Shimizu T, Shirasawa T, Katsumata A, Shi-raishi L, Sugita S, Ogawa Y, Miura S, Ezaki O (2009) Overex-pression of FOXO1 in skeletal muscle does not alter longevity in mice. Mech Ageing Dev 130:420–428

    45. Bousiges O, Vasconcelos AP, Neidl R, Cosquer B, Herbeaux K, Panteleeva I, Loeffler JP, Cassel JC, Boutillier AL (2010) Spa-tial memory consolidation is associated with induction of sev-eral lysine-acetyltransferase (histone acetyltransferase) expres-sion levels and H2B/H4 acetylation-dependent transcriptional events in the rat hippocampus. Neuropsychopharmacology 35:2521–2537

    46. Wang J, Xiao X, Zhang Y, Shi D, Chen W, Fu L, Liu L, Xie F, Kang T, Huang W, Deng W (2012) Simultaneous modulation of COX-2, p300, Akt, and Apaf-1 signaling by melatonin to inhibit proliferation and induce apoptosis in breast cancer cells. J Pineal Res 53:77–90

    47. Shi D, Xiao X, Wang J, Liu L, Chen W, Fu L, Xie F, Huang W, Deng W (2012) Melatonin suppresses proinflammatory media-tors in lipopolysaccharide-stimulated CRL1999 cells via target-ing MAPK, NF-kappaB, c/EBPbeta, and p300 signaling. J Pineal Res 53:154–165

    48. Anacker C, Zunszain PA, Cattaneo A, Carvalho LA, Garabedian MJ, Thuret S, Price J, Pariante CM (2011) Antidepressants increase human hippocampal neurogenesis by activating the glu-cocorticoid receptor. Mol Psychiatry 16:738–750

    49. Andreu Z, Khan MA, Gonzalez-Gomez P, Negueruela S, Hor-tiguela R, San Emeterio J, Ferron SR, Martinez G, Vidal A, Farinas I, Lie DC, Mira H (2015) The cyclin-dependent kinase inhibitor p27 kip1 regulates radial stem cell quiescence and neu-rogenesis in the adult hippocampus. Stem Cells 33:219–229

    50. Heine VM, Maslam S, Joels M, Lucassen PJ (2004) Increased P27KIP1 protein expression in the dentate gyrus of chronically stressed rats indicates G1 arrest involvement. Neuroscience 129:593–601

    51. Berschneider B, Konigshoff M (2011) WNT1 inducible signal-ing pathway protein 1 (WISP1): a novel mediator linking devel-opment and disease. Int J Biochem Cell Biol 43:306–309

    52. Wang S, Zhong Chong Z, Chen Shang Y, Maiese K (2013) WISP1 neuroprotection requires FoxO3a post-translational mod-ulation with autoregulatory control of SIRT1. Curr Neurovasc Res 10:54–69

    53. Su F, Overholtzer M, Besser D, Levine AJ (2002) WISP-1 attenuates p53-mediated apoptosis in response to DNA damage through activation of the Akt kinase. Genes Dev 16:46–57

    54. Sequea DA, Sharma N, Arias EB, Cartee GD (2013) Greater fil-amin C, GSK3alpha, and GSK3beta serine phosphorylation in insulin-stimulated isolated skeletal muscles of calorie restricted 24 month-old rats. Mech Ageing Dev 134:60–63

    55. Tajes Orduna M, Pelegri Gabalda C, Vilaplana Hortensi J, Pal-las Lliberia M, Camins Espuny A (2009) An evaluation of the neuroprotective effects of melatonin in an in  vitro experi-mental model of age-induced neuronal apoptosis. J Pineal Res 46:262–267

    56. Gutierrez-Cuesta J, Sureda FX, Romeu M, Canudas AM, Cabal-lero B, Coto-Montes A, Camins A, Pallas M (2007) Chronic administration of melatonin reduces cerebral injury biomarkers in SAMP8. J Pineal Res 42:394–402

    57. Peng CX, Hu J, Liu D, Hong XP, Wu YY, Zhu LQ, Wang JZ (2013) Disease-modified glycogen synthase kinase-3beta inter-vention by melatonin arrests the pathology and memory deficits in an Alzheimer’s animal model. Neurobiol Aging 34:1555–1563

    58. Kwon KJ, Kim HJ, Shin CY, Han SH (2010) Melatonin poten-tiates the neuroprotective properties of resveratrol against beta-amyloid-induced neurodegeneration by modulating AMP-acti-vated protein kinase pathways. J Clin Neurol 6:127–137

    59. Dietrich MO, Antunes C, Geliang G, Liu ZW, Borok E, Nie Y, Xu AW, Souza DO, Gao Q, Diano S, Gao XB, Horvath TL (2010) Agrp neurons mediate Sirt1’s action on the melanocortin system and energy balance: roles for Sirt1 in neuronal firing and synaptic plasticity. J Neurosci 30:11815–11825

    60. Simonneaux V, Ribelayga C (2003) Generation of the melatonin endocrine message in mammals: a review of the complex regu-lation of melatonin synthesis by norepinephrine, peptides, and other pineal transmitters. Pharmacol Rev 55:325–395

    61. Díaz E, Debeljuk L, Arce A, Esquifino A, Díaz B (2000) Pre-natal melatonin exposure affects luteinizing hormone and hypo-thalamic and striatal neuropeptide Y in the male rat offspring. Neurosci Lett 292:143–146

    62. Aydin M, Canpolat S, Kuloglu T, Yasar A, Colakoglu N, Kelestimur H (2008) Effects of pinealectomy and exogenous melatonin on ghrelin and peptide YY in gastrointestinal system and neuropeptide Y in hypothalamic arcuate nucleus: immuno-histochemical studies in male rats. Regul Pept 146:197–203

    Comparing the Effects of Melatonin with Caloric Restriction in the Hippocampus of Aging Mice: Involvement of Sirtuin1 and the FOXOs PathwayAbstract IntroductionExperimental ProcedureAnimals and Experimental DesignRNA Isolation and Reverse TranscriptionQuantitative Real-time PCR (qPCR)Statistical Analysis

    ResultsMelatonin, CR and NpyKO in Sirt1 ExpressionMelatonin, CR and NpyKO in FoxOs ExpressionMelatonin, CR and NpyKO in Cell Cycle Arrest SignalsMelatonin, CR and NpyKO in Wisp1 Expression

    DiscussionConclusionAcknowledgements References


Recommended