Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Neurochem Res ; 43(1): 153-161, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28770437

ABSTRACT

It has been suggested that age-related neurodegeneration might be associated with neuropeptide Y (NPY); sirtuin1 (SIRT1) and forkhead box transcription factors O subfamily (FOXOs) pathways. Melatonin, a hormone mainly secreted by the pineal gland, is another anti-aging agent associated with the SIRT1-FOXOs pathway. 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 previously treated either with food ad libitum or CR for 16 months. WT old animals were divided into four groups: control, CR, Mel and CR+Mel treated groups. The Mel and CR+Mel were treated with melatonin 10 mg/kg, daily, subcutaneously for 7 consecutive days. Mel treatment upregulated 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 expression 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 mimicked 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 melatonin, 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 melatonin to mediate the effect, possibly, on life extension.


Subject(s)
Caloric Restriction , Forkhead Transcription Factors/metabolism , Hippocampus/drug effects , Melatonin/pharmacology , Sirtuin 1/metabolism , Aging/genetics , Animals , Caloric Restriction/methods , Gene Expression/drug effects , Hippocampus/metabolism , Humans , Melatonin/metabolism , Neuropeptide Y/genetics
2.
EXCLI J ; 16: 340-353, 2017.
Article in English | MEDLINE | ID: mdl-28507478

ABSTRACT

Sirtuin1 (SIRT1) and forkhead box transcription factor O subfamily 1 (FOXO1) play vital roles in the maintenance of hippocampal neuronal homeostasis during aging. Our previous study showed that melatonin, a hormone mainly secreted by the pineal gland, restored the impaired memory of aged mice. Age-related neuronal energy deficits contribute to the pathogenesis of several neurodegenerative disorders. An attempt has been made to determine whether the effect of melatonin is mediated through the SIRT1-FOXO1 pathways. The present results showed that aged mice (22 months old) exhibited significantly downregulated SIRT1, FOXO1, and melatonin receptors MT1 and MT2 protein expression but upregulated tumor suppressor protein 53 (p53), acetyl-p53 protein (Ac-p53), mouse double minute 2 homolog (MDM2), Dickkopf-1 (DKK1) protein expression in mouse hippocampus compared with the young group. Melatonin treatment (10 mg/kg, daily in drinking water for 6 months) in aged mice significantly attenuated the age-induced downregulation of SIRT1, FOXO1, MT1 and MT2 protein expression and attenuated the age-induced increase in p53, ac-p53, MDM2, and DKK1 protein and mRNA expression. Melatonin decreased p53 and MDM2 expression, which led to a decrease in FOXO1 degradation. These present results suggest that melatonin may help the hippocampal neuronal homeostasis by increasing SIRT1, FOXO1 and melatonin receptors expression while decreasing DKK1 expression in the aging hippocampus. DKK1 can be induced by the accumulation of amyloid ß (Aß) which is the major hallmark of Alzheimer's disease.

3.
Int J Mol Sci ; 15(9): 16848-84, 2014 Sep 22.
Article in English | MEDLINE | ID: mdl-25247581

ABSTRACT

Brain aging is linked to certain types of neurodegenerative diseases and identifying new therapeutic targets has become critical. Melatonin, a pineal hormone, associates with molecules and signaling pathways that sense and influence energy metabolism, autophagy, and circadian rhythms, including insulin-like growth factor 1 (IGF-1), Forkhead box O (FoxOs), sirtuins and mammalian target of rapamycin (mTOR) signaling pathways. This review summarizes the current understanding of how melatonin, together with molecular, cellular and systemic energy metabolisms, regulates epigenetic processes in the neurons. This information will lead to a greater understanding of molecular epigenetic aging of the brain and anti-aging mechanisms to increase lifespan under healthy conditions.


Subject(s)
Aging/physiology , Autophagy/physiology , Circadian Rhythm/physiology , Energy Metabolism/physiology , Epigenesis, Genetic/physiology , Melatonin/physiology , Nerve Degeneration/physiopathology , Animals , Brain/growth & development , Brain/metabolism , Caloric Restriction , Circadian Rhythm/genetics , Diabetes Mellitus/physiopathology , Forkhead Transcription Factors/physiology , Gene Expression Regulation/physiology , Humans , Inflammation/physiopathology , Insulin/physiology , Insulin Resistance/physiology , Insulin-Like Growth Factor I/physiology , Longevity/physiology , Memory/physiology , Obesity/physiopathology , Signal Transduction/physiology , Sirtuins/physiology
4.
J Pharmacol Sci ; 123(1): 9-24, 2013 Sep 20.
Article in English | MEDLINE | ID: mdl-23985544

ABSTRACT

Neural aging as a progressive loss of function involves central and peripheral post-mitotic neurons and neural stem cells (NSCs). It promotes neurodegeneration, impairs neurogenesis, and can be considered a cause of cognitive impairment and sensory and motor deficits in the elderly. Age-related morphological atrophic changes and cellular alterations are addressed by neural aging mechanisms. Neurogenesis declines during aging through several mechanisms such as an increase in quiescence state, changes in lineage fate, telomerase dysfunction, the failure of the DNA repair system, increased apoptosis, and the impairment of self-renewal. The self-renewal transcriptional factor Sox2 has been correlated with retrotransposon L1 and certain cell-cycle- and epigenetic-related factors, which are sometimes considered age-related factors in NSC aging. As neurogenesis decreases, non-mitotic neurons undergo neurodegeneration by oxidative stress, sirtuin, insulin signaling and mTOR alteration, mitochondrial dysfunction, and protein misfolding and aggregation. As neurodegeneration and impaired neurogenesis promote the nervous system aging process, the identification of neuronal anti-aging is required to raise life expectancy. The role of melatonin in increasing neurogenesis and protecting against neurodegeneration has been investigated. Here, we review nervous system aging that is correlated with mechanisms of neurodegeneration and the impairment of neurogenesis and evaluate the effects of melatonin on these processes.


Subject(s)
Aging/drug effects , Aging/genetics , Melatonin/pharmacology , Melatonin/physiology , Nerve Degeneration/prevention & control , Neurogenesis/drug effects , Neuroprotective Agents , Aged , Aging/pathology , Aging/physiology , Animals , Apoptosis/genetics , Cognition Disorders/etiology , Humans , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Neural Stem Cells/pathology , Neural Stem Cells/physiology , Neurogenesis/genetics , Neurogenesis/physiology , Neurons/pathology , Neurons/physiology , Oxidative Stress/genetics , SOXB1 Transcription Factors/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...