How does resveratrol influence the genesis of some neurodegenerative diseases?

2016-12-01 12:37EsterTellone,AntonioGaltieri,AnnamariaRusso
关键词:主观变量检验

PERSPECTIVE

How does resveratrol influence the genesis of some neurodegenerative diseases?

Advancing age and increased lifespan of human populations worldwide in the mid-nineteenth century, have led to a significant increase in the incidence of neurodegenerative diseases, one of the major cause of disability and death for most of those affected. Neurodegeneration is one of the biggest public health problems in modern society also because effective pharmacological interventions for prevention and treatment of the disease, are lacking. Based on the premise that oxidative stress underlies a number of neurodegenerative diseases, the identification of novel antioxidants as potential therapeutics is a prolific area of neuroscience research. There is also a close relationship between specific therapies and the induction of oxidative stress in chronic neurodegeneration, such as the case of levodopa in patients with Parkinson’s disease (Müller et al., 2014).

Although the different neurodegenerative diseases manifest in distinct neuronal cell types, oxidative stress and suppression of neuronal survival signals are common to many of these pathological conditions and appear to be highly relevant targets for treatment. Growing evidences suggest that at the pathological level, almost all neurodegenerative diseases share common features such as the iron accumulation and the generation of misfolded protein deposits. Furthermore, it was shown that the oxidative stress markers precede the pathological lesions of neurodegeneration, see Figure 1.

Recently, we have focused on the potential therapeutic effects of an antioxidant compound as resveratrol (RV) or 3,5,4′-trihidroxy-trans-stilbene an antifungal molecule of the stilbene family produced in a variety of plant species. RV chemical structure, is characterized by two phenol groups in which the presence of conjugated double bond makes the electrons more delocalized and easily transferable. Just the ability to transfer hydrogen atoms or electrons to the free radicals makes RV an efficient free radical scavenger and a potent antioxidant, see Figure 2 (Hussein, 2011; Iuga et al., 2012).

Interestingly, RV location in the cellular compartments is strictly related and increases the antioxidant properties of the drug, because the polyphenol interaction with the membrane bilayer prevents lipid peroxidation and in blood, intraerythrocyte RV by interacting with hemoglobin, may protect the protein against oxidative damage (Tellone et al., 2014). In addition, RV mediates the activation of sirtuin-1 (SIRT1) a deacetylase protein with potential therapeutic targets in a variety of human diseases (Herskovits and Guarente, 2013).

In Tellone et al. (2015), we collected knowledge on many RV molecular targets and tried to explain or partly support the effectiveness of the drug a therapeutic agent for neurodegenerative diseases, see Figure 3.

In this light, the interaction of the drug with SIRT1 certainly produces several beneficial effects against Alzheimer’s disease (AD). In fact RV, potentiating SIRT1 activity positively regulates α-secretase promoter transcription, a protease which processes the amyloid precursor protein (APP) along a non amyloidogenic pathway precluding Aβ generation. Additionally, SIRT1 can directly deacetylate Tau protein, the main constituent of neurofibrillary tangles and primary marker of AD. The removal of these acetyl groups exposes Lys residues to ubiquitin ligases so that Tau protein could be marked for proteasomal degradation. A further protective role of RV-SIRT1 is the deacetylation and the subsequent inhibition of p53 tumor suppressor gene, whose upregulation and overexpression is commonly associated with Tau hyperphosphorylation, neuronal damage and cell death (Cohen et al., 2011). Besides, the p53 reduced activity affects and in turn inhibits GSK-3β, one of the major glycogen synthase kinase involved in the Tau modification and neurofibrillary degeneration. Tau pathophysiological phosphorylation is also limited by activation of calcium/calmodulin-dependent protein kinase kinase-β (CamKKβ) promoted by RV through the increase of intracellular calcium levels and activation of AMP protein kinase (AMPK).

Inhibition of p53 and the enhancement of SIRT1 activity by RV is also important for the protection of neuronal cells against the mutant polyglutamine huntingtin protein (m-htt) toxic effects occurring in Huntington’s disease (HD). People with HD have an abnormally high number of DNA sequence called“CAG”, in the coding region of the gene on chromosome 4. The expanded CAG segment leads to the production of an abnormally long version of the htt. The elongated protein is cut into smaller, toxic fragments that bind together and accumulate in neurons. Overexpression of these htt fragments and p53 activation in HD increase mitochondrial oxidation and result in a gain of function mechanosensory defect that is the main cause of the pathology. RV treatment can effectively counteract the progression of HD also improving the mitochondrial function via a pathway in which SIRT1-AMPK and peroxisome proliferator-activated receptor-α coactivator 1α (PGC-1α) play a pivotal role (Tellone et al., 2015). RV stimulates PGC-1α, the peroxisome proliferator-activated receptor gamma coactivator-1 alpha a potent stimulator of mithocondrial biogenesis and respiration through its interaction with SIRT1. PGC1α when stimulated, regulates the expression and activities of ROS scavenging antioxidant enzymes and therefore counteracts oxidative stress (Higashida et al., 2013).

Besides, RV inducing activation and expression of SIRT1 also protects against pathological α-synuclein aggregation in Parkinson’s disease (PD). α-Synuclein is a little protein encoded by a gene located in chromosome 4. Mutation in this gene leads to the overexpression of the α-synuclein protein, and its aggregates were found to be the major components of Lewy bodies, thehallmarks of PD.

Figure 1 Oxidative stress and main neurological lesions.

Figure 2 Scavenger ability of RV for hydroxyl radical (rate constant) compared with natural antioxidant SOD.

Figure 3 RV main targets for neuronal protection.

In detail, SIRT1 activates heat shock factor 1 (HSF1), which in turns affects heat shock proteins 70 (hsp70); hsp70 regulates homeostasis of cellular proteins decreasing the formation of abnormal α-synuclein aggregates. Another path of RV beneficial modulation in cellular model of PD is implemented through the downregulation and partial inhibition of GSK-3β because α-synuclein is a substrate for GSK-3β phosphorylation (Li et al., 2014).

利用SPSS19.0软件对主观加权后的数据进行主成分分析,在分析之前,需要进行KMO检验和球形度检验,来验证所选取的12个指标变量是否适合做主成分分析。

Activation of SIRT1 by RV treatment has been shown to decrease proteotoxic stress derived from misfolded superoxide dismutase 1 (SOD1) aggregates in amyotrophic lateral sclerosis (ALS) (Herskovits and Guarente, 2013). Zhao et al. (2011) reported that RV through the overexpression of PGC1α improved motor performance and survival in a mouse model of ALS .

In conclusions, all these studies provide the proof that the RV has a strong scientific support to develop as a new therapy treatment for neurodegenerative diseases. Given that, the drug is not only active scavenger of free radicals but also acts as modulator of pro-survival or pro-apoptotic signaling pathways. As a result, this compound may have a greater potential for therapeutic success than drugs with only one mechanism of action.

Ester Tellone*, Antonio Galtieri, Annamaria Russo, Silvana Ficarra

Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy

*Correspondence to: Ester Tellone, Ph.D., etellone@unime.it.

Accepted: 2015-11-06

orcid: 0000-0003-2379-3392 (Ester Tellone)

Cohen TJ, Guo JL, Hurtado DE, Kwong LK, Mills IP, Trojanowski JQ, Lee VM (2011) The acetylation of tau inhibits its function and promotes pathological tau aggregation. Nat Commun 2:252.

Galtieri A, Tellone E, Ficarra S, Russo A, Bellocco E, Barreca D, Scatena R, Laganà G, Leuzzi U, Giardina B (2010) Resveratrol treatment induces redox stress in red blood cells: A possible role of caspase 3 in metabolism and anion transport. Biol Chem 391:1057-1065.

Herskovits AZ, Guarente L (2013) Sirtuin deacetylases in neurodegenerative diseases of aging. Cell Res 23:746-758.

Higashida K, Kim SH, Jung SR, Asaka M, Holloszy JO, Han DH (2013) Effects of resveratrol and SIRT1 on PGC-1α activity and mitochondrial biogenesis: a reevaluation. PLoS Biol 11:7.

Hussein MA (2011) A convenient mechanism for the free radical scavenging activity of resveratrol. Int J Phytomed 3:459-469.

Iuga C, Alvarez-Idaboy JR, Russo N (2012) Antioxidant activity of trans-resveratrol toward hydroxyl and hydroperoxyl radicals: a quantum chemical and computational kinetics study. J Org Chem 77:3868-3877.

Li DW, Liu ZQ, Chen W, Yao M, Li GR (2014) Association of glycogen synthase kinase-3 with Parkinson’s disease (Review). Mol Med Rep 9:2043-2050.

Müller T, Muhlack S (2014) Levodopa-related cysteinyl-glycine and cysteine reduction with and without catechol-O-methyltransferase inhibition in Parkinson’s disease patients. J Neural Transm 121:643-648.

Tellone E, de Rosa MC, Pirolli D, Russo A, Giardina B, Galtieri A, Ficarra S (2014) Molecular interactions of hemoglobin with resveratrol: potential protective antioxidant role and metabolic adaptations of the erythrocyte. Biol Chem 395:347-354.

Tellone E, Galtieri A, Russo A, Giardina B, Ficarra S (2015) Resveratrol: a focus on several neurodegenerative diseases. Oxid Med Cell Longev 2015:392169.

Zhao W, Varghese M, Yemul S, Pan Y, Cheng A, Marano P, Hassan S, Vempati P, Chen F, Qian X, Pasinetti GM (2011) Peroxisome proliferator activator receptor gamma coactivator-1alpha (PGC-1α) improves motor performance and survival in a mouse model of amyotrophic lateral sclerosis. Mol Neurodegener 6:51.

10.4103/1673-5374.175047 http∶//www.nrronline.org/

How to cite this article: Tellone E, Galtieri A, Russo A, Ficarra S (2016) How does resveratrol influence the genesis of some neurodegenerative diseases? Neural Regen Res 11(1):86-87.

猜你喜欢
主观变量检验
“美好生活”从主观愿望到执政理念的历史性提升
抓住不变量解题
也谈分离变量
加一点儿主观感受的调料
电梯检验中限速器检验的常见问题及解决对策探究
关于锅炉检验的探讨
小议离子的检验与共存
对立与存在
期末综合复习测试卷
分离变量法:常见的通性通法