Skip to content
BY 4.0 license Open Access Published by De Gruyter Open Access September 9, 2020

BDMC protects AD in vitro via AMPK and SIRT1

  • Chenlin Xu , Zijian Xiao , Heng Wu , Guijuan Zhou , Duanqun He , Yunqian Chang , Yihui Li , Gang Wang EMAIL logo and Ming Xie EMAIL logo

Abstract

Background

Alzheimer’s disease (AD) is a common neurodegenerative disorder without any satisfactory therapeutic approaches. AD is mainly characterized by the deposition of β-amyloid protein (Aβ) and extensive neuronal cell death. Curcumin, with anti-oxidative stress (OS) and cell apoptosis properties, plays essential roles in AD. However, whether bisdemethoxycurcumin (BDMC), a derivative of curcumin, can exert a neuroprotective effect in AD remains to be elucidated.

Methods

In this study, SK-N-SH cells were used to establish an in vitro model to investigate the effects of BDMC on the Aβ1–42-induced neurotoxicity. SK-N-SH cells were pretreated with BDMC and with or without compound C and EX527 for 30 min after co-incubation with rotenone for 24 h. Subsequently, western blotting, cell viability assay and SOD and GSH activity measurement were performed.

Results

BDMC increased the cell survival, anti-OS ability, AMPK phosphorylation levels and SIRT1 in SK-N-SH cells treated with Aβ1–42. However, after treatment with compound C, an AMPK inhibitor, and EX527, an SIRT1inhibitor, the neuroprotective roles of BDMC on SK-N-SH cells treated with Aβ1–42 were inhibited.

Conclusion

These results suggest that BDMC exerts a neuroprotective role on SK-N-SH cells in vitro via AMPK/SIRT1 signaling, laying the foundation for the application of BDMC in the treatment of neurodegenerative diseases related to AMPK/SIRT1 signaling.

1 Introduction

Alzheimer’s disease (AD), a severe and progressive brain disorder, is predicted to increasingly affect a significant portion of the aging human population worldwide [1]. AD is characterized by the accumulation of β-amyloid protein (Aβ) plaques, intracellular tangles and the loss of neurons in selective brain regions [2]. To date, available therapies such as cholinesterase inhibitors (including donepezil, galantamine and rivastigmine) and N-methyl-d-receptor antagonists (including memantine) [3] only alleviate disease symptoms of AD, and no effective therapeutic approaches were found to address the underlying pathological processes [4]. Consequently, discovering more effective drugs and uncovering the pathological mechanisms are urgent medical needs related to the treatment of AD.

Attempts to target individual molecules that may reduce the pathological impact of AD have been made to counteract cell death by the introduction of survival-proactive molecules. One of these molecules that we have studied is bisdemethoxycurcumin (BDMC), a natural derivative of curcumin, due to its ability to protect against Aβ neurotoxicity in AD [5,6]. However, its therapeutic potential is limited due to poor bioavailability, low solubility in aqueous media and poor pharmacokinetic profiles [7,8,9,10,11]. BDMC, the most potent and stable curcuminoid in biological systems [12,13,14], is produced based on the curcumin matrix by removing the 3-position methoxy group on the bilateral benzene ring and retaining the 4-positionhydroxyl group [15]. It was reported that BDMC exerts more promising pharmacological and biological effects than curcumin [16]. BDMC also shows a more potent apoptotic effect via downregulating the levels of heme oxygenase-1 and BCL-2 and upregulating the production of reactive oxygen species (ROS) compared with curcumin [17]. BDMC, with its higher polarity, hydrophilicity, water solubility, increased stability and improved nuclear cellular uptake than the parent curcumin, owns considerable anti-oxidant, anti-inflammatory and antiproliferative effects [14,18], which may be a more promising drug for clinical application.

In AD, Aβ is neurotoxic and initiates the hyperphosphorylation of tau, resulting in the dysfunction and the death of neurons [19]. In addition, there is an increase in oxidative stress (OS) in response to the increased Aβ levels [20], and this is considered to be an early event in the AD development [21,22]. Numerous types of signaling pathways may be associated with the pathological process of AD. Attempts to target signaling pathways that are associated with the pathological process of AD have been made to counteract OS and Aβ-induced cell death. One of the signaling pathways is AMP-activated protein kinase (AMPK), a key kinase associated with regulating cell energy metabolism, modulating the generation of Aβ [23] and exerting an essential role in energy homeostasis via responding to low cellular energy [24]. AMPK has also been revealed to have beneficial functions in cells including anti-oxidant, antitumor and anti-inflammation activities [25,26]. Another signaling pathway involved is silent information regulator 1 (SIRT1), an NAD+-dependent histone deacetylase, which has been shown to decrease aging and the progression of age-related disorders. An increase in OS during aging could also decrease the catabolic activity of SIRT1, possibly by ROS production [27]. Moreover, activated SIRT1 can control OS and generate neuroprotective effects [28]. Therefore, the AMPK-SIRT1 pathway may be potentially involved in the pathogenesis of AD.

The aim of this study is to investigate the effects of BDMC on SK-N-SH cells treated with Aβ1–42, the major component of amyloid plaques that is prominently increased in the human AD brain [29], and the underlying mechanisms. The results indicate that BDMC may exert a cell-protective role in SK-N-SH cells against Aβ1–42-induced neurotoxicity via AMPK/SIRT1 signaling.

2 Materials and methods

2.1 Synthesis of BDMC

Bisdemethoxycurcumin (BDMC, containing >80% curcumin and >94% curcuminoid content purity) purchased from Sigma-Aldrich (St. Louis, MO, USA) was dissolved in dimethyl sulfoxide (DMSO) and added to the cell culture medium for drug treatment in in vitro experiments according to our previous study [30].

2.2 Oligomeric Aβ1–42

Oligomeric Aβ1–42 was prepared in our lab according to the previous study [31]. Briefly, 1 mg of synthetic Aβ1–42 powder (1932-2-15, Shanghai Qiangyao Biological Technology, Shanghai, China) was dissolved in 22 µL DMSO and then diluted into Dulbecco’s modified Eagle’s medium (DMEM/F-12, Thermo Scientific Hyclone, Beijing, China) to produce a nontoxic concentration of 0.1% DMSO. The diluted solutions were incubated for 24 h at 4°C and centrifuged at 14,000 × g for 10 min. The supernatant was collected and used as 1 mM oligomeric Aβ1–42 for cell culture experiments.

2.3 Cell culture and treatments

SK-N-SH cells were cultured as previously described [32]. SK-N-SH cells purchased from Shanghai FUHENG Biotechnology Co., Ltd (FH0164, Shanghai, China) were cultured in DMEM/F-12 supplemented with 10% fetal bovine serum (Sijiqing Biotech Corp.) and 100 U/mpenicillin/streptomycin (ps) mixture (Solarbio Biotech Corp.) in 75 cm2 cell culture plates (Corning Inc.) at 37°C in a humidified 5% CO2 atmosphere.

To investigate the effects of BDMC on SK-N-SH cells treated with Aβ1–42, 1 × 104 SK-N-SH cells were seeded into 96-well cell culture plates (for cell viability assays) or 24-well cell culture plates (for SOD, GSH and western blot assays) and treated as follows: (1) cells were treated with 15 µM BDMC for 30 min following 24 h co-culture with Aβ1–42; (2) cells were treated with 10 µM compound C and 15 µM BDMC for 30 min following a 24 h co-culture with Aβ1–42; (3) cells were treated with 100 nM EX527 and 15 µM BDMC for 30 min following a 24 h co-culture with Aβ1–42. Subsequently, cell viability, SOD, GSH and western blot assays were performed.

2.4 Western blotting

Western blotting was performed as described in the previous studies [33,34,35] with minor modifications. Protein samples were separated via 10% SDS-PAGE and electroblotted onto polyvinylidene difluoride membranes (Millipore) for 3 h at 300 mA. The membranes were blocked with 5% nonfat dry milk or BSA dissolved in Tris–HCl saline buffer containing 0.1% Tween-20 (TBS-T, PH 7.4). Subsequently, the blots were incubated overnight at 4°C with one of the following antibodies: rabbit anti-p-AMPK (1:500; cat. no. ab23875; Abcam, USA) and rabbit anti-sirt1 (1:500; cat. no. ab220807; Abcam, USA). Membranes were washed three times for 5 min each time in TBS-T. HRP-coupled goat antirabbit secondary antibodies (1:1,000; Boster, Wuhan, China) diluted in TBS-T were then applied for 1 h. Membranes were washed three times in TBS-T for 5 min each time at room temperature. Immunoreactive signals were then visualized with the enhanced chemiluminescence solution (Bio-Rad, USA). Signal intensities were quantified by densitometric analysis using ImageJ software (Dental Diagnosis Science, San Antonio, TX).

2.5 MTT assay

MTT assay was performed as previously described [36,37,38] with minor modifications. At the indicated time points, SK-N-SH cells were maintained in culture medium supplemented with 10 µL 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT, 500 µg/mL) (cat. no. M1020; Solarbio, China) for 4 h. Following aspiration of the culture medium, 100 µL DMSO was added to each well in the culture plates, and the cells were incubated at 37°C for 30 min. Optical density was measured spectrophotometrically at a wavelength of 410 nm.

2.6 Measurement of SOD activity

The WST-1 Cell Proliferation Assay kit was used to detect the SOD activity according to the manufacturer’s instructions (cat. no. A001-3-2; Jiancheng Biotech Ltd, Nanjing, China) [39,40]. The xanthine–xanthine oxidase system was applied to produce superoxide ions, which can react with 2-(4-iodophenyl)-3-(4-nitrophenol-5-phenlyltetrazolium chloride) to form a red formazan dye. The absorbance was determined at a wavelength of 550 nm. Protein concentration was determined using a BCA protein assay kit (QPBCA, Sigma-Aldrich, USA). The values were expressed as units per mg protein. One unit of SOD was defined as the amount of SOD inhibiting the rate of reaction by 50% at 25°C.

2.7 Measurement of GSH

GSH measurement was performed according to the previous method with minor modifications [41]. SK-N-SH cells in culture medium were centrifuged at 500 × g for 10 min and washed twice with PBS. The collected SK-N-SH cells were resuspended in the protein removal reagent and vigorously vortexed. Subsequently, the samples were rapidly frozen and thawed with liquid nitrogen twice at 37°C followed by incubation at 4°C for 5 min. After centrifugation at 10,000 × g for 10 min, the supernatant was collected. The GSH levels were determined using a GSH and GSSG assay kit (cat. no. S0053; Beyotime Institute of Biotechnology, Shanghai, China) according to the manufacturer’s instructions.

2.8 Statistics

Data were represented as the mean ± SEM. Comparison between groups was performed using Student’s t-test for independent samples using SPSS 18.0. Statistical significance was considered when P < 0.05.

3 Results

3.1 BDMC enhances AMPK phosphorylation and SIRT1 expression levels in SK-N-SH cells treated with Aβ1–42

To investigate the effects of BDMC on AMPK phosphorylation and SIRT expression in SK-N-SH cells induced by Aβ1–42, western blotting was performed after the cells were pretreated with BDMC and co-cultured with Aβ1–42 for 24 h.

Compared with the control group, BDMC treatment did not increase the AMPK phosphorylation levels, and the AMPK phosphorylation levels were decreased in response to Aβ1–42 treatment. Compared with the Aβ1–42-induced group, BDMC significantly increased the AMPK phosphorylation levels (Figure 1a and b). A similar pattern for SITR1 expression levels was observed (Figure 1c and d).

Figure 1 
            Effects of BDMC on AMPK phosphorylation and SIRT1 expression levels in SK-N-SH cells induced by Aβ1–42. A total of 1 × 104 SK-N-SH cells were pretreated with BDMC at a concentration of 15 µM for 30 min before 24 h co-culture with Aβ1–42. Western blotting was then performed to assess the AMPK phosphorylation and SIRT1 expression levels. (a and b) AMPK phosphorylation (c) and SIRT1 (d) levels were upregulated after BDMC treatment. *P < 0.05 vs control group; #P < 0.05 vs Aβ1–42-induced group from five independent experiments.
Figure 1

Effects of BDMC on AMPK phosphorylation and SIRT1 expression levels in SK-N-SH cells induced by Aβ1–42. A total of 1 × 104 SK-N-SH cells were pretreated with BDMC at a concentration of 15 µM for 30 min before 24 h co-culture with Aβ1–42. Western blotting was then performed to assess the AMPK phosphorylation and SIRT1 expression levels. (a and b) AMPK phosphorylation (c) and SIRT1 (d) levels were upregulated after BDMC treatment. *P < 0.05 vs control group; #P < 0.05 vs Aβ1–42-induced group from five independent experiments.

3.2 BDMC enhances the cell survival rate of SK-N-SH cells treated with Aβ1–42 via AMPK and SIRT1

To investigate the effects of BDMC on the cell survival of SK-N-SH cells induced by Aβ1–42, the cell viability was evaluated after the cells were pretreated with BDMC and with or without compound C or EX527 and co-cultured with Aβ1–42 for 24 h.

Compared with the control group, the cell survival rate decreased in response to Aβ1–42 treatment. Compared with the Aβ1–42-induced group, BDMC significantly increased the cell survival rate. However, following AMPK and SIRT1 inhibition, BDMC did not increase the cell survival rate (Figure 2).

Figure 2 
            Effects of BDMC on the cell survival in SH-SY5Y cells treated with SK-N-SH cells induced by Aβ1–42 after inhibition of the AMPK/SIRT1 signaling pathway. A total of 1 × 104 SH-SY5Y cells were pretreated with 10 µM compound C (an AMPK signaling pathway inhibitor) or 100 nM EX527 and 15 µM BDMC for 30 min before 24 h co-culture with Aβ1–42. The cell survival rate is upregulated in SK-N-SH cells induced by Aβ1–42 in response to BDMC treatment, but did not increase upon inhibition of the AMPK/SIRT1 signaling pathway. **p < 0.01 and ***P < 0.01 from five independent experiments.
Figure 2

Effects of BDMC on the cell survival in SH-SY5Y cells treated with SK-N-SH cells induced by Aβ1–42 after inhibition of the AMPK/SIRT1 signaling pathway. A total of 1 × 104 SH-SY5Y cells were pretreated with 10 µM compound C (an AMPK signaling pathway inhibitor) or 100 nM EX527 and 15 µM BDMC for 30 min before 24 h co-culture with Aβ1–42. The cell survival rate is upregulated in SK-N-SH cells induced by Aβ1–42 in response to BDMC treatment, but did not increase upon inhibition of the AMPK/SIRT1 signaling pathway. **p < 0.01 and ***P < 0.01 from five independent experiments.

3.3 BDMC enhances the SOD levels of SK-N-SH cells treated with Aβ1–4 via AMPK and SIRT1

To investigate the effects of BDMC on the SOD activity of SK-N-SH cells induced by Aβ1–42, SOD activity was evaluated after the cells were pretreated with BDMC and with or without compound C or EX527 and co-cultured with Aβ1–42 for 24 h.

Compared with the control group, SOD levels were decreased in response to Aβ1–42 treatment. Compared with the Aβ1–42-induced group, BDMC significantly increased the SOD levels. However, after AMPK and SIRT1 inhibition, BDMC did not increase the SOD levels (Figure 3).

Figure 3 
            Effects of BDMC on SOD levels in SH-SY5Y cells treated with SK-N-SH cells induced by Aβ1–42 after inhibition of the AMPK/SIRT1 signaling pathway. A total of 1 × 104 SH-SY5Y cells were pretreated with 10 µM compound C (an AMPK signaling pathway inhibitor) or 100 nM EX527 and 15 µM BDMC for 30 min before a 24 h co-culture with Aβ1–42. SOD levels are upregulated in SK-N-SH cells induced by Aβ1–42 in response BDMC treatment, but did not increase upon inhibition of the AMPK/SIRT1 signaling pathway.***P < 0.01 from five independent experiments.
Figure 3

Effects of BDMC on SOD levels in SH-SY5Y cells treated with SK-N-SH cells induced by Aβ1–42 after inhibition of the AMPK/SIRT1 signaling pathway. A total of 1 × 104 SH-SY5Y cells were pretreated with 10 µM compound C (an AMPK signaling pathway inhibitor) or 100 nM EX527 and 15 µM BDMC for 30 min before a 24 h co-culture with Aβ1–42. SOD levels are upregulated in SK-N-SH cells induced by Aβ1–42 in response BDMC treatment, but did not increase upon inhibition of the AMPK/SIRT1 signaling pathway.***P < 0.01 from five independent experiments.

3.4 BDMC enhances the GSH levels of SK-N-SH cells treated with Aβ1–42 via AMPK and SIRT1

To investigate the effects of BDMC on the GSH levels of SK-N-SH cells induced by Aβ1–42, GSH levels were evaluated after the cells were pretreated with BDMC and with or without compound C or EX527 and co-cultured with Aβ1–42 for 24 h.

Compared with the control group, the GSH levels were decreased in response to Aβ1–42 treatment. Compared with the Aβ1–42-induced group, BDMC significantly increased the GSH levels. However, after AMPK and SIRT1 inhibition, BDMC did not increase the GSH levels (Figure 4).

Figure 4 
            Effects of BDMC on GSH levels in SH-SY5Y cells treated with SK-N-SH cells induced by Aβ1–42 after inhibition of the AMPK/SIRT1 signaling pathway. A total of 1 × 104 SH-SY5Y cells were pretreated with 10 µM compound C (an AMPK signaling pathway inhibitor) or 100 nM EX527 and 15 µM BDMC for 30 min before 24 h co-culture with Aβ1–42. GSH levels are upregulated in SK-N-SH cells induced by Aβ1–42 in response to the treatment of BDMC but did not increase upon inhibition of the AMPK/SIRT1 signaling pathway. ***P < 0.01 from five independent experiments.
Figure 4

Effects of BDMC on GSH levels in SH-SY5Y cells treated with SK-N-SH cells induced by Aβ1–42 after inhibition of the AMPK/SIRT1 signaling pathway. A total of 1 × 104 SH-SY5Y cells were pretreated with 10 µM compound C (an AMPK signaling pathway inhibitor) or 100 nM EX527 and 15 µM BDMC for 30 min before 24 h co-culture with Aβ1–42. GSH levels are upregulated in SK-N-SH cells induced by Aβ1–42 in response to the treatment of BDMC but did not increase upon inhibition of the AMPK/SIRT1 signaling pathway. ***P < 0.01 from five independent experiments.

4 Discussion

In this study, we revealed that BDMC enhanced the cell survival, anti-OS ability and the levels of AMPK phosphorylation and SIRT1 in SK-N-SH cells treated with Aβ1–42. However, after inhibition of the AMPK/SIRT1 signaling pathway, BDMC cannot exert these neuroprotective roles. This study demonstrated that BDMC protected SK-N-SH cells from the neurotoxicity induced by the Aβ1–42 treatment via the AMPK/SIRT1 signaling pathway.

The accumulation of Aβ peptides was identified as a key step in the multiple pathogenic changes involved in neurodegeneration and dementia [42,43]. Aβ peptide fragments were observed to directly or indirectly induce neuronal cell death [44,45,46]. In vivo, small, stable oligomers of Aβ1–42 have been isolated from the brain, plasma and cerebrospinal fluid [47,48,49] and correlated with the severity of neurodegeneration in AD [50,51]. Previous studies demonstrated that neurotoxicity induced by Aβ1–42 can lead to the apoptotic cell death [52,53]. Therefore, the inhibition of neuronal apoptosis induced by Aβ protein provides a feasible method for the prevention and treatment of AD. In the present study, we observed that BDMC can decrease the cell death of SK-N-SH cells induced by Aβ1–42.

One of the most well-known and studied effects of Aβ is its capacity to induce, and be induced by, OS; thus, Aβ induces OS in vivo and in vitro [54,55,56,57]. OS is able to induce the increased production of Aβ [58,59,60]. A previous study reported that the accumulation of intraneuronal Aβ oligomers resulted in the dysfunction of mitochondria and synapses after neurotoxicity; moreover, mitochondria-targeted antioxidants can protect against the neurotoxicity of Aβ in AD [61,62,63]. SOD and GSH are important antioxidants by scavenging oxygen free radicals against OS. SOD is a metal enzyme that can catalyze the dismutation of superoxide anions, scavenge O2 and repair damaged cells. It is an important enzyme that can scavenge free radicals in the brains of AD mice [64]. SOD levels directly reflect the degree of senescence in mice brain cells [65]. The profound role of GSH as a detoxifying agent in the brain is critically important [66]. GSH is also involved in other cellular processes such as neuroinflammation and ferroptosis, which brings the attention of pharmacologists pertaining to medical interventions for therapeutic benefits. Depleted levels of GSH trigger ROS generation implicated in the cell death that causes various neurological diseases like AD [67]. In the present study, we observed that BDMC can increase SOD and GSH levels of SK-N-SH cells induced by Aβ1–42.

AMPK can modulate α and β-secretases expression, thus influencing Aβ generation and APP processing [68]. Upon inflammation, the combination of AMPK with SIRT1could exert synergistic effects to jointly maintain energy homeostasis [69,70]. A decline in the SIRT1 activity in mice could be related to oxidative damage [71]. In this study, we observed that the phosphorylation of the AMPK/SIRT1 signaling pathway was upregulated in response to BDMC treatment, whereas BDMC cannot exert any functional roles after inhibiting this pathway.

To conclude, this study found that BDMC alleviated the neurotoxicity of rotenone in SK-N-SH cells by improving cell survival and anti-OS. These effects might be exerted via inhibiting the AMPK/SIRT1 signaling pathway. This may lay the foundation for BDMC to be a novel strategy for the treatment of AD.

Despite the results, this study still has some limitations. Further studies, including TUNEL assays, are required to investigate the modulating roles of BDMC on free radicals and cell death in treated cells under the pathological conditions of AD.


These authors contributed equally to this study.


Acknowledgments

We thank the Hunan Provincial Natural Science Foundation of China (grant no. 2019JJ40267, 2017JJ3273, 2018JJ2358), Scientific Research Fund Project of Hunan Provincial Health Commission (grant no. 20201911, 20200469, 20201963, C20180238) and Hengyang Science and Technology Project (grant no. 2016KJ38) for support. We also thank Dr Shuangxi Chen in our lab for the kind help in the production of oligomeric Aβ1–42.

  1. Conflict of interest: The authors state no conflict of interest.

References

[1] Ferri CP, Prince M, Brayne C, Brodaty H, Fratiglioni L, Ganguli M, et al. Global prevalence of dementia: a Delphi consensus study. Lancet. 2005;366:2112–7. 10.1016/s0140-6736(05)67889-0.Search in Google Scholar

[2] Zhang J, Zhen YF, Pu Bu Ci R, Song LG, Kong WN, Shao TM, et al. Salidroside attenuates beta amyloid-induced cognitive deficits via modulating oxidative stress and inflammatory mediators in rat hippocampus. Behav Brain Res. 2013;244:70–81. 10.1016/j.bbr.2013.01.037.Search in Google Scholar PubMed

[3] Rygiel K. Novel strategies for Alzheimer’s disease treatment: an overview of anti-amyloid beta monoclonal antibodies. Indian J Pharmacol. 2016;48:629–36. 10.4103/0253-7613.194867.Search in Google Scholar PubMed PubMed Central

[4] Sadowski M, Wisniewski T. Disease modifying approaches for Alzheimer’s pathology. Curr Pharm Des. 2007;13:1943–54.10.2174/138161207781039788Search in Google Scholar PubMed

[5] Reddy PH, Manczak M, Yin X, Grady MC, Mitchell A, Kandimalla R, et al. Protective effects of a natural product, curcumin, against amyloid β induced mitochondrial and synaptic toxicities in Alzheimer’s disease. J Investig Med. 2016;64:1220–34. 10.1136/jim-2016-000240.Search in Google Scholar PubMed PubMed Central

[6] Reddy PH, Manczak M, Yin X, Grady MC, Mitchell A, Tonk S, et al. Protective Effects of Indian Spice Curcumin Against Amyloid-β in Alzheimer’s Disease. J Alzheimers Dis. 2018;61:843–66. 10.3233/jad-170512.Search in Google Scholar PubMed PubMed Central

[7] Prasad S, Gupta SC, Tyagi AK, Aggarwal BB. Curcumin, a component of golden spice: from bedside to bench and back. Biotechnol Adv. 2014;32:1053–64. 10.1016/j.biotechadv.2014.04.004.Search in Google Scholar PubMed

[8] Anand P, Kunnumakkara AB, Newman RA, Aggarwal BB. Bioavailability of curcumin: problems and promises. Mol Pharm. 2007;4:807–18. 10.1021/mp700113r.Search in Google Scholar PubMed

[9] Schneider C, Gordon ON, Edwards RL, Luis PB. Degradation of curcumin: from mechanism to biological implications. J Agric Food Chem. 2015;63:7606–14. 10.1021/acs.jafc.5b00244.Search in Google Scholar PubMed PubMed Central

[10] Ghosh S, Banerjee S, Sil PC. The beneficial role of curcumin on inflammation, diabetes and neurodegenerative disease: a recent update. Food Chem Toxicol. 2015;83:111–24. 10.1016/j.fct.2015.05.022.Search in Google Scholar PubMed

[11] Nelson KM, Dahlin JL, Bisson J, Graham J, Pauli GF, Walters MA. The essential medicinal chemistry of curcumin. J Med Chem. 2017;60:1620–37. 10.1021/acs.jmedchem.6b00975.Search in Google Scholar PubMed PubMed Central

[12] Cashman JR, Ghirmai S, Abel KJ, Fiala M. Immune defects in Alzheimer’s disease: new medications development. BMC Neurosci. 2008;9(Suppl 2):S13. 10.1186/1471-2202-9-s2-s13.Search in Google Scholar

[13] Fiala M, Liu PT, Espinosa-Jeffrey A, Rosenthal MJ, Bernard G, Ringman JM, et al. Innate immunity and transcription of MGAT-III and Toll-like receptors in Alzheimer’s disease patients are improved by bisdemethoxycurcumin. Proc Natl Acad Sci U S A. 2007;104:12849–54. 10.1073/pnas.0701267104.Search in Google Scholar PubMed PubMed Central

[14] Sandur SK, Pandey MK, Sung B, Ahn KS, Murakami A, Sethi G, et al. Curcumin, demethoxycurcumin, bisdemethoxycurcumin, tetrahydrocurcumin and turmerones differentially regulate anti-inflammatory and anti-proliferative responses through a ROS-independent mechanism. Carcinogenesis. 2007;28:1765–73. 10.1093/carcin/bgm123.Search in Google Scholar PubMed

[15] Zhang J, Han H, Shen M, Zhang L, Wang T. Comparative studies on the antioxidant profiles of curcumin and bisdemethoxycurcumin in erythrocytes and broiler chickens. Animals. 2019;9:953. 10.3390/ani9110953.Search in Google Scholar PubMed PubMed Central

[16] Ramezani M, Hatamipour M, Sahebkar A. Promising anti-tumor properties of bisdemethoxycurcumin: a naturally occurring curcumin analogue. J Cell Physiol. 2018;233:880–7. 10.1002/jcp.25795.Search in Google Scholar PubMed

[17] Lee AY, Fan CC, Chen YA, Cheng CW, Sung YJ, Hsu CP, et al. Curcumin inhibits invasiveness and epithelial-mesenchymal transition in oral squamous cell carcinoma through reducing matrix metalloproteinase 2, 9 and modulating p53-E-cadherin pathway. Integr Cancer Ther. 2015;14:484–90. 10.1177/1534735415588930.Search in Google Scholar PubMed

[18] Basile V, Ferrari E, Lazzari S, Belluti S, Pignedoli F, Imbriano C. Curcumin derivatives: molecular basis of their anti-cancer activity. Biochem Pharmacol. 2009;78:1305–15. 10.1016/j.bcp.2009.06.105.Search in Google Scholar PubMed

[19] Yankner BA. Mechanisms of neuronal degeneration in Alzheimer’s disease. Neuron. 1996;16:921–32. 10.1016/s0896-6273(00)80115-4.Search in Google Scholar PubMed

[20] Yu W, Bonnet M, Farso M, Ma K, Chabot JG, Martin E, et al. The expression of apoptosis inducing factor (AIF) is associated with aging-related cell death in the cortex but not in the hippocampus in the TgCRND8 mouse model of Alzheimer’s disease. BMC Neurosci. 2014;15:73. 10.1186/1471-2202-15-73.Search in Google Scholar PubMed PubMed Central

[21] Nunomura A, Perry G, Aliev G, Hirai K, Takeda A, Balraj EK, et al. Oxidative damage is the earliest event in Alzheimer disease. J Neuropathol Exp Neurol. 2001;60:759–67. 10.1093/jnen/60.8.759.Search in Google Scholar PubMed

[22] Cutler RG, Kelly J, Storie K, Pedersen WA, Tammara A, Hatanpaa K, et al. Involvement of oxidative stress-induced abnormalities in ceramide and cholesterol metabolism in brain aging and Alzheimer’s disease. Proc Natl Acad Sci U S A. 2004;101:2070–5. 10.1073/pnas.0305799101.Search in Google Scholar PubMed PubMed Central

[23] Barnes DE, Yaffe K. The projected effect of risk factor reduction on Alzheimer’s disease prevalence. Lancet Neurol. 2011;10:819–28. 10.1016/s1474-4422(11)70072-2.Search in Google Scholar

[24] Emerling BM, Weinberg F, Snyder C, Burgess Z, Mutlu GM, Viollet B, et al. Hypoxic activation of AMPK is dependent on mitochondrial ROS but independent of an increase in AMP/ATP ratio. Free Radic Biol Med. 2009;46:1386–91. 10.1016/j.freeradbiomed.2009.02.019.Search in Google Scholar PubMed PubMed Central

[25] Kim YW, Lee SM, Shin SM, Hwang SJ, Brooks JS, Kang HE, et al. Efficacy of sauchinone as a novel AMPK-activating lignan for preventing iron-induced oxidative stress and liver injury. Free Radic Biol Med. 2009;47:1082–92. 10.1016/j.freeradbiomed.2009.07.018.Search in Google Scholar PubMed

[26] Shin SM, Kim SG. Inhibition of arachidonic acid and iron-induced mitochondrial dysfunction and apoptosis by oltipraz and novel 1,2-dithiole-3-thione congeners. Mol Pharmacol. 2009;75:242–53. 10.1124/mol.108.051128.Search in Google Scholar PubMed

[27] Durairajan SS, Huang YY, Yuen PY, Chen LL, Kwok KY, Liu LF, et al. Effects of Huanglian-Jie-Du-Tang and its modified formula on the modulation of amyloid-β precursor protein processing in Alzheimer’s disease models. PLoS One. 2014;9:e92954. 10.1371/journal.pone.0092954.Search in Google Scholar PubMed PubMed Central

[28] Ma X, Sun Z, Han X, Li S, Jiang X, Chen S, et al. Neuroprotective effect of resveratrol via activation of Sirt1 signaling in a rat model of combined diabetes and Alzheimer’s disease. Front Neurosci. 2019;13:1400. 10.3389/fnins.2019.01400.Search in Google Scholar PubMed PubMed Central

[29] LaFerla FM, Green KN, Oddo S. Intracellular amyloid-beta in Alzheimer’s disease. Nat Rev Neurosci. 2007;8:499–509. 10.1038/nrn2168.Search in Google Scholar PubMed

[30] He D, Chen S, Xiao Z, Wu H, Zhou G, Xu C, et al. Bisdemethoxycurcumin exerts a cell-protective effect via JAK2/STAT3 signaling in a rotenone-induced Parkinson’s disease model in vitro. Folia Histochem Cytobiol. 2020;58:127-34. 10.5603/FHC.a2020.0011.Search in Google Scholar PubMed

[31] Chen S, Jiang Q, Huang P, Hu C, Shen H, Schachner M, et al. The L1 cell adhesion molecule affects protein kinase D1 activity in the cerebral cortex in a mouse model of Alzheimer’s disease. Brain Res Bull. 2020;162:141–50. 10.1016/j.brainresbull.2020.06.004.Search in Google Scholar PubMed

[32] Liu Y, Yu Y, Schachner M, Zhao W. Neuregulin 1-β regulates cell adhesion molecule L1 expression in the cortex and hippocampus of mice. Biochem Biophys Res Commun. 2013;441:7–12. 10.1016/j.bbrc.2013.09.102.Search in Google Scholar PubMed

[33] Chen SX, Hu CL, Liao YH, Zhao WJ. L1 modulates PKD1 phosphorylation in cerebellar granule neurons. Neurosci Lett. 2015;584:331–6. 10.1016/j.neulet.2014.11.012.Search in Google Scholar PubMed

[34] Jiang Q, Chen S, Hu C, Huang P, Shen H, Zhao W. Neuregulin-1 (Nrg1) signaling has a preventive role and is altered in the frontal cortex under the pathological conditions of Alzheimer’s disease. Mol Med Rep. 2016;14:2614–24. 10.3892/mmr.2016.5542.Search in Google Scholar PubMed PubMed Central

[35] Xu J, Hu C, Chen S, Shen H, Jiang Q, Huang P, et al. Neuregulin-1 protects mouse cerebellum against oxidative stress and neuroinflammation. Brain Res. 2017;1670:32–43. 10.1016/j.brainres.2017.06.012.Search in Google Scholar PubMed

[36] Chen S, Hou Y, Zhao Z, Luo Y, Lv S, Wang Q, et al. Neuregulin-1 accelerates functional motor recovery by improving motoneuron survival after brachial plexus root avulsion in mice. Neuroscience. 2019;404:510–8. 10.1016/j.neuroscience.2019.01.054.Search in Google Scholar PubMed

[37] Li J, Chen S, Zhao Z, Luo Y, Hou Y, Li H, et al. Effect of VEGF on inflammatory regulation, neural survival, and functional improvement in rats following a complete spinal cord transection. Front Cell Neurosci. 2017;11:381. 10.3389/fncel.2017.00381.Search in Google Scholar PubMed PubMed Central

[38] Chen SX, He JH, Mi YJ, Shen HF, Schachner M, Zhao WJ. A mimetic peptide of α2,6-sialyllactose promotes neuritogenesis. Neural Regen Res. 2020;15:1058–65. 10.4103/1673-5374.270313.Search in Google Scholar PubMed PubMed Central

[39] Yuhai GU, Zhen Z. Significance of the changes occurring in the levels of interleukins, SOD and MDA in rat pulmonary tissue following exposure to different altitudes and exposure times. Exp Ther Med. 2015;10:915–20. 10.3892/etm.2015.2604.Search in Google Scholar PubMed PubMed Central

[40] Mao GX, Zheng LD, Cao YB, Chen ZM, Lv YD, Wang YZ, et al. Antiaging effect of pine pollen in human diploid fibroblasts and in a mouse model induced by d-galactose. Oxid Med Cell Longev. 2012;2012:750963. 10.1155/2012/750963.Search in Google Scholar PubMed PubMed Central

[41] Zhang B, Chen N, Chen H, Wang Z, Zheng Q. The critical role of redox homeostasis in shikonin-induced HL-60 cell differentiation via unique modulation of the Nrf2/ARE pathway. Oxid Med Cell Longev. 2012;2012:781516. 10.1155/2012/781516.Search in Google Scholar PubMed PubMed Central

[42] Hardy JA, Higgins GA. Alzheimer’s disease: the amyloid cascade hypothesis. Science. 1992;256:184–5.10.1126/science.1566067Search in Google Scholar PubMed

[43] Karran E, Mercken M, De Strooper B. The amyloid cascade hypothesis for Alzheimer’s disease: an appraisal for the development of therapeutics. Nat Rev Drug Discovery. 2011;10:698–712. 10.1038/nrd3505.Search in Google Scholar PubMed

[44] Su JH, Anderson AJ, Cummings BJ, Cotman CW. Immunohistochemical evidence for apoptosis in Alzheimer’s disease. Neuroreport. 1994;5:2529–33.10.1097/00001756-199412000-00031Search in Google Scholar PubMed

[45] Yagami T, Ueda K, Asakura K, Sakaeda T, Nakazato H, Kuroda T, et al. Gas6 rescues cortical neurons from amyloid beta protein-induced apoptosis. Neuropharmacology. 2002;43:1289–96.10.1016/S0028-3908(02)00333-7Search in Google Scholar

[46] Wang DM, Li SQ, Zhu XY, Wang Y, Wu WL, Zhang XJ. Protective effects of hesperidin against amyloid-beta (Abeta) induced neurotoxicity through the voltage dependent anion channel 1 (VDAC1)-mediated mitochondrial apoptotic pathway in PC12 cells. Neurochem Res. 2013;38:1034–44. 10.1007/s11064-013-1013-4.Search in Google Scholar PubMed

[47] Kuo YM, Emmerling MR, Vigo-Pelfrey C, Kasunic TC, Kirkpatrick JB, Murdoch GH, et al. Water-soluble Abeta (N-40, N-42) oligomers in normal and Alzheimer disease brains. J Biol Chem. 1996;271:4077–81. 10.1074/jbc.271.8.4077.Search in Google Scholar PubMed

[48] Roher AE, Baudry J, Chaney MO, Kuo YM, Stine WB, Emmerling MR. Oligomerizaiton and fibril asssembly of the amyloid-beta protein. Biochim Biophys Acta. 2000;1502:31–43. 10.1016/s0925-4439(00)00030-2.Search in Google Scholar PubMed

[49] Roher AE, Chaney MO, Kuo YM, Webster SD, Stine WB, Haverkamp LJ, et al. Morphology and toxicity of Abeta-(1-42) dimer derived from neuritic and vascular amyloid deposits of Alzheimer’s disease. J Biol Chem. 1996;271:20631–5. 10.1074/jbc.271.34.20631.Search in Google Scholar PubMed

[50] Lue LF, Kuo YM, Roher AE, Brachova L, Shen Y, Sue L, et al. Soluble amyloid beta peptide concentration as a predictor of synaptic change in Alzheimer’s disease. Am J Pathol. 1999;155:853–62. 10.1016/s0002-9440(10)65184-x.Search in Google Scholar PubMed PubMed Central

[51] McLean CA, Cherny RA, Fraser FW, Fuller SJ, Smith MJ, Beyreuther K, et al. Soluble pool of Abeta amyloid as a determinant of severity of neurodegeneration in Alzheimer’s disease. Ann Neurol. 1999;46:860–6. 10.1002/1531-8249(199912)46:6<860:aid-ana8>3.0.co;2-m.Search in Google Scholar

[52] Thangnipon W, Puangmalai N, Chinchalongporn V, Jantrachotechatchawan C, Kitiyanant N, Soi-Ampornkul R, et al. N-Benzylcinnamide protects rat cultured cortical neurons from beta-amyloid peptide-induced neurotoxicity. Neurosci Lett. 2013;556:20–5. 10.1016/j.neulet.2013.09.071.Search in Google Scholar

[53] Thangnipon W, Suwanna N, Kitiyanant N, Soi-Ampornkul R, Tuchinda P, Munyoo B, et al. Protective role of N-trans-feruloyltyramine against beta-amyloid peptide-induced neurotoxicity in rat cultured cortical neurons. Neurosci Lett. 2012;513:229–32. 10.1016/j.neulet.2012.02.047.Search in Google Scholar

[54] Hensley K, Carney JM, Mattson MP, Aksenova M, Harris M, Wu JF, et al. A model for beta-amyloid aggregation and neurotoxicity based on free radical generation by the peptide: relevance to Alzheimer disease. Proc Natl Acad Sci U S A. 1994;91:3270–4. 10.1073/pnas.91.8.3270.Search in Google Scholar

[55] Mark RJ, Lovell MA, Markesbery WR, Uchida K, Mattson MP. A role for 4-hydroxynonenal, an aldehydic product of lipid peroxidation, in disruption of ion homeostasis and neuronal death induced by amyloid beta-peptide. J Neurochem. 1997;68:255–64. 10.1046/j.1471-4159.1997.68010255.x.Search in Google Scholar

[56] Murakami K, Irie K, Ohigashi H, Hara H, Nagao M, Shimizu T, et al. Formation and stabilization model of the 42-mer Abeta radical: implications for the long-lasting oxidative stress in Alzheimer’s disease. J Am Chem Soc. 2005;127:15168–74. 10.1021/ja054041c.Search in Google Scholar

[57] Tabner BJ, El-Agnaf OM, Turnbull S, German MJ, Paleologou KE, Hayashi Y, et al. Hydrogen peroxide is generated during the very early stages of aggregation of the amyloid peptides implicated in Alzheimer disease and familial British dementia. J Biol Chem. 2005;280:35789–92. 10.1074/jbc.C500238200.Search in Google Scholar

[58] Paola D, Domenicotti C, Nitti M, Vitali A, Borghi R, Cottalasso D, et al. Oxidative stress induces increase in intracellular amyloid beta-protein production and selective activation of betaI and betaII PKCs in NT2 cells. Biochem Biophys Res Commun. 2000;268:642–6. 10.1006/bbrc.2000.2164.Search in Google Scholar

[59] Tamagno E, Bardini P, Obbili A, Vitali A, Borghi R, Zaccheo D, et al. Oxidative stress increases expression and activity of BACE in NT2 neurons. Neurobiol Dis. 2002;10:279–88. 10.1006/nbdi.2002.0515.Search in Google Scholar

[60] Tong Y, Zhou W, Fung V, Christensen MA, Qing H, Sun X, et al. Oxidative stress potentiates BACE1 gene expression and Abeta generation. J Neural Transm. 2005;112:455–69. 10.1007/s00702-004-0255-3.Search in Google Scholar

[61] Calkins MJ, Manczak M, Mao P, Shirendeb U, Reddy PH. Impaired mitochondrial biogenesis, defective axonal transport of mitochondria, abnormal mitochondrial dynamics and synaptic degeneration in a mouse model of Alzheimer’s disease. Hum Mol Genet. 2011;20:4515–29. 10.1093/hmg/ddr381.Search in Google Scholar PubMed PubMed Central

[62] Manczak M, Mao P, Calkins MJ, Cornea A, Reddy AP, Murphy MP, et al. Mitochondria-targeted antioxidants protect against amyloid-beta toxicity in Alzheimer’s disease neurons. J Alzheimers Dis. 2010;20(Suppl 2):S609–31. 10.3233/jad-2010-100564.Search in Google Scholar PubMed PubMed Central

[63] Reddy PH, Manczak M, Yin X, Reddy AP. Synergistic protective effects of mitochondrial division inhibitor 1 and mitochondria-targeted small peptide SS31 in Alzheimer’s disease. J Alzheimers Dis. 2018;62:1549–65. 10.3233/jad-170988.Search in Google Scholar

[64] Ji D, Wu X, Li D, Liu P, Zhang S, Gao D, et al. Protective effects of chondroitin sulphate nano-selenium on a mouse model of Alzheimer’s disease. Int J Biol Macromol. 2020;154:233–45. 10.1016/j.ijbiomac.2020.03.079.Search in Google Scholar PubMed

[65] Perry JJ, Shin DS, Getzoff ED, Tainer JA. The structural biochemistry of the superoxide dismutases. Biochim Biophys Acta. 2010;1804:245–62. 10.1016/j.bbapap.2009.11.004.Search in Google Scholar PubMed PubMed Central

[66] Kety SS. The circulation and energy metabolism of the brain. Clin Neurosurg. 1963;9:56–66. 10.1093/neurosurgery/9.cn_suppl_1.56.Search in Google Scholar PubMed

[67] Lovell MA, Ehmann WD, Butler SM, Markesbery WR. Elevated thiobarbituric acid-reactive substances and antioxidant enzyme activity in the brain in Alzheimer’s disease. Neurology. 1995;45:1594–601. 10.1212/wnl.45.8.1594.Search in Google Scholar PubMed

[68] Lu J, Wu DM, Zheng YL, Hu B, Zhang ZF, Shan Q, et al. Quercetin activates AMP-activated protein kinase by reducing PP2C expression protecting old mouse brain against high cholesterol-induced neurotoxicity. J Pathol. 2010;222:199–212. 10.1002/path.2754.Search in Google Scholar PubMed

[69] Seixas da Silva GS, Melo HM, Lourenco MV, Lyra ESNM, de Carvalho MB, Alves-Leon SV, et al. Amyloid-β oligomers transiently inhibit AMP-activated kinase and cause metabolic defects in hippocampal neurons. J Biol Chem. 2017;292:7395–406. 10.1074/jbc.M116.753525.Search in Google Scholar PubMed PubMed Central

[70] Peairs A, Radjavi A, Davis S, Li L, Ahmed A, Giri S, et al. Activation of AMPK inhibits inflammation in MRL/lpr mouse mesangial cells. Clin Exp Immunol. 2009;156:542–51. 10.1111/j.1365-2249.2009.03924.x.Search in Google Scholar PubMed PubMed Central

[71] Braidy N, Guillemin GJ, Mansour H, Chan-Ling T, Poljak A, Grant R. Age related changes in NAD+ metabolism oxidative stress and Sirt1 activity in wistar rats. PLoS One. 2011;6:e19194. 10.1371/journal.pone.0019194.Search in Google Scholar PubMed PubMed Central

Received: 2020-05-22
Revised: 2020-08-04
Accepted: 2020-08-13
Published Online: 2020-09-09

© 2020 Chenlin Xu et al., published by De Gruyter

This work is licensed under the Creative Commons Attribution 4.0 International License.

Downloaded on 24.4.2024 from https://www.degruyter.com/document/doi/10.1515/tnsci-2020-0140/html
Scroll to top button