Open Access

PNU282987 inhibits amyloid‑β aggregation by upregulating astrocytic endogenous αB‑crystallin and HSP‑70 via regulation of the α7AChR, PI3K/Akt/HSF‑1 signaling axis

  • Authors:
    • Zhenkui Ren
    • Zhihui Dong
    • Peng Xie
    • Ju Lv
    • Yumei Hu
    • Zhizhong Guan
    • Chunlin Zhang
    • Wenfeng Yu
  • View Affiliations

  • Published online on: May 5, 2020     https://doi.org/10.3892/mmr.2020.11132
  • Pages: 201-208
  • Copyright: © Ren et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Alzheimer's disease (AD) is a chronic and irreversible neurodegenerative disorder. Abnormal aggregation of the neurotoxic amyloid‑β (Aβ) peptide is an early event in AD. The activation of astrocytic α7 nicotinic acetylcholine receptor (α7 nAChR) can inhibit Aβ aggregation; thus, the molecular mechanism between α7 nAChR activation and Aβ aggregation warrants further investigation. In the present study, Aβ oligomer levels were assessed in astrocytic cell lysates after treatment with PNU282987 (a potent agonist of α7 nAChRs) or co‑treatment with LY294002, a p‑Akt inhibitor. The levels of heat shock factor‑1 (HSF‑1), heat shock protein 70 (HSP‑70), and αB‑crystallin (Cryab) in astrocytes treated with PNU282987 at various time‑points or co‑treated with methyllycaconitine (MLA), a selective α7 nAChR antagonist, as well as co‑incubated with LY294002 were determined by western blotting. HSP‑70 and Cryab levels were determined after HSF‑1 knockdown (KD) in astrocytes. PNU282987 markedly inhibited Aβ aggregation and upregulated HSF‑1, Cryab, and HSP‑70 in primary astrocytes, while the PNU282987‑mediated neuroprotective effect was reversed by pre‑treatment with MLA or LY294002. Moreover, the HSF‑1 KD in astrocytes effectively decreased Cryab, but not HSP‑70 expression. HSF‑1 is necessary for the upregulation of Cryab expression, but not for that of HSP‑70. HSF‑1 and HSP‑70 have a neuroprotective effect. Furthermore, the neuroprotective effect of PNU282987 against Aβ aggregation was mediated by the canonical PI3K/Akt signaling pathway activation.

Introduction

Alzheimer's disease (AD), a chronic but irreversible neurodegenerative disorder with the highest incidence of age-related dementia, is mainly caused by abnormal aggregation of the neurotoxic amyloid-β (Aβ) peptide, a product that forms following proteolysis of the amyloid precursor protein (APP) (1). Pathological characteristics of AD often include neuronal apoptosis, synaptic loss and cognitive impairment, which are considered to be associated with abnormal aggregation of Aβ (2,3). Furthermore, it has been revealed that increased Aβ formation results in high levels of reactive oxygen species and an increased activation of proinflammatory cytokines, associated with neuroinflammation in AD (4). Because abnormal accumulation of Aβ plays a key role in the progression of AD, understanding how to inhibit its aggregation warrants further study.

Astrocytes can provide energy to neurons through the astrocyte-neuron shuttle (5), thereby, affecting their metabolism and synaptic activity, which is known to be important for memory formation (6,7). Notably, Aβ can promote the expression of inflammatory factors and inhibit the activity of Aβ-cleaving serine proteases in astrocytes (8,9), which is associated with AD severity (10).

The α7 nicotinic acetylcholine receptors (nAChRs) are pentameric molecules that belong to the ligand-gated ion channel family involved in regulating the permeability of Ca2+, associated with neuroprotective and anti-inflammatory effects in the AD brain (11). α7 nAChRs are abundantly expressed in the central and peripheral nervous system, and spinal cord (12). α7 nAChRs can form a stable complex with Aβ in neuritic plaques and neurons that are associated with the pathogenesis of AD, which results in attenuation of Aβ neurotoxicity (13). The expression of α7 nAChRs is significantly increased in the cerebral cortex and hippocampus of patients with AD, which is closely associated with Aβ deposits (14,15). However, it is not clear whether the physiological functions of α7 nAChRs are related to Aβ aggregation and deposition in astrocytes.

Heat shock proteins (HSPs), a conservative protein family induced by heat shock and heavy metals, among others, are widely expressed in prokaryotes and eukaryotes (3). HSPs, such as HSP-70 and αB-crystallin (Cryab), can effectively suppress the aberrant accumulation of Aβs (16). Moreover, Cryab combines with Aβs, significantly inhibiting their accumulation and neurotoxicity (17,18). Heat shock factor 1 (HSF-1) is activated through multiple steps; it then translocates to the nucleus, where it induces HSPs in response to stress by binding to the heat shock element (HSE). HSF-1 protects neurons from death by regulating the expression of HSPs (19). However, it is still not known whether this protective effect is mediated by α7 nAChRs stimulation.

In the present study, it was reported that PNU282987, a potent agonist of α7 nAChRs, can significantly enhance suppression of Aβ aberrant accumulation via the upregulation of endogenous Cryab and HSP-70 in cultured astrocytes. In fact, it was demonstrated that HSF-1 expression was necessary for the upregulation of Cryab, but not for that of HSP-70. Furthermore, the neuroprotective effect of PNU282987 against Aβ aggregation was mediated via activation of the canonical PI3K/Akt signaling pathway after α7 nAChRs.

Materials and methods

Materials

Fetal bovine serum (FBS) and Dulbecco's modified Eagle's medium (DMEM) were obtained from Gibco; Thermo Fisher Scientific, Inc. Synthetic drugs included Aβ1-42, PNU282987, dimethylsulfoxide (DMSO) and methyllycaconitine (MLA) (Sigma-Aldrich; Merck KGaA). The following antibodies were used in the present study: Mouse polyclonal anti-Aβ, 1–16 (cat. no. 803015; BioLegend, Inc.), rabbit polyclonal anti-HSP-70 (cat. no. AB9920; Sigma-Aldrich; Merck KGaA), anti-Cryab (cat. no. ab13496; Abcam), anti-β-actin (cat. no. sc-47778; Santa Cruz Technology, Inc.), rabbit monoclonal anti-phospho-(cat. no. 7252C) and non-phospho-specific Akt (cat. no. 2920) and anti-HSF-1 (cat. no. 12972; Cell Signaling Technology, Inc.), and peroxidase-conjugated secondary antibodies (cat. no. sc-2357; Santa Cruz Technology, Inc.). Phosphate-buffered saline (PBS), trypsin and penicillin-streptomycin were obtained from HyClone; GE Healthcare Life Sciences, Lipofectamine® 2000 was purchased from Invitrogen; Thermo Fisher Scientific, Inc., and the LY294002 inhibitor was obtained from Cell Signaling Technology, Inc. Culture flasks were purchased from Corning, Inc. The enhanced chemiluminescence system (ECL) was obtained from Amersham; GE Healthcare, and polyvinylidene fluoride (PVDF) membranes were purchased from EMD Millipore. Sprague-Dawley (SD) pregnant rats were purchased from the Animal Experiment Center of Guizhou Medical University (Guizhou, China).

Primary astrocyte cultures

All animal experiments in the present study were approved by the Animal Care Committee of Guizhou Medical University and were implemented in strict accordance with the relevant guidelines (no. 1503008). A total of 20 SD rats (8 weeks old, weight 240–280 g) were randomly divided into two groups, 10 (5 males and 5 females) in each group. Rats had access to food and water ad libitum and were maintained at 25°C (relative humidity 65%) with a 12-h light/dark cycle. Astrocytes were separated from the cerebral cortex of newborn SD rats according to a previously described method (20). Briefly, the cerebral cortex was separated and cut, a 10X volume of trypsin digestion buffer was added for 15 min at 37°C. Samples were centrifuged at 1,006.2 × g, 25°C for 5 min in complete medium (DMEM with 10% FBS and 1% penicillin-streptomycin). Cells were then resuspended with complete medium and transferred to 25-cm2 culture flasks for ~9 days. Cells were then purified by centrifugal methods (83.8 × g, 37°C, 6 h). Purified cells were sub-cultured in 25 cm2 culture flasks (37°C, 5% CO2). In this way, ~95% of the astrocytes could be purified, and they were identified using immunostaining with rabbit anti-GFAP antibody (1:200) and anti-rabbit IgG Cy3 (1:200). Rats were placed into enclosed flow cages for 5 min, and then 100% CO2 was infused at a 30% volume per minute displacement, in order to euthanize the animals. Subsequently, rats were rendered unconscious by CO2 inhalation (confirmed by slow deep breathing and absence of response to toe-pinch).

Preparation of Aβ oligomers

Aβ oligomers were prepared according to a previous method (21). Briefly, Aβ peptides were dissolved in hexafluoroisopropanol (HFIP) at a concentration of 1 mM. After evaporating HFIP, the peptide membrane was stored at −80°C. Aβ peptides were dissolved with DMSO and diluted to 1 µm with DMEM under sterile conditions. The quality of Aβ oligomers was identified by 12 alkyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE), as previously described (22).

Treatment of astrocytes

To evaluate the role of PNU282987 in HSP regulation, cultured astrocytes were treated with PNU282987 (5 µM) at different time-points (6, 12, 18, 24 h) in an incubator at 37°C. The regulatory effect of PNU282987 was partly inhibited by MLA (100 nM,), an α7 nAChR antagonist, at 37°C for 2 h. The concentration of MLA used was based on previous studies (23,24). To explore whether the regulatory effect of PNU282987 was mediated by the PI3K signaling pathway, astrocytes were pretreated with LY294002 (10 µM) for 2 h at 37°C before PNU282987 treatment. The concentration of LY294002 was based on previous studies (19,20). The medium was replaced by DMEM containing PNU282987 (5 µM), and the astrocytic cultures were incubated for 18 h at 37°C. After removing the medium, astrocytes were further exposed to DMEM containing Aβ oligomers (1 µM) for 24 h at 37°C.

Western blot analysis

Cells were washed three times with pre-chilled PBS in a six-well plate; then, cells were incubated on ice for 3 h with 60 µl/well of RIPA buffer containing protease and phosphatase inhibitors (cat. no. 9806S; Cell Signaling Technology., Inc.). The supernatant was collected after centrifugation at 4°C for 15 min at 4,024.8 × g, and the concentration of the supernatant protein was assessed with a BCA assay (Thermo Fisher Scientific, Inc.), according to the manufacturer's instructions. Proteins were loaded (10 µg/lane) and resolved in 12% SDS-PAGE gels. Proteins were transferred to nitrocellulose membranes, and then blocked with 5% non-fat milk at room temperature (RT) for 2 h. Membranes were washed three times with TBS buffer with 0.1% Tween-20 (TBST); subsequently, the proteins were incubated with various primary antibodies overnight at 4°C. Antibodies included: Anti-phospho-Akt (Ser 473; 1:1,000); anti-HSF-1 (1:1,000); anti-Cryab (1:1,000); anti-β-Amyloid 1–16 (1:1,000); anti-HSP-70 (1:1,000); or anti-non-phospho-Akt (1:1,000). Following the overnight incubation, the membranes were rinsed three times with TBST (5 min each) and proteins were incubated with the appropriate secondary antibody for 1 h at RT. GAPDH (1:1,000; cat. no. MA5-157381; Thermo Fisher Scientific, Inc.) or β-actin (1:1,000; cat. no. sc-47778; Santa Cruz Biotechnology, Inc.) were used as internal controls. Protein bands were detected with an ECL kit, according to the manufacturer's instructions. Quantification was performed using ImageJ software (version 1.46; National Institutes of Health).

Transfections

Cell transfections were carried out according to previously described protocols (25,26). Briefly, cells were cultured in six-well plates for 24 h at a density of 5×105 cells and allowed to adhere for 24 h. The target sequence of the rat HSF-1 shRNA plasmid (Qiagen, Inc.) was 5′-TGTCAACAAGCTCATCCAATT-3′. Scrambled shRNA (Qiagen, Inc.) or HSF-1-specific shRNA (2 µg), plus reagents (2 µl), and Lipofectamine 2000 (6 µl) were mixed with Opti-MEM medium (500 µl; cat. no. 31985062; Thermo Fisher Scientific, Inc.) for 20 min. Cells were incubated at 37°C for 48 h in this medium containing shRNA. Subsequently, the cells were washed 3 times with PBS and fresh medium was added. The transfected cells were seeded in the selection medium (containing Hygromycin 340 µg/ml) for 12 days. Well-separated antibiotic resistant clone cells were identified and expanded for subsequent experiments. The identified cells were treated with PNU (5 µM) for 18 h at 37°C. The time interval between transfection and subsequent experimentation was 15 days.

Statistical analysis

To compare differences among groups, one-way ANOVA was used with Tukey's post hoc test for multiple comparisons. Data results were analyzed with SPSS 22 software (IBM Corp.). P<0.05 was considered to indicate a statistically significant difference.

Results

PNU282987 can enhance astrocytes to inhibit Aβ aggregation

Pretreatment with 5 µM of the potent agonist of α7 nAChRs PNU282987 for 18 h followed by treatment with l µM Aβ1-42 for 24 h significantly inhibited Aβ accumulation, indicating that PNU282987 enhanced astrocytes to inhibit Aβ aggregation (Fig. 1). The control was used as the healthy control (without any treatment.), and the Aβ1-42 treatment group was employed as the PNU282987-treated control.

PNU282987 upregulates HSF-1, Cryab and HSP-70 expression in astrocytes

To evaluate the effect of PNU282987 on the HSP response, astrocytes were treated with 5 µM PNU282987 at various time-points (0, 6, 12, 18 and 24 h). This resulted in the upregulation of HSF-1, Cryab and HSP-70 (Fig. 2A and B). The upregulatory effect of PNU282987 was time-dependent and peaked at 18 h. Therefore, this time-point was selected for subsequent experiments. PNU282987-mediated upregulation of endogenous HSF-1, Cryab and HSP-70 was significantly antagonized by the α7 nAChR antagonist MLA (0.1 µM), indicating that α7 nAChR was involved in the upregulatory effect of PNU282987 (Fig. 2C and D). Western blot analysis revealed that the HSF-1 knockdown (KD) effectively decreased Cryab expression at 18 h after PNU282987 treatment. However, HSP-70 expression did not exhibit a significant change in HSF-1-KD cells compared with negative control (NC; scrambled shRNA) cells (Fig. 2E and F).

PNU282987 upregulates HSF-1, Cryab and HSP-70 via the PI3K/Akt signaling pathway

PNU282987 upregulated p-Akt levels at various time-points (0, 5, 10, 20 and 30 min) (Fig. 3A). Moreover, pre-treatment with the PI3K inhibitor LY294002 (10 µM) or the α7 nAChRs inhibitor MLA (0.1 µM) for 2 h, followed by co-incubation with PNU282987 (5 µM), indicated that MLA and LY294002 could significantly inhibit p-Akt upregulation during PNU282987 treatment of astrocytes (Fig. 3B and C). Moreover, pre-treatment with LY294002 for 2 h significantly inhibited the upregulation of HSF-1, Cryab and HSP-70 during PNU282987 treatment of astrocytes (Fig. 3D and E), indicating the involvement of the PI3K pathway in the upregulatory effect of PNU282987 (PNU282987 upregulated the expression of HSF-1, Cryab and HSP-70).

PNU282987 inhibits Aβ aggregation via the PI3K/Akt signaling pathway

Next, it was explored whether the PI3K/Akt pathway was involved in the inhibition of Aβ accumulation during PNU282987-treatment of astrocytes. The results revealed that PNU282987 treatment led to the inhibition of Aβ aggregation in astrocytes; however, pre-treatment with LY294002 for 2 h significantly attenuated this effect (Fig. 4A and B). The proposed pathway by which PNU282987 regulates Aβ accumulation in astrocytes is presented in Fig. 4C.

Discussion

PNU282987 can inhibit Aβ aggregation in astrocytes; however, the specific mechanism underlying this effect remains to be clarified. The present results indicated that the PNU282987-mediated neuroprotection against Aβ accumulation was associated with upregulation of endogenous Cryab and HSP-70 in astrocyte cultures. Moreover, upregulation of astrocytic Cryab depended on HSF-1, and HSF-1 KD effectively decreased Cryab expression. These effects were regulated by α7 nAChRs and the PI3K/Akt signal transduction pathway in astrocyte cultures.

α7 nAChRs are ion channel receptors associated with memory and cognitive function in AD and Parkinson's disease (PD) (27,28). These receptors are widely expressed in the human cerebral cortex, especially in astrocytes and microglia. α7 nAChRs are activated in neurons with simvastatin via the calmodulin-kinase II signaling pathway (29). Activation of α7 nAChRs has been revealed to protect neurons from the neurotoxic anticancer drug oxaliplatin (30). Hence, PNU282987, an α7 nAChR agonist, was employed to explore its neuroprotective effect in Aβ-treated astrocytes. PNU282987 enhanced astrocytes to inhibit Aβ aggregation, a process closely associated to the upregulation of HSP.

Aβ aggregates and deposits are associated with cognitive dysfunction, neuronal death and synaptic impairment in patients with AD (3,31,32). Previous studies have revealed a significant upregulation of α7 nAChR in astrocytes of AD brains that is closely associated with Aβ accumulation (14,15). Moreover, α7 nAChRs have been revealed to interact with Aβ (33). However, the relationship between α7 nAChRs and Aβ aggregation is still not clear in astrocytes. The present results indicated the involvement of α7 nAChRs and the PI3K/Akt signaling pathway in PNU282987-mediated protection against Aβ neurotoxicity, and the upregulation of endogenous Cryab and HSP-70 to inhibit Aβ aggregation in astrocytes. Moreover, HSF-1 KD effectively decreased Cryab expression. Therefore, HSF-1 may specifically activate the expression of Cryab. Cryab can effectively prevent the accumulation and cytotoxicity of Aβs by directly combining with them; in addition HSP-70 can inhibit the production of Aβs by combining directly with APP (17,34). Subsequently, HSF-1 becomes activated in in vitro models of PNU282987 neuroprotective activity, causing the upregulation of Cryab. Furthermore, in the present study, HSF-1 KD was associated with a notable decrease in Cryab in both control and PNU282987-treated astrocytes but did not affect HSP-70 expression. Collectively, the present results indicated that the increase of Cryab and HSP-70 may play a key role in the PNU282987-induced neuroprotection against Aβ aggregation in in vitro models, and the upregulation of Cryab seems to be mediated by HSF-1. The present results indicated that Cryab could directly combine with Aβs, preventing their aggregation and cytotoxicity, which is consistent with past research (17).

Previous studies have suggested an increased activation of the PI3K/Akt pathway during Tau hyperphosphorylation in AD mice (35). However, it is unclear whether the PI3K/Akt pathway is activated as a result of a neuroprotective heat shock response elicited by PNU282987. The present results revealed an increase in Akt phosphorylation in astrocytes during PNU282987 treatment. However, pre-treatment with the PI3K inhibitor LY294002 weakened the upregulatory effect of PNU282987. Moreover, pre-treatment with the PI3K inhibitor for 2 h significantly inhibited the PNU282987-mediated upregulation of endogenous HSF-1, Cryab and HSP-70 in astrocytes. In addition, HSF-1 KD was associated with a marked decrease in Cryab in both the negative control and HSF-1-KD cells, which supports a role of Cryab in regulating the protective effect of HSF-1 during the treatment of cells with PNU282987.

In conclusion, in response to stress or injury induced by Aβ, astrocytes may activate heat shock response for adaptation and cell survival. The present study provided evidence that PNU282987 can significantly enhance astrocytes to inhibit Aβ accumulation by the activation of α7 nAChRs and the PI3K/Akt signaling pathway. It was further verified that upregulation of endogenous HSF-1, Cryab and HSP-70 is one of the heat shock response mechanisms for astrocyte protection or adaption during PNU282987 treatment. Collectively, these results demonstrated the neuroprotective actions of PNU282987 in vitro, which will guide future studies exploring this mechanism in AD animal models.

Acknowledgements

Not applicable.

Funding

The present study was supported by grants from the National Natural Science Foundation of China (Project no. 81360199), a Special Grant of the Central Government Supporting Local Science and Technology Development, the Science and Technology Department of Guizhou Province [Guizhou specific grant (2019) 4008], the Science and Technology Fund Project of Guizhou Health and Health Commission (grant no. gzwjkj2019-1-039), and the Science and Technology Fund Project of Southwest Guizhou Autonomous Prefecture (2019-1-10).

Availability of data and materials

The datasets used during the present study are available from the corresponding author upon reasonable request.

Authors' contributions

The first two authors contributed equally to this work. WY and ZR designed the study, ZR wrote and revised the manuscript. ZD analyzed the data and revised the manuscript. JL, PX and YH performed the experiments. CZ and ZG helped perform the analysis of the data with constructive discussions. All authors read and approved the final manuscript.

Ethics approval and consent to participate

All animal experiments in the present study were approved by the Animal Care Committee of Guizhou Medical University and were implemented in strict accordance with the relevant guidelines (no. 1503008).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

α7 nAChR

α7 nicotinic acetylcholine receptor

amyloid-β

AD

Alzheimer's disease

APP

amyloid precursor protein

Cryab

αB-crystallin

HSF-1

heat shock factor 1

HSE

heat shock element

HSP

heat shock protein

KD

knock-down

MLA

methyllycaconitine

References

1 

Schilling LP, Pascoal TA, Zimmer ER, Mathotaarachchi S, Shin M, de Mello Rieder CR, Gauthier S, Palmini A and Rosa-Neto P; Alzheimer's Disease Neuroimaging Initiative, : Regional Amyloid-β load and white matter abnormalities contribute to hypometabolism in Alzheimer's dementia. Mol Neurobiol. 56:4916–4924. 2019. View Article : Google Scholar : PubMed/NCBI

2 

Karran E, Mercken M and De Strooper B: The amyloid cascade hypothesis for Alzheimer's disease: An appraisal for the development of therapeutics. Nat Rev Drug Discov. 10:698–712. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Glabe CG and Kayed R: Common structure and toxic function of amyloid oligomers implies a common mechanism of pathogenesis. Neurology. 66 (2 Suppl 1):S74–S78. 2006. View Article : Google Scholar : PubMed/NCBI

4 

Saito T, Hisahara S, Iwahara N, Emoto MC, Yokokawa K, Suzuki H, Manabe T, Matsumura A, Suzuki S, Matsushita T, et al: Early administration of galantamine from preplaque phase suppresses oxidative stress and improves cognitive behavior in APPswe/PS1dE9 mouse model of Alzheimer's disease. Free Radic Biol Med. 145:20–32. 2019. View Article : Google Scholar : PubMed/NCBI

5 

Sidoryk-Wegrzynowicz M, Wegrzynowicz M, Lee E, Bowman AB and Aschner M: Role of astrocytes in brain function and disease. Toxicol Pathol. 39:115–123. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Alberini CM, Cruz E, Descalzi G, Bessieres B and Gao V: Astrocyte glycogen and lactate: New insights into learning and memory mechanisms. Glia. 66:1244–1262. 2018. View Article : Google Scholar : PubMed/NCBI

7 

Steinman MQ, Gao V and Alberini CM: The role of lactate-mediated metabolic coupling between astrocytes and neurons in long-term memory formation. Front Integr Neurosci. 10:102016. View Article : Google Scholar : PubMed/NCBI

8 

Nielsen HM, Veerhuis R, Holmqvist B and Janciauskiene S: Binding and uptake of A beta1-42 by primary human astrocytes in vitro. Glia. 57:978–988. 2009. View Article : Google Scholar : PubMed/NCBI

9 

Pihlaja R, Koistinaho J, Malm T, Sikkilä H, Vainio S and Koistinaho M: Transplanted astrocytes internalize deposited beta-amyloid peptides in a transgenic mouse model of Alzheimer's disease. Glia. 56:154–163. 2008. View Article : Google Scholar : PubMed/NCBI

10 

Jabbari Azad F, Talaei A, Rafatpanah H, Yousefzadeh H, Jafari R, Talaei A and Farid Hosseini R: Association between Cytokine production and disease severity in Alzheimer's disease. Iran J Allergy Asthma Immunol. 13:433–439. 2014.PubMed/NCBI

11 

Hoskin JL, Al-Hasan Y and Sabbagh MN: Nicotinic acetylcholine receptor agonists for the treatment of Alzheimer's dementia: An update. Nicotine Tob Res. 21:370–376. 2019. View Article : Google Scholar : PubMed/NCBI

12 

Broide RS, Winzer-Serhan UH, Chen Y and Leslie FM: Distribution of α7 nicotinic acetylcholine receptor subunit mRNA in the developing mouse. Front Neuroanat. 13:762019. View Article : Google Scholar : PubMed/NCBI

13 

Parri HR, Hernandez CM and Dineley KT: Research update: Alpha7 nicotinic acetylcholine receptor mechanisms in Alzheimer's disease. Biochem Pharmacol. 82:931–942. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Yu WF, Guan ZZ, Bogdanovic N and Nordberg A: High selective expression of alpha7 nicotinic receptors on astrocytes in the brains of patients with sporadic Alzheimer's disease and patients carrying Swedish APP 670/671 mutation: A possible association with neuritic plaques. Exp Neurol. 192:215–225. 2005. View Article : Google Scholar : PubMed/NCBI

15 

Yu W, Mechawar N, Krantic S, Chabot JG and Quirion R: Upregulation of astrocytic α7 nicotinic receptors in Alzheimer's disease brain-possible relevant to amyloid pathology. Mol Neurodegener. 7 (Suppl 1):O72012. View Article : Google Scholar

16 

Shammas SL, Waudby CA, Wang S, Buell AK, Knowles TP, Ecroyd H, Welland ME, Carver JA, Dobson CM and Meehan S: Binding of the molecular chaperone αB-crystallin to Aβ amyloid fibrils inhibits fibril elongation. Biophys J. 101:1681–1689. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Wilhelmus MM, Boelens WC, Otte-Höller I, Kamps B, de Waal RM and Verbeek MM: Small heat shock proteins inhibit amyloid-beta protein aggregation and cerebrovascular amyloid-beta protein toxicity. Brain Res. 1089:67–78. 2006. View Article : Google Scholar : PubMed/NCBI

18 

Raman B, Ban T, Sakai M, Pasta SY, Ramakrishna T, Naiki H, Goto Y and Rao ChM: AlphaB-crystallin, a small heat-shock protein, prevents the amyloid fibril growth of an amyloid beta-peptide and beta2-microglobulin. Biochem J. 392:573–581. 2005. View Article : Google Scholar : PubMed/NCBI

19 

Volovik Y, Moll L, Marques FC, Maman M, Bejerano-Sagie M and Cohen E: Differential regulation of the heat shock factor 1 and DAF-16 by neuronal nhl-1 in the nematode C. elegans. Cell Rep. 9:2192–2205. 2014. View Article : Google Scholar : PubMed/NCBI

20 

McCarthy KD and de Vellis J: Preparation of separate astroglial and oligodendroglial cell cultures from rat cerebral tissue. J Cell Biol. 85:890–902. 1980. View Article : Google Scholar : PubMed/NCBI

21 

Klein WL: Abeta toxicity in Alzheimer's disease: Globular oligomers (ADDLs) as new vaccine and drug targets. Neurochem Int. 41:345–352. 2002. View Article : Google Scholar : PubMed/NCBI

22 

Rönicke R, Mikhaylova M, Rönicke S, Meinhardt J, Schröder UH, Fändrich M, Reiser G, Kreutz MR and Reymann KG: Early neuronal dysfunction by amyloid β oligomers depends on activation of NR2B-containing NMDA receptors. Neurobiol Aging. 32:2219–2228. 2011. View Article : Google Scholar : PubMed/NCBI

23 

Kihara T, Shimohama S, Urushitani M, Sawada H, Kimura J, Kume T, Maeda T and Akaike A: Stimulation of alpha4beta2 nicotinic acetylcholine receptors inhibits beta-amyloid toxicity. Brain Res. 792:331–334. 1998. View Article : Google Scholar : PubMed/NCBI

24 

Steiner RC, Heath CJ and Picciotto MR: Nicotine-induced phosphorylation of ERK in mouse primary cortical neurons: Evidence for involvement of glutamatergic signaling and CaMKII. J Neurochem. 103:666–678. 2007. View Article : Google Scholar : PubMed/NCBI

25 

Valle-Casuso JC, Gonzalez-Sanchez A, Medina JM and Tabernero A: HIF-1 and c-Src mediate increased glucose uptake induced by endothelin-1 and connexin43 in astrocytes. PLoS One. 7:e324482012. View Article : Google Scholar : PubMed/NCBI

26 

Herrero-González S, Valle-Casuso JC, Sánchez-Alvarez R, Giaume C, Medina JM and Tabernero A: Connexin43 is involved in the effect of endothelin-1 on astrocyte proliferation and glucose uptake. Glia. 57:222–233. 2009. View Article : Google Scholar : PubMed/NCBI

27 

Yang T, Xiao T, Sun Q and Wang K: The current agonists and positive allosteric modulators of α7 nAChR for CNS indications in clinical trials. Acta Pharm Sin B. 7:611–622. 2017. View Article : Google Scholar : PubMed/NCBI

28 

Kalkman HO and Feuerbach D: Modulatory effects of α7 nAChRs on the immune system and its relevance for CNS disorders. Cell Mol Life Sci. 73:2511–2530. 2016. View Article : Google Scholar : PubMed/NCBI

29 

Chen T, Wang Y, Zhang T, Zhang B and Chen L, Zhao L and Chen L: Simvastatin Enhances activity and trafficking of α7 nicotinic acetylcholine receptor in hippocampal neurons through PKC and CaMKII signaling pathways. Front Pharmacol. 9:3622018. View Article : Google Scholar : PubMed/NCBI

30 

Mannelli LDC, Tenci B, Zanardelli M, Failli P and Ghelardini C: α7 nicotinic receptor promotes the neuroprotective functions of astrocytes against oxaliplatin neurotoxicity. Neural Plasticity. 2015:1–10. 2015. View Article : Google Scholar

31 

Hardy JA and Higgins GA: Alzheimer's disease: The amyloid cascade hypothesis. Science. 256:184–185. 1992. View Article : Google Scholar : PubMed/NCBI

32 

Gendron R, Plamondon P and Grenier D: Binding of pro-matrix metalloproteinase 9 by Fusobacterium nucleatum subsp. nucleatum as a mechanism to promote the invasion of a reconstituted basement membrane. Infect Immun. 72:6160–6163. 2004. View Article : Google Scholar : PubMed/NCBI

33 

Gustavo D, Glogowski CM, Eliezer M and Heinemann SF: Deletion of the α7 nicotinic acetylcholine receptor gene improves cognitive deficits and synaptic pathology in a mouse model of Alzheimer's disease. J Neurosci. 29:8805–8815. 2009. View Article : Google Scholar : PubMed/NCBI

34 

Maiti P, Manna J, Veleri S and Frautschy S: Molecular chaperone dysfunction in neurodegenerative diseases and effects of curcumin. Biomed Res Int. 2014:4950912014. View Article : Google Scholar : PubMed/NCBI

35 

Qi Y, Dou DQ, Jiang H, Zhang BB, Qin WY, Kang K, Zhang N and Jia D: Arctigenin attenuates learning and memory deficits through PI3k/Akt/GSK-3β pathway reducing tau hyperphosphorylation in Aβ-induced AD mice. Planta Med. 83:51–56. 2017.PubMed/NCBI

Related Articles

Journal Cover

July-2020
Volume 22 Issue 1

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ren Z, Dong Z, Xie P, Lv J, Hu Y, Guan Z, Zhang C and Yu W: PNU282987 inhibits amyloid‑β aggregation by upregulating astrocytic endogenous αB‑crystallin and HSP‑70 via regulation of the α7AChR, PI3K/Akt/HSF‑1 signaling axis. Mol Med Rep 22: 201-208, 2020
APA
Ren, Z., Dong, Z., Xie, P., Lv, J., Hu, Y., Guan, Z. ... Yu, W. (2020). PNU282987 inhibits amyloid‑β aggregation by upregulating astrocytic endogenous αB‑crystallin and HSP‑70 via regulation of the α7AChR, PI3K/Akt/HSF‑1 signaling axis. Molecular Medicine Reports, 22, 201-208. https://doi.org/10.3892/mmr.2020.11132
MLA
Ren, Z., Dong, Z., Xie, P., Lv, J., Hu, Y., Guan, Z., Zhang, C., Yu, W."PNU282987 inhibits amyloid‑β aggregation by upregulating astrocytic endogenous αB‑crystallin and HSP‑70 via regulation of the α7AChR, PI3K/Akt/HSF‑1 signaling axis". Molecular Medicine Reports 22.1 (2020): 201-208.
Chicago
Ren, Z., Dong, Z., Xie, P., Lv, J., Hu, Y., Guan, Z., Zhang, C., Yu, W."PNU282987 inhibits amyloid‑β aggregation by upregulating astrocytic endogenous αB‑crystallin and HSP‑70 via regulation of the α7AChR, PI3K/Akt/HSF‑1 signaling axis". Molecular Medicine Reports 22, no. 1 (2020): 201-208. https://doi.org/10.3892/mmr.2020.11132