Open Access

Ginsenoside Rg1 reduces β‑amyloid levels by inhibiting CDΚ5‑induced PPARγ phosphorylation in a neuron model of Alzheimer's disease

  • Authors:
    • Qiankun Quan
    • Xi Li
    • Jianjun Feng
    • Jixing Hou
    • Ming Li
    • Bingwei Zhang
  • View Affiliations

  • Published online on: August 7, 2020     https://doi.org/10.3892/mmr.2020.11424
  • Pages: 3277-3288
  • Copyright: © Quan 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

The accumulation of β‑amyloid peptides (Aβ) in the brain is a hallmark of Alzheimer's disease (AD). Studies have indicated that ginsenoside Rg1, a primary component of ginseng (Panax ginseng), reduces brain Aβ levels in an AD model through peroxisome proliferator‑activated receptor γ (PPARγ), thereby regulating the expression of insulin‑degrading enzyme (Ide) and β‑amyloid cleavage enzyme 1 (Bace1), which are PPARγ target genes. However, the effects of ginsenoside Rg1 on PPARγ remain unclear. Since cyclin‑dependent kinase 5 (CDK5) mediates PPARγ phosphorylation in adipose tissue, this study aimed to investigate whether ginsenoside Rg1 regulates PPARγ target genes and reduces Aβ levels by inhibiting PPARγ phosphorylation through the CDK5 pathway. In the present study, a model of AD was established by treating primary cultured rat hippocampal neurons with Aβ1‑42. The cells were pretreatment with ginsenoside Rg1 and roscovitine, a CDK5‑inhibitor, prior to the treatment with Aβ1‑42. Neuronal apoptosis was detected using TUNEL staining. PPARγ phosphorylation and protein expression levels of PPARγ, CDK5, IDE, BACE1, amyloid precursor protein (APP) and Aβ1‑42 were measured by western blotting. The mRNA expression levels of PPARγ, CDK5, IDE, BACE1 and APP were assessed using reverse transcription‑quantitative PCR. The results of the present study demonstrated that in an AD model induced by Aβ1‑42, ginsenoside Rg1 significantly decreased CDK5 expression, inhibited PPARγ phosphorylation at serine 273, elevated IDE expression, downregulated BACE1 and APP expression, decreased Aβ1‑42 levels and attenuated neuronal apoptosis. The CDK5 inhibitor, roscovitine, demonstrated similar effects. These results suggest that ginsenoside Rg1 has neuroprotective properties and has potential for use in the treatment of AD.

Introduction

Ginseng (Panax ginseng), which is documented in Shennong's Classic of Materia Medica as having intellect-enhancing effects, has been widely used in China for millennia (1). Ginsenoside Rg1, a primary ginseng components, has multiple neuroprotective effects against Alzheimer's disease (AD), including the improvement of memory impairment (2), inhibition of neuronal apoptosis (3), amelioration of oxidative stress (4) and attenuation of mitochondrial dysfunction (5). The accumulation of β-amyloid peptides (Aβ) is the most prominent pathological feature of AD and significantly influences the pathogenesis of the disease (6). The aggregation and accumulation of Aβ in the brain may lead to neuroinflammatory responses (7), neurofibrillary tangle formation (8), synaptic loss (9), oxidative stress (10), neuronal apoptosis (11), cholinergic dysfunction (12) and tau phosphorylation (13). It has been reported that ginsenoside Rg1 has Aβ-scavenging effects, for example, ginsenoside Rg1 inhibits the transcription and translation of β-amyloid cleavage enzyme 1 (Bace1), a target gene of peroxisome proliferator-activated receptor γ (PPARγ), by enhancing the binding of PPARγ to the Bace1 promoter, thereby decreasing BACE1 activity, which ultimately attenuates Aβ production (14). Additionally, ginsenoside Rg1 upregulates PPARγ expression in the rat hippocampus AD model, thereby increasing the expression of insulin-degrading enzyme (Ide) (another PPARγ target gene) and promoting the clearance of Aβ1-42 from the hippocampus (15). These studies suggest that PPARγ is involved in the reduction of Aβ levels caused by ginsenoside Rg1; however, the mechanisms by which ginsenoside Rg1 affects PPARγ remain unclear.

Cyclin-dependent kinase 5 (CDK5), a cyclin-dependent kinase, is a proline-directed serine/threonine kinase predominantly activated in post-mitotic cells and has various activities including cytoskeletal dynamics, signaling cascades, gene expression, cell survival, neurodevelopment and brain function (1619). Phosphorylation is an important post-translational modification of PPARγ. It has been demonstrated that in the adipose tissue, CDK5 induces PPARγ phosphorylation at serine 273 (Ser273), which is located in the hinge region between the DNA- and the ligand-binding domains in vivo and in vitro (20). Furthermore, our previous study demonstrated that in rat primary hippocampal neurons CDK5 regulates the expression of IDE and BACE1 by mediating the phosphorylation of PPARγ, resulting in decreased Aβ clearance and increased Aβ production (21). The present study aimed to investigate whether ginsenoside Rg1 inhibits the phosphorylation of PPARγ through the downregulation of the CDK5 pathway. The findings of this study will deepen the understanding of the neuroprotective properties of ginsenoside Rg1 and its potential use in the treatment of AD.

Materials and methods

Reagents

Ginsenoside Rg1 (molecular formula: C42H72O14; molecular weight: 801.01; HPLC purity: 98%) was purchased from Baoji Herbest Bio-Tech Co., Ltd. Aβ1-42 and roscovitine were purchased from Sigma Aldrich; Merck KGaA. Rabbit anti-rat IDE (cat. no. ab133561), BACE1 (cat. no. ab10716) and amyloid precursor protein (APP; cat. no. ab15272) polyclonal antibodies were purchased from Abcam. Rabbit anti-rat p-PPARγ-Ser273 polyclonal antibody (cat. no. bs-4888R) was purchased from BIOSS Antibodies. Rabbit anti-rat CDK5 (cat. no. WL01673), PPARγ (cat. no. WL0269) and Aβ1-42 (cat. no. WL01427) polyclonal antibodies, anti-β-actin antibody (cat. no. WL01845), goat anti-rabbit secondary horseradish peroxidase-conjugated antibody (cat. no. WLA023), TUNEL assay kit, total protein extraction kit, bicinchoninic acid (BCA) protein assay kit and enhanced chemiluminescence (ECL) reagent were purchased from Wanleibio Co., Ltd. Dulbecco's modified Eagle's medium (DMEM) was purchased from Gibco; Thermo Fisher Scientific, Inc. Fetal bovine serum (FBS) was purchased from Biological Industries. Trypsin and 4′,6-diamidino-2-phenylindole (DAPI) were purchased from Beyotime Institute of Biotechnology. TRIzol® and 2X Power Taq PCR MasterMix were purchased from BioTeke Corporation. SYBR Green master mix was purchased from Beijing Solarbio Science & Technology Co., Ltd.

Isolation and culture of rat hippocampal neurons

Rat hippocampal neurons were isolated and cultured using the methods previously described (22). In total, 150 2-day-old Sprague Dawley rats (weight, 8±2 g; males, 75; females, 75) were used in the study. These rats were obtained from the Experimental Animal Center of Xi'an Jiaotong University Health Science Center [License no. SCXK (Shaan) 2018-001], which were housed in a specific-pathogen free facility maintained at 23°C with a 12-h light-dark cycle and 24-h atmosphere purification, and were allowed free access to breastmilk from their mother. In brief, brain tissues were isolated from these rats under aseptic conditions. The hippocampal tissues were then dissected on an ultra-clean bench, cut into pieces, and digested by trypsin. Subsequently, DMEM containing 10% FBS and penicillin/streptomycin was added for trypsin neutralization. Cell suspension was repeatedly digested five-six times after centrifugation (168 × g, 7 min, 25°C). After filtration and centrifugation, the hippocampal neurons were placed in a poly-lysine-coated 6-well plate at a density of 5×105 cells/ml and cultured for 8 h at 37°C with 5% CO2 and saturated humidity. Subsequently, the culture medium was changed to neurobasal medium containing 2% B27, 0.5 mM glutamine, 100 U/ml penicillin, and 100 U/ml streptomycin and the cells were cultured for 48 h. Cytarabine was then added to the medium (final concentration, 10 µM) to inhibit the growth of glial cells. The media were changed once every 3 days until maturation and network formation of the hippocampal neurons in ~15 days. All experimental procedures in this study were approved by the Ethics Committee of The Second Affiliated Hospital of Xi'an Jiaotong University (Shaanxi, China).

Drug treatment

The cultured neurons were divided into the following three groups: Control, model and ginsenoside Rg1 (Rg1) groups. The control group was used as the vehicle treated group, in which no drugs were added to the culture medium; in the model group, cultured neurons were treated with 8 µM Aβ1-42 (23) for 24 h at 37°C; in the Rg1 group, the cultured neurons were exposed to 60 µM ginsenoside Rg1 (24) for 1 h at 37°C and then to 8 µM Aβ1-42 for 24 h at 37°C. To confirm whether ginsenoside Rg1 regulates PPARγ phosphorylation by acting on CDK5, the effects of ginsenoside Rg1 on cultured neurons that were treated with Aβ1-42 after CDK5 expression was inhibited using the CDK5 inhibitor roscovitine were investigated. Neurons were divided into the three following groups: Model, roscovitine and roscovitine+Rg1 groups. In the model group, the cultured neurons were treated with 8 µM Aβ1-42 for 24 h at 37°C; in the roscovitine group, the cultured neurons were first exposed to 25 µM roscovitine (25) for 1 h at 37°C and then to 8 µM Aβ1-42 for 24 h at 37°C; in the roscovitine+Rg1 group, cultured neurons were first treated with 25 µM roscovitine for 0.5 h at 37°C followed by 60 µM ginsenoside Rg1 for 1 h at 37°C and subsequently 8 µM Aβ1-42 for 24 h at 37°C.

TUNEL staining

TUNEL staining was performed using an assay kit according to the manufacturer's instructions. After the slides were fixed with 4% paraformaldehyde at 25°C for 10 min, the cells were permeabilized with 50 µl of 0.1% Triton X-100 for 15 min at 25°C. After being washed with phosphate-buffered saline (PBS), the cells were incubated with the TUNEL reaction mixture (formulated by mixing the enzyme and label solutions at a ratio of 1:9) in a wet chamber in the dark at 37°C for 60 min. Subsequently, the cells were washed with PBS and counterstained with DAPI in the dark at 25°C for 5 min. After washing with PBS again, the slides were mounted using mounting medium with anti-fluorescent quenchers. The neurons were counted in a blind manner by two pathologists under a BX 53 fluorescence microscope (Olympus Corporation) at ×400 magnification.

The average number of neurons from four random fields of view was used as the final result for each pathologist. The results from the two pathologists were averaged and used to calculate the percentage of TUNEL-positive neurons.

Western blotting

Total proteins of the neurons were extracted using the total protein extraction kit according to the manufacturer's instructions and the protein concentration was measured using the BCA protein assay method. After denaturation at 95°C for 5 min, 20 µl protein sample (including 40 µg protein) was added into each electrophoretic lane, separated in an 8% sodium dodecyl sulfate-polyacrylamide gel electrophoresis and transferred to a nitrocellulose (NC) membrane. The membrane was blocked with 5% non-fat powdered milk at 37°C for 1 h and was then incubated with rabbit anti-rat CDK5 (1:500), p-PPARγ-Ser273 (1:500), PPARγ (1:400), IDE (1:900), BACE1 (1:600), APP (1:800), and Aβ1-42 (1:500) polyclonal antibodies overnight at 4°C. After being washed with Tris-buffered saline buffer with 0.05% Tween 20, the NC membrane was incubated with the secondary horseradish peroxidase-conjugated antibody (goat anti-rabbit, 1:1,000) at 37°C for 45 min. Subsequently, the NC membrane was developed using ECL reagent and the blots were scanned. The optical densities of the target bands were analyzed using Gel-Pro Analyzer software (version 4.0, Media Cybernetics, Inc.).

Reverse transcription-quantitative (RT-q)PCR

Total RNA from the neurons of the three groups was extracted using TRIzol and the concentrations were measured using a UV spectrophotometer. Reverse transcription was performed according to the manufacturer's instructions. In brief, each RNA sample was added into a nuclease-free centrifuge tube in an ice bath based on the concentration of the extracted RNA sample (consistent RNA concentrations during sample loading), followed by the addition of 1 µl of oligo (dT)15, 1 µl of random primers, and a sufficient volume of double-distilled water to reach a total volume of 12.5 µl. The mixture was incubated at 70°C for 5 min and was then rapidly cooled on ice for 2 min. After centrifugation (671 × g, 1 min, 4°C), the reaction mixture was mixed with 2 µl of deoxynucleoside triphosphate (2.5 mM each), 4 µl of 5X buffer, 0.5 µl of RNase inhibitor and 1 µl of Moloney-murine leukemia virus (200 U), and was then sequentially subjected to the following conditions: 25°C for 10 min, 42°C for 50 min and 80°C for 10 min to terminate the reaction. The resultant cDNA was stored at −20°C for further use. Primers (Table I) of rat Cdk5, Pparγ, Ide, Bace1 and App genes were designed using Primer Premier 5.0 (Premier Biosoft International). All primers were synthesized by Sangon Biotech Co., Ltd. Fluorescence-based RT-qPCR was performed using a 2X Power Taq PCR Master Mix kit with a Exicycler™ 96 real-time PCR instrument (Bioneer Corp.). The PCR reaction was performed according to the manufacturer's instructions in a 20 µl mixture including 1 µl of cDNA, 0.5 µl of the forward primer (10 µM), 0.5 µl of the reverse primer (10 µM), 10 µl of the SYBR Green Master Mix and sufficient double-distilled water. Reaction conditions were as follows: Initial denaturation at 94°C for 5 min, followed by denaturation at 94°C for 10 sec, annealing at 60°C for 20 sec and extension at 72°C for 30 sec for 40 cycles. Relative mRNA expression levels were calculated using the 2−ΔΔCq method (26). β-actin was used as the internal control.

Table I.

Primers used for reverse transcription-quantitative PCR.

Table I.

Primers used for reverse transcription-quantitative PCR.

GenePrimer sequence (5′→3′)Primer lengthTemperature, °CPCR product length, bp
Cyclin-dependent kinase 5F: GGACACCGACTGAGGAAC1852.0103
R: TTGGGCACGACATTCAC1752.5
Peroxisome proliferator-activatedF: TACCACGGTTGATTTCTC1847.7155
receptor γR: AATAATAAGGCGGGGACG1855.3
Insulin-degrading enzymeF: TCCCGTGAAGCGACTGT1754.3180
R: GACTTGTCCGTGGTGGG1753.6
β-amyloid cleavage enzyme 1F: TCCGCATCACCATCCTT1754.0123
R: TGACCGCTCCCATAACG1755.1
Amyloid precursor proteinF: ACTCTGTGCCAGCCAATA1851.2158
R: TGAATCATGTCCGAACTCC1953.0
β-actinF: GGAGATTACTGCCCTGGCTCCTAGC2560.1155
R: GGCCGGACTCATCGTACTCCTGCTT2562.0

[i] F, Forward; R, Reverse; bp, base pairs.

Data analysis

All data are expressed as the mean ± SEM. One-way analysis of variance (ANOVA) followed by a Least Significant Difference post hoc test was performed for multiple comparisons. Statistical analyses were conducted using SPSS (version 16.0, SPSS, Inc.). P<0.05 was considered to indicate a statistically significant difference.

Results

Ginsenoside Rg1 inhibits PPARγ phosphorylation in the AD model

In primary cultured rat hippocampal neurons Aβ1-42 treatment significantly enhanced PPARγ phosphorylation at Ser273, increased the p-PPARγ/PPARγ ratio and decreased PPARγ protein and mRNA expression levels compared with those in the control group (P<0.05; Fig. 1). These results suggested the presence of PPARγ phosphorylation in the AD neuron model induced by Aβ1-42. In addition, pretreatment with ginsenoside Rg1 significantly attenuated the aforementioned Aβ1-42-induced effects in these neurons (P<0.05; Fig. 1). Notably, after inhibiting CDK5 expression using roscovitine, the results demonstrated that compared with those in the model group PPARγ phosphorylation was significantly inhibited and PPARγ expression levels were significantly increased in the Aβ1-42-treated neurons (P<0.05; Fig. 2). Additionally, PPARγ phosphorylation and the expression levels of PPARγ protein in the Aβ1-42-treated neurons were not further affected by ginsenoside Rg1 treatment after CDK5 inhibition (P>0.05; Fig. 2), suggesting that CDK5 may be involved in the inhibition of PPARγ phosphorylation induced by ginsenoside Rg1.

Ginsenoside Rg1 decreases CDK5 expression levels in the AD model

The effects of ginsenoside Rg1 on CDK5 expression in the neuron model of AD were investigated. Western blotting and RT-qPCR analyses demonstrated that in the primary cultured rat hippocampal neurons Aβ1-42 treatment significantly increased the protein and mRNA expression levels of CDK5 compared with those in the control group (P<0.05); but, ginsenoside Rg1 pretreatment significantly attenuated the Aβ1-42-induced increase in the protein and mRNA expression levels of CDK5 (P<0.05; Fig. 3).

Ginsenoside Rg1 regulates the expression of PPARγ target genes and decreases intracellular Aβ1-42 levels in the AD model

The effects of ginsenoside Rg1 on the expression of PPARγ target genes and intracellular Aβ1-42 level when CDK5 was inhibited were examined. The results demonstrated that compared with the rat hippocampal neurons in the control group, those in the model group exhibited significantly decreased IDE protein and mRNA expression levels (P<0.05), but significantly increased BACE1, APP and enhanced intracellular Aβ1-42 levels (P<0.05; Fig. 4). Additionally, pretreatment with ginsenoside Rg1 significantly attenuated Aβ1-42-induced effects in these neurons (P<0.05; Fig. 4). Additionally, compared with those in the model group inhibition of CDK5 expression using roscovitine significantly increased the expression levels of IDE and reduced the expression levels of BACE1, APP and Aβ1-42 in rat hippocampal neurons treated with Aβ1-42 (P<0.05; Fig. 5). In addition, no significant differences were observed in the expression levels of IDE, BACE1, APP and Aβ1-42 after ginsenoside Rg1 treatment following CDK5 inhibition with roscovitine (P>0.05; Fig. 5).

Ginsenoside Rg1 attenuates Aβ1-42-induced apoptosis in rat hippocampal neurons

In the present study, TUNEL staining was performed to determine the effects of Aβ1-42 and ginsenoside Rg1 on the apoptosis of rat hippocampal neurons. Compared with that in the control group, Aβ1-42 treatment significantly increased neuronal apoptosis in the model group (P<0.05; Fig. 6), but pretreatment with ginsenoside Rg1 significantly decreased Aβ1-42-induced neuronal apoptosis (P<0.05; Fig. 6). In addition, the neuronal apoptosis rate in the roscovitine group was significantly lower compared with that in the model group (P<0.05; Fig. 7). Additionally, no significant difference was observed in the in the neuronal apoptosis rate between the roscovitine group and the roscovitine+Rg1 group (P>0.05; Fig. 7).

Discussion

This study investigated whether ginsenoside Rg1 inhibits the phosphorylation of PPARγ through the downregulation of the CDK5 pathway, thereby affecting the expression of PPARγ target genes Bace1 and Ide and reducing Aβ levels (Fig. 8). PPARs are ligand-activated nuclear transcription factors, which regulate the transcription of target genes by binding to the peroxisome proliferator response element (PPRE) located on the promoters of these genes (27). Different types of fatty acids (FAs), including docosahexaenoic acid, activate PPARs in adipose tissues (28,29). FA binding to PPARs control the transcription of specific genes including those encoding for various metabolic and cellular processes such as FA β-oxidation and adipogenesis, making them key mediators of lipid homeostasis (29). PPARγ is one of PPARs superfamily members (along with PPARα, PPARβ/δ). A previous study has reported that PPARγ inhibits BACE1 expression by binding to the PPRE on the Bace1 gene promoter in N2a/APP695 cells (30), which is an essential enzyme in the generation of Aβ as it hydrolyzes APP to form Aβ (31). Additionally, PPARγ also bind to the PPRE in the Ide promoter, thereby regulating Ide gene transcription and promoting IDE protein expression (32), which has been demonstrated to degrade Aβ (33). These findings indicate that PPARγ serves a key role in the inhibition of Aβ generation and the promotion of Aβ degradation. Moreover, PPARγ has anti-inflammatory effects through inhibiting the generation of certain proinflammatory cytokines (such as tumor necrosis factor, interleukin-1β and interleukin-6), the production of nitric oxide, and the expression of matrix metalloproteinase 9 and macrophage scavenger receptor 1 (34). Studies have reported that these molecules are closely associated with the onset of AD (3540). Furthermore, oxidative stress usually occurs during early stages of AD and elevates with increased AD severity (41); PPARγ inhibits Aβ-induced oxidative stress (42). Therefore, PPARγ agonists may provide neuroprotective effects against AD.

Several clinical and experimental studies have investigated the role of thiazolidinediones (TZDs), PPARγ agonists, in AD, for example, pioglitazone reduces brain Aβ levels (43) and improves learning and memory in APP/PS1 mice (44), and decreases tau phosphorylation (45) and neuronal apoptosis (46) in an AD cell model; rosiglitazone decreases Aβ1-40 and Aβ1-42 levels, reduces tau phosphorylation, alleviates memory impairment (47), and inhibits Aβ-induced oxidative stress (48), inflammatory responses (49) and mitochondrial dysfunction (50) in both in vitro and in vivo systems. Oral administration of rosiglitazone improves cognitive function in patients with mild-to-moderate AD (51). Troglitazone and ciglitazone prevent Aβ-induced microglial- and monocyte-mediated neurotoxicity and inhibit Aβ-induced increased expression of interleukin 6, tumor necrosis factor α, and cyclooxygenase-2 (52). Recently, a study demonstrated that pioglitazone inhibits the phosphorylation of PPARγ at Ser273 in vitro by inhibiting CDK5 expression, which in turn affected the expression of PPARγ target genes Ide and Bace1, thereby promoting Aβ degradation and reducing Aβ production. This reduced Aβ levels in the brain, thereby exerting neuroprotective effects in an AD model (53). These findings indicate that TZDs have neuroprotective effects against AD. However, as a long-term medication is required for AD, which is a chronic disease with complex pathogenic mechanisms and a long disease course, the side effects of TZDs will substantially limit their application in AD treatment. Studies have demonstrated that rosiglitazone may lead to an increased risk of fractures (54), heart failure (55,56) and increased incidence of stroke (56); pioglitazone may lead to fractures and bladder cancer (54,57); and troglitazone may induce severe hepatotoxicity (58). Therefore, there is a need for TZD replacement with safer and effective drugs presenting mild side effects and PPARγ-agonistic effects in the study of AD therapies.

Ginseng is a natural herbal remedy that has been used in China over several millennia (1). Ginseng has multi-target therapeutic and pharmacological effects in central nervous system, which have been well-demonstrated in the clinical practice (59). Ginsenosides, which mainly include Rb1, Rb2, Rb3, Rc, Rd, Re, Rg1 and Rg2, are the main components of ginseng (60). In particular, ginsenoside Rg1 is one of the most studied and representative ginsenoside components in the field of AD treatment (61). Previous studies have observed that ginsenoside Rg1 may potentially activate PPARγ and facilitate Aβ removal by enhancing the binding of PPARγ to target genes or by upregulating PPARγ expression (14,15). Accordingly, the present study further elucidated the mechanisms by which ginsenoside Rg1 affects PPARγ. The results of the present study suggested that ginsenoside Rg1 inhibits PPARγ phosphorylation by downregulating the expression of CDK5, thereby affecting the expression of PPARγ target genes.

Extracellular and intracellular Aβ serve essential roles in the onset of AD (62). BACE1-mediated APP proteolysis produces a soluble β-fragment of the amyloid precursor protein and a C-terminal fragment containing 99 amino acids (C99). Subsequently, C99 is enzymatically cleaved by γ-secretase to generate APP intracellular domain and Aβ; the latter is released into the extracellular matrix (63,64) (Fig. 8). APP proteolysis also occurs in the endoplasmic reticulum and golgi apparatus (64,65), producing intracellular Aβ. Gouras et al (66) reported that intraneuronal Aβ immunoreactivity appeared to precede the deposition of both neurofibrillary tangles and senile plaques, indicating that intraneuronal accumulation of Aβ is an early event in the onset of AD. The neuron model of AD in this study was established with the addition of exogenous Aβ, so exogenous and neuro-secreted Aβ could not be distinguished, and this study only observed the effect of ginsenoside Rg1 on the intracellular Aβ and did not investigate the changes in extracellular Aβ levels.

This study demonstrated that Rg1 inhibited CDK5 expression. However, the mechanism remains unclear. The results of the present study suggested that ginsenoside Rg1 may affect CDK5 directly, or through other mechanisms; this needs to be investigated. The results of the present also demonstrated that exogenously added Aβ increased CDK5 expression levels, aggravated PPARγ phosphorylation, decreased PPARγ expression levels, and affected the expression levels of the downstream PPARγ target genes Ide and Bace1. The exact mechanisms of these phenomena remain unclear and are subjects for future studies. Previous studies have observed a negative correlation between IDE expression and Aβ levels in AD brains (67), and a decrease in IDE expression levels were also observed in an AD rat model established through the injection of Aβ1-42 in the hippocampus (15). These findings are consistent with those of the present study, which demonstrated decreased IDE expression levels in Aβ-treated primary cultured hippocampal neurons. After treatment with ginsenoside Rg1, IDE expression levels increased and Aβ levels decreased. Several possible reasons may account for these findings: i) The consumption of IDE through its effects on degradation of Aβ exceeds the compensatory generation of IDE; ii) decreased IDE expression may consequently promote Aβ generation; and iii) mutual interactions may exist between IDE and Aβ, i.e., generated Aβ affects IDE expression, whereas decreased IDE expression promotes an increase in Aβ levels. Accordingly, further studies are required to verify these explanations. This study demonstrated that CDK5 mediates the ginsenoside Rg1 reduction of Aβ levels by phosphorylating PPARγ. In addition to CDK5, other CDKs such as CDK7 and CDK9 also take part in PPARγ phosphorylation. A previous study has reported that CDK7 phosphorylates PPARγ at Ser112 to inhibit the activity of PPARγ (68). However, CDK9 increases PPARγ activity after phosphorylating PPARγ at Ser112 (69). Thus, further investigation is necessary into whether the phosphorylation of PPARγ by CDK7 and CDK9 is involved in the ginsenoside Rg1 reduction of Aβ levels. Additionally, the results of the present study demonstrated that the protein and mRNA expression levels of PPARγ in the Rg1 group were higher compared with those in the control group. It was speculated this is due to the high concentrations of ginsenoside Rg1 used in the study, which were able to stimulate a significant response compared with the control.

There are several limitations to this study. In the study, no experiments such as chromatin immunoprecipitation RT-qPCR, were performed to confirm whether CDK5 directly regulates PPARγ. However, our previous study reported that CDK5 regulates PPARγ (21), and performed co-immunoprecipitation experiments to confirm that Aβ promotes the binding of CDK5 to PPARγ (53). In addition, our previous study demonstrated that the PPARγ agonist pioglitazone inhibits PPARγ phosphorylation by inhibiting CDK5 expression, thereby promoting Aβ degradation and reducing Aβ production (53). Therefore, no PPARγ agonist was used for comparison with ginsenoside Rg1 in this study. Since a CDK5 inhibitor was used in the study, no in vivo studies were conducted and only in vitro experiments were performed; therefore, the effects of ginsenoside Rg1 in vivo are unknown. Furthermore, since there are no available drugs that target CDK5, no positive-control drug was used in the study. A set of CDK5 overexpressing cells would allow for testing of the results of this study; unfortunately, CDK5 overexpression experiments were not be performed due to limitations on time and funding.

In conclusion, the results of the present study suggested that ginsenoside Rg1 inhibits PPARγ phosphorylation possibly through the downregulation of CDK5 expression, thereby affecting the expression of PPARγ target genes (Ide and Bace1) and decreasing Aβ levels through the promotion of Aβ degradation and reduction of Aβ synthesis, which ultimately provides neuroprotective effects against AD.

Acknowledgements

Not applicable.

Funding

This study was supported by the Chinese Postdoctoral Science Foundation (grant no. 2017M623191), the Natural Science Foundation of Shaanxi Province (grant no. 2017JQ8039) and the Foundation of The Second Affiliated Hospital of the Xi'an Jiaotong University [grant no. YJ(ZD)201517].

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

QQ, XL and JF designed the study and wrote the manuscript. QQ, JH, ML and BZ performed the experiments. QQ and JH collected and analyzed the data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

All experimental procedures in the present study were approved by the Ethics Committee of The Second Affiliated Hospital of Xi'an Jiaotong University.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Chen CF, Chiou WF and Zhang JT: Comparison of the pharmacological effects of Panax ginseng and Panax quinquefolium. Acta Pharmacol Sin. 29:3277–1108. 2008. View Article : Google Scholar

2 

Nie L, Xia J, Li H, Zhang Z, Yang Y, Huang X, He Z, Liu J and Yang X: Ginsenoside Rg1 ameliorates behavioral abnormalities and modulates the hippocampal proteomic change in triple transgenic mice of Alzheimer's disease. Oxid Med Cell Longev. 2017:64735062017. View Article : Google Scholar : PubMed/NCBI

3 

Mu JS, Lin H, Ye JX, Lin M and Cui XP: Rg1 exhibits neuroprotective effects by inhibiting the endoplasmic reticulum stress-mediated c-Jun N-terminal protein kinase apoptotic pathway in a rat model of Alzheimer's disease. Mol Med Rep. 12:3862–3868. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Liu QA, Kou JP and Yu BY: Ginsenoside Rg1 protects against hydrogen peroxide-induced cell death in PC12 cells via inhibiting NF-κB activation. Neurochem Int. 58:119–125. 2011. View Article : Google Scholar : PubMed/NCBI

5 

Huang T, Fang F, Chen L, Zhu Y, Zhang J, Chen X and Yan SS: Ginsenoside Rg1 attenuates oligomeric Aβ(1–42)-induced mitochondrial dysfunction. Curr Alzheimer Res. 9:388–395. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Tanzi RE, Moir RD and Wagner SL: Clearance of Alzheimer's Abeta peptide: The many roads to perdition. Neuron. 43:605–608. 2004. View Article : Google Scholar : PubMed/NCBI

7 

Álvarez-Arellano L, Pedraza-Escalona M, Blanco-Ayala T, Camacho-Concha N, Cortés-Mendoza J, Pérez-Martínez L and Pedraza-Alva G: Autophagy impairment by caspase-1-dependent inflammation mediates memory loss in response to β-amyloid peptide accumulation. J Neurosci Res. 96:234–246. 2018. View Article : Google Scholar : PubMed/NCBI

8 

Seino Y, Kawarabayashi T, Wakasaya Y, Watanabe M, Takamura A, Yamamoto-Watanabe Y, Kurata T, Abe K, Ikeda M, Westaway D, et al: Amyloid β accelerates phosphorylation of tau and neurofibrillary tangle formation in an amyloid precursor protein and tau double-transgenic mouse model. J Neurosci Res. 88:3547–3554. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Vargas LM, Leal N, Estrada LD, González A, Serrano F, Araya K, Gysling K, Inestrosa NC, Pasquale EB and Alvarez AR: EphA4 activation of c-Abl mediates synaptic loss and LTP blockade caused by amyloid-β oligomers. PLoS One. 9:e923092014. View Article : Google Scholar : PubMed/NCBI

10 

Ibi D, Tsuchihashi A, Nomura T and Hiramatsu M: Involvement of GAT2/BGT-1 in the preventive effects of betaine on cognitive impairment and brain oxidative stress in amyloid β peptide-injected mice. Eur J Pharmacol. 842:57–63. 2019. View Article : Google Scholar : PubMed/NCBI

11 

Hooshmandi E, Ghasemi R, Iloun P and Moosavi M: The neuroprotective effect of agmatine against amyloid β-induced apoptosis in primary cultured hippocampal cells involving ERK, Akt/GSK-3β, and TNF-α. Mol Biol Rep. 46:489–496. 2019. View Article : Google Scholar : PubMed/NCBI

12 

Schirinzi T, Di Lorenzo F, Sancesario GM, Di Lazzaro G, Ponzo V, Pisani A, Mercuri NB, Koch G and Martorana A: Amyloid-mediated cholinergic dysfunction in motor impairment related to Alzheimer's disease. J Alzheimers Dis. 64:525–532. 2018. View Article : Google Scholar : PubMed/NCBI

13 

Ali T, Yoon GH, Shah SA, Lee HY and Kim MO: Osmotin attenuates amyloid beta-induced memory impairment, tau phosphorylation and neurodegeneration in the mouse hippocampus. Sci Rep. 5:117082015. View Article : Google Scholar : PubMed/NCBI

14 

Chen LM, Lin ZY, Zhu YG, Lin N, Zhang J, Pan XD and Chen XC: Ginsenoside Rg1 attenuates β-amyloid generation via suppressing PPARγ-regulated BACE1 activity in N2a-APP695 cells. Eur J Pharmacol. 675:15–21. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Quan Q, Wang J, Li X and Wang Y: Ginsenoside Rg1 decreases Aβ1-42 level by upregulating PPARγ and IDE expression in the hippocampus of a rat model of Alzheimer's disease. PLoS One. 8:e591552013. View Article : Google Scholar : PubMed/NCBI

16 

Liu C, Zhai X, Zhao B, Wang Y and Xu Z: Cyclin I-like (CCNI2) is a Cyclin-dependent kinase 5 (CDK5) activator and is involved in cell cycle regulation. Sci Rep. 7:409792017. View Article : Google Scholar : PubMed/NCBI

17 

Su SC and Tsai LH: Cyclin-dependent kinases in brain development and disease. Annu Rev Cell Dev Biol. 27:465–491. 2011. View Article : Google Scholar : PubMed/NCBI

18 

Lopes JP and Agostinho P: Cdk5: Multitasking between physiological and pathological conditions. Prog Neurobiol. 94:49–63. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Wilkaniec A, Czapski GA and Adamczyk A: Cdk5 at crossroads of protein oligomerization in neurodegenerative diseases: Facts and hypotheses. J Neurochem. 136:222–233. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Choi JH, Banks AS, Estall JL, Kajimura S, Boström P, Laznik D, Ruas JL, Chalmers MJ, Kamenecka TM, Blüher M, et al: Anti-diabetic drugs inhibit obesity-linked phosphorylation of PPARγ by Cdk5. Nature. 466:451–456. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Quan Q, Qian Y, Li X and Li M: CDK5 participates in amyloid-β production by regulating PPARγ phosphorylation in primary rat hippocampal neurons. J Alzheimers Dis. 71:443–460. 2019. View Article : Google Scholar : PubMed/NCBI

22 

Vadukul DM, Gbajumo O, Marshall KE and Serpell LC: Amyloidogenicity and toxicity of the reverse and scrambled variants of amyloid-β 1-42. FEBS Lett. 591:822–830. 2017. View Article : Google Scholar : PubMed/NCBI

23 

Yang EJ, Ahn S, Ryu J, Choi MS, Choi S, Chong YH, Hyun JW, Chang MJ and Kim HS: Phloroglucinol attenuates the cognitive deficits of the 5XFAD mouse model of Alzheimer's sisease. PLoS One. 10:e01356862015. View Article : Google Scholar : PubMed/NCBI

24 

Li Y, Guan Y, Wang Y, Yu CL, Zhai FG and Guan LX: Neuroprotective effect of the ginsenoside Rg1 on cerebral ischemic injury in vivo and in vitro is mediated by PPARγ-regulated antioxidative and anti-inflammatory pathways. Evid Based Complement Alternat Med. 2017:78420822017.PubMed/NCBI

25 

Manser C, Vagnoni A, Guillot F, Davies J and Miller CC: Cdk5/p35 phosphorylates lemur tyrosine kinase-2 to regulate protein phosphatase-1C phosphorylation and activity. J Neurochem. 121:343–348. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Mandrekar-Colucci S, Karlo JC and Landreth GE: Mechanisms underlying the rapid peroxisome proliferator-activated receptor-γ-mediated amyloid clearance and reversal of cognitive deficits in a murine model of Alzheimer's disease. J Neurosci. 32:10117–10128. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Houseknecht KL, Cole BM and Steele PJ: Peroxisome proliferator-activated receptor gamma (PPARgamma) and its ligands: A review. Domest Anim Endocrinol. 22:1–23. 2002. View Article : Google Scholar : PubMed/NCBI

28 

Echeverría F, Valenzuela R, Catalina Hernandez-Rodas M and Valenzuela A: Docosahexaenoic acid (DHA), a fundamental fatty acid for the brain: New dietary sources. Prostaglandins Leukot Essent Fatty Acids. 124:1–10. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Echeverría F, Ortiz M, Valenzuela R and Videla LA: Long-chain polyunsaturated fatty acids regulation of PPARs, signaling: Relationship to tissue development and aging. Prostaglandins Leukot Essent Fatty Acids. 114:28–34. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Lin N, Chen LM, Pan XD, Zhu YG, Zhang J, Shi YQ and Chen XC: Tripchlorolide attenuates β-amyloid generation via suppressing PPARγ-regulated BACE1 activity in N2a/APP695 cells. Mol Neurobiol. 53:6397–6406. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Sadleir KR, Eimer WA, Cole SL and Vassar R: Aβ reduction in BACE1 heterozygous null 5XFAD mice is associated with transgenic APP level. Mol Neurodegener. 10:12015. View Article : Google Scholar : PubMed/NCBI

32 

Du J, Zhang L, Liu SB, Zhang C, Huang XQ, Li J, Zhao NM and Wang Z: PPARgamma transcriptionally regulates the expression of insulin-degrading enzyme in primary neurons. Biochem Biophys Res Commun. 383:485–490. 2009. View Article : Google Scholar : PubMed/NCBI

33 

Vingtdeux V, Chandakkar P, Zhao H, Blanc L, Ruiz S and Marambaud P: CALHM1 ion channel elicits amyloid-β clearance by insulin-degrading enzyme in cell lines and in vivo in the mouse brain. J Cell Sci. 128:2330–2338. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Delerive P, Fruchart JC and Staels B: Peroxisome proliferator-activated receptors in inflammation control. J Endocrinol. 169:453–459. 2001. View Article : Google Scholar : PubMed/NCBI

35 

Decourt B, Lahiri DK and Sabbagh MN: Targeting tumor necrosis factor alpha for Alzheimer's disease. Curr Alzheimer Res. 14:412–425. 2017.PubMed/NCBI

36 

Mendiola AS and Cardona AE: The IL-1β phenomena in neuroinflammatory diseases. J Neural Transm (Vienna). 125:781–795. 2018. View Article : Google Scholar : PubMed/NCBI

37 

Haddick PC, Larson JL, Rathore N, Bhangale TR, Phung QT, Srinivasan K, Hansen DV, Lill JR; Alzheimer's Disease Genetic Consortium (ADGC); Alzheimer's Disease Neuroimaging Initiative (ADNI), ; et al: A common variant of IL-6R is associated with elevated IL-6 pathway activity in Alzheimer's disease brains. J Alzheimers Dis. 56:1037–1054. 2017. View Article : Google Scholar : PubMed/NCBI

38 

Cifuentes D, Poittevin M, Bonnin P, Ngkelo A, Kubis N, Merkulova-Rainon T and Lévy BI: Inactivation of nitric oxide synthesis exacerbates the development of Alzheimer disease pathology in APPPS1 mice (Amyloid Precursor Protein/Presenilin-1). Hypertension. 70:613–623. 2017. View Article : Google Scholar : PubMed/NCBI

39 

Moussa C, Hebron M, Huang X, Ahn J, Rissman RA, Aisen PS and Turner RS: Resveratrol regulates Neuro-inflammation and induces adaptive immunity in Alzheimer's disease. J Neuroinflammation. 14:12017. View Article : Google Scholar : PubMed/NCBI

40 

Spitzer P, Weinbeer J, Herrmann M, Oberstein TJ, Condic M, Lewczuk P, Kornhuber J and Maler JM: Analysis of surface levels of IL-1 receptors and macrophage scavenger receptor I in peripheral immune cells of patients with Alzheimer's disease. J Geriatr Psychiatry Neurol. 32:211–220. 2019. View Article : Google Scholar : PubMed/NCBI

41 

de la Monte SM and Wands JR: Molecular indices of oxidative stress and mitochondrial dysfunction occur early and often progress with severity of Alzheimer's disease. J Alzheimers Dis. 9:167–181. 2006. View Article : Google Scholar : PubMed/NCBI

42 

Zhang ZX, Li YB and Zhao RP: Epigallocatechin gallate attenuates β-Amyloid generation and oxidative stress involvement of PPARγ in N2a/APP695 cells. Neurochem Res. 42:468–480. 2017. View Article : Google Scholar : PubMed/NCBI

43 

Silva-Abreu M, Calpena AC, Andrés-Benito P, Aso E, Romero IA, Roig-Carles D, Gromnicova R, Espina M, Ferrer I, García ML and Male D: PPARγ agonist-loaded PLGA-PEG nanocarriers as a potential treatment for Alzheimer's disease: In vitro and in vivo studies. Int J Nanomedicine. 13:5577–5590. 2018. View Article : Google Scholar : PubMed/NCBI

44 

Searcy JL, Phelps JT, Pancani T, Kadish I, Popovic J, Anderson KL, Beckett TL, Murphy MP, Chen KC, Blalock EM, et al: Long-term pioglitazone treatment improves learning and attenuates pathological markers in a mouse model of Alzheimer's disease. J Alzheimers Dis. 30:943–961. 2012. View Article : Google Scholar : PubMed/NCBI

45 

Hamano T, Shirafuji N, Makino C, Yen SH, Kanaan NM, Ueno A, Suzuki J, Ikawa M, Matsunaga A, Yamamura O, et al: Pioglitazone prevents tau oligomerization. Biochem Biophys Res Commun. 478:1035–1042. 2016. View Article : Google Scholar : PubMed/NCBI

46 

Dehghani L, Meamar R, Askari G, Khorvash F, Shaygannejad V, Pour AF and Javanmard SH: The effect of pioglitazone on the Alzheimer's disease-induced apoptosis in human umbilical vein endothelial cells. Int J Prev Med. 4 (Suppl 2):S205–S210. 2013.PubMed/NCBI

47 

Escribano L, Simón AM, Gimeno E, Cuadrado-Tejedor M, de Maturana RL, García-Osta A, Ricobaraza A, Pérez-Mediavilla A, Del Río J and Frechilla D: Rosiglitazone rescues memory impairment in Alzheimer's transgenic mice: Mechanisms involving a reduced amyloid and tau pathology. Neuropsychopharmacology. 35:1593–1604. 2010. View Article : Google Scholar : PubMed/NCBI

48 

Chiang MC, Nicol CJ, Cheng YC, Lin KH, Yen CH and Lin CH: Rosiglitazone activation of PPARγ-dependent pathways is neuroprotective in human neural stem cells against amyloid-beta-induced mitochondrial dysfunction and oxidative stress. Neurobiol Aging. 40:181–190. 2016. View Article : Google Scholar : PubMed/NCBI

49 

Toledo EM and Inestrosa NC: Activation of Wnt signaling by lithium and rosiglitazone reduced spatial memory impairment and neurodegeneration in brains of an APPswe/PSEN1ΔE9 mouse model of Alzheimer's disease. Mol Psychiatry. 15:272–285, 228. 2010. View Article : Google Scholar : PubMed/NCBI

50 

Strum JC, Shehee R, Virley D, Richardson J, Mattie M, Selley P, Ghosh S, Nock C, Saunders A and Roses A: Rosiglitazone induces mitochondrial biogenesis in mouse brain. J Alzheimers Dis. 11:45–51. 2007. View Article : Google Scholar : PubMed/NCBI

51 

Risner ME, Saunders AM, Altman JF, Ormandy GC, Craft S, Foley IM, Zvartau-Hind ME and Hosford DA; Rosiglitazone in Alzheimer's disease study group, : Efficacy of rosiglitazone in a genetically defined population with mild-to-moderate Alzheimer's disease. Pharmacogenomics J. 6:246–254. 2006. View Article : Google Scholar : PubMed/NCBI

52 

Combs CK, Johnson DE, Karlo JC, Cannady SB and Landreth GE: Inflammatory mechanisms in Alzheimer's disease: Inhibition of β-amyloid-stimulated proinflammatory responses and neurotoxicity by PPARγ agonists. J Neurosci. 20:558–567. 2000. View Article : Google Scholar : PubMed/NCBI

53 

Quan Q, Qian Y, Li X and Li M: Pioglitazone reduces β amyloid levels via inhibition of PPARγ phosphorylation in a neuronal model of Alzheimer's disease. Front Aging Neurosci. 11:1782019. View Article : Google Scholar : PubMed/NCBI

54 

Aubert RE, Herrera V, Chen W, Haffner SM and Pendergrass M: Rosiglitazone and pioglitazone increase fracture risk in women and men with type 2 diabetes. Diabetes Obes Metab. 12:716–721. 2010. View Article : Google Scholar : PubMed/NCBI

55 

Home PD, Pocock SJ, Beck-Nielsen H, Gomis R, Hanefeld M, Jones NP, Komajda M and McMurray JJ; RECORD study group, : Rosiglitazone evaluated for cardiovascular outcomes-an interim analysis. New Engl J Med. 357:28–38. 2007. View Article : Google Scholar : PubMed/NCBI

56 

Wang AT and Smith SA: ACP Journal Club. In older patients, rosiglitazone was associated with increased risk for stroke, heart failure, and mortality compared with pioglitazone. Ann Intern Med. 153:JC6–JC11. 2010. View Article : Google Scholar : PubMed/NCBI

57 

Lewis JD, Ferrara A, Peng T, Hedderson M, Bilker WB, Quesenberry CP, Vaughn DJ, Nessel L, Selby J and Strom BL: Risk of bladder cancer among diabetic patients treated with pioglitazone interim report of a longitudinal cohort study. Diabetes Care. 34:916–922. 2011. View Article : Google Scholar : PubMed/NCBI

58 

Toyota T and Ueno Y: Clinical effect and side effect of troglitazone. Nihon Rinsho. 58:376–382. 2000.PubMed/NCBI

59 

Kim HJ, Kim P and Shin CY: A comprehensive review of the therapeutic and pharmacological effects of ginseng and ginsenosides in central nervous system. J Ginseng Res. 37:8–29. 2013. View Article : Google Scholar : PubMed/NCBI

60 

Zhang S, Sun H, Wang C, Zheng X, Jia X, Cai E and Zhao Y: Comparative analysis of active ingredients and effects of the combination of Panax ginseng and Ophiopogon japonicus at different proportions on chemotherapy-induced myelosuppression mouse. Food Funct. 10:1563–1570. 2019. View Article : Google Scholar : PubMed/NCBI

61 

Kim MH, Kim SH and Yang WM: Mechanisms of action of phytochemicals from medicinal herbs in the treatment of Alzheimer's disease. Planta Med. 80:1249–1258. 2014. View Article : Google Scholar : PubMed/NCBI

62 

LaFerla FM, Green KN and Oddo S: Intracellular amyloid-beta in Alzheimer's disease. Nat Rev Neurosci. 8:499–509. 2007. View Article : Google Scholar : PubMed/NCBI

63 

Vassar R, Bennett BD, Babu-Khan S, Kahn S, Mendiaz EA, Denis P, Teplow DB, Ross S, Amarante P, Loeloff R, et al: Beta-secretase cleavage of Alzheimer's amyloid precursor protein by the transmembrane aspartic protease BACE. Science. 286:735–741. 1999. View Article : Google Scholar : PubMed/NCBI

64 

Yu G, Nishimura M, Arawaka S, Levitan D, Zhang L, Tandon A, Song YQ, Rogaeva E, Chen F, Kawarai T, et al: Nicastrin modulates presenilin-mediated notch/glp-1 signal transduction and betaAPP processing. Nature. 407:48–54. 2000. View Article : Google Scholar : PubMed/NCBI

65 

Xu H, Sweeney D, Wang R, Thinakaran G, Lo AC, Sisodia SS, Greengard P and Gandy S: Generation of Alzheimer β-amyloid protein in the trans-Golgi network in the apparent absence of vesicle formation. Proc Natl Acad Sci USA. 94:3748–3752. 1997. View Article : Google Scholar : PubMed/NCBI

66 

Gouras GK, Tsai J, Naslund J, Vincent B, Edgar M, Checler F, Greenfield JP, Haroutunian V, Buxbaum JD, Xu H, et al: Intraneuronal Abeta42 accumulation in human brain. Am J Pathol. 156:15–20. 2000. View Article : Google Scholar : PubMed/NCBI

67 

Pérez A, Morelli L, Cresto JC and Castano EM: Degradation of soluble amyloid β-peptides 1–40, 1–42, and the Dutch variant 1–40Q by insulin degrading enzyme from Alzheimer disease and control brains. Neurochem Res. 25:247–255. 2000. View Article : Google Scholar : PubMed/NCBI

68 

Helenius K, Yang Y, Alasaari J and Mäkelä TP: Mat1 inhibits peroxisome proliferator-activated receptor γ-mediated adipocyte differentiation. Mol Cell Biol. 29:315–323. 2009. View Article : Google Scholar : PubMed/NCBI

69 

Iankova I, Petersen RK, Annicotte JS, Chavey C, Hansen JB, Kratchmarova I, Sarruf D, Benkirane M, Kristiansen K and Fajas L: Peroxisome proliferator-activated receptor gamma recruits the positive transcription elongation factor b complex to activate transcription and promote adipogenesis. Mol Endocrinol. 20:1494–1505. 2006. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2020
Volume 22 Issue 4

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
Quan Q, Li X, Feng J, Hou J, Li M and Zhang B: Ginsenoside Rg1 reduces &beta;‑amyloid levels by inhibiting CD&Kappa;5‑induced PPAR&gamma; phosphorylation in a neuron model of Alzheimer's disease. Mol Med Rep 22: 3277-3288, 2020
APA
Quan, Q., Li, X., Feng, J., Hou, J., Li, M., & Zhang, B. (2020). Ginsenoside Rg1 reduces &beta;‑amyloid levels by inhibiting CD&Kappa;5‑induced PPAR&gamma; phosphorylation in a neuron model of Alzheimer's disease. Molecular Medicine Reports, 22, 3277-3288. https://doi.org/10.3892/mmr.2020.11424
MLA
Quan, Q., Li, X., Feng, J., Hou, J., Li, M., Zhang, B."Ginsenoside Rg1 reduces &beta;‑amyloid levels by inhibiting CD&Kappa;5‑induced PPAR&gamma; phosphorylation in a neuron model of Alzheimer's disease". Molecular Medicine Reports 22.4 (2020): 3277-3288.
Chicago
Quan, Q., Li, X., Feng, J., Hou, J., Li, M., Zhang, B."Ginsenoside Rg1 reduces &beta;‑amyloid levels by inhibiting CD&Kappa;5‑induced PPAR&gamma; phosphorylation in a neuron model of Alzheimer's disease". Molecular Medicine Reports 22, no. 4 (2020): 3277-3288. https://doi.org/10.3892/mmr.2020.11424