Open Access

Proteomic analysis of the effect of the polyphenol pentagalloyl glucose on proteins involved in neurodegenerative diseases in activated BV‑2 microglial cells

  • Authors:
    • Patricia Mendonca
    • Equar Taka
    • Karam F. A. Soliman
  • View Affiliations

  • Published online on: June 19, 2019     https://doi.org/10.3892/mmr.2019.10400
  • Pages: 1736-1746
  • Copyright: © Mendonca 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

Neuroinflammation and microglial activation are two important hallmarks of neurodegenerative diseases. Continuous microglial activation may cause the release of several cytotoxic molecules, including many cytokines that are involved in the inflammatory process. Therefore, attenuating inflammation caused by activated microglia may be an approach for the therapeutic management of neurodegenerative diseases. In addition, many studies have reported that polyphenol pentagalloyl glucose (1,2,3,4,6‑penta‑O‑galloyl‑β‑D‑glucose; PGG) is a molecule with potent anti‑inflammatory effects, such as inhibiting the release of proinflammatory cytokines. Our previous studies revealed that PGG attenuated the expression of two inflammatory cytokines (murine monocyte chemoattractant protein‑5 and pro‑metalloproteinase‑9) in lipopolysaccharide/interferon γ‑activated BV‑2 microglial cells. Additionally, PGG modulated the NF‑κB and MAPK signaling pathways by altering genes and proteins, which may affect the MAPK cascade and NF‑κB activation. The aim of the present study was to investigate the ability of PGG to modulate the expression of proteins released in activated BV‑2 microglial cells, which may be involved in the pathological process of inflammation and neurodegeneration. Proteomic analysis of activated BV‑2 cells identified 17 proteins whose expression levels were significantly downregulated by PGG, including septin‑7, ataxin‑2, and adenylosuccinate synthetase isozyme 2 (ADSS). These proteins were previously described as being highly expressed in neurodegenerative diseases and/or involved in the signaling pathways associated with the formation and growth of neuronal connections and the control of Alzheimer's disease pathogenesis. The inhibitory effect of PGG on ataxin‑2, septin‑7 and ADSS was further confirmed at the protein and transcriptional levels. Therefore, the obtained results suggest that PGG, with its potent inhibitory effects on ataxin‑2, septin‑7 and ADSS, may have potential use in the therapeutic management of neurodegenerative diseases.

Introduction

Microglia represent immune cells of the central nervous system (CNS) and are the main cells responsible for the intrinsic brain immune system (1,2). Under normal conditions, microglia monitor the surroundings continuously to identify the presence of CNS injury or damage, and they seem to preserve homeostasis through several interactions with neurons. However, continuous microglial activation increases the release of proinflammatory cytokines (1), which may cause detrimental inflammatory effects (3). This chronic inflammatory process enlists effector cells and can create a feedback loop, perpetuating inflammation and finally damaging the neurons (4,5). Microglial production of cytokines occurs as a response to injury, trauma or infection (6). Thus, the chronic activation of microglia is a critically important step in the development of neurodegeneration, leading to neuronal malfunction or complete neuron disability (1).

The knowledge of the detrimental effects caused by inflammation in the development of neurodegeneration shows that reducing inflammation may assist in delaying disease advancement. Compounds that can modulate microglial activity and decrease inflammation to controllable levels could be beneficial for neuronal survival (7). Moreover, the role of neuroinflammation in Alzheimer's disease (AD) progression was previously reported, showing the effects of NSAIDs on AD progression (8,9). Furthermore, several AD models were used to demonstrate the neuroprotective effects of different compounds with anti-inflammatory potential (10). Consequently, attenuating inflammatory mediators can help reduce or retard the advancement of AD.

Various compounds extracted from plants have shown strong anti-inflammatory activity. The polyphenol pentagalloyl glucose (1,2,3,4,6-penta-O-galloyl-β-D-glucose, PGG) is found in many medicinal plants, including Rhus chinensis Mill and Paeonia suffruticosa (5,11). This compound has been reported in several in vivo and in vitro studies showing its potential in the therapy and prevention of several inflammatory diseases. Our earlier work showed that PGG inhibited the release of MCP-5 (monocyte chemoattractant protein-5) and pro-MMP-9 (pro-metalloproteinase-9) in activated BV-2 cells (12). PGG also modulated genes and proteins involved in the nuclear-factor-κB (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways, which may affect the MAPK cascade, NF-κB activation, and the subsequent release of MCP-5 and pro-MMP-9 (13). The current work investigated the effect of PGG on the expression of proteins that may be involved in the pathogenesis of neuroinflammation and neurodegenerative diseases using an LPS/IFNγ-activated BV-2 microglial cell model.

Materials and methods

Chemicals and reagents

Polyphenol pentagalloyl glucose (1,2,3,4,6-penta-O-galloyl-β-D-glucose-purity 96.8%), bovine serum albumin (BSA), dimethyl sulfoxide (DMSO), lipopolysaccharide from Salmonella enterica (LPS), interferon γ (IFNγ), urea, Tris/HCl, iodoacetamide, trypsin, NaCl, trifluoracetic acid (CF3COOH), ammonium bicarbonate (NH4HCO3), and general chemicals were purchased from Sigma-Aldrich Co. and VWR International. Dulbecco's modified Eagle's medium-high glucose medium (DMEM), penicillin/streptomycin, fetal bovine serum heat inactivated (FBS-HI), trypsin-EDTA, and Hank's Balanced Salt Solution (HBSS) were purchased from Genesee Scientific. All reagents and plates for western blot assays were purchased from ProteinSimple. Bradford reagent, PCR primers, and reagents were from Bio-Rad, and primary antibodies were from Thermo Fisher Scientific (Table I).

Table I.

List of primary antibodies.

Table I.

List of primary antibodies.

AntibodyCatalog no.TypeSpecies reactivityHost/Isotype
Septin-7PA5-56181Polyclonalhuman, mouse, ratRabbit/IgG
Ataxin-2PA5-54336Polyclonalhuman, mouse, ratRabbit/IgG
ADSSPA5-55070Polyclonalhuman, mouse, ratRabbit/IgG

[i] All antibodies were purchased from Thermo Fisher Scientific, Inc. IgG, immunoglobulin G; ADSS, adenylosuccinate synthetase isozyme 2.

Cell culture and treatments

The immortalized murine microglial BV-2 cell line was provided by Dr Elizabetta Blasi (14). Cells were grown in T-75 flasks using DMEM supplemented with 10% heat-inactivated FBS and 1% penicillin (100 U/ml)/streptomycin (0.1 mg/ml) in an incubator set for 5% CO2. at 37°C. For the experimental media, only 2.5% of heat-inactivated FBS was added. LPS and IFNγ were stored at −20°C at concentrations of 1 mg/ml and 200 ng/ml, respectively. The working concentrations of 0.2 µg/ml and ٠.٢ ng/ml were then used in the experiments. DMSO was used to dissolve PGG, and the final concentration of DMSO did not exceed 0.1% (15). The selected concentration of PGG was based on the cell viability results from our prior study (12), which showed >80% of viable cells after treatment with 25 µM PGG. The count of cells in all the experiments was ٥x١٠5/ml, and the treatments consisted of the control (DMSO), PGG, LPS/IFNγ, and PGG followed by the combination of LPS/IFNγ after 1 h.

Proteomic assay

In this experiment, BV-2 cells were plated in experimental media and incubated overnight. The next day, the cells were treated as described before. After 24 h, cells were harvested using 0.25% trypsin-EDTA by first aspirating the medium, centrifuging, and washing the cells twice with PBS. Once the pellet was ready, using the Filter Aided Sample Preparation (FASP) protocol, samples were prepared by mixing 30 µl of cell lysate with 200 µl of UA (8 M urea in 0.1 M Tris/HCl pH 8.5) in the filter unit and centrifuged twice. The flow through from the collection tube was discarded. Then, 100 µl of IAA (0.05 M iodoacetamide in UA) solution was added and mixed at 600 rpm in a thermomixer for 1 min and incubated without mixing for 20 min. After centrifuging, 100 µl of UA was added to the filter unit and centrifuged. Next, 100 µl of ABC (0.05 M NH4HCO3 in water) was added to the filter unit and centrifuged. Then, 40 µl ABC with trypsin (enzyme to protein ratio 1:100) was added and mixed at 600 rpm in a thermomixer for 1 min. The units were incubated at 37°C for 18 h, and the filter units were transferred to new collection tubes. Then, 50 µl of NaCl (٠.٥ M) was pipetted into the filter and centrifuged. Using CF3COOH, the filtrate was acidified, desalted and sent to the Translational Science Laboratory for Orbitrap/QExactive Proteomic LC-MS/MS (liquid chromatography-mass spectrometry) for complex mixture analysis. Using ‘Scaffold version 4.4’ software, the results were then analyzed, and the fold change of each protein of interest was calculated.

Protein assay

Cells were appropriately treated and kept in the incubator for 24 h. The next day, cells were harvested, washed twice with PBS, and lysed with buffer containing protease inhibitor cocktail. Using Bradford reagent, the concentration of protein was measured. Five µl of standards in concentrations of ٠ to ٢ mg/ml or 5 µl of samples and ٢٠٠ µl of protein assay reagent were added to the ٩٦-well plate. Using a Synergy HTX Multi-Reader (BioTek), the concentration of proteins was measured at 595 nm wavelength.

Capillary electrophoresis western blot analysis

Total protein expression was determined using western blot analysis (Wes, ProteinSimple). The protein concentration was determined as described above. ProteinSimple provided the reagents and protocol for the assay. The concentrations of antibody and protein for the experiments were first optimized by being tested at 3 different concentrations. The best concentration was then selected for further tests. Briefly, the protein samples (concentration: 0.2 mg/ml) were mixed with sample buffer, fluorescent molecular weight markers, and dithiothreitol and left in a heat block at 95°C for 5 min. The microplate provided with the kit was then loaded with blocking buffer, antibodies (in dilutions ranging from 1:5 to 1:25), secondary antibody, chemiluminescent substrate, separation and stacking matrices. The microplate was then placed in the instrument and through the capillary system; the electrophoresis and immunodetection occurred. The reaction identifies specific proteins by using primary and secondary antibodies and a chemiluminescent substrate. The Wes instrument provided us with real-time results of the experiment. By using a charge-coupled device camera, the chemiluminescence reaction and the digital image were analyzed by the software (ProteinSimple Compass).

Reverse transcriptase-polymerase chain reaction (RT-qPCR)
RNA extraction and cDNA synthesis

Cell pellets from the different treatments were initially lysed with 1 ml of TRIzol reagent. Chloroform (0.2 ml) was added, vortexed, incubated at 15–30°C for 2–3 min, and centrifuged at 10,000 g for 15 min at 2–8°C. The aqueous phase of the lysed samples was transferred to a fresh tube, and the RNA was precipitated by mixing with isopropyl alcohol (0.5 ml). After centrifugation, 75% ethanol was used to wash the RNA pellet. After centrifugation at 7,500 g for 5 min at 2–8°C, the pellet was left to dry for 15 min and then dissolved in water (RNase free). The purity and RNA concentration were measured using a NanoDrop (Thermo Fischer Scientific). Using iScript advanced reverse transcriptase from Bio-Rad, the cDNA strands were synthesized based on the mRNA. The solutions were loaded into 0.2 ml tubes and included 5X iScript advanced reaction mix (4 µl) (containing primers), reverse transcriptase (١ µl), sample (١.٥ µg/٧.٥ µl), and water (٧.٥ µl) in a total of ٢٠ µl. The thermal cycling for reverse transcription steps included ٣٠ min at 42°C and 5 min at 85°C.

RT-PCR

PCR amplification followed the Bio-Rad protocol. A 1 µl aliquot of the sample (٢٠٠ ng cDNA/reaction), 10 µl of SsoAdvanced™ Universal SYBR®-Green Supermix (Bio-Rad), 1 µl of primer, and ٨ µl of water were pipetted into each well. Using the CFX٩٦ Real-Time System from Bio-Rad, the protocol provided by the company was followed: Initial 95°C for 2 min (hold step) and 95°C for 15 sec (denaturation), followed by 40 cycles of 60°C for 30 sec (annealing/extension) and 65–95°C for ٥ sec/step (melting curve). Primers were selected according to the specific genes of interest. The identification of each primer was provided by the manufacturer (Bio-Rad).

– Septin-7-UniqueAssayID: qMmuCID0017359

– Ataxin-2-UniqueAssayID: qMmuCID0025396

– ADSS-UniqueAssayID: qMmuCID0005512

Data analysis
Statistical analysis

Statistical analysis of the obtained results included a minimum of 3 biological replicates and 3 technical replicates for each one of the assays. For the proteomics assay, data analysis was performed using Scaffold software, and the significance of the difference between the groups was assessed using Student's t-test (P<0.05). The results from the western blot assay were analyzed using ProteinSimple Compass software from the manufacturer. For the RT-PCR assays, Bio-Rad CFX manager software was used. The software automatically calculates the differences in mRNA expression for each of the treatments compared to the reference gene (GAPDH). All data are expressed as the mean ± standard error of the mean, and the significance of the difference between the groups was assessed using a one-way analysis of variance, followed by Dunnett's multiple comparison post hoc tests. P<0.05 was considered to indicate a statistically significant difference.

Bioinformatics analysis

The NCBI (National Center for Biotechnology Information-ncbi.nlm.nih.gov) (16) and PANTHER (Protein Analysis Through Evolutionary Relationships-pantherdb.org) (17) databases were used to identify biological and molecular functions, as well as signaling pathways associated with the proteins.

Results

A proteomic approach was used to profile the effect of PGG on global protein expression in LPS/IFNγ-activated BV-2 microglia cells. A total of 1895 proteins were identified as being differentially expressed after cells were treated with PGG, LPS/IFNγ or the combinations of PGG + LPS/IFNγ. Among those, specific proteins were selected according to the fold change. Proteins with at least a 7-fold change in inhibition after PGG treatment were chosen, resulting in a total of 24 proteins. Then, statistical analysis was used to select those that showed significant inhibition after PGG treatment. A total of 17 proteins were identified for further studies, as shown in the volcano plot (Fig. 1). The fold change was calculated by comparing the control vs. LPS/IFNγ groups (Table II) and the LPS/IFNγ vs. PGG + LPS/IFNγ groups (Table III). The data obtained show that even for those proteins where LPS/IFNγ did not induce an augmentation in protein expression, PGG pretreatment downregulated their expression up to 53-fold (Tables II and III). Some of these highly downregulated proteins are known to be involved in cancer progression.

Table II.

Effect of LPS/IFNγ on the protein expression of BV-2 cells.

Table II.

Effect of LPS/IFNγ on the protein expression of BV-2 cells.

Protein nameProtein IDMolecular weight, kDaP-value (t-test)Fold change
Ataxin-2-like proteinATX2L1110.047+3
Septin-7SEPT7510.011+2
SNW domain-containing proteinSNW1610.048+5
WAS/WAS/L-interacting proteinWIPF1500.0501
Thymidylate synthaseTYSY35<0.000101
Acidic leucine-rich nuclear phosphoprotein 32 family memberAN32B310.0111
5′-3′ exoribonuclease 2XRN21090.00181
Protein-L-isoaspartate (D-aspartate) O-methyltransferasePIMT250.00361
Puromycin-sensitive aminopeptidasePSA103<0.00014+3.4
Coronin-1BCOR1B540.0171
Bleomycin hydrolaseBLMH530.040+1.6
EmerinEMD290.013+1.7
Nuclear transport factor 2NTF214<0.000951
Treacle proteinTCOF135<0.000981
Adenylosuccinate synthetase isozyme 2PURA250<0.000101
Protein phosphatasePPM1G59<0.000101
Glyoxalase domain-containing protein 4GLOD433<0.00010+3.2

[i] Table of the proteins identified by Orbitrap/QExactive Proteomic LC-MS/MS and the respective molecular weight. The fold change corresponds to the increase in the protein expression when BV-2 cells were activated by LPS/IFNγ, tested in three independent studies (n=3). Statistically significant differences between control and LPS/IFNγ were evaluated by a Student's t-test, P<0.05.

Table III.

Effect of PGG on LPS/IFNγ-activated BV-2 Cells

Table III.

Effect of PGG on LPS/IFNγ-activated BV-2 Cells

Protein nameProtein IDMolecular weight, kDaP-value (t-Test)Fold change
Ataxin-2-like proteinATX2L1110.015−7.2
Septin-7SEPT7510.017−8
SNW domain-containing proteinSNW1610.048−8.4
WAS/WAS/L-interacting proteinWIPF1500.022−9.4
Thymidylate synthaseTYSY350.01−9.7
Acidic leucine-rich nuclearphosphoprotein 32 family memberAN32B310.025−10
5′-3′ exoribonuclease 2XRN21090.015−14
Protein-L-isoaspartate (D-aspartate) O-methyltransferasePIMT250.0013−14
Puromycin-sensitive aminopeptidasePSA1030.008−15
Coronin-1BCOR1B540.029−16
Bleomycin hydrolaseBLMH530.0089−17
EmerinEMD290.046−18
Nuclear transport factor 2NTF2140.0075−20
Treacle proteinTCOF1350.012−23
Adenylosuccinate synthetase isozyme 2PURA2500.0028−23
Protein phosphatasePPM1G590.041−30
Glyoxalase domain-containing protein 4GLOD4330.022−53

[i] Table of proteins identified by Orbitrap/QExactive Proteomic LC-MS/MS and the respective molecular weight. The fold change corresponds to the inhibition of the protein expression when activated BV-2 cells were pre-treated with PGG, tested in three independent studies (n=3). Statistically significant differences between LPS/IFNγ and PGG + LPS/IFNγ were evaluated by a Student's t-test, P<0.05.

Annotation of the function of the different proteins was performed using the NCBI data file to identify the biological relevance of the differentially expressed proteins in response to PGG treatment. The proteins were placed into 15 biological processes (biological regulations, cellular and developmental processes, establishment of localization, growth, localization, locomotion, metabolic process, multi-organism and multicellular organism processes, reproduction, reproductive process, response to stimulus, rhythmic process and viral reproduction) (Fig. 2) and 5 molecular functions (binding, catalytic activity, molecular function, transcription regulator activity, and transporter activity).

The PANTHER classification system was used to classify the proteins according to the signaling pathway that they are associated with. The database found that 3 proteins (adenylosuccinate synthase isozyme 2 (ADSS), thymidylate synthase, and 5′-3′ exoribonuclease) were involved in 6 signaling pathways: DNA replication, de novo purine biosynthesis, de novo pyrimidine deoxyribonucleotide biosynthesis, formyltetrahydrofolate biosynthesis, tetrahydroformate biosynthesis, and the Wnt signaling pathway.

Among the 17 proteins significantly downregulated by PGG, septin-7, ataxin-2 and ADSS were of special interest because these proteins were previously described as being highly expressed in neurodegenerative diseases and/or involved in the development of neuronal circuits and control of AD and Parkinson's disease (PD) pathogenesis. To validate the proteomic results, we performed western blot assays with specific antibodies against Septin-7, Ataxin-2 and ADSS. The results showed that LPS/IFNγ induced the expression of these proteins in BV-2 microglial cells and significantly reduced their expression when activated BV-2 microglial cells were pretreated with PGG (Figs. 35).

In this investigation, to confirm whether the changes observed in protein expression were the result of transcriptional regulation, we used RT-PCR to determine the mRNA levels of the three proteins studied. The RT-PCR assay results were consistent with the results obtained from the proteomics and western blot assays. PGG significantly suppressed the expression of the ataxin-2, septin-7 and ADSS genes (Fig. 6A-C). Therefore, these results confirmed the inhibitory effect of PGG on related proteins and genes involved in neurodegeneration.

Discussion

Neuroinflammation is a prominent trait common to many neurodegenerative diseases (7), such as AD, PD, and several other disorders, such as amyotrophic lateral sclerosis, multiple sclerosis, spinal cord injury, and traumatic brain (18). Persistent activation of innate immunity, including responses mediated by microglial activation, is a common association among these diseases, causing activation of neurotoxic pathways that may lead to progressive neurodegeneration (18).

The current study indicates that PGG has profound effects on the proteome, causing up to a 53-fold inhibition of protein expression. PGG has proven to be a very potent antioxidant agent (19,20). While gallic acid (GA) is the primary product of tannic acid hydrolysis (21), PGG showed higher antioxidant activity (22). Moreover, oral PGG was found to reduce the accumulation of human amyloid β (Aβ) protein in the brains of transgenic mice overexpressing this protein (23). PGG inhibited accumulation at doses more than 10 times lower than that of the plant extract. The study showed that PGG suppresses Aβ fibril accumulation in treated animals, implying that PGG treatment could offer a therapeutic aid for AD patients with moderate cognitive injury (23). Additionally, by applying the combined technology of ion mobility associated with mass spectrometry, transmission electron microscopy, and molecular kinetics, studies indicated that PGG interacts with the N-terminal metal binding segment and the first central hydrophobic core, interfering with Aβ1-40 and Aβ1-42 amyloid assembly. These results showed that PGG is a potential therapeutic drug to mitigate Aβ oligomer neurotoxicity (24).

Currently, there are many cell models used to examine neuroinflammation, including primary microglial cultures and immortalized microglial cell cultures (25). In the present work, we used BV-2 microglial cells that showed similar properties to primary microglia (2,25). Henn et al (2) showed that in response to LPS, 90% of genes induced in BV-2 cells were also induced in primary microglia; however, the upregulation of genes in BV-2 was less pronounced. Although BV-2 cells have an inflammatory response that is not identical to primary cultures, they are still an important and widely used cell model for neuroinflammation. Our prior studies showed that PGG inhibited the expression of MCP-5 and pro-MMP-9 in LPS/IFNγ-activated BV-2 microglial cells. These proinflammatory cytokines may have a connection with the development of neurofibrillary tangles and senile plaques in AD patients. Our earlier studies also showed that PGG can modulate genes that participate in NF-κB and MAPK signaling, which are key components in the process of neuroinflammation. PGG modulated the expression of CDK2, CHUK, IRAK1 and NF-κB1 at the transcriptional and protein levels, which may help to explain how PGG downregulates the release of MCP-5 and pro-MMP-9 in stimulated BV-2 microglial cells (12,13).

In the present study, the data obtained indicate that PGG was very effective at inhibiting the expression of proteins and genes that are critical to neurodegeneration, including septin-7, ataxin-2 and ADSS. These proteins have been found to be involved in the pathogenesis of neurodegenerative diseases. Kinoshita and colleagues reported that septins were concentrated in the brains of AD patients, specifically in the intracellular neurofibrillary tangles, dystrophic neurites in senile plaques, and neuropil threads (26). Septins were shown to be involved in neurodegeneration and neurobehavioral conditions. Reports have shown that the septin family is found in postsynaptic densities when analyzed by mass spectrometry (27,28), indicating a possible involvement in neurodegenerative diseases and cognitive impairment. Septins may aid in the development of cellular aggregates, although their function in neurodegeneration is still unknown. Additionally, the septin filaments that maintain the structure and shape of the cells become unfolded, which may facilitate the formation of aggregates that interrupt cell function, leading to cell death (29). The septin family provides attractive candidates that may be involved in the essential mechanisms of synaptic dysfunction and neurodegeneration in neurodegenerative diseases. Several reports have described the association of septins with many diseases, including AD, PD, Huntington's disease, frontotemporal lobar degeneration, and Down's syndrome (26,29,3035), indicating that these proteins are involved in the pathogenic mechanism of neurodegeneration.

Another critical protein whose expression was attenuated by PGG in this study is ataxin 2, which is an RNA-binding protein found in the body with multiple roles in RNA metabolism and is broadly expressed in the mammalian nervous system (36). Ataxin 2 was found to be involved in neurodegenerative diseases, such as amyotrophic lateral sclerosis (ALS) and spinocerebellar ataxia type 2 (SCA2). In mouse models, reduced ataxin 2 levels improved motor performance of the animals in both diseases. In addition, this protein could alter the toxicity produced by the RNA-binding protein TDP43 (encoded by TARDBP), which participates in the formation of aggregates in the brain and spinal cords of ALS patients. A decrease in disease progression was observed when TARDBP transgenic mice were crossed with Atxn2-knockout mice, increasing the median lifespan in 80% of the animals studied. This work suggests that neurodegenerative conditions could be improved by targeting ataxin-2 (36,37).

Furthermore, adenylosuccinate synthetase isozyme 2 (ADSS) was another protein of interest, which was inhibited by PGG in the current study. ADSS is an enzyme that participates in the de novo and salvage pathways for purine and nucleotide biosynthesis, both of which play a role in the formation of nucleotide inosine monophosphate. Vertebrates present two isozymes, one basic (ADSS1) and the other acidic (ADSS2), which respond in a different way to inhibitors, indicating that they differ in their regulation (٣٨-41). The regulation of ADSS is also implicated in the maintenance of AMP/GMP ratios in the cell (٣٨). ADSS was identified as one of the altered proteins in MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine)-treated animal models of PD (42). Since mitochondrial dysfunction is likely to be one of the mechanisms that leads to PD, an investigation using quantitative proteomic analysis for mitochondrial protein showed that MPTP-treated animals also presented an increase in the expression of ADSS, from 50 to 100% (43). The present study, together with previous proteomic studies in the literature, revealed that proteins such as ADSS may be connected to PD pathogenesis and could be regulated by PGG, indicating that this compound may have potential use in targeted therapy.

Since ataxin-2, septin-7 and ADSS were found to be concentrated in neurofibrillary tangles and/or involved in synapse impairment induced by Aβ protein (26,44), these proteins are attractive candidates that may be associated with the fundamental mechanisms of synaptic damage in neurodegenerative diseases. Thus, by inhibiting the expression of these proteins, PGG may provide a useful target compound for neurodegenerative disease therapies.

The proteomic results not only revealed the inhibitory effect of PGG on proteins overexpressed in neurodegenerative diseases but also showed that PGG modulates many proteins involved in different cellular functions, which participate in several signaling pathways. PGG exhibited a significant fold change inhibition in the expression of WAS/WASL-interacting protein family member 1 (WIPF1) (−9.4-fold↓), thymidylate synthase (−9.7-fold↓), and glyoxalase (−53-fold↓), whose overexpression has been shown to be associated with cancer development and progression. The expression of WIPF1 had a critical role in thyroid cancer cell migration and invasion, suggesting WIPF1 as a novel therapeutic target for thyroid cancer (45). Additionally, high levels of thymidylate synthase expression have been correlated with poor prognosis in breast cancer (46), gastric cancer and colorectal cancers (47,48).

In the present study, the most remarkable effect of PGG was on the expression of glyoxalase 4 (GLOD4), with a 53-fold inhibition. The overexpression of glyoxalases has been reported in numerous types of malignant tumors, such as colon, breast, ovarian, and prostate cancers (4954). Furthermore, glyoxalases I and II are involved in the detoxification of methylglyoxal, a cytotoxic product of glycolysis, whose overexpression was observed in 79% of tumors (55). Methylglyoxal and glycation end-products are associated with an increase in proinflammatory markers and affect AD, Aβ peptide depositions and neurofibrillary tangles (5658). In tumor cells, increased cellular levels of toxic methylglyoxal and S-d-lactoylglutathione metabolites are present due to a higher metabolism compared to normal cells. In response to the increase in the production of these cytotoxic products, tumor cells augment the activity of the detoxifying glyoxalase system to minimize the intracellular concentrations of toxic metabolites (59). Methylglyoxal also accumulates with age, and it is associated with age-associated pathologies, such as diabetic complications and neurodegenerative disorders (60). Previous studies using mutant GLOD-4/GLO1-animals (Caenorhabditis elegans model) showed that animals quickly exhibited several pathogenic phenotypes reminiscent of α-dicarbonyl stress-induced age-associated disorders (61). Therefore, glyoxalase inhibitors, such as PGG, may have potential as antitumor agents as well as regulatory mediators in neurodegenerative diseases. While the role of glyoxalase 4 is not well established in humans, our results suggest that PGG may regulate the expression of distinct but overlapping genes and proteins involved in neurodegeneration and cancer progression.

Although the current study did not directly confirm the effect of PGG on the expression of CDK2, CHUK, IRAK1, and NF-κB1 observed in our previous manuscript (13), our current results indirectly showed that there may be an association among all the proteins that were downregulated by PGG. CDK2, CHUK, IRAK1 and NF-κB1 are proteins involved in the NF-κB and MAPK signaling pathways, which are both activated by pathogenic and noxious stimuli. Evidence suggests that MAPKs can participate in the regulation of NF-κB transcriptional activity, demonstrating that both the c-Jun N-terminal kinase and the p38 pathways are implicated in the activation of NF-κB in the cytoplasm, as well as in modulation of its transactivation potential in the nucleus, confirming the association between these signaling pathways (62). Moreover, Septin 7 was identified as a novel ERK3-interacting protein by using a Ras recruitment system, and evidence showed that Sept7, ERK3 and MK5 exist in the same complex and, together with Kal7, are of physiological relevance in neuronal plasticity by regulating dendritic spine formation (63). Furthermore, Ataxin-2 was described as a positive regulator of Notch signaling (64), which may maintain NF-κB activity by direct interaction with p50/c-Rel in the nucleus (65). Thus, we can conclude that there is an indirect association with the current and previous results due to the overlapping functions and interactions of multiple proteins involved in the NF-κB and MAPK signaling pathways.

The results of this work provide evidence that the polyphenolic compound PGG can modulate the expression of several proteins and transcripts whose expression is increased in LPS/IFNγ-stimulated BV-2 microglial cells. Among the proteins, PGG significantly inhibited the expression of ataxin-2, septin-7 and ADSS, which play a critical role in synapse impairment and pathogenesis of neurodegenerative diseases. Therefore, this study suggests that PGG may have therapeutic potential for neuroinflammation and neurodegeneration by targeting ataxin-2, septin-7 and ADSS.

Acknowledgements

Not applicable.

Funding

The present study was supported by NIH NIMHD (grant nos. G12 MD007582 and P20 MD 006738).

Availability of data and materials

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

Authors' contributions

PM and KFAS conceived and designed the study. PM and ET performed the experiments. PM analyzed and interpreted the data, and wrote the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

amyloid-β

AD

Alzheimer's disease

ADSS

adenylosuccinate synthetase isozyme 2

ALS

amyotrophic lateral sclerosis

CNS

central nervous system

IFNγ

interferon γ

FASP

Filter Aided Sample Preparation

GA

gallic acid

GLOD4

Glyoxalase 4

LPS

lipopolysaccharide

MAPK

mitogen-activated protein kinase

MCP-5

monocyte chemoattractant protein-5

NCBI

National Center for Biotechnology Information

NF-κB

nuclear factor-κB

PANTHER

Protein Analysis Through Evolutionary Relationships

PD

Parkinson's disease

PGG

1,2,3,4,6-penta-O-galloyl-β-D-glucose

Pro MMP-9

pro-metalloproteinase-9

SCA2

spinocerebellar ataxia type 2

TLR4

Toll-like receptor 4

WIPF1

WAS/WASL-interacting protein family member 1

References

1 

Street WJ, Mark RE and Griffin WS: Microglia and neuroinflammation: A pathological perspective. J Neuroinflammation. 1:142004. View Article : Google Scholar : PubMed/NCBI

2 

Henn A, Lund S, Hedtjärn M, Schrattenholz A, Pörzgen P and Leist M: The suitability of BV-2 cells as an alternative model system for primary microglia cultures for animal experiments examining brain inflammation. ALTEX. 26:83–94. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Schwartz M and Baruch K: The resolution of neuroinflammation in neurodegeneration: Leukocyte recruitment via the choroid plexus. EMBO J. 33:7–22. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Gendelman HE: Neural immunity: Friend or foe? J Neurovirol. 8:474–479. 2002. View Article : Google Scholar : PubMed/NCBI

5 

Oh GS, Pae HO, Choi BM, Lee HS, Kim IK, Yun YG, Kim JD and Chung HT: Penta-O-galloyl-Beta-D-glucose inhibits phorbol myristate acetate-induced interleukin-8 [correction of interleukin-8] gene expression in human monocytic U937 cells through its inactivation of nuclear factor-kappa B. Int Immunopharmacol. 4:377–386. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Hanisch UK: Microglia as a source and target of cytokines. Glia. 40:140–155. 2002. View Article : Google Scholar : PubMed/NCBI

7 

Gao HM and Hong JS: Why neurodegenerative diseases are progressive: Uncontrolled inflammation drives disease progression. Trends Immunol. 29:357–365. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Etminan M, Gill S and Samii A: Effect of non-steroidal anti-inflammatory drugs on risk of Alzheimer's disease: Systematic review and meta-analysis of observational studies. BMJ. 327:1282003. View Article : Google Scholar : PubMed/NCBI

9 

McGeer PL and McGeer EG: The amyloid cascade-inflammatory hypothesis of Alzheimer disease: Implications for therapy. Acta Neuropathol. 126:479–497. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Heneka MT, Kummer MP, Weggen S, Bulic B, Multhaup G, Münter L, Hüll M, Pflanzner T and Pietrzik CU: Molecular mechanisms and therapeutic application of NSAIDs and derived compounds in Alzheimer's disease. Curr Alzheimer Res. 8:115–131. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Cao Y, Himmeldirk KB, Qian Y, Ren Y, Malki A and Chen X: Biological and biomedical functions of Penta-O-Galloyl-D-Glucose and its derivatives. J Nat Med. 68:465–472. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Mendonca P, Taka E, Bauer D, Cobourne-Duval M and Soliman KF: The attenuating effects of 1,2,3,4,6 Penta-O-Galloyl-β-D-glucose on inflammatory cytokines release from activated BV-2 microglial cells. J Neuroimmunol. 305:9–15. 2017. View Article : Google Scholar : PubMed/NCBI

13 

Mendonca P, Taka E, Bauer D, Reams RR and Soliman KFA: The attenuating effects of 1,2,3,4,6 penta-O-galloyl-β-D-glucose on pro-inflammatory responses of LPS/IFNγ-activated BV-2 microglial cells through NFκB and MAPK signaling pathways. J Neuroimmunol. 324:43–53. 2018. View Article : Google Scholar : PubMed/NCBI

14 

Blasi E, Barluzzi R, Bocchini V, Mazzolla R and Bistoni F: Immortalization of murine microglial cells by a v-raf/v-myc carrying retrovirus. J Neuroimmunol. 27:229–237. 1990. View Article : Google Scholar : PubMed/NCBI

15 

Elsisi N, Darling-Reed S, Lee EY, Oriaku ET and Soliman KF: Ibuprofen and apigenin induce apoptosis and cell cycle arrest in activated microglia. Neurosci Lett. 375:91–96. 2005. View Article : Google Scholar : PubMed/NCBI

16 

NCBI Resource Coordinators: Database resources of the National Center for Biotechnology Information. Nucleic Acids Res. 44:D7–D19. 2016. View Article : Google Scholar : PubMed/NCBI

17 

Thomas PD, Campbell MJ, Kejariwal A, Mi H, Karlak B, Daverman R, Diemer K, Muruganujan A and Narechania A: PANTHER: A library of protein families and subfamilies indexed by function. Genome Res. 13:2129–2141. 2003. View Article : Google Scholar : PubMed/NCBI

18 

Amor S, Peferoen LA, Vogel DY, Breur M, van der Valk P, Baker D and van Noort JM: Inflammation in neurodegenerative diseases-an update. Immunology. 142:151–166. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Abdelwahed A, Bouhlel I, Skandrani I, Valenti K, Kadri M, Guiraud P, Steinman R, Mariotte AM, Ghedira K, Laporte F, et al: Study of antimutagenic and antioxidant activities of Gallic acid and 1,2,3,4,6 penta galloyl glucose from Pistacia lentiscus. Confirmation by microarray expression profiling. Chem Biol Interact. 165:1–13. 2007. View Article : Google Scholar : PubMed/NCBI

20 

Kim BH, Choi MS, Lee HG, Lee SH, Noh KH, Know S, Jeong AJ, Lee H, Yi EH, Park JY, et al: The photoprotective potential of penta-O-galloyl-β-D-glucose by targeting NF-κB and MAPK signaling in UVB radiation-induced human dermal fibroblasts and mouse skin. Mol Cells. 38:982–990. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Aithal M and Belur P: Enhancement of propyl gallate yield in a nonaqueous medium using novel cell-associated tannase of Bacillus massiliensis. Prep Biochem Biotechnol. 43:445–455. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Wang KJ, Yang CR and Zhang YJ: Phenolic antioxidants from Chinese toon (fresh young leaves and shoots of Toona sinensis). Food Chemistry. 101:365–371. 2006. View Article : Google Scholar

23 

Fujiwara H, Tabuchi M, Yamaguchi T, Iwasaki K, Furukawa K, Sekiguchi K, Ikarashi Y, Kudo Y, Higuchi M, Saido TC, et al: Traditional medicinal herb Paeonia suffruticosa and its active constituent 1,2,3,4,6-Penta-O-Galloyl-Beta-D-Glucopyranose have potent anti-aggregation effects on Alzheimer's amyloid beta proteins in vitro and in vivo. J Neurochem. 109:1648–1657. 2009. View Article : Google Scholar : PubMed/NCBI

24 

De Almeida NEC, Do TD, LaPointe NE, Tro M, Feinstein SC, Shea JE and Bowers MT: 1,2,3,4,6-Penta-O-Galloyl-β-D-glucopyranose binds to the N-terminal metal binding region to inhibit amyloid β-protein oligomer and fibril formation. Int J Mass Spectrom. 420:24–34. 2017. View Article : Google Scholar : PubMed/NCBI

25 

Stansley B, Post J and Hensley K: A comparative review of cell culture systems for the study of microglial biology in Alzheimer's disease. J Neuroinflammation. 9:1152012. View Article : Google Scholar : PubMed/NCBI

26 

Kinoshita A, Kinoshita M, Akiyama H, Tomimoto H, Akiguchi I, Kumar S, Noda M and Kimura J: Identification of septins in neurofibrillary tangles in Alzheimer's disease. Am J Pathol. 153:1551–1560. 1998. View Article : Google Scholar : PubMed/NCBI

27 

Tada T, Simonetta A, Batterton M, Kinoshita M, Edbauer D and Sheng M: Role of septin cytoskeleton in spine morphogenesis and dendrite development in neurons. Curr Biol. 17:1752–1758. 2007. View Article : Google Scholar : PubMed/NCBI

28 

Walikonis RS, Jensen ON, Mann M, Provance DW Jr, Mercer JA and Kennedy MB: Identification of proteins in the postsynaptic density fraction by mass spectrometry. J Neurosci. 20:4069–4080. 2000. View Article : Google Scholar : PubMed/NCBI

29 

Barr AM, Young CE, Sawada K, Trimble WS, Phillips AG and Honer WG: Abnormalities of presynaptic protein CDCrel-1 in the striatum of rats reared in social isolation: Relevance to neural connectivity in schizophrenia. Eur J Neurosci. 20:303–307. 2004. View Article : Google Scholar : PubMed/NCBI

30 

Ageta-Ishihara N, Yamakado H, Morita T, Hattori S, Takao K, Miyakawa T, Takahashi R and Kinoshita M: Chronic overload of SEPT4, a parkin substrate that aggregates in Parkinson's disease, causes behavioral alterations but not neurodegeneration in mice. Mol Brain. 6:352013. View Article : Google Scholar : PubMed/NCBI

31 

Dong Z, Ferger B, Paterna JC, Vogel D, Furler S, Osinde M, Feldon J and Büeler H: Dopamine-dependent neurodegeneration in rats induced by viral vector-mediated overexpression of the parkin target protein, CDCrel-1. Proc Natl Acad Sci USA. 100:12438–12443. 2003. View Article : Google Scholar : PubMed/NCBI

32 

Gozal YM, Seyfried NT, Gearing M, Glass JD, Heilman CJ, Wuu J, Duong DM, Cheng D, Xia Q, Rees HD, et al: Aberrant septin 11 is associated with sporadic frontotemporal lobar degeneration. Mol Neurodegener. 6:822011. View Article : Google Scholar : PubMed/NCBI

33 

Ihara M, Yamasaki N, Hagiwara A, Tanigaki A, Kitano A, Hikawa R, Tomimoto H, Noda M, Takanashi M, Mori H, et al: Sept4, a component of presynaptic scaffold and Lewy bodies, is required for the suppression of alpha-synuclein neurotoxicity. Neuron. 53:519–533. 2007. View Article : Google Scholar : PubMed/NCBI

34 

Pissuti Damalio JC, Garcia W, Alves Macedo JN, De Almeida Marques I, Andreu JM, Giraldo R, Garratt RC and Ulian Araújo AP: Self-assembly of human septin 2 into amyloid filaments. Biochimie. 94:628–636. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Zhang Y, Gao J, Chung KK, Huang H, Dawson VL and Dawson TM: Parkin functions as an E2-dependent ubiquitin-protein ligase and promotes the degradation of the synaptic vesicle-associated protein, CDCrel-1. Proc Natl Acad Sci USA. 97:13354–13359. 2000. View Article : Google Scholar : PubMed/NCBI

36 

Crunkhorn S: Neurodegenerative disorders: Ataxin 2 reduction rescues motor defects. Nat Rev Drug Discov. 16:384–385. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Schmidt EE, Pelz O, Buhlmann S, Kerr G, Horn T and Boutros M: RNAi: A database for cell-based and in vivo RNAi phenotypes, 2013 update. Nucleic Acids Res. 41:D1021–D1026. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Stayton MM, Rudolph FB and Fromm HJ: Regulation, genetics, and properties of adenylosuccinate synthetase. Curr Top Cell Regul. 22:103–141. 1983. View Article : Google Scholar : PubMed/NCBI

39 

Borza T, Iancu CV, Pike E, Honzatko RB and Fromm HJ: Variations in the response of mouse isozymes of adenylosuccinate synthetase to inhibitors of physiological relevance. J Biol Chem. 278:6673–6679. 2003. View Article : Google Scholar : PubMed/NCBI

40 

Oshchepkova-Nedosekina EA and Likhoshvai VA: A mathematical model for the adenylosuccinate synthetase reaction involved in purine biosynthesis. Theor Biol Med Model. 4:112007. View Article : Google Scholar : PubMed/NCBI

41 

Nomura Y, Nozawa A and Tozawa Y: Biochemical analyses of ppGpp effect on adenylosuccinate synthetases, key enzymes in purine biosynthesis in rice. Biosci Biotechnol Biochem. 78:1022–1025. 2014. View Article : Google Scholar : PubMed/NCBI

42 

Sowell RA, Owen JB and Butterfield DA: Proteomics in animal models of Alzheimer's and Parkinson's diseases. Ageing Res Rev. 8:1–17. 2009. View Article : Google Scholar : PubMed/NCBI

43 

Jin J, Meredith GE, Chen L, Zhou Y, Xu J, Shie FS, Lockhart P and Zhang J: Quantitative proteomic analysis of mitochondrial proteins: Relevance to Lewy body formation and Parkinson's disease. Brain Res. Mol. Brain Res. 134:119–138. 2005.

44 

Wan W, Xia S, Kalionis B, Liu L and Li Y: The role of Wnt signaling in the development of Alzheimer's disease: A potential therapeutic target? Biomed Res Int. 2014:3015752014. View Article : Google Scholar : PubMed/NCBI

45 

Zhang T, Shen X, Liu R, Zhu G, Bishop J and Xing M: Epigenetically upregulated WIPF1 plays a major role in BRAF V600E-promoted papillary thyroid cancer aggressiveness. Oncotarget. 8:900–914. 2017.PubMed/NCBI

46 

Pestalozzi BC, Peterson HF, Gelber RD, Goldhirsch A, Gusterson BA, Trihia H, Lindtner J, Cortés-Funes H, Simmoncini E, Byrne MJ, et al: Prognostic importance of thymidylate synthase expression in early breast cancer. J Clin Oncol. 15:1923–1931. 1997. View Article : Google Scholar : PubMed/NCBI

47 

Johnston PG, Lenz HJ, Leichman CG, Danenberg KD, Allegra CJ, Danenberg PV and Leichman L: Thymidylate synthase gene and protein expression correlate and are associated with response to 5-fluorouracil in human colorectal and gastric tumors. Cancer Res. 55:1407–1412. 1995.PubMed/NCBI

48 

Nimmagadda S and Shields AF: The role of DNA synthesis imaging in cancer in the era of targeted therapeutics. Cancer Metastasis Rev. 27:575–587. 2008. View Article : Google Scholar : PubMed/NCBI

49 

Ranganathan S and Tew KD: Analysis of Glyoxalase-I from normal and tumor tissue from human colon. Biochim Biophys Acta. 1182:311–316. 1993. View Article : Google Scholar : PubMed/NCBI

50 

Ranganathan S, Walsh ES and Tew KD: Glyoxalase I in detoxification: Studies using glyoxalase I transfectant cell line. Biochem J. 309:127–131. 1995. View Article : Google Scholar : PubMed/NCBI

51 

Rulli A, Carli L, Romani R, Baroni T, Giovannini E, Rosi G and Talesa V: Expression of glyoxalase I and II in normal and breast cancer tissues. Breast Cancer Res Treat. 66:67–72. 2001. View Article : Google Scholar : PubMed/NCBI

52 

Jones MB, Krutzsch H, Shu H, Zhao Y, Liotta LA, Kohn EC and Petricoin EF III: Proteomic analysis and identification of new biomarkers and therapeutic targets for invasive ovarian cancer. Proteomics. 2:76–84. 2002. View Article : Google Scholar : PubMed/NCBI

53 

Di Ilio C, Angelucci S, Pennelli A, Zezza A, Tenaglia R and Sacchetta P: Glyoxalase activities in tumor and non-tumor human urogenital tissues. Cancer Lett. 96:189–193. 1995. View Article : Google Scholar : PubMed/NCBI

54 

Davidson SD, Cherry JP, Choudhury MS, Tazaki H, Mallouh C and Konno S: Glyoxalase I activity in human prostate cancer: A potential marker and importance in chemotherapy. J Urol. 161:690–691. 1999. View Article : Google Scholar : PubMed/NCBI

55 

Fonseca-Sánchez MA, Rodríguez-Cuevas S, Mendoza-Hernández G, Bautista-Piña V, Arechaga Ocampo E, Hidalgo Miranda A, Quintanar Jurado V, Marchat LA, Alvarez-Sánchez E, Pérez Plasencia C and López-Camarillo C: Breast cancer proteomics reveals a positive correlation between glyoxalase 1 expression and high tumor grade. Int J Oncol. 41:670–680. 2012. View Article : Google Scholar : PubMed/NCBI

56 

Ledesma MD, Bonay P and Avila J: Tau protein from Alzheimer's disease patients is glycated at its tubulin-binding domain. J Neurochem. 65:1658–1664. 1995. View Article : Google Scholar : PubMed/NCBI

57 

Vitek MP, Bhattacharya K, Glendening JM, Stopa E, Vlassara H, Bucala R, Manogue K and Cerami A: Advanced glycation end products contribute to amyloidosis in Alzheimer disease. Proc Natl Acad Sci USA. 91:4766–4770. 1994. View Article : Google Scholar : PubMed/NCBI

58 

Angeloni C, Zambonin L and Hrelia S: Role of methylglyoxal in Alzheimer's disease. Biomed Res Int. 2014:2384852014. View Article : Google Scholar : PubMed/NCBI

59 

Al-Balas QA, Hassan MA, Al-Shar'i NA, Mhaidat NM, Almaaytah AM, Al-Mahasneh FM and Isawi IH: Novel glyoxalase-I inhibitors possessing a ‘zinc-binding feature’ as potential anticancer agents. Drug Des Devel Ther. 10:2623–2629. 2016. View Article : Google Scholar : PubMed/NCBI

60 

Thornalley PJ: Methylglyoxal, glyoxalases and the development of diabetic complications. Amino Acids. 6:15–23. 1994. View Article : Google Scholar : PubMed/NCBI

61 

Chaudhuri J, Bose N, Gong J, Hall D, Rifkind A, Bhaumik D, Peiris TH, Chamoli M, Le CH, Liu J, et al: A Caenorhabditis elegans model elucidates a conserved role for TRPA1-Nrf signaling in reactive α-Dicarbonyl detoxification. Curr Biol. 26:3014–3025. 2016. View Article : Google Scholar : PubMed/NCBI

62 

Schulze-Osthoff K, Ferrari D, Riehemann K and Wesselborg S: Regulation of NF-kappa B activation by MAP kinase cascades. Immunobiology. 198:35–49. 1997. View Article : Google Scholar : PubMed/NCBI

63 

Brand F, Schumacher S, Kant S, Menon MB, Simon R, Turgeon B, Britsch S, Meloche S, Gaestel M and Kotlyarov A: The extracellular signal-regulated kinase 3 (mitogen-activated protein kinase 6 [MAPK6])-MAPK-activated protein kinase 5 signaling complex regulates septin function and dendrite morphology. Mol Cell Biol. 32:2467–2478. 2012. View Article : Google Scholar : PubMed/NCBI

64 

Shukla JP, Deshpande G and Shashidhara LS: Ataxin 2-binding protein 1 is a context-specific positive regulator of Notch signaling during neurogenesis in Drosophila melanogaster. Development. 144:905–915. 2017. View Article : Google Scholar : PubMed/NCBI

65 

Shin HM, Minter LM, Cho OH, Gottipati S, Fauq AH, Golde TE, Sonenshein GE and Osborne BA: Notch1 augments NF-kappaB activity by facilitating its nuclear retention. EMBO J. 25:129–138. 2006. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2019
Volume 20 Issue 2

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
Mendonca P, Taka E and Soliman KF: Proteomic analysis of the effect of the polyphenol pentagalloyl glucose on proteins involved in neurodegenerative diseases in activated BV‑2 microglial cells. Mol Med Rep 20: 1736-1746, 2019
APA
Mendonca, P., Taka, E., & Soliman, K.F. (2019). Proteomic analysis of the effect of the polyphenol pentagalloyl glucose on proteins involved in neurodegenerative diseases in activated BV‑2 microglial cells. Molecular Medicine Reports, 20, 1736-1746. https://doi.org/10.3892/mmr.2019.10400
MLA
Mendonca, P., Taka, E., Soliman, K. F."Proteomic analysis of the effect of the polyphenol pentagalloyl glucose on proteins involved in neurodegenerative diseases in activated BV‑2 microglial cells". Molecular Medicine Reports 20.2 (2019): 1736-1746.
Chicago
Mendonca, P., Taka, E., Soliman, K. F."Proteomic analysis of the effect of the polyphenol pentagalloyl glucose on proteins involved in neurodegenerative diseases in activated BV‑2 microglial cells". Molecular Medicine Reports 20, no. 2 (2019): 1736-1746. https://doi.org/10.3892/mmr.2019.10400