2023 Impact Factor
In the present study, we investigated the anti-inflammatory properties of
Microglia are immune cells resident in the central nervous system (CNS) that respond to extracellular stimuli and play a crucial role in the progress of neuro-inflammation (Rock
Inflammation is the organized response of an organism to injuries of multiple pathologies. The inflammatory response involves the rapid up-regulation of many genes. Nuclear factor-kappa B (NF-κB) is a critical transcription factor involved in the inflammatory response, and is also known to play an important role in neurodegenerative diseases (O’Neill and Kaltschmidt, 1997). NF-κB is normally bound to inhibitor of nuclear factor kappa B alpha (IκBα) in the cytosol in an inactive form. However, in response to stress, phosphorylated IκBα is degraded through selective ubiquitination, resulting in NF-κB activation. Activated NF-κB then translocates into the nucleus and binds to the promoter regions of pro-inflammatory molecules, thereby upregulating the expression of multiple target genes (Nomura, 2001). Ultimately, NF-κB induces the expression of many inflammatory mediators, including inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), and TNF-α, via the binding of NF-κB to specific promoter regions. Therefore, the modulation of NF-κB activation is a promising strategy for the treatment of many neuropathologies (Baima
Nuclear factor erythroid 2-related factor 2 (Nrf2) is a redox-sensitive transcription factor that is normally sequestered in the cytoplasm by the regulatory protein Kelch-like ECH-associated protein 1 (Keap1). Under conditions of oxidative or xenobiotic stress, Keap1 undergoes structural modification. Nrf2 is then released from Keap1, translocates to the nucleus, and subsequently binds to a promoter sequence known as the antioxidant response element (ARE). Nuclear accumulation of Nrf2 thus results in the up-regulation of phase II detoxifying anti-oxidant enzymes such as NAD(P)H, quinone oxidoreductase, heme oxygenase-1 (HO-1), the glutamate-cysteine ligase catalytic subunit, and the glutamate-cysteine ligase modifier subunit (Kensler
In the present study, we investigated whether EUE exerts anti-inflammatory effects. Specifically, we studied the mechanisms of action of EUE on LPS-stimulated pro-inflammatory responses in BV-2 microglial cells. We investigated whether EUE inhibits NF-κB activation and whether EUE activates Nrf2-dependent signaling, which leads to HO-1 up-regulation in an event that is downstream of ARE-responsive proteins. We also determined whether EUE-induced activation of Nrf2-dependent HO-1 signaling is the major cellular mechanism mediating the anti-inflammatory effects of EUE.
2,7′-Dichlorofluorescin diacetate (DCFH-DA), dimethyl sulfoxide (DMSO), Hoechst 33258, 3-(4,5-dimethyl thiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT), lipopolysaccharide (LPS,
The dried stem bark of
BV-2 microglial cells were grown in DMEM supplemented with 10% heat-inactivated FBS (v/v) and 0.1% penicillin/streptomycin (v/v) in a humidified atmosphere of 5% CO2 and 95% air at 37°C. LPS was prepared immediately before use as a 10 μg/ml stock and diluted in PBS to the indicated final concentration, while EUE was dissolved in DMSO. All stock solutions were added directly to the culture medium. Control cells were treated with DMSO only. The final concentration of solvent was always <0.1% (v/v). No significant cytotoxicity was observed in any of the experiments (data not shown). In all experiments, cells were treated with the indicated concentrations of EUE in the presence or absence of LPS (100 ng/ml) in serum-free DMEM.
Cell viability was measured based on the formation of formazan, a blue product resulting from the metabolism of a colorless substrate (MTT) by mitochondrial dehydrogenases, which are active only in live cells. BV-2 microglial cells (2.5×105 cells/well in 24-well plates) were incubated at 37°C with LPS for 24 h with or without EUE pretreatment and then treated with an MTT solution (5 mg/ml) for 2 h. The dark blue formazan crystals formed in intact cells were dissolved in DMSO and the resultant absorbances were measured at 540 nm with a microplate reader (SpectraMax 250, Molecular Device, Sunnyvale, CA, USA). Results are expressed as the percentage of metabolized MTT relative to that of control cells as determined by absorbance measurements.
NO release into culture supernatants was measured by the Griess reaction. In brief, BV-2 microglial cells (2.5×105 cells/well in 24-well plates) were incubated at 37°C with LPS for 24 h with or without EUE pretreatment. 100 μl of culture supernatant from each sample was then mixed with an equal volume of Griess reagent [0.1% N-(1-naphthyl)-ethylenediamine dihydrochloride and 1% sulfanilamide in 5% phosphoric acid] in 96-well plates for 10 min at room temperature in the dark. Nitrite concentrations were determined by reference to standard solutions of sodium nitrite prepared in cell culture medium. The absorbances at 540 nm were determined using a microplate reader.
The levels of PGE2, TNF-α, and IL-1β were determined using specific ELISA kits (PGE2 kit, Cayman Chemical, Ann Arbor, MI, USA; TNF-α and IL-1β kits, KOMA Biotech, Seoul, Korea) as per the manufacturers’ instructions.
BV-2 microglial cells (1×106 cells/well) were seeded on 6-well plates or poly-D-lysine coated slides and cultured overnight. The levels of intracellular ROS were measured using DCFH-DA. After pretreatment with or without EUE for 30 min, the cells were incubated with LPS for 24 h. The cells were rinsed with PBS and treated with 10 μM DCFH-DA (for 30 min) and 5 μg/ml Hoechst 33258 (for 5 min) at 37°C, washed twice with PBS, and the resultant fluorescence was measured at 530 nm with a microplate reader (SpectraMax M2, Molecular Device, Sunnyvale, CA, USA) with excitation at 488 nm. DCFH-DA fluorescence images were captured with a fluorescence microscope (20×).
BV-2 microglial cells (1×106 cells/well in 6-well plates) were incubated at 37°C with LPS, with or without pretreatment with EUE. Total RNA was isolated using Trizol® reagent (Invitrogen). Reverse transcription was carried out with a Superscript®-III kit (Invitrogen) using 5 μg total RNA and oligo dT according to the manufacturer’s instructions. Specific primer sequences were as follows: iNOS (forward, 5′-CTGCAGCACTTGGATCAGGAACCTG-3′ and reverse, 5′-GGGAGTAGCCTGTGTGCACCTGGAA-3′), COX-2 (forward, 5′-TTGAAGACCAGGAGTACAGC-3′ and reverse, 5′-GGTACAGTTCCATGACATCG-3′), TNF-α (forward, 5′-CGTCAGCCGATTTGCTATCT-3′ and reverse, 5′-CGGACTCCGCAAAGTCTAAG-3′), IL-1β (forward, 5′-GCCCATCCTCTGTGACTCAT-3′ and reverse, 5′-AGGCCACAGGTATTTTGTCG-3′), HO-1 (forward, 5′-CAAGCCGAGAATGCTGAGTTCATG-3′ and reverse, 5′-GCAAGGGATGATTTCCTGCCAG-3′), and β-actin (forward, 5′-AGCCATGTACGTAGCCATCC-3′ and reverse, 5′-GCTGTGGTGGTGAAGCTGTA-3′). PCR amplification of the resulting cDNA template was conducted for 25 (iNOS, COX-2, HO-1, and β-actin) or 30 (TNF-α and IL-1β) cycles. Briefly, after an initial denaturation step at 95°C for 2 min, thermal cycling was initiated. Each cycle consisted of denaturation at 95°C for 1 min, annealing at 65°C (iNOS), 55°C (COX-2), 57°C (TNF-α and IL-1β), 60°C (HO-1), or 58°C (β-actin) for 1 min, extension at 72°C for 1 min, and a final extension at 72°C for 5 min. PCR products were analyzed by staining with ethidium bromide after electrophoresis for 30 min at 100 V on 1.5% agarose gels in Tris borate/EDTA buffer (890 mM Tris base, 890 mM boric acid, 20 mM EDTA, pH 8.3). Amplicons were visualized on a UV transilluminator and their intensities quantified by densitometric analysis using ImageJ software (NIH Image, public domain, USA).
BV-2 microglial cells were seeded at 5×106 cells/well in 100 mm2 cell culture dishes. After pretreatment with EUE, the cells were incubated either with or without LPS. To evaluate the nuclear translocation of NF-κB p65, nuclear and cytosolic fractions were prepared using NE-PER nuclear and cytoplasmic extraction reagents for cultured cells (Pierce, Rockford, IL, USA) according to the manufacturer’s instructions. The levels of NF-κB p65 and Nrf2 were determined by Western blot analysis and electrophoretic mobility shift assays (EMSA) were performed as described below.
BV-2 microglial cells were seeded at 1×106 cells/well in 6-well plates. After pretreatment with EUE, the cells were incubated either with or without LPS. Cells were then washed with ice-cold PBS and harvested by scraping with 100 μl ice-cold TPER tissue protein extraction buffer (Thermo Scientific, Rockford, IL, USA) containing a protease and phosphatase inhibitor cocktail (Roche Diagnostics GmbH, Mannheim, Germany). Lysates were incubated on ice for 30 min. After centrifugation at 10,000×
NF-κB and ARE reporter constructs were purchased from SABiosiences (Qiagen, Valencia, CA, USA). In brief, BV-2 microglial cells were plated in 24-well plates at a density of 2.5×105 cells/well and grown overnight. Cells were cotransfected with 5 μg/ml of an NF-κB reporter plasmid, an ARE reporter plasmid, or a negative control plasmid along with an internal control plasmid to measure transfection efficiency. Transfections were performed for 6 h using lipofectamine. After transfection, cells were cultured in medium with 10% FBS for 24 h. Twenty-four hours after transfection, the cells were incubated with EUE and then treated either with or without LPS. Luciferase activity was assayed using a dual-luciferase assay kit (Promega, Madison, WI, USA) according to the manufacturer’s instructions. Luminescence was measured using a single tube luminometer (FB12, Berthold Detection Systems GmbH, Pforzheim, Germany).
Nuclear extracts were prepared using the NE-PER nuclear and cytoplasmic extraction reagents as described above. Synthetic complementary NF-κB-binding (5′-AGTTGAGGGG ACTTTCCCAGGC-3′) and Nrf2-binding (5′-GCTCTTCCGGTGCTCTTCCGGT-3′) oligonucleotides (Affimetrix Inc., CA, USA) were 5′-biotinylated using a biotin 5′-end DNA labeling EMSA kit (Affimetrix Inc.) according to the manufacturer’s protocol. The binding reactions contained 10 μg of nuclear extract proteins, binding buffer, 1 μg of poly d(I-C), and 10 ng of biotin-labeled DNA. The reactions were incubated for 5 min at room temperature in a final volume of 10 μl. The protein-DNA complexes were separated from the DNA probes by electrophoresis on a native 6% polyacrylamide gel that had been pre-electrophoresed for 1 h in 0.5× Tris borate/EDTA buffer (50 mM Tris base, 18 mM boric acid, 500 mM EDTA, pH 8.3). Complexes were then transferred to a positively charged nylon membrane (Pall Corporation, Pensacola, FL, USA) in 0.5× Tris borate/EDTA buffer at 300 mA for 30 min, after which the DNA was hybridized to the nylon membrane in a drying oven at 80°C for 1 h. Horseradish peroxidase-conjugated streptavidin was used according to the manufacturer’s instructions to detect the transferred DNA.
BV-2 microglial cells (5×105 cells/well) were seeded on poly-D-lysine coated culture slides and incubated for 24 h. After pretreatment with EUE, the cells were incubated either with or without LPS. Cells were then washed with PBS and fixed with 4% paraformaldehyde for 15 min. After washing, cells were permeabilized with 0.1% Triton X-100 in PBS for 10 min. Cells were blocked in 5% BSA/PBS for 1 h and then incubated overnight with anti-NF-κB p65 and anti-Nrf2 antibodies (1:250 each). Cells were then washed with PBS and incubated for 1 h with Texas Red®-conjugated goat anti-rabbit IgG antibodies (1:250 for NF-κB p65) or Alexa Fluor® 488-conjugated goat anti-rabbit IgG antibodies (1:250 for Nrf2) and Hoechst 33258 (5 μg/ml) for 5 min. Cells were washed in PBS and mounted on glass slides in Permafluor aqueous mounting medium. All procedures were performed at room temperature. Cells were observed under a fluorescence microscope (100×). The results shown are representative of three independent experiments.
Data were analyzed with Prism 5.0 software (GraphPad Software, Inc., San Diego, CA, USA) and are expressed as means ± S.E.M. Statistical analyses were performed using one-way analysis of variance (ANOVA) followed by the Newman-Keuls test. Statistical significance was set at
To exclude the possibility that the decreases in NO and cytokine levels were simply due to cell death, the MTT assay was used to assess the cytotoxicity of EUE in BV-2 microglial cells in the absence or presence of LPS. EUE was not cytotoxic at any of the concentrations (2.5, 5, 10, 25, 50, and 100 μg/ml) used in this study (Fig. 1A, 1B). Thus, these concentrations of EUE were used in subsequent experiments.
We initially evaluated the effects of EUE on NO and PGE2 production in LPS-stimulated BV-2 microglial cells. NO production was assessed by measuring the amount of nitrite released into the culture medium using the Griess reagent. Treatment with LPS significantly increased NO and PGE2 production by 37.63 ± 0.51 μM and 564.60 ± 59.07 pg/ml more than the control values, respectively (Fig. 1C, 1D,
Since EUE inhibited the production of NO and PGE2, we next examined the relationship between the concentration of EUE and the expression of iNOS and COX-2. Western blot analysis showed that LPS treatment significantly increased the protein levels of iNOS and COX-2 to 430.60 ± 32.22% and 160.50 ± 3.80% of the control values, respectively (Fig. 2A, 2B,
Treatment with LPS also significantly increased the mRNA expression levels of iNOS and COX-2 to 296.00 ± 34.47% and 258.20 ± 4.01% of the control values, respectively (Fig. 2C, 2D,
We next investigated whether EUE inhibits LPS-induced production of TNF-α and IL-1β by RT-PCR and ELISA. Treatment with LPS significantly increased the mRNA levels of these pro-inflammatory cytokines to 361.70 ± 5.59% and 174.40 ± 23.46% of the control values, respectively (Fig. 3A, 3B,
As a complementary approach, we also performed ELISAs to determine whether EUE inhibits the release of these cytokines
We next examined the effect of EUE on the production of ROS, which are known to be early signaling inducers in microglial inflammation that contribute to neuronal cell death and neurodegeneration. Intracellular ROS formation was investigated with DCFH-DA, a fluorescent ROS-sensitive probe. Treatment with LPS significantly increased intracellular ROS production to 222.70 ± 3.19% of the control value (Fig. 4A,
To evaluate the effects of EUE on the upstream signaling pathways associated with NF-κB translocation, we examined changes in the activation of intracellular signaling proteins such as JNK, ERK 1/2, p38 MAPKs, PI3K/Akt, and GSK-3β in BV-2 microglial cells. As shown in Fig. 5, treatment with LPS rapidly increased the phosphorylation of JNK, p38 MAPKs, ERK 1/2, PI3K/Akt, and GSK-3β to 649.90 ± 47.70%, 207.50 ± 5.44%, 575.70 ± 69.37%, 215.40 ± 14.56%, and 274.90 ± 10.35% of the control values, respectively (Fig. 5,
The pro-inflammatory response and cytokine production are tightly regulated by multiple signaling molecules, such as IκBα. We thus evaluated the cytoplasmic levels of IκBα after LPS stimulation and EUE-mediated inhibition by Western blotting. Treatment with LPS significantly decreased degradation of IκBα to 32.57 ± 9.43% of the control value (Fig. 6A,
Given the inhibitory effects of EUE on LPS-induced nuclear translocation of NF-κB, we measured NF-κB-driven transcription using a luciferase reporter assay. In this assay, BV-2 microglial cells were transfected with an NF-κB reporter construct and its activity was assessed after various treatments. As shown in Fig. 6F, treatment with LPS significantly increased NF-κB activation to 405.30 ± 18.61% of the control value (
We next examined the effects of EUE on HO-1 expression, which is directly linked to Nrf2-dependent activation. As shown in Fig. 7A, 7B, treatment with EUE significantly induced the nuclear translocation of Nrf2 in a time-dependent (Fig. 7A) and concentration-dependent (Fig. 7B) manner. The levels of nuclear Nrf2 were significantly increased 2 h after treatment with 100 μg/ml EUE, peaked at 6 h, and then decreased at 12 h. Thus, treatment with 100 μg/ml EUE for 6 h was used to induce the nuclear translocation of Nrf2 in all subsequent experiments. Immunocytochemical analysis clearly revealed Nrf2 accumulation in the nuclei of EUE-treated BV-2 microglial cells (Fig. 7C). To investigate whether ARE binding by Nrf2 plays a role in EUE-mediated induction of HO-1 expression, cells were transfected with luciferase reporters under the control of the ARE promoter. As shown in Fig. 7D, EUE potently induced ARE-mediated transcription of luciferase in BV-2 microglial cells transfected with the ARE reporter construct. Moreover, ARE-driven transcription was significantly increased by treatment with 25, 50, and 100 μg/ml EUE to 176.70 ± 4.44%, 214.80 ± 12.85%, and 359.30 ± 11.64% of the control value, respectively (
To determine whether the observed anti-neuroinflammatory effects of EUE were accompanied by activation of the Nrf2-ARE pathway in BV-2 microglial cells, we analyzed the protein and mRNA levels of HO-1, a downstream target of Nrf2, by Western blotting and RT-PCR. Treatment with EUE resulted in a time-dependent (Fig. 8A, 8C) and concentration-dependent (Fig. 8B, 8D) increase in the levels of HO-1 protein and mRNA. Both the mRNA and protein levels of HO-1 were significantly increased at 2 h after treatment with 100 μg/ml EUE and peaked at 6 h and 12 h, respectively. We next investigated whether HO-1 is the downstream target of Nrf2 that mediates the anti-inflammatory activity of EUE against LPS-stimulated inflammatory responses in BV-2 microglial cells. To this end, we tested the effects of ZnPP, an HO-1-specific inhibitor, on the anti-inflammatory effects of EUE. As shown in Fig. 8E and 8F, treatment with LPS significantly increased the levels of secreted NO and PGE2 by 33.42 ± 0.48 μM and 564.60 ± 59.07 pg/ml more than the control values, respectively (
The main finding of this study is that EUE potently inhibits pro-inflammatory responses in microglia, most likely by inhibiting NF-κB activation and thus preventing Nrf2-mediated up-regulation of HO-1 expression. To the best of our knowledge, this is the first report to demonstrate that NF-κB activation and Nrf2-dependent HO-1 expression mediate the anti-neuroinflammatory effects of EUE. It is also the first report to identify the molecular mechanisms by which EUE modulates pro-inflammatory molecule production in BV-2 microglial cells.
NO and PGE2 are important mediators of inflammation. These two molecules are thought to be responsible for some of the damage that is seen in the CNS in brain injuries and diseases such as AD, PD, and stroke.(Minghetti and Levi, 1998) NO is synthesized from L-arginine by NO synthase, which is comprised of three enzymes. One of these enzymes is iNOS, which is produced mainly by activated glial cells (Lee
Pro-inflammatory cytokines such as TNF-α and IL-1β have been shown to promote neuronal injury by activated microglia (Block
Elevated ROS levels have been found to be associated with neurodegenerative diseases such as AD, PD, and stroke (Block
Various intracellular signaling pathways are involved in mediating the inflammatory response. Many signaling proteins such as MAPKs, PI3K/Akt, and GSK-3β are phosphorylated in response to LPS treatment. These proteins regulate the pro-inflammatory response and cytokine production in microglial cells (Kaminska, 2005; Wang
Activation of NF-κB in microglia contributes to neuronal injury and promotes the development of neurodegenerative diseases such as AD, PD, and stroke (Mattson, 2005). NF-κB is also a central regulator of microglial responses to various stimuli such as LPS and cytokines (O’Neill and Kaltschmidt, 1997). The mechanism by which NF-κB acts is well characterized: NF-κB is normally inactive when bound to IκBα in the cytoplasm in microglial cells. However, in response to stress, phosphorylated IκBα is degraded through selective ubiquitination, resulting in the activation of NF-κB. Activated NF-κB then translocates to the nucleus and binds to the promoter regions of pro-inflammatory genes, thereby upregulating the expression of these molecules (Nomura, 2001). Therefore, inhibition of these signaling pathways may explain the potent EUE-mediated suppression of the inflammatory response observed in the present study. We found that EUE inhibited LPS-induced phosphorylation of IκBα, degradation of IκBα, and nuclear translocation of NF-κB in BV-2 microglial cells. Furthermore, EUE also significantly repressed DNA binding by NF-κB and NF-κB-driven transcription upon LPS-mediated activation of microglial cells. These findings suggest that EUE-mediated down-regulation of inflammatory mediators results from the inhibition of NF-κB signaling pathways, ultimately resulting in anti-neuro-inflammatory effects.
Nrf2 is expressed in most cell types of the brain, including microglia. Activation of Nrf2 in microglia following brain injury is known to play a role in inhibiting microglial hyperactivation and in preventing neuronal death caused by microgliosis. Moreover, recent studies have reported that ROS are generated in response to inflammatory signals and participate in microglial activation (Fremond
HO-1 is an enzyme with potent anti-inflammatory and anti-oxidant effects. A large body of evidence suggests that HO-1 plays a key role in maintaining anti-oxidant homeostasis during cellular stress (Maines, 1988). The induction of HO-1 expression is primarily regulated at the transcriptional level by various transcription factors and is also linked to Nrf2. In the light of growing evidence indicating that HO-1 provides neuroprotection, elevating HO-1 expression by a pharmacologic modulator may represent a valid strategy for therapeutic intervention. In particular, the identification of a non-cytotoxic inducer of HO-1 expression could enhance the anti-oxidant potential of cells. Therefore, since HO-1 is an important component of cellular defenses against the inflammatory response, we assessed whether EUE could induce HO-1 expression to strengthen the anti-inflammatory response. We found that EUE upregulates HO-1 expression via Nrf2-mediated ARE activation. Moreover, many studies have suggested that HO-1 plays a pivotal protective role in inflammatory responses due to its ability to inhibit pro-inflammatory responses such as NO production (Takagi
Considering the current literature and the data reported here, inhibition of pro-inflammatory responses mediated by EUE appears to be associated with suppression of NF-κB and activation of HO-1/Nrf2, which are signaling molecules involved in neuro-inflammation. The primary compounds from EUE reported to be responsible for various pharmacological actions, particularly chlorogenic acid (CGA), were also evaluated previously for neuroprotective effects against oxidative stress-induced cell death in neuronal cells (Kwon
In summary, the present study showed the potential anti-inflammatory effects of EUE in BV-2 microglial cells. In LPS-stimulated BV-2 microglial cells, EUE significantly inhibited the production of two inflammatory mediators, NO and PGE2, and suppressed the expression and release of multiple molecules involved in inflammation, including iNOS, COX-2, TNF-α, and IL-1β. EUE also significantly attenuated intracellular ROS accumulation in LPS-stimulated BV-2 microglial cells. These inhibitory effects of EUE were associated with reduced phosphorylation of MAPKs, PI3K/Akt, and GSK-3β. They were also associated with reduced activation of NF-κB. In addition, EUE induces HO-1 expression in BV-2 microglial cells, which confers protection against the LPS-induced inflammatory response. EUE also induces Nrf2 nuclear translocation, which occurs upstream of EUE-induced HO-1 expression. Although we did not evaluate whether EUE protects against inflammation-related neuronal damage
This research was supported by a grant from the Basic Science Research Program through the National Research Foundation of Korea (NRF-2012R1A5A2A28671860 and NRF-2011-00503), which is funded by the Ministry of Education, Science and Technology, Republic of Korea.
The authors have no conflicts of interest to declare.