Open Access

Luteolin inhibits SH-SY5Y cell apoptosis through suppression of the nuclear transcription factor-κB, mitogen‑activated protein kinase and protein kinase B pathways in lipopolysaccharide-stimulated cocultured BV2 cells

  • Authors:
    • Lihong Zhu
    • Wei Bi
    • Dan Lu
    • Chanjuan Zhang
    • Xiaoming Shu
    • Daxiang Lu
  • View Affiliations

  • Published online on: February 20, 2014     https://doi.org/10.3892/etm.2014.1564
  • Pages: 1065-1070
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Microglial activation is one of the causative factors for neuroinflammation, which is associated with the pathophysiology of neurodegenerative diseases. Our previous study showed that the flavonoid luteolin inhibited several pro-inflammatory enzymes and pro-inflammatory cytokines that are induced by activated microglia; however, its effect on signaling pathways is currently unknown. The present study examined the effects of luteolin on signaling pathways stimulated by lipopolysaccharide (LPS), including Toll-like receptor-4 (TLR-4), nuclear transcription factor-κB (NF-κB), mitogen-activated protein kinase (MAPK) family and protein kinase B (Akt) pathways in murine microglial BV2 cells. In addition, BV2 microglia and SH-SY5Y neuroblastoma cells were cocultured to observe the indirect neuroprotective effects of luteolin. Luteolin inhibited the LPS-stimulated expression of TLR-4. In addition, luteolin blocked LPS-induced NF-κB, p38, JNK and Akt activation, but had no effect on ERK. When SH-SY5Y cells were cocultured with LPS-stimulated BV2 microglia, pretreatment with luteolin increased neuronal viability and reduced the number of apoptotic cells. These data suggest that luteolin has a beneficial effect on neuroinflammatory events in neurodegenerative diseases via suppression of the NF-κB, MAPK and Akt pathways in activated microglial cells.

Introduction

Microglia are the cells responsible for innate immunity in the central nervous system and accumulating evidence suggests that activated microglia modulate the development and/or progression of Alzheimer’s disease, Parkinson’s disease and amyotrophic lateral sclerosis, among other diseases (13). Exposure to lipopolysaccharide (LPS), β-amyloid or interferon (IFN)-γ activates microglia, inducing the secretion of a variety of pro-inflammatory mediators and potentially neurotoxic compounds (4,5). Receptor binding of cytokines stimulates a variety of intracellular signaling pathways that have been implicated in neurodegenerative disorders, including activation of nuclear transcription factor-κB (NF-κB), the mitogen-activated protein kinase (MAPK) family and protein kinase B (Akt). These are the most important molecules that control the synthesis and release of pro-inflammatory substances from activated microglia (68). Epidemiological studies suggest that inhibition of microglial activation attenuates the severity of neurotoxicity (9,10). The inhibition of activated microglia is therefore an important therapeutic target for neurodegenerative disorders.

Luteolin (3′,4′,5,7-tetrahydroxyflavone), a flavone that has been identified in celery, green pepper, perilla leaves and seeds, and at high concentrations in chamomile, exhibits strong anti-inflammatory, antioxidant and free-radical scavenging properties. In addition, luteolin has been shown to inhibit the LPS-induced production of tumor necrosis factor alpha (TNF-α) and nitric oxide (NO) in an activated macrophage-like cell line (11). Luteolin also reduces the production of LPS-induced pro-inflammatory cytokines in intestinal epithelial cells, mouse bone marrow-derived dendritic cells (12), rat fibroblasts (13) and human gingival fibroblasts (14). Furthermore, in a previous study we observed that luteolin inhibited the secretion of several pro-inflammatory enzymes and pro-inflammatory cytokines by activated microglia (15). However, the mechanism by which luteolin inhibits microglial inflammation is not completely understood. Much less is known about the role of luteolin in neuroprotection and regulation of the underlying signaling pathways.

In the present study, the effects of luteolin on Toll-like receptor-4 (TLR-4) expression and the NF-κB, MAPK and Akt signaling pathways were investigated using LPS-stimulated BV2 cells, a murine microglial cell line. In further experiments using a microglial-neuronal coculture system, the protective effects of luteolin against microglial-mediated LPS neurotoxicity, and therefore its potential role in the prevention of neurodegenerative diseases were investigated.

Materials and methods

Materials

Luteolin (purity > 98%; molecular weight, 286.24; chemical formula C15H10O6), LPS, dimethylsulfoxide (DMSO) and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) were purchased from Sigma (St. Louis, MO, USA). Antibodies against NF-κB p65, p38, phosphorylated p38 (p-p38), JNK, phosphorylated JNK (p-JNK), ERK, phosphorylated ERK (p-ERK), Akt and phosphorylated Akt (p-Akt) were obtained from Cell Signaling Technology (Beverly, MA, USA). Antibodies against TLR-4 were obtained from Santa Cruz Biotechnology (Santa Cruz, CA, USA). Mouse anti-β-actin antibody was purchased from Sigma.

Cell culture

BV2 immortalized murine microglia were provided by the Cell Culture Center of the Chinese Academy of Medical Sciences (Beijing, China). The human neuroblastoma cell line SH-SY5Y was donated by Dr Enxiang Tao. The cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% fetal bovine serum (FBS), 100 U/ml penicillin and 100 μg/ml streptomycin in a humidified atmosphere of 5% CO2 at 37°C. In all experiments, the BV2 microglia were pretreated with the indicated concentrations of luteolin for 1 h prior to the addition of LPS (1.0 μg/ml) in serum-free DMEM.

Immunofluorescence staining

For immunofluorescence staining, the cells were fixed with 4% paraformaldehyde for 15 min, permeabilized with 0.1% Triton X-100 for 10 min and blocked with 5% bovine serum albumin (BSA) for 30 min. The cells were then incubated with primary antibody to NF-κB p65 (1:100 dilution) overnight at 4°C. After washing three times with PBS, the cells were incubated with secondary antibody conjugated to rhodamine for 1 h (Cell Signaling Technology). The nuclei were stained with Hoechst 33258. Fluorescent images were captured using a laser scanning confocal microscope (LSM 510 META; Carl Zeiss, Stuttgart, Germany).

Total RNA isolation and quantitative PCR (qPCR) analysis

Total RNA was isolated with TRIzol reagent (Invitrogen Life Technologies, Carlsbad, CA, USA) according to the manufacturer’s instructions. Total RNA (1.0 μg) was reverse transcribed using M-MLV reverse transcriptase (Promega, Madison, WI, USA) to produce cDNA. The primers used for qPCR were as follows: TLR-4, 5′-GCT TTC ACC TCT GCC TTC AC-3′ and 5′-CCA ACG GCT CTG AAT AAA GTG-3′; and GAPDH, 5′-TCA CCA CCA TGG AGA AGG C-3′ and 5′-GCT AAG CAG TTG GTG GTG CA-3′. The following qPCR conditions were used: 40 cycles of denaturation at 94°C for 20 sec, annealing at 62°C for 30 sec and extension at 72°C for 30 sec. SYBR Green qPCR Master mix 2 (Takara Bio, Inc., Shiga, Japan) was used in all samples and the reactions were carried out in a 20-μl reaction volume using a LightCycler LC480 qPCR instrument (Roche Diagnostics, Basel, Switzerland). The mRNA expression levels of target genes relative to glyceraldehyde-3-phosphate dehydrogenase (GAPDH, a housekeeping gene used as an endogenous control) were calculated according to the standard curves.

Western blot analysis

BV2 microglia were harvested and lysed in RIPA buffer [1 mM ethylenediaminetetraacetic acid (EDTA), 150 mM NaCl, 1% igepal (CA-630), 0.1% sodium dodecyl sulfate (SDS), 0.5 % sodium deoxycholate and 50 mM Tris HCl; pH 8.0 (Sigma)]. Equal amounts of protein were separated by 8–12% sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE), transferred to polyvinylidene fluoride (PVDF) membranes, blocked with 5% nonfat milk for 2 h and incubated with primary antibodies at 4°C overnight. Following incubation with appropriate secondary antibodies conjugated to horseradish peroxidase (goat anti-rabbit secondary antibody was obtained from Cell Signaling Technology), immunoblots were exposed on film using electrochemiluminescence (ECL) western detection reagent (Amersham Pharmacia Biotech, Amersham, UK). The bands were quantified by the optical density ratio using β-actin as a control.

Cytotoxicity assay in a coculture of microglia and neurons

SH-SY5Y cells were grown in the bottom of wells, BV2 cells were then grown in culture inserts (pore size 0.4 μm; Corning, New York, NY, USA) and 1.0 μg/ml LPS was added to the culture insert. In this coculture system, the microglia were able to communicate with the neurons through a semipermeable membrane, which avoids direct contact between the two cellular systems (16). After coculture for 24 h, the SH-SY5Y cells were incubated with MTT solution (0.5 mg/ml in PBS) for 4 h at 37°C. The culture supernatants were then removed, the resulting formazan crystals were dissolved in DMSO and the absorbance was read at 570 nm with a microplate reader (ReTiSoft Inc., Mannedorf, Switzerland). Cell survival was expressed as the ratio of absorbance (percentage survival) compared with a DMSO control.

Detection of apoptosis in a coculture of microglia and neurons

In the coculture system described above, apoptotic SH-SY5Y cells were detected by the terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) assay. Following each treatment, the TUNEL assay was performed according to manufacturer’s instructions (Roche Diagnostics Corporation, Indianapolis, IN, USA) and all nuclei were counterstained with 5 mg/ml Hoechst 33342 for 10 min at 37°C. The labeled SH-SY5Y cells were examined with a laser scanning confocal microscope. SH-SY5Y cells were considered to be apoptotic when their nuclei were costained with Hoechst 33342 and TUNEL. The number of apoptotic cells was counted among 100 randomly chosen neurons observed on several optic fields.

Statistical analysis

Quantitative data are presented as the mean ± standard error of the mean (SEM) of at least three independent experiments. Comparisons between two groups were analyzed using the Student’s t-test. P<0.05 was considered to indicate a statistically significant result.

Results

Luteolin suppresses TLR-4 expression in LPS-stimulated BV2 microglia

To examine the effect of luteolin on TLR-4 expression, the levels of TLR-4 mRNA and protein in LPS-stimulated BV2 microglia were measured. The BV2 microglia were pretreated with luteolin for 1 h and then stimulated with LPS for 1 h prior to qPCR or for 24 h prior to western blotting. As shown in Figs. 1 and 2, TLR-4 mRNA and protein levels increased in LPS-stimulated BV2 microglia, but both were markedly suppressed by treatment with luteolin. This result indicates that luteolin suppressed TLR-4 expression and therefore may lead to inhibition of activation of the NF-κB, MAPK and Akt pathways.

Effects of luteolin on the NF-κB signaling pathway

Activation of NF-κB leads to its translocation to the nucleus where it mediates the transcriptional regulation of pro-inflammatory genes. The activation and nuclear translation of NF-κB is a key step in LPS-stimulated microglial activation. The regulation of NF-κB by luteolin was investigated using immunofluorescence staining. As shown in Fig. 3, the NF-κB p65 subunit was primarily retained in the cytoplasm in unstimulated cells; however, following stimulation with LPS, cytoplasmic NF-κB p65 levels were reduced, with a corresponding increase in nuclear NF-κB p65. Treatment with 20 μM luteolin significantly blocked the activation of NF-κB p65 nuclear translocation in LPS-stimulated BV-2 cells. This result suggests that luteolin suppresses pro-inflammatory enzymes and pro-inflammatory cytokines by inhibiting NF-κB activation.

Effects of luteolin on the MAPK signaling pathway

To investigate whether the inhibition of NF-κB activation by luteolin is mediated via the MAPK pathway, the phosphorylation of three MAPK molecules, p38 MAPK, JNK and ERK1/2, was examined in LPS-stimulated BV-2 cells. As shown in Fig. 4, LPS rapidly activated MAPKs within 15 min of LPS stimulation, while luteolin at 20 μM markedly inhibited the LPS-induced phosphorylation of p38 and JNK, but had no effect on ERK phosphorylation. The levels of non-phosphorylated p38, JNK and ERK were unaffected by LPS or luteolin treatment.

Effects of luteolin on the Akt signaling pathway

The effect of luteolin on Akt was then examined. As shown in Fig. 5, luteolin significantly inhibited the LPS-induced activation of Akt. The results suggest that inhibition of Akt by luteolin may contribute to the suppression of LPS-induced NF-κB activation and the expression of inflammatory mediators in BV2 cells.

Luteolin decreases microglial-induced SH-SY5Y cell death in a coculture system

In order to investigate whether luteolin protects against the neuronal death induced by microglial activation, a coculture system with SH-SY5Y neuronal cells and BV2 microglia was used. LPS at concentrations of 0.1, 1.0 or 10.0 μg/ml were not observed to induce cell death in the SH-SY5Y cells (data not shown). Next, the SH-SY5Y cell viability following coculture with LPS-activated BV2 microglia was examined using the MTT assay. As shown in Fig. 6, SH-SY5Y cells in control inserts in the absence of LPS-stimulated BV2 microglia did not undergo cell death. By contrast, LPS treatment alone led to a high level of SH-SY5Y cell death in the coculture, suggesting that the LPS-activated microglia secreted pro-inflammatory cytokines that were able to migrate through the insert, inducing the death of the neuronal cells. Treatment with luteolin markedly reduced the death of the SH-SY5Y cells; cell viability was increased by ~28.6% when the LPS-stimulated BV2 microglia were pretreated with luteolin (Fig. 6).

Apoptosis was determined by the TUNEL assay

As shown in Fig. 7, SH-SY5Y nuclei were stained with Hoechst 33342 (blue) and apoptotic neurons were stained using the TUNEL technique (green). Coculture with BV2 microglia exposed to LPS alone resulted in a significant increase in the number of apoptotic SH-SY5Y cells compared with the number of untreated cells. The administration of luteolin reduced the number of apoptotic SH-SY5Y cells (Fig. 8). These results clearly demonstrate that luteolin protected neurons from microglial-mediated LPS neurotoxicity, supporting its potential role in the prevention of neurodegenerative diseases.

Discussion

The flavonoid luteolin has been shown to inhibit LPS-induced IL-6 production in the brain by inhibiting the JNK signaling pathway and the activation of AP-1 in microglia (17). Luteolin also suppressed microglial TNF-α and IL-6 production stimulated by IFN-γ in the presence of CD40 ligation, and markedly inhibited the IFN-γ-induced phosphorylation of STAT1 (18). These data suggest that this flavonoid is a potent modulator of microglial activation and affects several signaling pathways, leading to a unique phenotype with anti-inflammatory, anti-oxidative and neuroprotective characteristics (19). Previous findings suggest dietary luteolin enhances spatial working memory by mitigating microglial-associated inflammation in the hippocampus (20). Our previous observations confirm the inhibitory effects of luteolin on pro-inflammatory cytokine expression in microglia (15). However, the mechanism by which luteolin mediates these anti-inflammatory effects on microglia is not completely understood and few studies have explored the impact of luteolin on neuroprotection.

In vitro, microglia may be activated experimentally with the bacterial cell wall component LPS. Internalization of TLR-4, rendering microglial cells less sensitive to activation by LPS, plays a role in inducing a reduction in TNF-α production by BV-2 microglial cells (21). TLR-4 is a member of the TLR family of pattern recognition receptors that generate innate immune responses to pathogens by activating a cascade of pro-inflammatory events (22). Therefore, treatments that attenuate TLR-4-associated inflammatory cascades may prove beneficial in ameliorating microglial activation and preventing neurodegenerative processes. The results of the present study indicate that luteolin pretreatment inhibited the upregulation of TLR-4 expression induced by LPS at the transcriptional and translational levels. It was therefore hypothesized that the underlying molecular mechanisms involved include interference with the LPS-triggered increase in TLR-4 expression. The results of the present study indicate that luteolin may inhibit NF-κB, p38, JNK, MAPK and Akt activation through the suppression of TLR-4 expression.

Activation of NF-κB leads to its translocation to the nucleus where it mediates the transcriptional regulation of pro-inflammatory genes (23). The activation and nuclear translation of NF-κB is a key step in LPS-stimulated microglial activation (24). In the current study, treatment with 20 μM luteolin significantly blocked NF-κB p65 nuclear translocation in LPS-stimulated BV-2 cells. In our previous study, a luciferase reporter assay was performed to investigate the possibility that luteolin inhibits NF-κB transcriptional activity, and the possibility that luteolin blocks the phosphorylation and subsequent degradation of IκB in LPS-induced BV2 cells was investigated; it was observed that luteolin causes a marked inhibition of NF-κB p65 nuclear translocation (15). These results, also confirmed by the current study, suggest that luteolin suppresses pro-inflammatory enzymes and pro-inflammatory cytokines through the inhibition of NF-κB activation.

There is evidence that MAPKs play a key role in the regulation of the synthesis and release of pro-inflammatory substances by activated microglia (25). LPS is known to activate various MAPKs, including p38, JNK and ERK. The MAPKs tested in this study (p38, JNK and ERK1/2) were activated in glia and neurons following LPS treatment, suggesting their involvement in glial activation and the neuronal response to diffusible, glia-derived neurotoxic molecules (26). To investigate whether the inhibition of NF-κB activation by luteolin is mediated via the MAPK pathway, the phosphorylation of three MAPK molecules, p38 MAPK, JNK and ERK1/2 in LPS-stimulated BV-2 cells was examined. LPS rapidly activated MAPKs within 15 min of LPS stimulation, while luteolin at 20 μM markedly inhibited the LPS-induced phosphorylation of p38 and JNK, but had no effect on ERK. However, further studies are necessary to support this conclusion.

Multiple signaling pathways, such as those involving MAPKs and Akt, are involved in LPS-stimulated signal transduction and lead to the activation of NF-κB and the subsequent induction of pro-inflammatory gene expression (27,28). Akt is activated via the phosphoinositide-3-OH kinase (PI3K) pathway, an important pathway regulating inflammation and immunity (29). The results of the present study indicate that luteolin also significantly inhibited the LPS-induced activation of Akt. The data suggest that inhibition of Akt by luteolin may contribute to the suppression of LPS-induced NF-κB activation and the expression of inflammatory mediators in BV2 cells.

Neurotoxic microglial-neuronal interactions have been implicated in the pathogenesis of various neurodegenerative diseases (30). Microglial activation has been shown to promote the production of inflammatory cytokines leading to neuronal apoptosis (31,32). In order to investigate whether luteolin is able to rescue neurons from death induced by microglial activation, SH-SY5Y cells and BV2 microglia in a coculture system were used in the present study. The results clearly indicate that when SH-SY5Y cells were cocultured with LPS-stimulated BV2 microglia, neuronal cell death increased by 60.2% and the number of apoptotic neurons increased by 57.0%. However, treatment with luteolin in this LPS-induced coculture system increased cell viability by 28.6% and reduced the apoptotic cell number by 27.0%. These data suggest that luteolin inhibited SH-SY5Y cell apoptosis via inhibition of microglial activation in the microglial-neuronal coculture system. These results provide strong evidence that luteolin protects neurons from microglial-mediated LPS neurotoxicity. However, further in vivo investigation of this activity is necessary in order to clarify the molecular mechanisms involved and assess the full medicinal potential of luteolin.

In conclusion, the present study demonstrated that luteolin inhibited the LPS-stimulated expression of TLR-4. Luteolin also blocked LPS-induced NF-κB, p38, JNK and Akt activation, but had no effect on ERK. When SH-SY5Y cells were cocultured with LPS-stimulated BV2 microglia, pretreatment with luteolin increased neuronal viability and reduced the number of apoptotic cells. These observations suggest that luteolin has a therapeutic application in the treatment of neurodegenerative diseases.

Acknowledgements

This study was supported by grants from the National Natural Science Foundation of China (Nos. 81102449 and 81200930), the Natural Science Foundation of Guangdong Province (Nos. S2011040003038 and S2012040007768) and the Fundamental Research Funds for the Central Universities (No. 21612423).

References

1 

Bernardi A, Frozza RL, Meneghetti A, Hoppe JB, Battastini AM, Pohlmann AR, Guterres SS and Salbego CG: Indomethacin-loaded lipid-core nanocapsules reduce the damage triggered by Aβ1–42 in Alzheimer’s disease models. Int J Nanomedicine. 7:4927–4942. 2012.PubMed/NCBI

2 

Yokoyama H, Uchida H, Kuroiwa H, Kasahara J and Araki T: Role of glial cells in neurotoxin-induced animal models of Parkinson’s disease. Neurol Sci. 32:1–7. 2011.PubMed/NCBI

3 

Dibaj P, Zschüntzsch J, Steffens H, Scheffel J, Göricke B, Weishaupt JH, Le Meur K, Kirchhoff F, Hanisch UK, Schomburg ED and Neusch C: Influence of methylene blue on microglia-induced inflammation and motor neuron degeneration in the SOD1(G93A) model for ALS. PLoS One. 7:e439632012. View Article : Google Scholar : PubMed/NCBI

4 

McGeer EG and McGeer PL: Neuroinflammation in Alzheimer’s disease and mild cognitive impairment: a field in its infancy. J Alzheimers Dis. 19:355–361. 2010.

5 

Mrak RE and Griffin WS: Glia and their cytokines in progression of neurodegeneration. Neurobiol Aging. 26:349–354. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Anisman H: Cascading effects of stressors and inflammatory immune system activation: implications for major depressive disorder. J Psychiatry Neurosci. 34:4–20. 2009.PubMed/NCBI

7 

Ha SK, Lee P, Park JA, Oh HR, Lee SY, Park JH, et al: Apigenin inhibits the production of NO and PGE2 in microglia and inhibits neuronal cell death in a middle cerebral artery occlusion-induced focal ischemia mice model. Neurochem Int. 52:878–886. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Lyons A, Downer EJ, Crotty S, Nolan YM, Mills KH and Lynch MA: CD 200 ligand receptor interaction modulates microglial activation in vivo and in vitro: a role for IL-4. J Neurosci. 27:8309–8313. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Qian L, Flood PM and Hong JS: Neuroinflammation is a key player in Parkinson’s disease and a prime target for therapy. J Neural Transm. 117:971–979. 2010.

10 

Krause DL and Müller N: Neuroinflammation, microglia and implications for anti-inflammatory treatment in Alzheimer’s disease. Int J Alzheimers Dis. 2010:7328062010.

11 

Park E, Kum S, Wang C, Park SY, Kim BS and Schuller-Levis G: Anti-inflammatory activity of herbal medicines: Inhibition of nitric oxide production and tumor necrosis factor-alpha secretion in an activated macrophage-like cell line. Am J Chin Med. 33:415–424. 2005. View Article : Google Scholar : PubMed/NCBI

12 

Kim JS and Jobin C: The flavonoid luteolin prevents lipopolysaccharide-induced NF-κB signaling and gene expression by blocking IκB kinase activity in intestinal epithelial cells and bone-marrow derived dendritic cells. Immunology. 115:375–387. 2005.PubMed/NCBI

13 

Kim SH, Shin KJ, Kim D, Kim YH, Han MS, Lee TG, et al: Luteolin inhibits the nuclear factor-κB transcriptional activity in Rat-1 fibroblasts. Biochem Pharmacol. 66:955–963. 2003.

14 

Gutiérrez-Venegas G, Kawasaki-Cárdenas P, Arroyo-Cruz SR and Maldonado-Frías S: Luteolin inhibits lipopolysaccharide actions on human gingival fibroblasts. Eur J Pharmacol. 541:95–105. 2006.PubMed/NCBI

15 

Zhu LH, Bi W, Qi RB, Wang HD and Lu DX: Luteolin inhibits microglial inflammation and improves neuron survival against inflammation. Int J Neurosci. 121:329–336. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Bureau G, Longpré F and Martinoli MG: Resveratrol and quercetin, two natural polyphenols, reduce apoptotic neuronal cell death induced by neuroinflammation. J Neurosci Res. 86:403–410. 2008. View Article : Google Scholar : PubMed/NCBI

17 

Jang S, Kelley KW and Johnson RW: Luteolin reduces IL-6 production in microglia by inhibiting JNK phosphorylation and activation of AP-1. Proc Natl Acad Sci USA. 105:7534–7539. 2008. View Article : Google Scholar : PubMed/NCBI

18 

Rezai-Zadeh K, Ehrhart J, Bai Y, Sanberg PR, Bickford P, Tan J and Shytle RD: Apigenin and luteolin modulate microglial activation via inhibition of STAT1-induced CD40 expression. J Neuroinflammation. 5:412008. View Article : Google Scholar : PubMed/NCBI

19 

Dirscherl K, Karlstetter M, Ebert S, Kraus D, Hlawatsch J, Walczak Y, Moehle C, Fuchshofer R and Langmann T: Luteolin triggers global changes in the microglial transcriptome leading to a unique anti-inflammatory and neuroprotective phenotype. J Neuroinflammation. 7:32010. View Article : Google Scholar : PubMed/NCBI

20 

Jang S, Dilger RN and Johnson RW: Luteolin inhibits microglia and alters hippocampal-dependent spatial working memory in aged mice. J Nutr. 140:1892–1898. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Willis LM, Bielinski DF, Fisher DR, Matthan NR and Joseph JA: Walnut extract inhibits LPS-induced activation of BV-2 microglia via internalization of TLR4: Possible involvement of phospholipase D2. Inflammation. 33:325–333. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Medzhitov R: Toll-like receptors and innate immunity. Nat Rev Immunol. 1:135–145. 2001. View Article : Google Scholar : PubMed/NCBI

23 

Ozato K, Tsujimura H and Tamura T: Toll-like receptor signaling and regulation of cytokine gene expression in the immune system. Biotechniques. 33(4 Suppl): S66–S75. 2002.PubMed/NCBI

24 

Bi W, Jing X, Zhu L, Liang Y, Liu J, Yang L, Xiao S, Xu A, Shi Q and Tao E: Inhibition of 26S protease regulatory subunit 7 (MSS1) suppresses neuroinflammation. Plos One. 7:e361422012. View Article : Google Scholar : PubMed/NCBI

25 

Bi W, Zhu L, Wang C, Liang Y, Liu J, Shi Q and Tao E: Rifampicin inhibits microglial inflammation and improves neuron survival against inflammation. Brain Res. 1395:12–20. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Xie Z, Smith CJ and Van Eldik LJ: Activated glia induce neuron death via MAP kinase signaling pathways involving JNK and p38. Glia. 45:170–179. 2004. View Article : Google Scholar : PubMed/NCBI

27 

Bhat NR, Zhang P, Lee JC and Hogan EL: Extracellular signal-regulated kinase and p38 subgroups of mitogen-activated protein kinases regulate inducible nitric oxide synthase and tumor necrosis factor-α gene expression in endotoxin-stimulated primary glial cultures. J Neurosci. 18:1633–1641. 1998.PubMed/NCBI

28 

Madrid LV, Wang CY, Guttridge DC, Schottelius AJ, Baldwin AS Jr and Mayo MW: Akt suppresses apoptosis by stimulating the transactivation potential of the Rel A/p65 subunit of NF-κB. Mol Cell Biol. 20:1626–1638. 2000.PubMed/NCBI

29 

Downward J: Lipid-regulated kinases: some common themes at last. Science. 279:673–674. 1998. View Article : Google Scholar : PubMed/NCBI

30 

Li Y, Liu L, Barger SW, Mrak RE and Griffin WS: Vitamin E suppression of microglial activation is neuroprotective. J Neurosci Res. 15:163–170. 2001. View Article : Google Scholar

31 

Brown GC and Neher JJ: Inflammatory neurodegeneration and mechanisms of microglial killing of neurons. Mol Neurobiol. 41:242–247. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Bi W, Zhu L, Jing X, Liang Y and Tao E: Rifampicin and Parkinson’s disease. Neurol Sci. 34:137–141. 2013.

Related Articles

Journal Cover

May-2014
Volume 7 Issue 5

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhu L, Bi W, Lu D, Zhang C, Shu X and Lu D: Luteolin inhibits SH-SY5Y cell apoptosis through suppression of the nuclear transcription factor-κB, mitogen‑activated protein kinase and protein kinase B pathways in lipopolysaccharide-stimulated cocultured BV2 cells. Exp Ther Med 7: 1065-1070, 2014
APA
Zhu, L., Bi, W., Lu, D., Zhang, C., Shu, X., & Lu, D. (2014). Luteolin inhibits SH-SY5Y cell apoptosis through suppression of the nuclear transcription factor-κB, mitogen‑activated protein kinase and protein kinase B pathways in lipopolysaccharide-stimulated cocultured BV2 cells. Experimental and Therapeutic Medicine, 7, 1065-1070. https://doi.org/10.3892/etm.2014.1564
MLA
Zhu, L., Bi, W., Lu, D., Zhang, C., Shu, X., Lu, D."Luteolin inhibits SH-SY5Y cell apoptosis through suppression of the nuclear transcription factor-κB, mitogen‑activated protein kinase and protein kinase B pathways in lipopolysaccharide-stimulated cocultured BV2 cells". Experimental and Therapeutic Medicine 7.5 (2014): 1065-1070.
Chicago
Zhu, L., Bi, W., Lu, D., Zhang, C., Shu, X., Lu, D."Luteolin inhibits SH-SY5Y cell apoptosis through suppression of the nuclear transcription factor-κB, mitogen‑activated protein kinase and protein kinase B pathways in lipopolysaccharide-stimulated cocultured BV2 cells". Experimental and Therapeutic Medicine 7, no. 5 (2014): 1065-1070. https://doi.org/10.3892/etm.2014.1564