Skip to main content
Erschienen in: Journal of Neuroinflammation 1/2011

Open Access 01.12.2011 | Research

Aspirin-triggered lipoxin A4attenuates LPS-induced pro-inflammatory responses by inhibiting activation of NF-κB and MAPKs in BV-2 microglial cells

verfasst von: Yan-Ping Wang, Yan Wu, Long-Yan Li, Jin Zheng, Ren-Gang Liu, Jie-Ping Zhou, Shi-Ying Yuan, You Shang, Shang-Long Yao

Erschienen in: Journal of Neuroinflammation | Ausgabe 1/2011

Abstract

Background

Microglial activation plays an important role in neurodegenerative diseases through production of nitric oxide (NO) and several pro-inflammatory cytokines. Lipoxins (LXs) and aspirin-triggered LXs (ATLs) are considered to act as 'braking signals' in inflammation. In the present study, we investigated the effect of aspirin-triggered LXA4 (ATL) on infiammatory responses induced by lipopolysaccharide (LPS) in murine microglial BV-2 cells.

Methods

BV-2 cells were treated with ATL prior to LPS exposure, and the effects of such treatment production of nitric oxide (NO), inducible nitric oxide synthase (iNOS), interleukin-1β (IL-1β) and tumour necrosis factor-α (TNF-α) were analysed by Griess reaction, ELISA, western blotting and quantitative RT-PCR. Moreover, we investigated the effects of ATL on LPS-induced nuclear factor-κB (NF-κB) activation, phosphorylation of mitogen-activated protein kinases (MAPKs) and activator protein-1 (AP-1) activation.

Results

ATL inhibited LPS-induced production of NO, IL-1β and TNF-α in a concentration-dependent manner. mRNA expressions for iNOS, IL-1β and TNF-α in response to LPS were also decreased by ATL. These effects were inhibited by Boc-2 (a LXA4 receptor antagonist). ATL significantly reduced nuclear translocation of NF-κB p65, degradation of the inhibitor IκB-α, and phosphorylation of extracellular signal-regulated kinase (ERK) and p38 MAPK in BV-2 cells activated with LPS. Furthermore, the DNA binding activity of NF-κB and AP-1 was blocked by ATL.

Conclusions

This study indicates that ATL inhibits NO and pro-inflammatory cytokine production at least in part via NF-κB, ERK, p38 MAPK and AP-1 signaling pathways in LPS-activated microglia. Therefore, ATL may have therapeutic potential for various neurodegenerative diseases.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1742-2094-8-95) contains supplementary material, which is available to authorized users.
Yan-Ping Wang, Yan Wu contributed equally to this work.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

YPW, YW and LYL performed the experiments and analyzed the data. JZ, RGL, and JPZ provided useful advice and reviewed the manuscript. YS conceived the study, participated in its design and coordination, and wrote the manuscript. SYY and SLY oversaw the experimental design and edited the manuscript. All authors of this paper have read and approved the final version the manuscript.
Abkürzungen
ALX
lipoxin A4 receptor
AP-1
activator protein-1
ATL
aspirin-triggered lipoxin A4
CNS
central nervous system
EMSA
Electrophoretic mobility shift assay
ERK
extracellular signal-regulated kinase
IL
interleukin
iNOS
inducible nitric oxide synthase
IκB
inhibitor of κB
JNK
c-jun N-terminal kinase
LPS
lipopolysaccharide
LX
lipoxin
LXA4
lipoxin A4
MAPK
mitogen-activated protein kinase
NF-κB
nuclear factor-κB
RIPA
radioimmune precipitation assay buffer

Background

There is increasing awareness that inflammation may play a role in various neurodegenerative disorders, including Alzheimer's disease, Parkinson's disease, HIV-associated dementia, trauma, multiple sclerosis and stroke [1, 2]. Microglial cells are generally considered to be the immune cells of the central nervous system (CNS). They respond to neuronal injury or immunologic challenges with a reaction termed microglial activation. Activated microglial cells can serve diverse beneficial functions essential to neuron survival, which include cellular maintenance and innate immunity [3, 4]. However, overactivated microglia can induce significant and highly detrimental neurotoxic effects through excess production of a large array of cytotoxic factors such as superoxide, nitric oxide (NO), tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) [1]. Overactivation of microglia followed by overproduction of neurotoxic factors results in deleterious and progressive neurotoxic consequences [5, 6]. In several studies it has been shown that reduction of pro-inflammatory mediators produced by microglia may attenuate the severity of neuronal damage [7]. Therefore, inhibiting inflammatory cytokine production by activated microglia may be useful for preventing neurodegeneration [810].
Lipoxins (LXs) are endogenous lipid mediators with potent anti-infiammatory and pro-resolving actions [11]. Of special interest, aspirin can also trigger transcellular biosynthesis of 15-epimers of LX, termed aspirin-triggered LX (ATL) [12], that share the potent anti-infiammatory actions of LX but are more resistant to metabolic inactivation [13]. LXs and ATL elicit multicellular responses via a specific G protein-coupled receptor termed the LXA4 receptor (ALX) that has been identified in human [14], mouse [15] and rat [16] tissues. In our previous papers, we evaluated the anti-inflammatory activity of an LXA4 analogue, 5(S), 6(R)-LXA4 methyl ester, in a rat model of permanent focal cerebral ischemia and focal cerebral ischemia reperfusion [17, 18]. Our results showed that this LXA4 analogue could attenuate focal ischemia-induced inflammatory responses and inhibit activation of microglia in vivo. Expression of functional ALXs was identified in neural stem cells, neurons, astrocytes and microglia [1923]. Microglial cells are key sensors and versatile effectors in normal and pathologic brain [24]. These findings suggest that microglia may be a target for LXs in brain. However, the effects of LXs on expression of inflammation-related genes and molecular mechanisms in microglia have not been demonstrated.
Lipopolysaccharide (LPS), a component of the outer membrane of Gram-negative bacteria, initiates a number of major cellular responses that play critical roles in the pathogenesis of inflammatory responses and has been commonly used to model proinflammatory and neurotoxic activation of microglia [25, 26]. We used LPS as a stimulant of the microglial reactivity in the current study.
In the present study, we investigated the impact of ATL on the infiammatory response induced by LPS in murine microglial BV-2 cells, as well as the signaling pathways involved in these processes. Our data suggest that ATL inhibits NO and pro-inflammatory cytokine production in LPS-activated microglia at least in part via NF-κB, ERK, p38 MAPK and AP-1 signaling pathways.

Methods

Cell culture

The immortalized murine microglia cell line BV-2 was purchased from Cell Resource Centre of Peking Union Medical College (Beijing, China) and maintained in Dulbecco's modified Eagle's medium with F12 supplement (DMEM/F12, Gibco, Grand Island, NY) supplemented with 10% fetal bovine serum (Gibco), 100 U/ml penicillin and 100 μg/ml streptomycin at 37°C in a humidified atmosphere of 95% air, 5% CO2. Confiuent cultures were passaged by trypsinization. BV-2 cells were seeded onto 96-well plates (104 cells/well for cell viability assay), 24-well-culture plates (105 cells/well for ELISA and NO measurement, 104 cells/well for immunofluorescence), 6-well plates (2.5 × 105 cells/well for PCR) or 100 mm culture dishes (1.2 × 106 cells/dish for western blotting and EMSA). Before each experiment, cells were serum-starved for 12 h. BV-2 cells were incubated in the initial experiments with different concentrations (1 nM, 10 nM or 100 nM) of ATL (Cayman Chemical, Ann Arbor, MI), leading to a concentration of 100 nM ATL used in further experiments or vehicle (0.035% ethanol) for 30 min before addition of 100 ng/ml LPS (Escherichia coli O26:B6, Sigma-Aldrich, St. Louis, MO) under serum-free conditions. To investigate the involvement of ALXs in the anti-inflammatory effects of ATL, the cells were treated with 100 μM Boc-2 (Phoenix Pharmaceuticals), a specific receptor antagonist, prior to the treatment with ATL for 30 min.

RNA isolation, reverse-transcriptase (RT) PCR and real-time PCR

Total RNA was extracted from BV-2 cells with TRIzol reagent (Invitrogen, Carlsbad, CA) according to the manufacturer's protocol. 1.0 μg of total RNA was subjected to oligo-dT-primed RT with ReverTra Ace Kit (Toyobo, Osaka, Japan).
Semi-quantitative PCR was carried out with DNA polymerase (Toyobo) by using specific primers (Invitrogen): 5'-GGCAACTCTGTTGAGGAAAG-3' and 5'-GGCTCTCGGTAGACGAGA-3', which amplify the 423 bp product for ALX1/FPR-rs1; and 5'-GTCAAGATCAACAGAAGAAACC-3' and 5'-GGGCTCTCTCAAGACTATAAGG-3', which amplify 298 bp product for ALX2/FPR2; and 5'-TGGAATCCTGTGGCATCCATGAAAC-3' and 5'-TAAAACGCAGCTCAGTAACAGTCCG-3', which amplify 349 bp product for β-actin. The amplified PCR products were resolved by 2% agarose gel electrophoresis.
Real-time PCR was performed for a quantitative analysis of iNOS, IL-1β and TNF-α mRNA expression using SYBR Green real-time PCR Master Mix (Toyobo) on an MX3000P real-time PCR system (Stratagene). The following primers were used (Invitrogen): 5'-CAGCTGGGCTGTACAAACCTT-3' and 5'- CATTGGAAGTGAAGCGTTTCG-3', which amplify the 95 bp product for iNOS; 5'-CAACCAACAAGTGATATTCTCCATG-3' and 5'- GATCCACACTCTCCAGCTGCA-3', which amplify the 152 bp product for IL-1β; 5'-CATCTTCTCAAAATTCGAGTGACAA-3' and 5'-TGGGAGTAGACAAGGTACAACCC-3', which amplify the 175 bp product for TNF-α; and 5'-TGTCCACCTTCCAGCAGATGT-3' and 5'-AGCTCAGTAACAGTCCGCCTAGA-3', which amplify the 101 bp product for β-actin. Relative gene expression was calculated by the 2-ΔΔCT method [27].

Cell viability assay

Cell viability was measured by quantitative colorimetric assay with MTT (Sigma-Aldrich), showing the mitochondrial activity of living cells. BV-2 cells in 96-well plates were pretreated with various concentrations of ATL for 30 min and incubated with or without LPS for 24 h in the continued presence of ATL. Upon termination of the experiments, the culture media were aspirated and MTT (0.5 mg/ml) was added to cells and then incubated at 37°C for 4 h. The supernatant was aspirated and dimethyl sulfoxide (Sigma-Aldrich) was added to the wells. Insoluble crystals were dissolved by mixing and the plates were read on an automated Tecan Sunrise absorbance reader, using a test wavelength of 570 nm and a reference wavelength of 630 nm.

Nitrite measurements

Production of NO was determined by measuring the level of accumulated nitrite, a metabolite of NO in the culture supernatant using Griess reagent (Sigma-Aldrich). After 24 h of treatment with LPS with or without ATL, the culture supernatants were collected and mixed with an equal volume of Griess reagent in 96-well culture plates and incubated at room temperature for 10 min. The absorbance was measured at 540 nm and nitrite concentrations were calculated by reference to a standard curve generated by known concentrations of sodium nitrite.

ELISA for IL-1β and TNF-α

BV-2 cells in 24-well plates were stimulated for 24 h, and then culture supernatants were harvested. Levels of IL-1β and TNF-α in 100 μl medium were measured by commercial ELISA kits (Boster Biological Technology, Wuhan, China) according to the manufacturer's instructions.

Immunofluorescence confocal microscopy

For the detection of intracellular location of NF-κB p65, BV-2 cells were cultured on sterile glass cover slips in 24 well plates and treated with ATL and LPS as described above. At various times after the LPS treatment, cells were fixed with 4% paraformaldehyde in PBS and permeabilized with 0.1% Triton X-100 in PBS. After rinsing, cells were blocked with 3% BSA in PBS for 1 h and incubated with rabbit anti-NF-κB p65 antibodies (1:200, Santa Cruz Biotechnology, Santa Cruz) overnight at 4°C. After washing, cells were incubated with FITC-conjugated goat anti-rabbit IgG (1:400, Pierce, Rockford, IL) for 1 h and counterstained with 4, 6-diamidino-2-phenylindole (DAPI, Roche, Shanghai, China) for the identification of nuclei. After washing with PBS, the cover slips were mounted with antifade mounting medium (Beyotime, China) on slides, and the cells were observed with a confocal microscope Olympus Fluoview FV500.

Protein extraction

For making whole cell lysates, the cells were lysed in radioimmune precipitation assay (RIPA) buffer supplemented with protease inhibitor cocktail (Roche). Nuclear and cytoplasmic fractionations were performed with Proteo JET™ Cytoplasmic and Nuclear Protein Extraction Kit (Fermentas Life Science) according to manufacturer's protocol.

Western blot analysis

Equal amounts of cytoplasmic, nuclear, or whole cell extracts were electrophoresed on sodium dodecyl sulfate-polyacrylamide gels, and then transferred onto a polyvinylidene difluoride membrane (Millipore). The transformed membrane was blocked for 1 h and incubated with indicated primary antibodies (Santa Cruz Biotechnology) at 4°C overnight. The primary antibodies usedwere as follows: rabbit anti-iNOS (1:500), β-actin (1:1000), p65 (1:1000), Lamin B (1:1000), IκB-α (1:500), ERK1/2 (1:1000), p38 (1:1000), JNK (1:1000) and mouse anti-phosphorylated ERK1/2, p38, JNK antibody (1:1000). The membrane was washed three times with Tris-bufffered saline containing 0.05% Tween 20 (TBST) for 10 min and incubated with anti-rabbit or anti-mouse IgG-horseradish peroxidase (1:5000, Pierce) at room temperature for 1 h. The Supersignal West Pico chemiluminescent substrate system (Pierce) was used to detect immunoreactive bands. The intensity of protein bands after western blotting were quantitated by using Quantity One Version 4.6.3 Image software (Bio-Rad) and normalized against proper loading controls.

Electrophoretic mobility shift assay (EMSA)

Nuclear extracts were prepared as described above. Oligonucleotides corresponding to the NF-κB (5'-AGTTGAGGGGACTTTCCCAGGC-3') and AP-1 (5'-CGCTTGATGAGTCAGCCGGAA-3') binding site consensus sequences were synthesized and end-labeled with biotin by Invitrogen. EMSAs were performed using the LightShift chemiluminescent EMSA kit (Pierce). Briefly, 20 fmol of biotin-labeled, double strand probe was incubated for 20 min at room temperature in 20 μl of EMSA binding buffer containing 2.5% glycerol, 5 mM MgCl2, 50 ng/μl poly (dI-dC), 0.05% Nonidet P-40, and 6 μg of nuclear proteins. For competition EMSA, 200-fold (4 pmol) excess unlabeled, double strand probe was added to the binding reaction. The DNA-nuclear protein complexes were resolved by electrophoresis in 6% nondenaturing polyacrylamide gel in 0.5 × Tris-borate-EDTA (TBE) buffer at 100 V. Gels were then electroblotted onto Hybond nylon membranes (GE Healthcare) at 380 mA for 50 min. The membranes were then cross-linked for 15 min with the membrane face down on a transilluminator at 312 nm, and the biotinylated protein-DNA bands were detected with HRP-conjugated streptavidin using the chemiluminescent nucleic acid detection system (Pierce).

Statistical analysis

Data are expressed as means ± SEM of the indicated number of independent experiments. Changes in IκB protein levels were analyzed by two-way ANOVA (treatment and time). All other data were analyzed by one-way ANOVA. Least significant difference (LSD) post hoc test was used for multiple comparisons. Statistical analysis was performed using the SPSS software version 17.0 (SPSS Inc., Chicago, IL, USA). P < 0.05 was considered statistically significant.

Results

ALXs are expressed in BV-2 microglial cells

Using RT-PCR, we showed that both ALX1/FPR-rs1 and ALX2/FPR2 were expressed in BV-2 microglial cells. The mRNA expression levels of these two receptors were significantly enhanced when the cells were exposed to LPS (100 ng/ml) for 6 h (Figure 1).

ATL inhibits LPS-induced NO, IL-1β and TNF-α production in BV-2 cells

Initially, we evaluated the effects of ATL on NO, IL-1β and TNF-α production in LPS-stimulated BV-2 microglia. BV-2 cells were incubated with vehicle or different concentrations of ATL (1, 10 and 100 nM) for 30 min and stimulated with 100 ng/ml LPS for 24 h. To determine NO production, we measured nitrite released into the culture medium using the Griess reagent. Stimulation of BV-2 cells with LPS markedly increased (about 7.5-fold) NO production, compared with that generated under control conditions. Pretreatment with ATL significantly inhibited this increase in a concentration-dependent manner (Figure 2A).
We then tested whether ATL reduces the production of LPS-induced pro-inflammatory cytokines IL-1β and TNF-α using ELISA. As shown in Figure 2B and 2C, stimulation of BV-2 cells with LPS led to a significant increase in the levels of IL-1β and TNF-α in the cell-conditioned media after 24 h. Pretreatment of BV-2 cells with ATL significantly inhibited the LPS-induced IL-1β and TNF-α production, concentration dependently.
To evaluate the role of the ALXs in the anti-inflammatory effects of ATL, BV-2 cells were treated with an ALX antagonist, Boc-2 (100 μM, 30 min) prior to treatment with ATL. Pretreatment with Boc-2 inhibited these effects in response to ATL (Figure 2).
To exclude the possibility that the decrease in the NO and cytokines levels was simply due to the cytotoxicity of the drug, cell viability was evaluated. The cytotoxic effects of ATL in BV-2 cells were evaluated in the absence or presence of LPS using MTT assays. ATL (1, 10 and 100 nM) and vehicle did not affect cell viability (Figure 2D). When cells were treated with 100 ng/ml LPS only, a decrease in viability was detected compared with the control cells. However, cells pretreated with ATL for 30 min showed no significant increase compared with cells that were treated with LPS only (Figure 2D). Therefore, the inhibitory effect of ATL on LPS-induced, inflammation-related responses in activated BV-2 cells was not the result of ATL effects on cell survival.

ATL inhibits mRNA expressions of iNOS, IL-1β, and TNF-α

To find out whether ATL suppresses iNOS, IL-1β and TNF-α expression at the transcriptional level, BV-2 cells were incubated for 30 min with the indicated concentrations of ATL and then incubated with 100 ng/ml LPS for 6 h. The relative amounts of iNOS, IL-1β and TNF-α mRNA were determined by real-time RT-PCR. As anticipated, LPS induced a marked increase in iNOS, IL-1β and TNF-α mRNA in BV-2 cells, about 20, 11, 26-fold increase, respectively (Figure 3). Pretreatment with ATL reduced LPS-induced up-regulation of iNOS, IL-1β and TNF-α mRNA levels in a dose-dependent manner (Figure 3). The inhibitory effects of ATL on LPS-induced iNOS mRNA up-regulation were accompanied by attenuation of iNOS protein induction (Figure 3B). ATL inhibition of LPS-induced expression of iNOS, IL-1β and TNF-α was reversed after pre-exposure of BV-2 cells to the ALX antagonist Boc-2 (100 μM) for 30 min (Figure 3). Taken together, our current data prove that ATL inhibits the inflammatory activation of BV-2 microglia cells with respect to NO production and pro-inflammatory cytokine expression.

ATL inhibits nuclear translocation of NF-κB and degradation of IκB-α

Because ATL reduced the transcriptional activation of iNOS, IL-1β and TNF-α genes, it is likely that it blocks signaling events involved in transcriptional activation of these genes. Expression of iNOS and cytokines genes requires NF-κB activation and nuclear translocation to interact with DNA. Therefore, the involvement of NF-κB nuclear translocation in ATL-induced suppression of NO and cytokines was examined by fluorescence microscopy. LPS stimulation caused obvious translocation of NF-κB p65 from the cytoplasm into the nucleus 60 min after activation (Figure 4A), whereas the presence of 100 nM ATL reduced this (Figure 4B). To further verify the p65 nuclear translocation data, we analyzed the cells by western blotting and found that pretreatment of cells with 100 nM ATL prevented p65 nuclear localization induced by LPS (Figure 4C and 4D).
To address the possibility that the impaired nuclear translocation of p65 was due to inhibition of degradation of IκB-α, we examined the effect of ATL on IκB-α degradation induced by LPS. Western blot analysis showed that LPS-induced degradation of IκB-α was significantly reversed by 100 nM ATL in BV-2 cells (Figure 4E).

ATL inhibits LPS-induced ERK and p38 MAPK activation

Along with NF-κB, MAPKs are known to play an important role in the signaling pathways that induce proinfiammatory cytokines and iNOS in glial cells [28]. To investigate whether the inhibition of infiammation by ATL is regulated by the MAPK pathway, we examined the effects of ATL on LPS-induced phosphorylation of ERK, p38 MAPK and JNK in BV-2 microglia by western blot analysis. Cells were pretreated with 100 nM ATL for 30 min and then incubated with 100 ng/ml LPS for 30 min. The 30-min treatment of LPS was determined to be optimal in a preliminary study that examined MAPK phosphorylation at 0, 10, 20, 30, and 60 min after LPS treatment (data not shown). ATL (100 nM) markedly inhibited ERK and p38 MAPK activation, while phosphorylation of JNK was not affected (Figure 5A-C). Strikingly, ATL could induce JNK phosphorylation without effect on ERK and p38 MAPK activity.

ATL inhibits LPS-induced NF-κB and AP-1 DNA binding activity

To determine the effects of ATL on transcription factor signaling pathways that might mediate LPS-induced proinfiammatory cytokines production, EMSA was performed. BV-2 cells were pretreated with vehicle and 100 nM ATL for 30 min before stimulation with LPS (100 ng/ml) for 1 h. NF-κB and AP-1 binding activities were induced by LPS treatment (Figure 6A and 6B, lane 3). Binding specificity was verified by incubating nuclear extracts from LPS-stimulated BV-2 cells with excess unlabeled specific competitor oligonucleotide probe (Figure 6A and 6B, lane 5). Pretreatment with ATL markedly reduced the LPS-induced DNA-binding activity of NF-κB and AP-1 (Figure 6A and 6B, lane 4).

Discussion

Our present data provide the first evidence that ATL inhibits the infiammatory activation of microglia. To date, two separate LXA4 receptors (ALX1/FPR-rs1 and ALX2/FPR2) have been identified in mice [15, 29]. Mouse ALX2/FPR2 is expressed by neutrophils, monocytes, macrophages, dendritic cells, and microglial cells, and its transcripts are detected at high levels in spleen and lung [30]. ALX1/FPR-rs1 and ALX2/FPR2 are both expressed in the mouse pituitary gland, hypothalamic tissue and vomeronasal organ [31, 32]. As demonstrated by RT-PCR analysis, ALX1/FPR-rs1 and ALX2/FPR2 are both expressed in BV-2 microglial cells. ATL reduced LPS-induced production of NO, IL-1β and TNF-α in BV-2 microglial cells. This is a receptor-mediated effect as it disappeared when microglial cells were pretreated with Boc-2 before ATL treatment. Quantitative PCR analysis showed that ATL markedly suppresses iNOS, IL-1β and TNF-α gene expression in BV-2 microglia cells. Similarly, this effect was abrogated by the use of Boc-2. NF-κB, ERK and p38 MAPK pathways are at least partly involved in the anti-infiammatory mechanisms of ATL in BV-2 cells. Thus, ATL is a promising agent for preventing and treating neuroinflammation and may be useful for mitigating a dysregulated linkage between the immune system and brain.
Although microglial activation has important repairative functions in the CNS, microglial cell activation in infection, infiammation, or injury may go beyond control and eventually produce detrimental effects that override the beneficial effects. Activation of microglia leads to release of various toxic molecules such as superoxide, NO, IL-1β and TNF-α, contributing to neuronal damage in various neurodegenerative disorders [1].
LX possesses dual anti-inflammatory and pro-resolution activities that have been demonstrated in a multitude of acute and chronic inflammatory conditions [11]. Previously, LXA4, ATL and their stable analogues have been shown to play a major role in important functional properties of the central nervous system, such as neural stem cell proliferation and differentiation, pain, and cerebral ischemia [1719, 33]. In primary murine microglia or N9 microglial cells, expression of ALX2/FPR2 has been identified and is up-regulated by inflammatory stimuli [20, 21]. In the present study, the expression of ALX2/FPR2 and another murine high-affinity ALX1/FPR-rs1 were confirmed in BV-2 microglial cells. These findings suggest that ATL could work as a modulator of the inflammatory reaction of the brain immune system, eventually acting as a microglial activation repressor.
NO and pro-infiammatory cytokines such as IL-1β and TNF-α are known to be important mediators in the process of infiammation. These proinfiammatory mediators are thought to be responsible for some of the harmful effects of brain injuries and diseases, including ischemia, Alzheimer's disease, Parkinson's disease and multiple sclerosis [34]. Under various pathological conditions associated with infiammation, large amounts of NO are produced in the brain as a result of the induced expression of iNOS in glial cells [35]. High levels of NO exert their toxic effects through multiple mechanisms, including lipid peroxidation, mitochondrial damage, protein nitration and oxidation, depletion of antioxidant reserves, activation or inhibition of various signaling pathways, and DNA damage [35]. Therefore, the effect of ATL on NO production and iNOS expression in LPS-stimulated microglia cells was examined. As shown in previous research [36, 37], NO is produced at low levels in unstimulated microglia. Stimulation of BV-2 microglial cells with LPS induced strong NO production and iNOS expression. The magnitude of the NO/iNOS response to LPS in BV-2 microglial cells is different in different studies with different concentrations as well as durations of LPS treatment. In the present study, ATL markedly reduced NO production and mRNA and protein expression of iNOS in dose-dependent manners without significant cytotoxicity. This indicates that inhibition of NO production by ATL is a result of inhibition of iNOS gene expression. Previous studies also have shown that LXA4 and ATL analogues inhibit LPS-induced NO production and peroxynitrite formation in human leukocytes [38] and in mouse lung [39].
Pro-infiammatory cytokines produced by activated microglia, including IL-1β and TNF-α, play an important role in the process of neuroinfiammatory diseases [34]. IL-1β is a potent pro-infiammatory cytokine that acts through IL-1 receptors found on numerous cell types, including neurons and microglia. TNF-α can cause cell death directly by binding to neuronal TNF receptors linked to death domains that activate caspase-dependent apoptosis [40] or by potentiating glutamate release, thereby enhancing excitotoxicity [41]. IL-1β and TNF-α also drive self-propagating cycles of microglial activation and neuroinflammation by inducing activation of NF-κB, cytokine generation and further activation of NF-κB. Thus, inhibition of cytokine production or function serves as a key mechanism in the control of neurodegeneration. Our results showed that ATL markedly attenuates the production of IL-1β and TNF-α, and their mRNA expressions; induced by LPS in BV-2 cells. Consistent with our findings, similar results have shown that LXA4 and ATL inhibit LPS-induced production of IL-1β and TNF-α in uvea and in macrophages and endothelial cells [4244].
In subsequent studies, we found that ATL has a strong inhibitory effect on infiammatory signaling pathways that include NF-κB and MAPK/AP-1. NF-κB activity increases in acute neurodegenerative disorders such as stroke, severe epileptic seizures, and traumatic brain injury; and in chronic neurodegenerative conditions, including Alzheimer's disease, Parkinson's disease, Huntington disease, and amyotrophic lateral sclerosis [45]. In general, activation of NF-κB in microglia contributes to neuronal injury and promotes the development of neurodegenerative disorders [45]. NF-κB is known as a pleiotropic regulator of various genes involved in the production of many proinfiammatory cytokines and enzymes. NF-κB is also a central regulator of microglial responses to activating stimuli, including LPS and cytokines [46]. In this study, ATL was able to inhibit the LPS-evoked degradation of IκB-α, nuclear translocation of NF-κB p65 and the DNA-binding activities of NF-κB in BV-2 cells. Previous studies have shown that LXs reduce nuclear translocation of NF-κB in human neutrophils, mononuclear leukocytes [38] and macrophages [43]. It has also been reported that ATLs reduce NF-κB-mediated transcriptional activation in an ALX-dependent manner, and inhibit the degradation of IκB [47]. Therefore, induction of anti-inflammatory responses by LXs may be dependent on the NF-κB signaling pathway.
In addition, LPS also activates MAPK pathways which lead to the induction of another transcription factor, AP-1. MAPKs are a group of signaling molecules that appear to play key roles in infiammatory processes [48]. We found that phosphorylation of ERK and p38 MAPK in response to LPS is decreased by ATL treatment. Our results also show that ATL treatment of BV-2 microglia results in decreased DNA-binding activities of AP-1 following LPS stimulation. This observation is in line with studies in mesangial cells, endothelial cells, neutrophils, fibroblasts and T cells, which have shown that ERK and/or p38 MAPK activation is attenuated in the presence of LXs [42, 4951]. In the present study, ATL failed to inhibit LPS-induced phosphorylation of JNK. A previous study in primary astrocytes found that an ATL analogue prevents ATP-evoked JNK phosphorylation, but has no effect on TNF-α-induced JNK phosphorylation [33]. Strikingly, our results show that ATL induces JNK phosphorylation, but has no effect on ERK and p38 MAPK activity. In another study, LXA4 attenuated microvascular fluid leaks caused by LPS partly mediated by the JNK signaling pathway [52]. LXA4 and ATL analogues could promote ERK phosphorylation in macrophages and monocytes [53, 54]. The reasons for these discrepancies are mainly due to differences in experimental models, cell types and stimulators.

Conclusions

In summary, our results show that ATL inhibits release of NO and pro-inflammatory cytokines in a concentration-dependent manner. Moreover, ATL acts at the level of transcription in LPS-stimulated microglia. A possible mechanism for this effect involves ATL's ability to activate a signaling cascade that results in repression of NF-κB, ERK and p38 MAPK activation in activated microglia. Given the fact that microglial activation contributes to the pathogenesis of neurodegenerative diseases, ATL may be considered as a potential therapeutic agent for neurodegenerative diseases involving neuroinflammation.

Acknowledgements

This study was supported by the grants from the National Natural Science Foundation of China (30700784 and 30900448) and Science Foundation for The Excellent Youth Scholars of Ministry of Education of China (20090142120047).
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

YPW, YW and LYL performed the experiments and analyzed the data. JZ, RGL, and JPZ provided useful advice and reviewed the manuscript. YS conceived the study, participated in its design and coordination, and wrote the manuscript. SYY and SLY oversaw the experimental design and edited the manuscript. All authors of this paper have read and approved the final version the manuscript.
Literatur
1.
Zurück zum Zitat Block ML, Zecca L, Hong JS: Microglia-mediated neurotoxicity: uncovering the molecular mechanisms. Nat Rev Neurosci. 2007, 8: 57-69. 10.1038/nrn2038.CrossRefPubMed Block ML, Zecca L, Hong JS: Microglia-mediated neurotoxicity: uncovering the molecular mechanisms. Nat Rev Neurosci. 2007, 8: 57-69. 10.1038/nrn2038.CrossRefPubMed
2.
Zurück zum Zitat Amor S, Puentes F, Baker D, van der Valk P: Inflammation in neurodegenerative diseases. Immunology. 2010, 129: 154-169. 10.1111/j.1365-2567.2009.03225.x.PubMedCentralCrossRefPubMed Amor S, Puentes F, Baker D, van der Valk P: Inflammation in neurodegenerative diseases. Immunology. 2010, 129: 154-169. 10.1111/j.1365-2567.2009.03225.x.PubMedCentralCrossRefPubMed
3.
Zurück zum Zitat Brandenburg LO, Jansen S, Wruck CJ, Lucius R, Pufe T: Antimicrobial peptide rCRAMP induced glial cell activation through P2Y receptor signalling pathways. Mol Immunol. 2010, 47: 1905-1913. 10.1016/j.molimm.2010.03.012.CrossRefPubMed Brandenburg LO, Jansen S, Wruck CJ, Lucius R, Pufe T: Antimicrobial peptide rCRAMP induced glial cell activation through P2Y receptor signalling pathways. Mol Immunol. 2010, 47: 1905-1913. 10.1016/j.molimm.2010.03.012.CrossRefPubMed
4.
Zurück zum Zitat Brandenburg LO, Varoga D, Nicolaeva N, Leib SL, Wilms H, Podschun R, Wruck CJ, Schroder JM, Pufe T, Lucius R: Role of glial cells in the functional expression of LL-37/rat cathelin-related antimicrobial peptide in meningitis. J Neuropathol Exp Neurol. 2008, 67: 1041-1054. 10.1097/NEN.0b013e31818b4801.CrossRefPubMed Brandenburg LO, Varoga D, Nicolaeva N, Leib SL, Wilms H, Podschun R, Wruck CJ, Schroder JM, Pufe T, Lucius R: Role of glial cells in the functional expression of LL-37/rat cathelin-related antimicrobial peptide in meningitis. J Neuropathol Exp Neurol. 2008, 67: 1041-1054. 10.1097/NEN.0b013e31818b4801.CrossRefPubMed
5.
Zurück zum Zitat Polazzi E, Contestabile A: Reciprocal interactions between microglia and neurons: from survival to neuropathology. Rev Neurosci. 2002, 13: 221-242. 10.1515/REVNEURO.2002.13.3.221.CrossRefPubMed Polazzi E, Contestabile A: Reciprocal interactions between microglia and neurons: from survival to neuropathology. Rev Neurosci. 2002, 13: 221-242. 10.1515/REVNEURO.2002.13.3.221.CrossRefPubMed
6.
Zurück zum Zitat Streit WJ, Conde JR, Fendrick SE, Flanary BE, Mariani CL: Role of microglia in the central nervous system's immune response. Neurol Res. 2005, 27: 685-691.PubMed Streit WJ, Conde JR, Fendrick SE, Flanary BE, Mariani CL: Role of microglia in the central nervous system's immune response. Neurol Res. 2005, 27: 685-691.PubMed
7.
Zurück zum Zitat Ramirez BG, Blazquez C, Gomez del Pulgar T, Guzman M, de Ceballos ML: Prevention of Alzheimer's disease pathology by cannabinoids: neuroprotection mediated by blockade of microglial activation. J Neurosci. 2005, 25: 1904-1913. 10.1523/JNEUROSCI.4540-04.2005.CrossRefPubMed Ramirez BG, Blazquez C, Gomez del Pulgar T, Guzman M, de Ceballos ML: Prevention of Alzheimer's disease pathology by cannabinoids: neuroprotection mediated by blockade of microglial activation. J Neurosci. 2005, 25: 1904-1913. 10.1523/JNEUROSCI.4540-04.2005.CrossRefPubMed
8.
Zurück zum Zitat Jin R, Yang G, Li G: Inflammatory mechanisms in ischemic stroke: role of inflammatory cells. J Leukoc Biol. 2010, 87: 779-789. 10.1189/jlb.1109766.PubMedCentralCrossRefPubMed Jin R, Yang G, Li G: Inflammatory mechanisms in ischemic stroke: role of inflammatory cells. J Leukoc Biol. 2010, 87: 779-789. 10.1189/jlb.1109766.PubMedCentralCrossRefPubMed
9.
Zurück zum Zitat Block ML, Hong JS: Microglia and inflammation-mediated neurodegeneration: multiple triggers with a common mechanism. Prog Neurobiol. 2005, 76: 77-98. 10.1016/j.pneurobio.2005.06.004.CrossRefPubMed Block ML, Hong JS: Microglia and inflammation-mediated neurodegeneration: multiple triggers with a common mechanism. Prog Neurobiol. 2005, 76: 77-98. 10.1016/j.pneurobio.2005.06.004.CrossRefPubMed
10.
11.
Zurück zum Zitat Serhan CN, Chiang N, Van Dyke TE: Resolving inflammation: dual anti-inflammatory and pro-resolution lipid mediators. Nat Rev Immunol. 2008, 8: 349-361. 10.1038/nri2294.PubMedCentralCrossRefPubMed Serhan CN, Chiang N, Van Dyke TE: Resolving inflammation: dual anti-inflammatory and pro-resolution lipid mediators. Nat Rev Immunol. 2008, 8: 349-361. 10.1038/nri2294.PubMedCentralCrossRefPubMed
12.
Zurück zum Zitat Claria J, Serhan CN: Aspirin triggers previously undescribed bioactive eicosanoids by human endothelial cell-leukocyte interactions. Proc Natl Acad Sci USA. 1995, 92: 9475-9479. 10.1073/pnas.92.21.9475.PubMedCentralCrossRefPubMed Claria J, Serhan CN: Aspirin triggers previously undescribed bioactive eicosanoids by human endothelial cell-leukocyte interactions. Proc Natl Acad Sci USA. 1995, 92: 9475-9479. 10.1073/pnas.92.21.9475.PubMedCentralCrossRefPubMed
13.
Zurück zum Zitat Serhan CN, Savill J: Resolution of inflammation: the beginning programs the end. Nat Immunol. 2005, 6: 1191-1197. 10.1038/ni1276.CrossRefPubMed Serhan CN, Savill J: Resolution of inflammation: the beginning programs the end. Nat Immunol. 2005, 6: 1191-1197. 10.1038/ni1276.CrossRefPubMed
14.
Zurück zum Zitat Fiore S, Maddox JF, Perez HD, Serhan CN: Identification of a human cDNA encoding a functional high affinity lipoxin A4 receptor. J Exp Med. 1994, 180: 253-260. 10.1084/jem.180.1.253.CrossRefPubMed Fiore S, Maddox JF, Perez HD, Serhan CN: Identification of a human cDNA encoding a functional high affinity lipoxin A4 receptor. J Exp Med. 1994, 180: 253-260. 10.1084/jem.180.1.253.CrossRefPubMed
15.
Zurück zum Zitat Takano T, Fiore S, Maddox JF, Brady HR, Petasis NA, Serhan CN: Aspirin-triggered 15-epi-lipoxin A4 (LXA4) and LXA4 stable analogues are potent inhibitors of acute inflammation: evidence for anti-inflammatory receptors. J Exp Med. 1997, 185: 1693-1704. 10.1084/jem.185.9.1693.PubMedCentralCrossRefPubMed Takano T, Fiore S, Maddox JF, Brady HR, Petasis NA, Serhan CN: Aspirin-triggered 15-epi-lipoxin A4 (LXA4) and LXA4 stable analogues are potent inhibitors of acute inflammation: evidence for anti-inflammatory receptors. J Exp Med. 1997, 185: 1693-1704. 10.1084/jem.185.9.1693.PubMedCentralCrossRefPubMed
16.
Zurück zum Zitat Chiang N, Takano T, Arita M, Watanabe S, Serhan CN: A novel rat lipoxin A4 receptor that is conserved in structure and function. Br J Pharmacol. 2003, 139: 89-98. 10.1038/sj.bjp.0705220.PubMedCentralCrossRefPubMed Chiang N, Takano T, Arita M, Watanabe S, Serhan CN: A novel rat lipoxin A4 receptor that is conserved in structure and function. Br J Pharmacol. 2003, 139: 89-98. 10.1038/sj.bjp.0705220.PubMedCentralCrossRefPubMed
17.
Zurück zum Zitat Wu Y, Ye XH, Guo PP, Xu SP, Wang J, Yuan SY, Yao SL, Shang Y: Neuroprotective effect of lipoxin A(4) methyl ester in a rat model of permanent focal cerebral ischemia. J Mol Neurosci. 2010, 42: 226-234. 10.1007/s12031-010-9355-8.CrossRefPubMed Wu Y, Ye XH, Guo PP, Xu SP, Wang J, Yuan SY, Yao SL, Shang Y: Neuroprotective effect of lipoxin A(4) methyl ester in a rat model of permanent focal cerebral ischemia. J Mol Neurosci. 2010, 42: 226-234. 10.1007/s12031-010-9355-8.CrossRefPubMed
18.
Zurück zum Zitat Ye XH, Wu Y, Guo PP, Wang J, Yuan SY, Shang Y, Yao SL: Lipoxin A4 analogue protects brain and reduces inflammation in a rat model of focal cerebral ischemia reperfusion. Brain Res. 2010, 1323: 174-183.CrossRefPubMed Ye XH, Wu Y, Guo PP, Wang J, Yuan SY, Shang Y, Yao SL: Lipoxin A4 analogue protects brain and reduces inflammation in a rat model of focal cerebral ischemia reperfusion. Brain Res. 2010, 1323: 174-183.CrossRefPubMed
19.
Zurück zum Zitat Wada K, Arita M, Nakajima A, Katayama K, Kudo C, Kamisaki Y, Serhan CN: Leukotriene B4 and lipoxin A4 are regulatory signals for neural stem cell proliferation and differentiation. FASEB J. 2006, 20: 1785-1792. 10.1096/fj.06-5809com.CrossRefPubMed Wada K, Arita M, Nakajima A, Katayama K, Kudo C, Kamisaki Y, Serhan CN: Leukotriene B4 and lipoxin A4 are regulatory signals for neural stem cell proliferation and differentiation. FASEB J. 2006, 20: 1785-1792. 10.1096/fj.06-5809com.CrossRefPubMed
20.
Zurück zum Zitat Cui YH, Le Y, Zhang X, Gong W, Abe K, Sun R, Van Damme J, Proost P, Wang JM: Up-regulation of FPR2, a chemotactic receptor for amyloid beta 1-42 (A beta 42), in murine microglial cells by TNF alpha. Neurobiol Dis. 2002, 10: 366-377. 10.1006/nbdi.2002.0517.CrossRefPubMed Cui YH, Le Y, Zhang X, Gong W, Abe K, Sun R, Van Damme J, Proost P, Wang JM: Up-regulation of FPR2, a chemotactic receptor for amyloid beta 1-42 (A beta 42), in murine microglial cells by TNF alpha. Neurobiol Dis. 2002, 10: 366-377. 10.1006/nbdi.2002.0517.CrossRefPubMed
21.
Zurück zum Zitat Chen K, Iribarren P, Hu J, Chen J, Gong W, Cho EH, Lockett S, Dunlop NM, Wang JM: Activation of Toll-like receptor 2 on microglia promotes cell uptake of Alzheimer disease-associated amyloid beta peptide. J Biol Chem. 2006, 281: 3651-3659.CrossRefPubMed Chen K, Iribarren P, Hu J, Chen J, Gong W, Cho EH, Lockett S, Dunlop NM, Wang JM: Activation of Toll-like receptor 2 on microglia promotes cell uptake of Alzheimer disease-associated amyloid beta peptide. J Biol Chem. 2006, 281: 3651-3659.CrossRefPubMed
22.
Zurück zum Zitat Braun BJ, Slowik A, Leib SL, Lucius R, Varoga D, Wruck CJ, Jansen S, Podschun R, Pufe T, Brandenburg LO: The formyl peptide receptor like-1 and scavenger receptor MARCO are involved in glial cell activation in bacterial meningitis. J Neuroinflammation. 2011, 8: 11-10.1186/1742-2094-8-11.PubMedCentralCrossRefPubMed Braun BJ, Slowik A, Leib SL, Lucius R, Varoga D, Wruck CJ, Jansen S, Podschun R, Pufe T, Brandenburg LO: The formyl peptide receptor like-1 and scavenger receptor MARCO are involved in glial cell activation in bacterial meningitis. J Neuroinflammation. 2011, 8: 11-10.1186/1742-2094-8-11.PubMedCentralCrossRefPubMed
23.
Zurück zum Zitat Decker Y, McBean G, Godson C: Lipoxin A4 inhibits IL-1beta-induced IL-8 and ICAM-1 expression in 1321N1 human astrocytoma cells. Am J Physiol Cell Physiol. 2009, 296: C1420-1427. 10.1152/ajpcell.00380.2008.CrossRefPubMed Decker Y, McBean G, Godson C: Lipoxin A4 inhibits IL-1beta-induced IL-8 and ICAM-1 expression in 1321N1 human astrocytoma cells. Am J Physiol Cell Physiol. 2009, 296: C1420-1427. 10.1152/ajpcell.00380.2008.CrossRefPubMed
24.
Zurück zum Zitat Hanisch UK, Kettenmann H: Microglia: active sensor and versatile effector cells in the normal and pathologic brain. Nat Neurosci. 2007, 10: 1387-1394. 10.1038/nn1997.CrossRefPubMed Hanisch UK, Kettenmann H: Microglia: active sensor and versatile effector cells in the normal and pathologic brain. Nat Neurosci. 2007, 10: 1387-1394. 10.1038/nn1997.CrossRefPubMed
25.
Zurück zum Zitat Qin L, Wu X, Block ML, Liu Y, Breese GR, Hong JS, Knapp DJ, Crews FT: Systemic LPS causes chronic neuroinflammation and progressive neurodegeneration. Glia. 2007, 55: 453-462. 10.1002/glia.20467.PubMedCentralCrossRefPubMed Qin L, Wu X, Block ML, Liu Y, Breese GR, Hong JS, Knapp DJ, Crews FT: Systemic LPS causes chronic neuroinflammation and progressive neurodegeneration. Glia. 2007, 55: 453-462. 10.1002/glia.20467.PubMedCentralCrossRefPubMed
26.
Zurück zum Zitat Wilms H, Sievers J, Rickert U, Rostami-Yazdi M, Mrowietz U, Lucius R: Dimethylfumarate inhibits microglial and astrocytic inflammation by suppressing the synthesis of nitric oxide, IL-1beta, TNF-alpha and IL-6 in an in-vitro model of brain inflammation. J Neuroinflammation. 2010, 7: 30-10.1186/1742-2094-7-30.PubMedCentralCrossRefPubMed Wilms H, Sievers J, Rickert U, Rostami-Yazdi M, Mrowietz U, Lucius R: Dimethylfumarate inhibits microglial and astrocytic inflammation by suppressing the synthesis of nitric oxide, IL-1beta, TNF-alpha and IL-6 in an in-vitro model of brain inflammation. J Neuroinflammation. 2010, 7: 30-10.1186/1742-2094-7-30.PubMedCentralCrossRefPubMed
27.
Zurück zum Zitat Livak KJ, Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001, 25: 402-408. 10.1006/meth.2001.1262.CrossRefPubMed Livak KJ, Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001, 25: 402-408. 10.1006/meth.2001.1262.CrossRefPubMed
28.
Zurück zum Zitat Koistinaho M, Koistinaho J: Role of p38 and p44/42 mitogen-activated protein kinases in microglia. Glia. 2002, 40: 175-183. 10.1002/glia.10151.CrossRefPubMed Koistinaho M, Koistinaho J: Role of p38 and p44/42 mitogen-activated protein kinases in microglia. Glia. 2002, 40: 175-183. 10.1002/glia.10151.CrossRefPubMed
29.
Zurück zum Zitat Vaughn MW, Proske RJ, Haviland DL: Identification, cloning, and functional characterization of a murine lipoxin A4 receptor homologue gene. J Immunol. 2002, 169: 3363-3369.CrossRefPubMed Vaughn MW, Proske RJ, Haviland DL: Identification, cloning, and functional characterization of a murine lipoxin A4 receptor homologue gene. J Immunol. 2002, 169: 3363-3369.CrossRefPubMed
30.
Zurück zum Zitat Ye RD, Boulay F, Wang JM, Dahlgren C, Gerard C, Parmentier M, Serhan CN, Murphy PM: International Union of Basic and Clinical Pharmacology. LXXIII. Nomenclature for the formyl peptide receptor (FPR) family. Pharmacol Rev. 2009, 61: 119-161. 10.1124/pr.109.001578.PubMedCentralCrossRefPubMed Ye RD, Boulay F, Wang JM, Dahlgren C, Gerard C, Parmentier M, Serhan CN, Murphy PM: International Union of Basic and Clinical Pharmacology. LXXIII. Nomenclature for the formyl peptide receptor (FPR) family. Pharmacol Rev. 2009, 61: 119-161. 10.1124/pr.109.001578.PubMedCentralCrossRefPubMed
31.
Zurück zum Zitat John CD, Sahni V, Mehet D, Morris JF, Christian HC, Perretti M, Flower RJ, Solito E, Buckingham JC: Formyl peptide receptors and the regulation of ACTH secretion: targets for annexin A1, lipoxins, and bacterial peptides. FASEB J. 2007, 21: 1037-1046. 10.1096/fj.06-7299com.PubMedCentralCrossRefPubMed John CD, Sahni V, Mehet D, Morris JF, Christian HC, Perretti M, Flower RJ, Solito E, Buckingham JC: Formyl peptide receptors and the regulation of ACTH secretion: targets for annexin A1, lipoxins, and bacterial peptides. FASEB J. 2007, 21: 1037-1046. 10.1096/fj.06-7299com.PubMedCentralCrossRefPubMed
32.
Zurück zum Zitat Liberles SD, Horowitz LF, Kuang D, Contos JJ, Wilson KL, Siltberg-Liberles J, Liberles DA, Buck LB: Formyl peptide receptors are candidate chemosensory receptors in the vomeronasal organ. Proc Natl Acad Sci USA. 2009, 106: 9842-9847. 10.1073/pnas.0904464106.PubMedCentralCrossRefPubMed Liberles SD, Horowitz LF, Kuang D, Contos JJ, Wilson KL, Siltberg-Liberles J, Liberles DA, Buck LB: Formyl peptide receptors are candidate chemosensory receptors in the vomeronasal organ. Proc Natl Acad Sci USA. 2009, 106: 9842-9847. 10.1073/pnas.0904464106.PubMedCentralCrossRefPubMed
33.
Zurück zum Zitat Svensson CI, Zattoni M, Serhan CN: Lipoxins and aspirin-triggered lipoxin inhibit inflammatory pain processing. J Exp Med. 2007, 204: 245-252. 10.1084/jem.20061826.PubMedCentralCrossRefPubMed Svensson CI, Zattoni M, Serhan CN: Lipoxins and aspirin-triggered lipoxin inhibit inflammatory pain processing. J Exp Med. 2007, 204: 245-252. 10.1084/jem.20061826.PubMedCentralCrossRefPubMed
34.
Zurück zum Zitat Tambuyzer BR, Ponsaerts P, Nouwen EJ: Microglia: gatekeepers of central nervous system immunology. J Leukoc Biol. 2009, 85: 352-370.CrossRefPubMed Tambuyzer BR, Ponsaerts P, Nouwen EJ: Microglia: gatekeepers of central nervous system immunology. J Leukoc Biol. 2009, 85: 352-370.CrossRefPubMed
35.
Zurück zum Zitat Pacher P, Beckman JS, Liaudet L: Nitric oxide and peroxynitrite in health and disease. Physiol Rev. 2007, 87: 315-424. 10.1152/physrev.00029.2006.PubMedCentralCrossRefPubMed Pacher P, Beckman JS, Liaudet L: Nitric oxide and peroxynitrite in health and disease. Physiol Rev. 2007, 87: 315-424. 10.1152/physrev.00029.2006.PubMedCentralCrossRefPubMed
36.
Zurück zum Zitat Ock J, Han HS, Hong SH, Lee SY, Han YM, Kwon BM, Suk K: Obovatol attenuates microglia-mediated neuroinflammation by modulating redox regulation. Br J Pharmacol. 2010, 159: 1646-1662. 10.1111/j.1476-5381.2010.00659.x.PubMedCentralCrossRefPubMed Ock J, Han HS, Hong SH, Lee SY, Han YM, Kwon BM, Suk K: Obovatol attenuates microglia-mediated neuroinflammation by modulating redox regulation. Br J Pharmacol. 2010, 159: 1646-1662. 10.1111/j.1476-5381.2010.00659.x.PubMedCentralCrossRefPubMed
37.
Zurück zum Zitat Kaushik DK, Gupta M, Das S, Basu A: Kruppel-like factor 4, a novel transcription factor regulates microglial activation and subsequent neuroinflammation. J Neuroinflammation. 2010, 7: 68-10.1186/1742-2094-7-68.PubMedCentralCrossRefPubMed Kaushik DK, Gupta M, Das S, Basu A: Kruppel-like factor 4, a novel transcription factor regulates microglial activation and subsequent neuroinflammation. J Neuroinflammation. 2010, 7: 68-10.1186/1742-2094-7-68.PubMedCentralCrossRefPubMed
38.
Zurück zum Zitat Jozsef L, Zouki C, Petasis NA, Serhan CN, Filep JG: Lipoxin A4 and aspirin-triggered 15-epi-lipoxin A4 inhibit peroxynitrite formation, NF-kappa B and AP-1 activation, and IL-8 gene expression in human leukocytes. Proc Natl Acad Sci USA. 2002, 99: 13266-13271. 10.1073/pnas.202296999.PubMedCentralCrossRefPubMed Jozsef L, Zouki C, Petasis NA, Serhan CN, Filep JG: Lipoxin A4 and aspirin-triggered 15-epi-lipoxin A4 inhibit peroxynitrite formation, NF-kappa B and AP-1 activation, and IL-8 gene expression in human leukocytes. Proc Natl Acad Sci USA. 2002, 99: 13266-13271. 10.1073/pnas.202296999.PubMedCentralCrossRefPubMed
39.
Zurück zum Zitat Jin SW, Zhang L, Lian QQ, Liu D, Wu P, Yao SL, Ye DY: Posttreatment with aspirin-triggered lipoxin A4 analog attenuates lipopolysaccharide-induced acute lung injury in mice: the role of heme oxygenase-1. Anesth Analg. 2007, 104: 369-377. 10.1213/01.ane.0000252414.00363.c4.CrossRefPubMed Jin SW, Zhang L, Lian QQ, Liu D, Wu P, Yao SL, Ye DY: Posttreatment with aspirin-triggered lipoxin A4 analog attenuates lipopolysaccharide-induced acute lung injury in mice: the role of heme oxygenase-1. Anesth Analg. 2007, 104: 369-377. 10.1213/01.ane.0000252414.00363.c4.CrossRefPubMed
40.
Zurück zum Zitat Zhao X, Bausano B, Pike BR, Newcomb-Fernandez JK, Wang KK, Shohami E, Ringger NC, DeFord SM, Anderson DK, Hayes RL: TNF-alpha stimulates caspase-3 activation and apoptotic cell death in primary septo-hippocampal cultures. J Neurosci Res. 2001, 64: 121-131. 10.1002/jnr.1059.CrossRefPubMed Zhao X, Bausano B, Pike BR, Newcomb-Fernandez JK, Wang KK, Shohami E, Ringger NC, DeFord SM, Anderson DK, Hayes RL: TNF-alpha stimulates caspase-3 activation and apoptotic cell death in primary septo-hippocampal cultures. J Neurosci Res. 2001, 64: 121-131. 10.1002/jnr.1059.CrossRefPubMed
41.
Zurück zum Zitat Zou JY, Crews FT: TNF alpha potentiates glutamate neurotoxicity by inhibiting glutamate uptake in organotypic brain slice cultures: neuroprotection by NF kappa B inhibition. Brain Res. 2005, 1034: 11-24. 10.1016/j.brainres.2004.11.014.CrossRefPubMed Zou JY, Crews FT: TNF alpha potentiates glutamate neurotoxicity by inhibiting glutamate uptake in organotypic brain slice cultures: neuroprotection by NF kappa B inhibition. Brain Res. 2005, 1034: 11-24. 10.1016/j.brainres.2004.11.014.CrossRefPubMed
42.
Zurück zum Zitat Wu SH, Liao PY, Dong L, Chen ZQ: Signal pathway involved in inhibition by lipoxin A(4) of production of interleukins induced in endothelial cells by lipopolysaccharide. Inflamm Res. 2008, 57: 430-437. 10.1007/s00011-008-7147-1.CrossRefPubMed Wu SH, Liao PY, Dong L, Chen ZQ: Signal pathway involved in inhibition by lipoxin A(4) of production of interleukins induced in endothelial cells by lipopolysaccharide. Inflamm Res. 2008, 57: 430-437. 10.1007/s00011-008-7147-1.CrossRefPubMed
43.
Zurück zum Zitat Kure I, Nishiumi S, Nishitani Y, Tanoue T, Ishida T, Mizuno M, Fujita T, Kutsumi H, Arita M, Azuma T, Yoshida M: Lipoxin A(4) reduces lipopolysaccharide-induced inflammation in macrophages and intestinal epithelial cells through inhibition of nuclear factor-kappaB activation. J Pharmacol Exp Ther. 2010, 332: 541-548. 10.1124/jpet.109.159046.CrossRefPubMed Kure I, Nishiumi S, Nishitani Y, Tanoue T, Ishida T, Mizuno M, Fujita T, Kutsumi H, Arita M, Azuma T, Yoshida M: Lipoxin A(4) reduces lipopolysaccharide-induced inflammation in macrophages and intestinal epithelial cells through inhibition of nuclear factor-kappaB activation. J Pharmacol Exp Ther. 2010, 332: 541-548. 10.1124/jpet.109.159046.CrossRefPubMed
44.
Zurück zum Zitat Medeiros R, Rodrigues GB, Figueiredo CP, Rodrigues EB, Grumman A, Menezes-de-Lima O, Passos GF, Calixto JB: Molecular mechanisms of topical anti-inflammatory effects of lipoxin A(4) in endotoxin-induced uveitis. Mol Pharmacol. 2008, 74: 154-161. 10.1124/mol.108.046870.CrossRefPubMed Medeiros R, Rodrigues GB, Figueiredo CP, Rodrigues EB, Grumman A, Menezes-de-Lima O, Passos GF, Calixto JB: Molecular mechanisms of topical anti-inflammatory effects of lipoxin A(4) in endotoxin-induced uveitis. Mol Pharmacol. 2008, 74: 154-161. 10.1124/mol.108.046870.CrossRefPubMed
45.
Zurück zum Zitat Mattson MP: NF-kappaB in the survival and plasticity of neurons. Neurochem Res. 2005, 30: 883-893. 10.1007/s11064-005-6961-x.CrossRefPubMed Mattson MP: NF-kappaB in the survival and plasticity of neurons. Neurochem Res. 2005, 30: 883-893. 10.1007/s11064-005-6961-x.CrossRefPubMed
46.
Zurück zum Zitat O'Neill LA, Kaltschmidt C: NF-kappa B: a crucial transcription factor for glial and neuronal cell function. Trends Neurosci. 1997, 20: 252-258. 10.1016/S0166-2236(96)01035-1.CrossRefPubMed O'Neill LA, Kaltschmidt C: NF-kappa B: a crucial transcription factor for glial and neuronal cell function. Trends Neurosci. 1997, 20: 252-258. 10.1016/S0166-2236(96)01035-1.CrossRefPubMed
47.
Zurück zum Zitat Gewirtz AT, Collier-Hyams LS, Young AN, Kucharzik T, Guilford WJ, Parkinson JF, Williams IR, Neish AS, Madara JL: Lipoxin a4 analogs attenuate induction of intestinal epithelial proinflammatory gene expression and reduce the severity of dextran sodium sulfate-induced colitis. J Immunol. 2002, 168: 5260-5267.CrossRefPubMed Gewirtz AT, Collier-Hyams LS, Young AN, Kucharzik T, Guilford WJ, Parkinson JF, Williams IR, Neish AS, Madara JL: Lipoxin a4 analogs attenuate induction of intestinal epithelial proinflammatory gene expression and reduce the severity of dextran sodium sulfate-induced colitis. J Immunol. 2002, 168: 5260-5267.CrossRefPubMed
48.
Zurück zum Zitat Lu YC, Yeh WC, Ohashi PS: LPS/TLR4 signal transduction pathway. Cytokine. 2008, 42: 145-151. 10.1016/j.cyto.2008.01.006.CrossRefPubMed Lu YC, Yeh WC, Ohashi PS: LPS/TLR4 signal transduction pathway. Cytokine. 2008, 42: 145-151. 10.1016/j.cyto.2008.01.006.CrossRefPubMed
49.
Zurück zum Zitat Wu SH, Wu XH, Lu C, Dong L, Zhou GP, Chen ZQ: Lipoxin A4 inhibits connective tissue growth factor-induced production of chemokines in rat mesangial cells. Kidney Int. 2006, 69: 248-256. 10.1038/sj.ki.5000025.CrossRefPubMed Wu SH, Wu XH, Lu C, Dong L, Zhou GP, Chen ZQ: Lipoxin A4 inhibits connective tissue growth factor-induced production of chemokines in rat mesangial cells. Kidney Int. 2006, 69: 248-256. 10.1038/sj.ki.5000025.CrossRefPubMed
50.
Zurück zum Zitat Ohira T, Bannenberg G, Arita M, Takahashi M, Ge Q, Van Dyke TE, Stahl GL, Serhan CN, Badwey JA: A stable aspirin-triggered lipoxin A4 analog blocks phosphorylation of leukocyte-specific protein 1 in human neutrophils. J Immunol. 2004, 173: 2091-2098.CrossRefPubMed Ohira T, Bannenberg G, Arita M, Takahashi M, Ge Q, Van Dyke TE, Stahl GL, Serhan CN, Badwey JA: A stable aspirin-triggered lipoxin A4 analog blocks phosphorylation of leukocyte-specific protein 1 in human neutrophils. J Immunol. 2004, 173: 2091-2098.CrossRefPubMed
51.
Zurück zum Zitat Serhan CN: Lipoxins and aspirin-triggered 15-epi-lipoxins are the first lipid mediators of endogenous anti-inflammation and resolution. Prostaglandins Leukot Essent Fatty Acids. 2005, 73: 141-162. 10.1016/j.plefa.2005.05.002.CrossRefPubMed Serhan CN: Lipoxins and aspirin-triggered 15-epi-lipoxins are the first lipid mediators of endogenous anti-inflammation and resolution. Prostaglandins Leukot Essent Fatty Acids. 2005, 73: 141-162. 10.1016/j.plefa.2005.05.002.CrossRefPubMed
52.
Zurück zum Zitat Ereso AQ, Cureton EL, Cripps MW, Sadjadi J, Dua MM, Curran B, Victorino GP: Lipoxin a(4) attenuates microvascular fluid leak during inflammation. J Surg Res. 2009, 156: 183-188. 10.1016/j.jss.2009.01.009.PubMedCentralCrossRefPubMed Ereso AQ, Cureton EL, Cripps MW, Sadjadi J, Dua MM, Curran B, Victorino GP: Lipoxin a(4) attenuates microvascular fluid leak during inflammation. J Surg Res. 2009, 156: 183-188. 10.1016/j.jss.2009.01.009.PubMedCentralCrossRefPubMed
53.
Zurück zum Zitat Prieto P, Cuenca J, Traves PG, Fernandez-Velasco M, Martin-Sanz P, Bosca L: Lipoxin A4 impairment of apoptotic signaling in macrophages: implication of the PI3K/Akt and the ERK/Nrf-2 defense pathways. Cell Death Differ. 2010, 17: 1179-1188. 10.1038/cdd.2009.220.CrossRefPubMed Prieto P, Cuenca J, Traves PG, Fernandez-Velasco M, Martin-Sanz P, Bosca L: Lipoxin A4 impairment of apoptotic signaling in macrophages: implication of the PI3K/Akt and the ERK/Nrf-2 defense pathways. Cell Death Differ. 2010, 17: 1179-1188. 10.1038/cdd.2009.220.CrossRefPubMed
54.
Zurück zum Zitat Simoes RL, Niconi-de-Almeida Y, da-Fe AR, Barja-Fidalgo C, Fierro IM: A synthetic analog of 15-epi-lipoxin A4 inhibits human monocyte apoptosis: involvement of ERK-2 and PI3-kinase. Prostaglandins Other Lipid Mediat. 2010, 91: 10-17. 10.1016/j.prostaglandins.2009.12.001.CrossRefPubMed Simoes RL, Niconi-de-Almeida Y, da-Fe AR, Barja-Fidalgo C, Fierro IM: A synthetic analog of 15-epi-lipoxin A4 inhibits human monocyte apoptosis: involvement of ERK-2 and PI3-kinase. Prostaglandins Other Lipid Mediat. 2010, 91: 10-17. 10.1016/j.prostaglandins.2009.12.001.CrossRefPubMed
Metadaten
Titel
Aspirin-triggered lipoxin A4attenuates LPS-induced pro-inflammatory responses by inhibiting activation of NF-κB and MAPKs in BV-2 microglial cells
verfasst von
Yan-Ping Wang
Yan Wu
Long-Yan Li
Jin Zheng
Ren-Gang Liu
Jie-Ping Zhou
Shi-Ying Yuan
You Shang
Shang-Long Yao
Publikationsdatum
01.12.2011
Verlag
BioMed Central
Erschienen in
Journal of Neuroinflammation / Ausgabe 1/2011
Elektronische ISSN: 1742-2094
DOI
https://doi.org/10.1186/1742-2094-8-95

Weitere Artikel der Ausgabe 1/2011

Journal of Neuroinflammation 1/2011 Zur Ausgabe

Neu in den Fachgebieten Neurologie und Psychiatrie

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Frühe Alzheimertherapie lohnt sich

25.04.2024 AAN-Jahrestagung 2024 Nachrichten

Ist die Tau-Last noch gering, scheint der Vorteil von Lecanemab besonders groß zu sein. Und beginnen Erkrankte verzögert mit der Behandlung, erreichen sie nicht mehr die kognitive Leistung wie bei einem früheren Start. Darauf deuten neue Analysen der Phase-3-Studie Clarity AD.

Viel Bewegung in der Parkinsonforschung

25.04.2024 Parkinson-Krankheit Nachrichten

Neue arznei- und zellbasierte Ansätze, Frühdiagnose mit Bewegungssensoren, Rückenmarkstimulation gegen Gehblockaden – in der Parkinsonforschung tut sich einiges. Auf dem Deutschen Parkinsonkongress ging es auch viel um technische Innovationen.

Demenzkranke durch Antipsychotika vielfach gefährdet

23.04.2024 Demenz Nachrichten

Wenn Demenzkranke aufgrund von Symptomen wie Agitation oder Aggressivität mit Antipsychotika behandelt werden, sind damit offenbar noch mehr Risiken verbunden als bislang angenommen.