Skip to main content
Erschienen in: Journal of Neuroinflammation 1/2010

Open Access 01.12.2010 | Research

Luteolin triggers global changes in the microglial transcriptome leading to a unique anti-inflammatory and neuroprotective phenotype

verfasst von: Konstantin Dirscherl, Marcus Karlstetter, Stefanie Ebert, Dominik Kraus, Julia Hlawatsch, Yana Walczak, Christoph Moehle, Rudolf Fuchshofer, Thomas Langmann

Erschienen in: Journal of Neuroinflammation | Ausgabe 1/2010

Abstract

Background

Luteolin, a plant derived flavonoid, exerts a variety of pharmacological activities and anti-oxidant properties associated with its capacity to scavenge oxygen and nitrogen species. Luteolin also shows potent anti-inflammatory activities by inhibiting nuclear factor kappa B (NFkB) signaling in immune cells. To better understand the immuno-modulatory effects of this important flavonoid, we performed a genome-wide expression analysis in pro-inflammatory challenged microglia treated with luteolin and conducted a phenotypic and functional characterization.

Methods

Resting and LPS-activated BV-2 microglia were treated with luteolin in various concentrations and mRNA levels of pro-inflammatory markers were determined. DNA microarray experiments and bioinformatic data mining were performed to capture global transcriptomic changes following luteolin stimulation of microglia. Extensive qRT-PCR analyses were carried out for an independent confirmation of newly identified luteolin-regulated transcripts. The activation state of luteolin-treated microglia was assessed by morphological characterization. Microglia-mediated neurotoxicity was assessed by quantifying secreted nitric oxide levels and apoptosis of 661W photoreceptors cultured in microglia-conditioned medium.

Results

Luteolin dose-dependently suppressed pro-inflammatory marker expression in LPS-activated microglia and triggered global changes in the microglial transcriptome with more than 50 differentially expressed transcripts. Pro-inflammatory and pro-apoptotic gene expression was effectively blocked by luteolin. In contrast, mRNA levels of genes related to anti-oxidant metabolism, phagocytic uptake, ramification, and chemotaxis were significantly induced. Luteolin treatment had a major effect on microglial morphology leading to ramification of formerly amoeboid cells associated with the formation of long filopodia. When co-incubated with luteolin, LPS-activated microglia showed strongly reduced NO secretion and significantly decreased neurotoxicity on 661W photoreceptor cultures.

Conclusions

Our findings confirm the inhibitory effects of luteolin on pro-inflammatory cytokine expression in microglia. Moreover, our transcriptomic 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. With the identification of several novel luteolin-regulated genes, our findings provide a molecular basis to understand the versatile effects of luteolin on microglial homeostasis. The data also suggest that luteolin could be a promising candidate to develop immuno-modulatory and neuroprotective therapies for the treatment of neurodegenerative disorders.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1742-2094-7-3) contains supplementary material, which is available to authorized users.
Konstantin Dirscherl, Marcus Karlstetter, Stefanie Ebert contributed equally to this work.
An erratum to this article is available at http://​dx.​doi.​org/​10.​1186/​1742-2094-9-118.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

KD, SE, and DK carried out all cell cultures and qRT-PCR experiments. MK, JH, and YW analyzed qRT-PCR and functional data. CM performed microarray hybridizations and raw data analyses. RF analyzed microarray data and critically read the manuscript. TL designed the study, obtained funding, carried out biostatistical analyses of microarrays and wrote the manuscript. All authors read and approved the final manuscript.

Background

Microglia, the resident macrophages of the nervous system, have important roles in immune regulation [1, 2] and neuronal homeostasis [3, 4]. Microglia belong to the mononuclear phagocyte system but their special localization in the fragile neuronal environment and their morphological features clearly distinguish them from other peripheral macrophages [5]. Ramified microglia perform a very active and continous surveillance function with their long protrusions [6, 7]. They receive permanent tonic inhibitory inputs from neurons to prevent microglial neurotoxicity [8, 9]. Loss of microglia-neuron cross-talk [10], local danger signals such as extracellular ATP [11], or neurotransmitter gradients [12] rapidly lead to a functional transformation of ramified microglia with a variety of effector functions.
Microglia activation is a protective mechanism regulating tissue repair and recovery in the early phase of neurodegeneration [4]. However, excessive or sustained activation of microglia often contributes to acute and chronic neuro-inflammatory responses in the brain and the retina [2]. Activated microglia in the vicinity of degenerating neurons have been identified in a broad spectrum of neurodegenerative disorders including Alzheimer's disease [13], Parkinson's disease [14], amyotrophic lateral sclerosis [15], multiple sclerosis [16], and inherited photoreceptor dystrophies [17, 18].
Macrophage heterogeneity and plasticity is very large and the set of marker combinations and sub-populations is essentially infinite [19]. To define a simplified conceptual framework, classification into polarized functional categories, called M1 and M2 macrophages has been proposed [20, 21]. M1 or "classically activated" macrophages produce high levels of oxidative metabolites and pro-inflammatory cytokines but also cause damage to healthy tissue as side effect [22]. M2 or "alternatively activated" macrophages promote tissue remodeling and generally suppress destructive immune reactions. Informations on microglial subsets in the nervous system are relatively scarce compared to other tissue macrophages. Nevertheless, recent findings from in vitro cultures of the murine microglial cell line MMGT12 [23] and hippocampal microglia from the PS1xAPP Alzheimer's mouse model [24] implicate that microglia have the ability to differentiate into M1 and M2 polarized phenotypes. A co-existence of neurotoxic M1 microglia and regenerative M2 microglia has been recently documented in the injured mouse spinal cord [25]. Microarray-based quantitation of M1 and M2 markers as well as functional tests on axonal regrowth after injury demonstrated that a transient anti-inflammatory and neuroprotective M2 response was rapidly overwhelmed by a neurotoxic M1 microglial response [25]. A similar but age-dependent switch from alternative to classical activation was shown in PS1xAPP Alzheimer's mice [24], indicating a common phenomenon in neurodegenerative disorders. Compounds that induce the switch of microglia from inflammatory M1 type to anti-inflammatory M2 type could therefore be a potential therapeutic agent to attenuate neuronal inflammation and boost neuronal recovery [26].
Several anti-inflammatory drugs have been shown to diminish neuroinflammation, but only a few direct functional effects on microglial activity have been elucidated [27]. Among the naturally occuring immuno-modulators, the flavonoid luteolin (3',4',5,7-tetrahydroxyflavone), abundant in parsley, green pepper, celery, perilla leaf, and chamomile tea, exerts prominent anti-inflammatory and anti-oxidant activities [28]. Luteolin suppressed pro-inflammatory cytokine production in macrophages by blocking nuclear factor kappa B (NFkB) and activator protein 1 (AP1) signaling pathways [29] and inhibited the production of nitric oxide [30] and pro-inflammatory eicosanoids [31]. Luteolin also diminshed the release of Tnf and superoxide anions in LPS or interferon-γ treated microglial cell cultures [32, 33] and reduced the LPS-induced Il6 production in brain microglia in vivo [34].
Although the inhibitory function of luteolin on NFkB and a few selected cytokines is well documented in macrophages, a genome-wide search for further molecular targets in microglia has not yet been published. Furthermore, the immuno-modulatory effects of luteolin related to the stimulation of distinct functional microglial phenotypes has not been investigated before. Therefore, this study investigated the global transcriptomic effects of luteolin at near physiological concentrations [35] alone or in combination with LPS in pure BV-2 microglial cultures. We further validated the luteolin-regulated expression of novel pro- and anti-inflammtory microglial transcripts, analyzed microglial morphology, and studied the consequences of microglia-conditioned media for photoreceptor viability.

Methods

Reagents

Luteolin (3',4',5,7-tetrahydroxyflavone) and E. coli 0111:B4 lipopolysaccharide were purchased from Sigma Aldrich (Steinheim, Germany). Luteolin was dissolved in DMSO and added in concentrations that did not exceed 0.05% of the total volume in any of the cell culture experiments.

Animals

C57BL/6 mice were purchased from Charles River Laboratories. Mice were kept in an air-conditioned barrier environment at constant temperature of 20-22°C on a 12-h light-dark schedule, and had free access to food and water. The health of the animals was regularly monitored, and all procedures were approved by the University of Regensburg animal rights committee and complied with the German Law on Animal Protection and the Institute for Laboratory Animal Research Guide for the Care and Use of Laboratory Animals, 1999.

Cell culture

Brain microglia were isolated and cultured as described earlier [36]. BV-2 microglia-like cells were provided by Professor Ralph Lucius (Clinic of Neurology, Christian Albrechts University, Kiel, Germany). BV-2 cells were cultured in RPMI/5% FCS supplemented with 2 mM L-Glutamine and 195 nM β-mercaptoethanol. Primary brain microglia or BV-2 cells were stimulated with 10 ng/ml or 50 ng/ml LPS and various concentrations of luteolin for 24 h. 661W photoreceptor-like cells were a gift from Prof. Muayyad Al-Ubaidi (University of Illinois, Chicago, IL) and the culture conditions have been described elsewhere [36].

Phalloidin staining

BV-2 cells were plated overnight on coverslips, fixed with 3.7% paraformaldehyde for 10 min at 37°C, permeabilized with 0.2% Triton X-100 for 5 min, blocked with 5% non-fat milk, 0.2% Triton X-100, and stained with DAPI for 10 min at room temperature (0.1 μg/ml in PBS, 4',6-diamidino-2-phenylindol, Molecular Probes). Filamentous actin was stained by addition of 1.5 μM TRITC-conjugated phalloidin (Sigma). The coverslips were mounted on microscopic glass slides and viewed with a Axioskop 2 fluorescence microscope equipped with an Eclipse digital analyzer (Carl Zeiss).

NO measurement

NO concentrations were determined by measuring the amount of nitrite secreted by BV-2 cells into the culture medium using the Griess reagent system (Promega). 50 μl cell culture supernatant was collected and an equal volume of Griess reagent was added to each well. After incubation for 15 min at room temperature, the absorbance was read at 540 nm on a BMG FluoStar Optima plate reader (Labtech, Offenburg, Germany). The concentration of nitrite for each sample was calculated from a sodium nitrite standard curve.

Apoptosis assay

Apoptotic cell death of 661W cells was determined with the Caspase-Glo® 3/7 Assay (Promega). Cells were lysed and incubated with a luminogenic caspase-3/7 substrate, which contains the tetrapeptide sequence DEVD. Luminescence was then generated by addition of recombinant luciferase and was proportional to the amount of caspase activity present. The luminescent signal was read on a BMG FluoStar Optima plate reader (Labtech, Offenburg, Germany). A blank reaction was used to measure background luminescence associated with the cell culture system and Caspase-Glo® 3/7 Reagent. The value for the blank reaction was subtracted from all experimental values. Negative control reactions were performed to determine the basal caspase activity of 661W cells.

RNA isolation and reverse transcription

Total RNA was extracted from cultured microglia according to the manufacturer's instructions using the RNeasy Protect Mini Kit (Qiagen, Hilden, Germany). Purity and integrity of the RNA was assessed on the Agilent 2100 bioanalyzer with the RNA 6000 Nano LabChip® reagent set (Agilent Technologies, Büblingen, Germany). The RNA was quantified spectrophotometrically and then stored at -80°C. First-strand cDNA synthesis was performed with RevertAid™ H Minus First Strand cDNA Synthesis Kit (Fermentas, St. Leon-Rot, Germany).

DNA microarray analysis

Triplicate microarrays were carried out with three independent RNAs from non-stimulated BV-2 microglia or cultures treated for 24 h with 50 μM luteolin, 50 ng/ml LPS, or 50 μM LPS + 50 ng/ml LPS, respectively. Generation of double-stranded cDNA, preparation and labeling of cRNA, hybridization to Affymetrix 430 2.0 mouse genome arrays, washing, and scanning were performed according to the Affymetrix standard protocol. Minimum information about a microarray experiment (MIAME) criteria were met [37]. The microarray datasets of this study are publicly available at the National Center for Biotechnology Information Gene Expression Omnibus http://​www.​ncbi.​nlm.​nih.​gov/​geo/​ as series record GSE18740.

Bioinformatic data analysis

The Affymetrix Expression Console Software Version 1.0 was used to create summarized expression values (CHP-files) from 3' expression array feature intensities (CEL-files) using the Robust Multichip Analysis (RMA) algorithm. Integrative analysis of genome-wide expression activities from BV-2 cells was performed with the Gene Expression Dynamics Inspector (GEDI), a Matlab (Mathworks, Natick, MA) freeware program which uses self-organizing maps (SOMs) to translate high-dimensional data into a 2D mosaic [38]. Each tile of the mosaic represents an individual SOM cluster and is color-coded to represent high or low expression of the cluster's genes, thus identifying the underlying pattern.
Differentially regulated transcrips in 24 h luteolin stimulated versus non-treated and luteolin + LPS versus LPS-treated BV-2 cells, respectively, were retrieved with the Genomatix ChipInspector program (Genomatix Software GmbH, Munich, Germany), applying the Significance Analysis of Microarray (SAM) algorithm using a false-discovery rate of 0.1% and a minimum coverage of 3 independent probes.
Functional annotation of transcripts was performed using the Database for Annotation, Visualization, and Integrated Discovery (DAVID) [39] and the Bibliosphere pathway edition (Genomatix).

Quantitative real-time RT-PCR

Amplifications of 50 ng cDNA were performed with an ABI7900HT machine (Applied Biosystems) in triplicates in 10 μl reaction mixtures containing 1 × TaqMan Universal PCR Master Mix (Applied Biosystems), 200 nM of primers and 0.25 μl dual-labeled probe (Roche ProbeLibrary). The reaction parameters were as follows: 2-min 50°C hold, 30-min 60°C hold, and 5-min 95°C hold, followed by 45 cycles of 20-s 94°C melt and 1-min 60°C anneal/extension. Measurements were performed in triplicate. Results were analyzed with an ABI sequence detector software version 2.3 using the ΔΔCt method for relative quantitation. Primer sequences and Roche Library Probe numbers are listed in Table 1.
Table 1
Primer pairs and Roche library probes for real time qRT-PCR validation
Gene
F-Primer (5'-3')
R-Primer (5'-3')
Roche Library Probe
AA467197
aaatggtggatcctactcaacc
gttgccctccggactttt
17
Blvrb
tcctcggagttctcagcttt
gcaccgtcacctcataacct
81
C3
accttacctcggcaagtttct
ttgtagagctgctggtcagg
76
CD36
ttgaaaagtctcggacattgag
tcagatccgaacacagcgta
6
CD83
gctctcctatgcagtgtcctg
ggatcgtcagggaataggc
2
Cfb
ctcgaacctgcagatccac
tcaaagtcctgcggtcgt
1
Cst7
atgtcagcaaagccctggta
ggtcttcctgcatgtagttcg
67
Cxcl10
gctgccgtcattttctgc
tctcactggcccgtcatc
3
Ddit3
ccaccacacctgaaagcag
tcctcataccaggcttcca
33
Gbp2
tgtagaccaaaagttccagacaga
gataaaggcatctcgcttgg
62
Gbp3
aagattgagctgggctacca
gaaactcttgagaacctcttttgc
73
Gclm
tggagcagctgtatcagtgg
caaaggcagtcaaatctggtg
18
Gusb
gtgggcattgtgctacctg
atttttgtcccggcgaac
25
Hmox1
ctgctagcctggtgcaaga
ccaacaggaagctgagagtga
25
Hp
ccctgggagctgttgtca
ctttgggcagctgtcatctt
15
Hprt1
tcctcctcagaccgctttt
cctggttcatcatcgctaatc
95
Ifi44
ctgattacaaaagaagacatgacagac
aggcaaaaccaaagactcca
78
Ifitm3
aacatgcccagagaggtgtc
accatcttccgatccctagac
84
Ifitm6
ccggatcacattacctggtc
catgtcgcccaccatctt
27
IL-6
gatggatgctaccaaactggat
ccaggtagctatggtactccaga
6
iNos
ctttgccacggacgagac
tcattgtactctgagggctga
13
Irf7
cttcagcactttcttccgaga
tgtagtgtggtgacccttgc
25
Kdr
cagtggtactggcagctagaag
acaagcatacgggcttgttt
68
Lcn2
atgtcacctccatcctggtc
cctgtgcatatttcccagagt
1
Lpcat1
aatgtgaggcgtgtcatgg
ggcagtcctcaaatgtatagtcg
81
Marco
cagagggagagcacttagcag
gccccgacaattcacatt
20
Mpeg1
cacagtgagcctgcacttaca
gcgctttcccaatagcttta
69
Nupr1-F
gatggaatcctggatgaatatga
gtccgacctttccgacct
64
Rnf145
catggacttctggcttctcat
aataaaaagtgttcccagaacctg
67
Saa3
atgctcgggggaactatgat
acagcctctctggcatcg
26
Slpi
gtgaatcctgttcccattcg
cctgagttttgacgcacctc
69
Srxn1
gctatgccacacagagaccata
gtgggaaagctggtgtcct
33
Trib3
gctatcgagccctgcact
acatgctggtgggtaggc
98

Statistical analysis

Statistical analysis were performed on ΔΔCt data using analysis of variance with a two-sample Student's t test (P < 0.05) unless otherwise indicated. Quantitative data are expressed as mean ± SEM. The levels of gene expression in treated BV-2 cells are shown relative to control cells.

Results

Effects of luteolin on selected pro-inflammatory markers

As a basis to study the genome-wide transcriptional effects of luteolin on activated microglia and to validate our cell culture system, we initially performed a dose-response curve for luteolin. Four pro-inflammatory microglia markers with different expression levels and ranges of induction were selected as positive controls for qRT-PCR analyses. Interleukin 6 (Il6) is a well known pro-inflammatory cytokine target of luteolin [34], chemokine (C-X-C motif) ligand 10 (Cxcl10), interferon-regulatory factor 7 (Irf7), and interferon-inducible protein 44 (Ifi44) are LPS-sensitive genes in microglia [36]. BV-2 microglia were pre-treated with different concentrations of luteolin (0, 5, 10, 25, and 50 μM) for 1 h and then stimulated with LPS (10 ng/ml and 50 ng/ml) for a further 24 h period. Neither LPS nor luteolin changed the proliferation rate or cell survival at the concentration levels applied to the cells (data not shown). mRNA levels of Il6 (Fig. 1A), Cxcl10 (Fig. 1B), Irf7 (Fig. 1C), and Ifi44 (Fig. 1D), which were all induced by 10 ng/ml LPS and further increased by 50 ng/ml LPS, were dose-dependently reduced by luteolin (Fig. 1A-D). Luteolin's effects on the four genes were most prominent at 50 μM (Fig. 1A-D) and therefore this concentration was used in all further experiments.

Luteolin triggers global changes in the microglial transcriptome

Our next goal was to capture and compare the transcriptional profile of non-activated and LPS-activated BV-2 microgli treated each with 50 μM luteolin for 24 h using Affymetrix Mouse Genome 430 2.0 GeneChips. Twelve microarray analyses from three independent stimulation experiments were performed and high stringency criteria with a minimal signal intensity of 50 fluorescence units were used. The complete microarray RMA datasets and all raw data (Affymetrix CEL-files) were stored in the NCBI GEO repository as record GSE18740. We used the Gene Expression Dynamics Inspector (GEDI) to determine the global patterns of gene expression in the four different conditions, untreated, luteolin-treated, LPS-treated, and luteolin + LPS-treated microglia. GEDI is based on self-organizing maps to identify genome-wide transcriptome activity via 'gestalt' recognition [38]. GEDI is sample-oriented rather than gene-oriented, which facilitates the identification of genome-wide patterns. Each mosaic tile in the GEDI map represents a gene group or cluster that is expressed at similar levels, with blue color indicating a low level and red corresponding to high expression. The four GEDI maps clearly show a dynamic regulation of gene expression in stimulated versus non-stimulated microglia (Fig. 2). Especially the upper right positions in the control dataset and more pronounced in the LPS-treated dataset display an inverse regulation of the gene clusters following luteolin stimulation (Fig. 2A, white circles). These results demonstrate that stimulation with luteolin has a major impact on the global pattern of gene expression notably in activated microglia and to a lower extend in resting cells.
We have previously shown that a single probe analysis of Affymetrix microarrays using the Genomatix ChipInspector tool with the Significance Analysis of Microarray (SAM) algorithm improves detection of regulated transcripts [40]. We therefore used ChipInspector to analyze our dataset applying a false discovery rate (FDR) of 0.1%, a minimal probe coverage of 3, and a minimum log2 ratio of 1 (fold change of 2.0). Thereby, 18 significantly regulated genes were identified in luteolin-treated versus non-treated microglia (Table 2) and 54 differentially expressed genes were detected in luteolin + LPS versus LPS-stimulated cells (Table 3). Comparison of the total numer of differentially expressed genes indicated that the effects of luteolin were more pronounced in LPS-activated BV-2 cells than in resting microglia. Also, the overall fold change range was higher in the LPS-treated microglia than in the resting BV-2 cells when stimulated with luteolin. These finding correlate with the gene cluster patterns observed in GEDI analysis and implicate that luteolin is an effective counter-player of pro-inflammatory microglial activation.
Table 2
Differentially expressed transcripts after 24 h stimulation of BV-2 cells with 50 μM luteolin
Nr
ID
Symbol
Gene Name
FC*
Cov
UP-REGULATED
1
74315
Rnf145
Ring finger protein 145
2.87
11
2
76650
Srxn1
Sulfiredoxin 1 homolog
2.38
5
3
19252
Dusp1
Dual specificity phosphatase 1
2.35
10
4
12267
C3ar1
Complement component 3a receptor 1
2.30
22
5
12491
Cd36
CD36 antigen
2.28
10
6
12475
Cd14
CD14 antigen
2.25
10
7
233016
Blvrb
Biliverdin reductase B
2.22
9
8
15368
Hmox1
Heme oxygenase 1
2.17
10
9
210992
Lpcat1
Lysophosphatidylcholine acyltransferase 1
2.13
11
DOWN-REGULATED
1
13198
Ddit3
DNA-damage inducible transcript 3
-5.21
11
2
12862
Cox6a2
Cytochrome c oxidase, subunit VI a, polypeptide 2
-2.89
9
3
228775
Trib3
Tribbles homolog 3
-2.62
22
4
56312
Nupr1
Nuclear protein 1
-2.36
15
5
13011
Cst7
Cystatin F
-2.19
9
6
223920
Soat2
Sterol O-acyltransferase 2
-2.16
8
7
16149
Cd74
CD74 antigen
-2.10
8
8
17064
Cd93
CD93 antigen
-2.08
11
9
213002
Ifitm6
Interferon induced transmembrane protein 6
-2.06
8
Significance analysis of triplicate microarrays was performed with a false discovery rate of 0.1% and a minimum probe coverage of 3. ID, Entrez Gene ID; FC: Fold change; Cov: probe coverage; * validated by Taqman realtime RT-PCR
Table 3
Differentially expressed transcripts after 24 h stimulation with 50 μM luteolin + 50 ng/ml LPS versus 50 ng/ml LPS
Nr
ID
Symbol
Gene Name
FC*
Cov
UP-REGULATED
1
74315
Rnf145
Ring finger protein 145
3.27
11
2
210992
Lpcat1
Lysophosphatidylcholine acyltransferase 1
2.55
11
3
12522
Cd83
CD83 antigen
2.51
11
4
14630
Gclm
Glutamate-cysteine ligase, modifier subunit
2.46
10
5
16889
Lipa
Lysosomal acid lipase A
2.31
6
6
56336
B4galt5
UDP-Gal:betaGlcNAc beta 1,4-galactosyltransferase, polypeptide 5
2.30
8
7
14950
H13
Histocompatibility 13
2.30
7
8
14104
Fasn
Fatty acid synthase
2.28
4
9
12125
Bcl2l11
BCL2-like 11
2.16
14
10
16542
Kdr
Kinase insert domain protein receptor
2.16
9
11
216345
Ccdc131
Coiled-coil domain containing 131
2.11
7
DOWN-REGULATED
1
66141
Ifitm3
Interferon induced transmembrane protein 3
-13.74
9
2
433470
AA467197
Expressed sequence AA467197, miR-147
-9.32
9
3
16819
Lcn2
Lipocalin 2
-6.87
10
4
14469
Gbp2
Guanylate binding protein 2
-6.41
16
5
14962
Cfb
Complement factor B
-5.94
8
6
13198
Ddit3
DNA-damage inducible transcript 3
-5.58
11
7
16181
Il1rn
Interleukin 1 receptor antagonist
-5.28
33
8
55932
Gbp3
Guanylate binding protein 3
-4.72
10
9
56312
Nupr1
Nuclear protein 1
-4.35
15
10
75345
Slamf7
SLAM family member 7
-4.14
9
11
20210
Saa3
Serum amyloid A 3
-3.78
7
12
17386
Mmp13
Matrix metallopeptidase 13
-3.63
11
13
15439
Hp
Haptoglobin
-3.56
11
14
17167
Marco
Macrophage receptor with collagenous structure
-3.34
8
15
12266
C3
Complement component 3
-3.29
6
16
12870
Cp
Ceruloplasmin
-3.05
5
17
13011
Cst7
Cystatin F
-3.03
9
18
23882
Gadd45g
Growth arrest and DNA-damage-inducible 45 gamma
-2.95
11
19
242341
Atp6v0d2
ATPase, H+ transporting, lysosomal V0 subunit D2
-2.81
13
20
68774
Ms4a6d
Membrane-spanning 4-domains, subfamily A, member 6D
-2.75
19
21
83673
Snhg1
Small nucleolar RNA host gene 1
-2.75
24
22
12494
Cd38
CD38 antigen
-2.73
6
23
19655
Rbmx
RNA binding motif protein, X chromosome
-2.73
11
24
56405
Dusp14
Dual specificity phosphatase 14
-2.68
10
25
213002
Ifitm6
Interferon induced transmembrane protein 6
-2.57
8
26
12517
Cd72
CD72 antigen
-2.55
9
27
14129
Fcgr1
Fc receptor, IgG, high affinity I
-2.55
11
28
14130
Fcgr2b
Fc receptor, IgG, low affinity IIb
-2.48
27
29
231532
Arhgap24
Rho GTPase activating protein 24
-2.46
8
30
29818
Hspb7
Heat shock protein family, member 7 (cardiovascular)
-2.45
5
31
17476
Mpeg1
Macrophage expressed gene 1
-2.43
6
32
66222
Serpinb1a
Serine peptidase inhibitor, clade B, member 1a
-2.41
10
33
78771
Mctp1
Multiple C2 domains, transmembrane 1
-2.39
8
34
20568
Slpi
Secretory leukocyte peptidase inhibitor
-2.39
9
35
12507
Cd5
CD5 antigen
-2.35
9
36
50778
Rgs1
Regulator of G-protein signaling 1
-2.35
11
37
21897
Tlr1
Toll-like receptor 1
-2.20
5
38
16149
Cd74
CD74 antigen
-2.17
5
39
73167
Arhgap8
Rho GTPase activating protein 8
-2.13
8
40
15064
Mr1
Major histocompatibility complex, class I-related
-2.06
7
41
347722
Centg2
Centaurin, gamma 2
-2.04
11
42
98365
Slamf9
SLAM family member 9
-2.04
8
43
72999
Insig2
Insulin induced gene 2
-2.00
8
Significance analysis of triplicate microarrays was performed with a false discovery rate of 0.1% and a minimum probe coverage of 3. ID, Entrez Gene ID; FC: Fold change; Cov: probe coverage; * validated by Taqman realtime RT-PCR

Luteolin regulates important immune pathways and changes the microglial transcriptional phenotype

Our next aim was to put the newly identified luteolin-regulated expression patterns into a specific biological context. We used all differentially expressed genes to perform a classification into functional categories with the Database for Annotation, Visualization, and Integrated Discovery (DAVID). The major enriched functional categories in luteolin-treated resting microglia were anti-oxidants, phagocytosis, and anti-inflammation for up-regulated genes and pro-inflammation and apoptosis for down-regulated genes (Fig. 2B, upper part). These pathways indicate that luteolin stimulated anti-oxidant and anti-inflammatory transcriptional programs which could potentially reflect M2 macrophage polarization. Moreover, basal pro-inflammatory and pro-apoptotic gene expression was blocked in non-activated BV-2 microglia. In luteolin + LPS co-treated versus LPS-treated cells, 11 induced transcripts represented pathways of lipid metabolism, antigen presentation, and anti-oxidants (Fig. 2B, bottom left). Interestingly, a large number of down-regulated genes covered the pathway categories pro-inflammation, toll-like receptor (TLR) signaling, acute phase response, apoptosis, and complement factors (Fig. 2B, bottom right). These results implicate that luteolin-induced transcriptomic changes promote a stable anti-inflammatory, anti-oxidant, and anti-apoptotic microglial phenotype reminescent of M2 macrophages. Our microarray data also identified a significant number of genes that could not be grouped into larger immune-related pathways and that have not been previously connected to microglial activation or flavonoid stimulation (Tables 2 and 3).

Microarray validation by qRT-PCR confirms novel luteolin-regulated target genes

To validate a significant portion of the DNA microarray results, real-time qRT-PCR analyses were carried out with RNA samples from three independent BV-2 replicate experiments. We focused on representative candidates from different biological pathway which have not been previously described as flavonoid targets. 22 genes fulfilled these criteria and were analzyed with qRT-PCR. In the first set of experiments, mRNA levels of genes up-regulated by luteolin treatment were assessed (Fig. 3). Transcripts of Sulfirexodin 1 (Srxn1), Biliverdin reductase B (Blvrb), Heme oxigenase 1 (Hmox1), and Glutamate-cysteine ligase (Gclm) are components of the cellular anti-oxidant response [4143] and were all induced by luteolin treatment (Fig. 3A-D). We could further validate increased levels of Lysophosphatidylcholine acyltransferase (Lpcat1), Ring finger protein 145 (Rnf145), Cd36 antigen, Kinase insert domain receptor (Kdr, alias Vegfr2), and Cd83 antigen. Lpcat1 catalyzes the inactivation of inflammatory lipids [44], whereas Cd36 and Kdr are surface receptors required for phagocytic uptake and chemotactic migration, respectively [45, 46]. The function of Rnf145 is currently unknown.
In the next set of qRT-PCR experiments, down-regulated transcripts involved in pro-inflammatory activation and acute phase response were analyzed. Complement factor C3 (C3), Complement factor b (Cfb), Secreted leukocyte peptidase inhibitor (Slpi), and the pro-inflammatory Guanylate binding proteins 2 (Gbp2) and Gbp3 [47] showed diminished mRNA levels in luteolin-treated samples (Fig. 4A-D). The same tendency was seen in the newly identified microRNA miR-147, which is involved in toll-like receptor signaling [48], the acute phase gene Haptoglobin (Hp), the stress-related gene Nuclear protein 1 (Nupr1, alias p8) [49], and Cystatin 7 (Cst7) (Fig. 4F-I).
As third group of genes validated by qRT-PCR, four luteolin-repressed genes related to apoptosis and microglial shape formation were studied (Fig. 5A-D). The apoptotic mediators DNA-damage inducible transcript 3 (Ddit3, alias Chop, Gadd153) and Tribbles homolog 3 [50, 51] were down-regulated by luteolin in non-activated as well as LPS-activated microglia (Fig. 5A, B), indicating anti-apoptotic protection mechanisms elicited by luteolin. In line with these findings, we also observed reduced expression of the Macrophage receptor with collagenous structure (Marco) and Lipocalin 2 (Lcn2), two molecules with dual roles in apoptosis and de-ramification of activated microglia [52, 53].

Luteolin changes microglial morphology and inhibits NO secretion

To assess whether the particular gene expression profiles measured in luteolin-stimulated microglia translate into detectable functional properties, phenotypic characterization was performed. The general activation state and morphological phenotype of microglia is particularly reflected by their cell shape and cytoskeletal organization. To detected morphological changes and for visualization of filopodia we performed phalloidin staining. Conventional BV-2 microglia cultures were low level activated cells with a flat shape and some filopodia (Fig. 6A). Culture of BV-2 cells in the presence of luteolin lead to considerable ramification and formation of long protrusions (Fig. 6B), indicating induction of a surveillance state. LPS-activation of BV-2 cells caused formation of a round cell shape with retracted filopodia (Fig. 6C). In contrast, co-incubation of LPS-treated cells with luteolin sustained the ramified microglial morphology (Fig. 6D). A similar effect was seen in primary mouse microglia, where luteolin increased the length of filopodia in non-activated cells (Fig. 6E, F) and caused flatening and ramification of LPS-treated cells (Fig. 6G, H). To test the direct effect of luteolin on microglial secretion of toxic metabolites, NO levels were determined in the supernatant of BV-2 cells. Treatment of BV-2 cells with luteolin alone did not result in increased NO concentrations (Fig. 6I), whereas stimulation with LPS markedly increased secreted NO levels. Co-incubation of LPS-activated cells with luteolin nearly completely abolished NO secretion (Fig. 6I). These data indicate that luteolin favors the ramified surveillance state of microglia and effectively inhibits cytotoxic NO formation. Kim et al. previously reported that luteolin triggered a blockade of NO secretion in LPS-stimulated BV-2 microglia, which was mediated by inhibition of inducible NO synthase (iNos) protein expression [54]. To indenpendently verify these data by qRT-PCR, we analyzed iNos mRNA levels in BV-2 cells treated with luteolin, LPS, or both simultaneously. LPS caused a more than 40-fold increase in iNos transcripts and luteolin co-treatment significantly reduced iNos levels more than 2-fold. Thus, our expression data corroborate the findings by Kim et al. and provide a reasonable explanation for reduced NO secretion levels.
To test the hypothesis that luteolin leads to decreased microglial neurotoxicity, a culture system of 661W photoreceptor cells with conditioned medium (CM) from BV-2 cells or primary microglia was established. 661W, a retina-derived cell line [55], represents a valuable model to study microglial neurotoxicity in the special context of retinal degeneration [36]. 661W cells were incubated for 24 h with culture supernatants from unstimulated, luteolin-, LPS- or LPS + luteolin-treated cells and 661W photoreceptor cell morphology was assessed by phase contrast microscopy. 661W cells in their own medium grew confluent after 24 h and the presence of CM from control- or luteolin-treated BV-2 cells did not affect cell morphology (Fig. 7A, B). As described previously [56], confluent 661W cells flattened out and often multiple cells were connected to each other through their projected extensions (Fig. 7A, B). In contrast, 661W cells co-incubated with LPS-stimulated BV-2 supernatant appeared elongated and smaller, leading to prominent cell-free areas present in the culture (Fig. 7C). When adding CM from LPS + luteolin-stimulated BV-2 cells, nearly normal cell characteristics were retained (Fig. 7D). Similar morphological changes of 661W cells were observed when cultured in the presence of primary microglia CM (Fig. 7E-H). Direct incubation of photoreceptor cells with LPS, luteolin, or both had no effects on cell morphology (data not shown), indicating that the observed changes in 661W cell growth arise from secreted microglial compounds.
To correlate the microscopic findings with functional data, we next studied the influence of microglia-secreted products on caspase-related apoptotic signaling in the neuronal cell model. 661W cells cultured in the presence of CM from LPS-stimulated BV-2 cells displayed a strong induction of caspase 3/7 activity (Fig. 7I). When CM from microglia co-treated with LPS + luteolin was used, 661W apoptosis was still present but was significantly diminished (Fig. 7I). Culture media from BV-2 cells kept in the presence of luteolin alone had no effect on 661W apoptosis. These findings implicate that luteolin either stimulates microglia to produce less pro-apoptotic substances or actively promotes the release of survival signals.

Discussion

Like other plant-derived flavonoids, luteolin has a variety of biological activities including well-known anti-mutagenic and anti-tumorigenic properties [57]. Moreover, this flavone possesses direct anti-oxidant activity, which is attributed to structural features of all flavonoids, which favor scavenging of reactive oxygen and nitrogen species [58]. Although the anti-oxidant and anti-inflammatory activities of luteolin may be also useful in the treatment of many chronic inflammatory diseases including neurodegeneration, only little information is available about luteolin-mediated transcriptional mechanisms or molecular targets in microglia [59].
We have therefore performed the first genome-wide study of luteolin-mediated transcriptional effects in microglia. To our surprise, luteolin treatment did not only change expression levels of a few transcripts but had a broad and strong impact on the transcriptome of resting and particularly of LPS-activated microglia. The microarray dataset and the qRT-PCR validations revealed several luteolin-regulated pathways. Luteolin caused simultaneous up-regulation of four important anti-oxidant enzymes Srxn1, Blvrb, Gclm, and Hmox1. These data are consistent with earlier findings demonstrating increased Hmox1 transcription in RAW264.7 macrophages after luteolin treatment [60]. Stimulation with the flavonoid induced binding of the transcription factor NF-E2-related factor 2 (Nrf2) to anti-oxidant response elements (ARE) in the Hmox1 promoter region [60]. Luteolin is a potent activator of Nrf2 [61] and the majority of anti-oxidant enzymes contain ARE in their regulatory regions, including Srxn1 [62]. Moreover, mouse embryonic fibroblasts derived from Nrf2 -/- mice showed significantly lower Blvrb and Gclm mRNA levels upon Diquat induction [63]. Therefore, we speculate that increased microglial expression of Srxn1, Blvrb, and Gclm is also mediated by activation of Nrf2. This hypothesis is further corroborated by the protective functions of Nrf2 in several microglia-related neurodegenerative disorders [64].
Luteolin significantly enhanced mRNA synthesis of five other genes involved in different biological pathways. Lpcat1 is a lysophospholipid acyltransferase implicated in anti-inflammatory responses by converting lyso-platelet activation factor (lyso-PAF) to PAF and lyso-phosphatidylcholine (lyso-PC) to PC [65]. LPC exerts considerable neuro-inflammatory reactivity in the brain and inhibition of LPC signaling in astrocytes and microglia confers neuroprotection [66]. Lpcat1 is also highly expressed in the retina [44], indicating that luteolin-induced Lpact1 levels could lead to diminished LPC levels in retinal microglia. Rnf145 was also up-regulated by luteolin but the function of this protein remains to be determined. Cd36 and Kdr (alias Vegfr2) were also significantly induced by luteolin in non-activated as well as activated microglia. The pattern recognition receptor Cd36 signals to the actin cytoskeleton and regulates microglial migration and phagocytosis [67], whereas Kdr is involved in the chemotactic response of microglia [46]. We thus speculate that luteolin-mediated expression of both genes could result in increased phagocytic responses of microglia without inducing inflammation.
Several reports have demonstrated that luteolin inhibits pro-inflammatory cytokine expression in various cell types by blocking NFkB (reviewed in [28]). Our microarray data confirmed these findings and revealed further NFkB target genes including the recently discovered microRNA miR-147 [48]. Recently, Jang et al. showed that luteolin reduced Il6 production mainly by inhibiting JNK signaling and AP1 activation [34]. Luteolin did not affect IkB-α degradation or NFkB DNA binding in brain microglia, implicating that luteolin-mediated effects in microglia are not solely dependent on NFkB blockade [34]. In line with this notion, our luteolin-regulated expression profiles identified several genes with NFkB-independent promoter control. Likewise, luteolin down-regulated complement factor 3, which is regulated by AP1 [68] and blocked expression of Slpi, which is a target of interferon regulatory factor 1 (IRF1) [69]. Luteolin also diminished mRNA levels of the pro-inflammatory GTPase Gbp2 and the acute phase protein Haptoglobin, which are both regulated by signal transducers and activators of transcription (STATs) [70, 71]. These data clearly show that luteolin dampens microglia activation by interfering with several divergent signaling pathways.
The luteolin-regulated differential expression patterns also revealed genes involved in microglial apoptosis and ramification. Microglia are more susceptible than macrophages to apoptosis [72] and recent evidence indicates that microglial apoptosis and senescence may precede neurodegeneration [73]. Ddit 3 and Trib3, which were both induced by LPS and suppressed by luteolin, support stress and NO-mediated apoptosis [50, 51]. We therefore hypothesize that luteolin could promote the survival of activated and stressed microglia in an environment of early neurodegeneration. Our expression data also revealed the unexpected finding that luteolin down-regulated Lcn2 and Marco, two molecules involved in microglial ramification and formation of filopodia. Lee et al. demonstrated that stable expression of Lcn in BV-2 microglia, the same cell line we used in our experiments, induced a round cell shape with a loss of processes [52]. In line with this, over-expression of the scavenger receptor Marco in dendritic cells caused rounding of cells and down-regulated antigen uptake [53]. Thus, we hypothesized that the observed changes in mRNA levels of both genes might also translate into different morphological characters.
The morphological and functional assays fully supported the implications from gene expression profiles and revealed a direct effect of luteolin on the microglial phenotype. Luteolin stimulated the formation of filopodia and caused ramification of BV-2 cells and primary microglia even in the setting of strong LPS activation. Moreover, NO secretion was completely blocked in LPS-activated microglia upon co-incubation with luteolin. We studied the effects of conditioned media from microglia on cultured photoreceptor-like 661W cells and demonstrated that luteolin-treatment effectively protected 661W cells from LPS-induced microglial toxicity. Since NO and other reactive oxygen species are the major radicals secreted from microglia, we speculate that luteolin directly inhibits the secretion of these cytotoxic radicals. Our hypothesis is corroborated by recent data demonstrating that luteolin concentration-dependently attenuated LPS-induced dopaminergic neuron loss by blocking NO secretion from cultured rat microglia [32].

Conclusions

We have shown that the flavonoid luteolin is a potent modulator of microglial activation, cell shape, and effector functions. Luteolin induced global changes in the transcriptome of resting or LPS-activated microglia leading to a polarized M2-like phenotype with anti-inflammatory and neuroprotective characteristics. Luteolin's mechanisms of action appear to target several independent pathways independent of NFkB. Our results provide a basis to develop immuno-modulatory and neuroprotective therapies for the treatment of neurodegenerative disoders.

Acknowledgements

This work was supported by grants from the German Research Foundation (FOR1075 Project 4), the Elite Network of Bavaria, and the Pro Retina Foundation. The authors thank Prof. Muayyad Al Ubaidi for providing the 661W photoreceptor cell line.
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

KD, SE, and DK carried out all cell cultures and qRT-PCR experiments. MK, JH, and YW analyzed qRT-PCR and functional data. CM performed microarray hybridizations and raw data analyses. RF analyzed microarray data and critically read the manuscript. TL designed the study, obtained funding, carried out biostatistical analyses of microarrays and wrote the manuscript. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Hanisch UK: Microglia as a source and target of cytokines. Glia. 2002, 40: 140-155. 10.1002/glia.10161.CrossRefPubMed Hanisch UK: Microglia as a source and target of cytokines. Glia. 2002, 40: 140-155. 10.1002/glia.10161.CrossRefPubMed
2.
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
3.
Zurück zum Zitat Streit WJ: Microglia as neuroprotective, immunocompetent cells of the CNS. Glia. 2002, 40: 133-139. 10.1002/glia.10154.CrossRefPubMed Streit WJ: Microglia as neuroprotective, immunocompetent cells of the CNS. Glia. 2002, 40: 133-139. 10.1002/glia.10154.CrossRefPubMed
4.
Zurück zum Zitat Streit WJ: Microglia and neuroprotection: implications for Alzheimer's disease. Brain Res Brain Res Rev. 2005, 48: 234-239. 10.1016/j.brainresrev.2004.12.013.CrossRefPubMed Streit WJ: Microglia and neuroprotection: implications for Alzheimer's disease. Brain Res Brain Res Rev. 2005, 48: 234-239. 10.1016/j.brainresrev.2004.12.013.CrossRefPubMed
5.
Zurück zum Zitat Giulian D, Li J, Bartel S, Broker J, Li X, Kirkpatrick JB: Cell surface morphology identifies microglia as a distinct class of mononuclear phagocyte. J Neurosci. 1995, 15: 7712-7726.PubMed Giulian D, Li J, Bartel S, Broker J, Li X, Kirkpatrick JB: Cell surface morphology identifies microglia as a distinct class of mononuclear phagocyte. J Neurosci. 1995, 15: 7712-7726.PubMed
6.
Zurück zum Zitat Davalos D, Grutzendler J, Yang G, Kim JV, Zuo Y, Jung S, Littman DR, Dustin ML, Gan WB: ATP mediates rapid microglial response to local brain injury in vivo. Nat Neurosci. 2005, 8: 752-758. 10.1038/nn1472.CrossRefPubMed Davalos D, Grutzendler J, Yang G, Kim JV, Zuo Y, Jung S, Littman DR, Dustin ML, Gan WB: ATP mediates rapid microglial response to local brain injury in vivo. Nat Neurosci. 2005, 8: 752-758. 10.1038/nn1472.CrossRefPubMed
7.
Zurück zum Zitat Nimmerjahn A, Kirchhoff F, Helmchen F: Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science. 2005, 308: 1314-1318. 10.1126/science.1110647.CrossRefPubMed Nimmerjahn A, Kirchhoff F, Helmchen F: Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science. 2005, 308: 1314-1318. 10.1126/science.1110647.CrossRefPubMed
8.
Zurück zum Zitat Broderick C, Hoek RM, Forrester JV, Liversidge J, Sedgwick JD, Dick AD: Constitutive retinal CD200 expression regulates resident microglia and activation state of inflammatory cells during experimental autoimmune uveoretinitis. Am J Pathol. 2002, 161: 1669-1677.PubMedCentralCrossRefPubMed Broderick C, Hoek RM, Forrester JV, Liversidge J, Sedgwick JD, Dick AD: Constitutive retinal CD200 expression regulates resident microglia and activation state of inflammatory cells during experimental autoimmune uveoretinitis. Am J Pathol. 2002, 161: 1669-1677.PubMedCentralCrossRefPubMed
9.
Zurück zum Zitat Cardona AE, Pioro EP, Sasse ME, Kostenko V, Cardona SM, Dijkstra IM, Huang D, Kidd G, Dombrowski S, Dutta R, Lee JC, Cook DN, Jung S, Lira SA, Littman DR, Ransohoff RM: Control of microglial neurotoxicity by the fractalkine receptor. Nat Neurosci. 2006, 9: 917-924. 10.1038/nn1715.CrossRefPubMed Cardona AE, Pioro EP, Sasse ME, Kostenko V, Cardona SM, Dijkstra IM, Huang D, Kidd G, Dombrowski S, Dutta R, Lee JC, Cook DN, Jung S, Lira SA, Littman DR, Ransohoff RM: Control of microglial neurotoxicity by the fractalkine receptor. Nat Neurosci. 2006, 9: 917-924. 10.1038/nn1715.CrossRefPubMed
10.
Zurück zum Zitat Dick AD, Carter D, Robertson M, Broderick C, Hughes E, Forrester JV, Liversidge J: Control of myeloid activity during retinal inflammation. J Leukoc Biol. 2003, 74: 161-166. 10.1189/jlb.1102535.CrossRefPubMed Dick AD, Carter D, Robertson M, Broderick C, Hughes E, Forrester JV, Liversidge J: Control of myeloid activity during retinal inflammation. J Leukoc Biol. 2003, 74: 161-166. 10.1189/jlb.1102535.CrossRefPubMed
11.
Zurück zum Zitat Haynes SE, Hollopeter G, Yang G, Kurpius D, Dailey ME, Gan WB, Julius D: Control of myeloid activity during retinal inflammation. Nat Neurosci. 2006, 9: 1512-1519. 10.1038/nn1805.CrossRefPubMed Haynes SE, Hollopeter G, Yang G, Kurpius D, Dailey ME, Gan WB, Julius D: Control of myeloid activity during retinal inflammation. Nat Neurosci. 2006, 9: 1512-1519. 10.1038/nn1805.CrossRefPubMed
12.
Zurück zum Zitat Ransohoff RM, Perry VH: Microglial physiology: unique stimuli, specialized responses. Annu Rev Immunol. 2009, 27: 119-145. 10.1146/annurev.immunol.021908.132528.CrossRefPubMed Ransohoff RM, Perry VH: Microglial physiology: unique stimuli, specialized responses. Annu Rev Immunol. 2009, 27: 119-145. 10.1146/annurev.immunol.021908.132528.CrossRefPubMed
13.
Zurück zum Zitat El Khoury J, Luster AD: Mechanisms of microglia accumulation in Alzheimer's disease: therapeutic implications. Trends Pharmacol Sci. 2008, 29: 626-632. 10.1016/j.tips.2008.08.004.CrossRefPubMed El Khoury J, Luster AD: Mechanisms of microglia accumulation in Alzheimer's disease: therapeutic implications. Trends Pharmacol Sci. 2008, 29: 626-632. 10.1016/j.tips.2008.08.004.CrossRefPubMed
14.
Zurück zum Zitat Orr CF, Rowe DB, Halliday GM: An inflammatory review of Parkinson's disease. Prog Neurobiol. 2002, 68: 325-340. 10.1016/S0301-0082(02)00127-2.CrossRefPubMed Orr CF, Rowe DB, Halliday GM: An inflammatory review of Parkinson's disease. Prog Neurobiol. 2002, 68: 325-340. 10.1016/S0301-0082(02)00127-2.CrossRefPubMed
15.
Zurück zum Zitat Sargsyan SA, Monk PN, Shaw PJ: Microglia as potential contributors to motor neuron injury in amyotrophic lateral sclerosis. Glia. 2005, 51: 241-253. 10.1002/glia.20210.CrossRefPubMed Sargsyan SA, Monk PN, Shaw PJ: Microglia as potential contributors to motor neuron injury in amyotrophic lateral sclerosis. Glia. 2005, 51: 241-253. 10.1002/glia.20210.CrossRefPubMed
16.
Zurück zum Zitat Raivich G, Banati R: Brain microglia and blood-derived macrophages: molecular profiles and functional roles in multiple sclerosis and animal models of autoimmune demyelinating disease. Brain Res Brain Res Rev. 2004, 46: 261-281. 10.1016/j.brainresrev.2004.06.006.CrossRefPubMed Raivich G, Banati R: Brain microglia and blood-derived macrophages: molecular profiles and functional roles in multiple sclerosis and animal models of autoimmune demyelinating disease. Brain Res Brain Res Rev. 2004, 46: 261-281. 10.1016/j.brainresrev.2004.06.006.CrossRefPubMed
17.
Zurück zum Zitat Langmann T: Microglia activation in retinal degeneration. J Leukoc Biol. 2007, 81: 1345-1351. 10.1189/jlb.0207114.CrossRefPubMed Langmann T: Microglia activation in retinal degeneration. J Leukoc Biol. 2007, 81: 1345-1351. 10.1189/jlb.0207114.CrossRefPubMed
18.
Zurück zum Zitat Schuetz E, Thanos S: Microglia-targeted pharmacotherapy in retinal neurodegenerative diseases. Curr Drug Targets. 2004, 5: 619-627. 10.2174/1389450043345164.CrossRefPubMed Schuetz E, Thanos S: Microglia-targeted pharmacotherapy in retinal neurodegenerative diseases. Curr Drug Targets. 2004, 5: 619-627. 10.2174/1389450043345164.CrossRefPubMed
19.
Zurück zum Zitat Hume DA: Differentiation and heterogeneity in the mononuclear phagocyte system. Mucosal Immunol. 2008, 1: 432-441. 10.1038/mi.2008.36.CrossRefPubMed Hume DA: Differentiation and heterogeneity in the mononuclear phagocyte system. Mucosal Immunol. 2008, 1: 432-441. 10.1038/mi.2008.36.CrossRefPubMed
20.
Zurück zum Zitat Gordon S, Taylor PR: Monocyte and macrophage heterogeneity. Nat Rev Immunol. 2005, 5: 953-964. 10.1038/nri1733.CrossRefPubMed Gordon S, Taylor PR: Monocyte and macrophage heterogeneity. Nat Rev Immunol. 2005, 5: 953-964. 10.1038/nri1733.CrossRefPubMed
21.
Zurück zum Zitat Mosser DM: Monocyte and macrophage heterogeneity. J Leukoc Biol. 2003, 73: 209-212. 10.1189/jlb.0602325.CrossRefPubMed Mosser DM: Monocyte and macrophage heterogeneity. J Leukoc Biol. 2003, 73: 209-212. 10.1189/jlb.0602325.CrossRefPubMed
22.
Zurück zum Zitat Martinez FO, Sica A, Mantovani A, Locati M: Macrophage activation and polarization. Front Biosci. 2008, 13: 453-461. 10.2741/2692.CrossRefPubMed Martinez FO, Sica A, Mantovani A, Locati M: Macrophage activation and polarization. Front Biosci. 2008, 13: 453-461. 10.2741/2692.CrossRefPubMed
23.
Zurück zum Zitat Michelucci A, Heurtaux T, Grandbarbe L, Morga E, Heuschling P: Characterization of the microglial phenotype under specific pro-inflammatory and anti-inflammatory conditions: Effects of oligomeric and fibrillar amyloid-beta. J Neuroimmunol. 2009, 210: 3-12. 10.1016/j.jneuroim.2009.02.003.CrossRefPubMed Michelucci A, Heurtaux T, Grandbarbe L, Morga E, Heuschling P: Characterization of the microglial phenotype under specific pro-inflammatory and anti-inflammatory conditions: Effects of oligomeric and fibrillar amyloid-beta. J Neuroimmunol. 2009, 210: 3-12. 10.1016/j.jneuroim.2009.02.003.CrossRefPubMed
24.
Zurück zum Zitat Jimenez S, Baglietto-Vargas D, Caballero C, Moreno-Gonzalez I, Torres M, Sanchez-Varo R, Ruano D, Vizuete M, Gutierrez A, Vitorica J: Inflammatory response in the hippocampus of PS1M146L/APP751SL mouse model of Alzheimer's disease: age-dependent switch in the microglial phenotype from alternative to classic. J Neurosci. 2008, 28: 11650-11661. 10.1523/JNEUROSCI.3024-08.2008.CrossRefPubMed Jimenez S, Baglietto-Vargas D, Caballero C, Moreno-Gonzalez I, Torres M, Sanchez-Varo R, Ruano D, Vizuete M, Gutierrez A, Vitorica J: Inflammatory response in the hippocampus of PS1M146L/APP751SL mouse model of Alzheimer's disease: age-dependent switch in the microglial phenotype from alternative to classic. J Neurosci. 2008, 28: 11650-11661. 10.1523/JNEUROSCI.3024-08.2008.CrossRefPubMed
25.
Zurück zum Zitat Kigerl KA, Gensel JC, Ankeny DP, Alexander JK, Donnelly DJ, Popovich PG: Identification of two distinct macrophage subsets with divergent effects causing either neurotoxicity or regeneration in the injured mouse spinal cord. J Neurosci. 2009, 29: 13435-13444. 10.1523/JNEUROSCI.3257-09.2009.PubMedCentralCrossRefPubMed Kigerl KA, Gensel JC, Ankeny DP, Alexander JK, Donnelly DJ, Popovich PG: Identification of two distinct macrophage subsets with divergent effects causing either neurotoxicity or regeneration in the injured mouse spinal cord. J Neurosci. 2009, 29: 13435-13444. 10.1523/JNEUROSCI.3257-09.2009.PubMedCentralCrossRefPubMed
26.
Zurück zum Zitat Zhang Z, Zhang ZY, Schluesener HJ: Compound A, a plant origin ligand of glucocorticoid receptors, increases regulatory T cells and M2 macrophages to attenuate experimental autoimmune neuritis with reduced side effects. J Immunol. 2009, 183: 3081-3091. 10.4049/jimmunol.0901088.CrossRefPubMed Zhang Z, Zhang ZY, Schluesener HJ: Compound A, a plant origin ligand of glucocorticoid receptors, increases regulatory T cells and M2 macrophages to attenuate experimental autoimmune neuritis with reduced side effects. J Immunol. 2009, 183: 3081-3091. 10.4049/jimmunol.0901088.CrossRefPubMed
27.
Zurück zum Zitat Lleo A, Galea E, Sastre M: Molecular targets of non-steroidal anti-inflammatory drugs in neurodegenerative diseases. Cell Mol Life Sci. 2007, 64: 1403-1418. 10.1007/s00018-007-6516-1.CrossRefPubMed Lleo A, Galea E, Sastre M: Molecular targets of non-steroidal anti-inflammatory drugs in neurodegenerative diseases. Cell Mol Life Sci. 2007, 64: 1403-1418. 10.1007/s00018-007-6516-1.CrossRefPubMed
28.
Zurück zum Zitat Lopez-Lazaro M: Distribution and biological activities of the flavonoid luteolin. Mini Rev Med Chem. 2009, 9: 31-59. 10.2174/138955709787001712.CrossRefPubMed Lopez-Lazaro M: Distribution and biological activities of the flavonoid luteolin. Mini Rev Med Chem. 2009, 9: 31-59. 10.2174/138955709787001712.CrossRefPubMed
29.
Zurück zum Zitat Chen CY, Peng WH, Tsai KD, Hsu SL: Luteolin suppresses inflammation-associated gene expression by blocking NF-kappaB and AP-1 activation pathway in mouse alveolar macrophages. Life Sci. 2007, 81: 1602-1614. 10.1016/j.lfs.2007.09.028.CrossRefPubMed Chen CY, Peng WH, Tsai KD, Hsu SL: Luteolin suppresses inflammation-associated gene expression by blocking NF-kappaB and AP-1 activation pathway in mouse alveolar macrophages. Life Sci. 2007, 81: 1602-1614. 10.1016/j.lfs.2007.09.028.CrossRefPubMed
30.
Zurück zum Zitat Hu C, Kitts DD: Luteolin and luteolin-7-O-glucoside from dandelion flower suppress iNOS and COX-2 in RAW264.7 cells. Mol Cell Biochem. 2004, 265: 107-113. 10.1023/B:MCBI.0000044364.73144.fe.CrossRefPubMed Hu C, Kitts DD: Luteolin and luteolin-7-O-glucoside from dandelion flower suppress iNOS and COX-2 in RAW264.7 cells. Mol Cell Biochem. 2004, 265: 107-113. 10.1023/B:MCBI.0000044364.73144.fe.CrossRefPubMed
31.
Zurück zum Zitat Harris GK, Qian Y, Leonard SS, Sbarra DC, Shi X: Luteolin and chrysin differentially inhibit cyclooxygenase-2 expression and scavenge reactive oxygen species but similarly inhibit prostaglandin-E2 formation in RAW 264.7 cells. J Nutr. 2006, 136: 1517-1521.PubMed Harris GK, Qian Y, Leonard SS, Sbarra DC, Shi X: Luteolin and chrysin differentially inhibit cyclooxygenase-2 expression and scavenge reactive oxygen species but similarly inhibit prostaglandin-E2 formation in RAW 264.7 cells. J Nutr. 2006, 136: 1517-1521.PubMed
32.
Zurück zum Zitat Chen HQ, Jin ZY, Wang XJ, Xu XM, Deng L, Zhao JW: Luteolin protects dopaminergic neurons from inflammation-induced injury through inhibition of microglial activation. Neurosci Lett. 2008, 448: 175-179. 10.1016/j.neulet.2008.10.046.CrossRefPubMed Chen HQ, Jin ZY, Wang XJ, Xu XM, Deng L, Zhao JW: Luteolin protects dopaminergic neurons from inflammation-induced injury through inhibition of microglial activation. Neurosci Lett. 2008, 448: 175-179. 10.1016/j.neulet.2008.10.046.CrossRefPubMed
33.
Zurück zum Zitat Rezai-Zadeh K, Ehrhart J, Bai Y, Sanberg PR, Bickford P, Tan J, Shytle RD: Apigenin and luteolin modulate microglial activation via inhibition of STAT1-induced CD40 expression. J Neuroinflammation. 2008, 5: 41-10.1186/1742-2094-5-41.PubMedCentralCrossRefPubMed Rezai-Zadeh K, Ehrhart J, Bai Y, Sanberg PR, Bickford P, Tan J, Shytle RD: Apigenin and luteolin modulate microglial activation via inhibition of STAT1-induced CD40 expression. J Neuroinflammation. 2008, 5: 41-10.1186/1742-2094-5-41.PubMedCentralCrossRefPubMed
34.
Zurück zum Zitat Jang S, Kelley KW, Johnson RW: Luteolin reduces IL-6 production in microglia by inhibiting JNK phosphorylation and activation of AP-1. Proc Natl Acad Sci USA. 2008, 105: 7534-7539. 10.1073/pnas.0802865105.PubMedCentralCrossRefPubMed Jang S, Kelley KW, Johnson RW: Luteolin reduces IL-6 production in microglia by inhibiting JNK phosphorylation and activation of AP-1. Proc Natl Acad Sci USA. 2008, 105: 7534-7539. 10.1073/pnas.0802865105.PubMedCentralCrossRefPubMed
35.
Zurück zum Zitat Shimoi K, Okada H, Furugori M, Goda T, Takase S, Suzuki M, Hara Y, Yamamoto H, Kinae N: Intestinal absorption of luteolin and luteolin 7-O-beta-glucoside in rats and humans. FEBS Lett. 1998, 438: 220-224. 10.1016/S0014-5793(98)01304-0.CrossRefPubMed Shimoi K, Okada H, Furugori M, Goda T, Takase S, Suzuki M, Hara Y, Yamamoto H, Kinae N: Intestinal absorption of luteolin and luteolin 7-O-beta-glucoside in rats and humans. FEBS Lett. 1998, 438: 220-224. 10.1016/S0014-5793(98)01304-0.CrossRefPubMed
36.
Zurück zum Zitat Ebert S, Schoeberl T, Walczak Y, Stoecker K, Stempfl T, Moehle C, Weber BH, Langmann T: Chondroitin sulfate disaccharide stimulates microglia to adopt a novel regulatory phenotype. J Leukoc Biol. 2008, 84: 736-740. 10.1189/jlb.0208138.CrossRefPubMed Ebert S, Schoeberl T, Walczak Y, Stoecker K, Stempfl T, Moehle C, Weber BH, Langmann T: Chondroitin sulfate disaccharide stimulates microglia to adopt a novel regulatory phenotype. J Leukoc Biol. 2008, 84: 736-740. 10.1189/jlb.0208138.CrossRefPubMed
37.
Zurück zum Zitat Brazma A, Hingamp P, Quackenbush J, Sherlock G, Spellman P, Stoeckert C, Aach J, Ansorge W, Ball CA, Causton HC, Gaasterland T, Glenisson P, Holstege FC, Kim IF, Markowitz V, Matese JC, Parkinson H, Robinson A, Sarkans U, Schulze-Kremer S, Stewart J, Taylor R, Vilo J, Vingron M: Minimum information about a microarray experiment (MIAME)-toward standards for microarray data. Nat Genet. 2001, 29: 365-371. 10.1038/ng1201-365.CrossRefPubMed Brazma A, Hingamp P, Quackenbush J, Sherlock G, Spellman P, Stoeckert C, Aach J, Ansorge W, Ball CA, Causton HC, Gaasterland T, Glenisson P, Holstege FC, Kim IF, Markowitz V, Matese JC, Parkinson H, Robinson A, Sarkans U, Schulze-Kremer S, Stewart J, Taylor R, Vilo J, Vingron M: Minimum information about a microarray experiment (MIAME)-toward standards for microarray data. Nat Genet. 2001, 29: 365-371. 10.1038/ng1201-365.CrossRefPubMed
38.
Zurück zum Zitat Eichler GS, Huang S, Ingber DE: Gene Expression Dynamics Inspector (GEDI): for integrative analysis of expression profiles. Bioinformatics. 2003, 19: 2321-2322. 10.1093/bioinformatics/btg307.CrossRefPubMed Eichler GS, Huang S, Ingber DE: Gene Expression Dynamics Inspector (GEDI): for integrative analysis of expression profiles. Bioinformatics. 2003, 19: 2321-2322. 10.1093/bioinformatics/btg307.CrossRefPubMed
39.
Zurück zum Zitat Dennis G, Sherman BT, Hosack DA, Yang J, Gao W, Lane HC, Lempicki RA: DAVID: Database for Annotation, Visualization, and Integrated Discovery. Genome Biol. 2003, 4: 3-10.1186/gb-2003-4-5-p3.CrossRef Dennis G, Sherman BT, Hosack DA, Yang J, Gao W, Lane HC, Lempicki RA: DAVID: Database for Annotation, Visualization, and Integrated Discovery. Genome Biol. 2003, 4: 3-10.1186/gb-2003-4-5-p3.CrossRef
40.
Zurück zum Zitat Weigelt K, Lichtinger M, Rehli M, Langmann T: Transcriptomic profiling identifies a PU.1 regulatory network in macrophages. Biochem Biophys Res Commun. 2009, 380: 308-312. 10.1016/j.bbrc.2009.01.067.CrossRefPubMed Weigelt K, Lichtinger M, Rehli M, Langmann T: Transcriptomic profiling identifies a PU.1 regulatory network in macrophages. Biochem Biophys Res Commun. 2009, 380: 308-312. 10.1016/j.bbrc.2009.01.067.CrossRefPubMed
41.
Zurück zum Zitat Wegiel B, Baty CJ, Gallo D, Csizmadia E, Scott JR, Akhavan A, Chin BY, Kaczmarek E, Alam J, Bach FH, Zuckerbraun BS, Otterbein LE: Cell surface biliverdin reductase mediates biliverdin-induced anti-inflammatory effects via phosphatidylinositol 3-kinase and Akt. J Biol Chem. 2009, 284: 21369-21378. 10.1074/jbc.M109.027433.PubMedCentralCrossRefPubMed Wegiel B, Baty CJ, Gallo D, Csizmadia E, Scott JR, Akhavan A, Chin BY, Kaczmarek E, Alam J, Bach FH, Zuckerbraun BS, Otterbein LE: Cell surface biliverdin reductase mediates biliverdin-induced anti-inflammatory effects via phosphatidylinositol 3-kinase and Akt. J Biol Chem. 2009, 284: 21369-21378. 10.1074/jbc.M109.027433.PubMedCentralCrossRefPubMed
42.
Zurück zum Zitat Doi Y, Mizuno T, Maki Y, Jin S, Mizoguchi H, Ikeyama M, Doi M, Michikawa M, Takeuchi H, Suzumura A: Microglia activated with the toll-like receptor 9 ligand CpG attenuate oligomeric amyloid {beta} neurotoxicity in in vitro and in vivo models of Alzheimer's disease. Am J Pathol. 2009, 175: 2121-2132. 10.2353/ajpath.2009.090418.PubMedCentralCrossRefPubMed Doi Y, Mizuno T, Maki Y, Jin S, Mizoguchi H, Ikeyama M, Doi M, Michikawa M, Takeuchi H, Suzumura A: Microglia activated with the toll-like receptor 9 ligand CpG attenuate oligomeric amyloid {beta} neurotoxicity in in vitro and in vivo models of Alzheimer's disease. Am J Pathol. 2009, 175: 2121-2132. 10.2353/ajpath.2009.090418.PubMedCentralCrossRefPubMed
43.
Zurück zum Zitat Bea F, Hudson FN, Chait A, Kavanagh TJ, Rosenfeld ME: Induction of glutathione synthesis in macrophages by oxidized low-density lipoproteins is mediated by consensus antioxidant response elements. Circ Res. 2003, 92: 386-393. 10.1161/01.RES.0000059561.65545.16.CrossRefPubMed Bea F, Hudson FN, Chait A, Kavanagh TJ, Rosenfeld ME: Induction of glutathione synthesis in macrophages by oxidized low-density lipoproteins is mediated by consensus antioxidant response elements. Circ Res. 2003, 92: 386-393. 10.1161/01.RES.0000059561.65545.16.CrossRefPubMed
44.
Zurück zum Zitat Cheng L, Han X, Shi Y: A Regulatory Role of LPCAT1 in the Synthesis of Inflammatory Lipids, PAF and LPC, in the Retina of Diabetic Mice. Am J Physiol Endocrinol Metab. 2009 Cheng L, Han X, Shi Y: A Regulatory Role of LPCAT1 in the Synthesis of Inflammatory Lipids, PAF and LPC, in the Retina of Diabetic Mice. Am J Physiol Endocrinol Metab. 2009
45.
Zurück zum Zitat Koenigsknecht J, Landreth G: Microglial phagocytosis of fibrillar beta-amyloid through a beta1 integrin-dependent mechanism. J Neurosci. 2004, 24: 9838-9846. 10.1523/JNEUROSCI.2557-04.2004.CrossRefPubMed Koenigsknecht J, Landreth G: Microglial phagocytosis of fibrillar beta-amyloid through a beta1 integrin-dependent mechanism. J Neurosci. 2004, 24: 9838-9846. 10.1523/JNEUROSCI.2557-04.2004.CrossRefPubMed
46.
Zurück zum Zitat Ryu JK, Cho T, Choi HB, Wang YT, McLarnon JG: Microglial VEGF receptor response is an integral chemotactic component in Alzheimer's disease pathology. J Neurosci. 2009, 29: 3-13. 10.1523/JNEUROSCI.2888-08.2009.CrossRefPubMed Ryu JK, Cho T, Choi HB, Wang YT, McLarnon JG: Microglial VEGF receptor response is an integral chemotactic component in Alzheimer's disease pathology. J Neurosci. 2009, 29: 3-13. 10.1523/JNEUROSCI.2888-08.2009.CrossRefPubMed
47.
Zurück zum Zitat Shenoy AR, Kim BH, Choi HP, Matsuzawa T, Tiwari S, MacMicking JD: Emerging themes in IFN-gamma-induced macrophage immunity by the p47 and p65 GTPase families. Immunobiology. 2007, 212: 771-784. 10.1016/j.imbio.2007.09.018.PubMedCentralCrossRefPubMed Shenoy AR, Kim BH, Choi HP, Matsuzawa T, Tiwari S, MacMicking JD: Emerging themes in IFN-gamma-induced macrophage immunity by the p47 and p65 GTPase families. Immunobiology. 2007, 212: 771-784. 10.1016/j.imbio.2007.09.018.PubMedCentralCrossRefPubMed
48.
Zurück zum Zitat Liu G, Friggeri A, Yang Y, Park YJ, Tsuruta Y, Abraham E: miR-147, a microRNA that is induced upon Toll-like receptor stimulation, regulates murine macrophage inflammatory responses. Proc Natl Acad Sci USA. 2009, 106: 15819-15824. 10.1073/pnas.0901216106.PubMedCentralCrossRefPubMed Liu G, Friggeri A, Yang Y, Park YJ, Tsuruta Y, Abraham E: miR-147, a microRNA that is induced upon Toll-like receptor stimulation, regulates murine macrophage inflammatory responses. Proc Natl Acad Sci USA. 2009, 106: 15819-15824. 10.1073/pnas.0901216106.PubMedCentralCrossRefPubMed
49.
Zurück zum Zitat Plant SR, Wang Y, Vasseur S, Thrash JC, McMahon EJ, Bergstralh DT, Arnett HA, Miller SD, Carson MJ, Iovanna JL, Ting JP: Upregulation of the stress-associated gene p8 in mouse models of demyelination and in multiple sclerosis tissues. Glia. 2006, 53: 529-537. 10.1002/glia.20297.PubMedCentralCrossRefPubMed Plant SR, Wang Y, Vasseur S, Thrash JC, McMahon EJ, Bergstralh DT, Arnett HA, Miller SD, Carson MJ, Iovanna JL, Ting JP: Upregulation of the stress-associated gene p8 in mouse models of demyelination and in multiple sclerosis tissues. Glia. 2006, 53: 529-537. 10.1002/glia.20297.PubMedCentralCrossRefPubMed
50.
Zurück zum Zitat Gotoh T, Oyadomari S, Mori K, Mori M: Nitric oxide-induced apoptosis in RAW 264.7 macrophages is mediated by endoplasmic reticulum stress pathway involving ATF6 and CHOP. J Biol Chem. 2002, 277: 12343-12350. 10.1074/jbc.M107988200.CrossRefPubMed Gotoh T, Oyadomari S, Mori K, Mori M: Nitric oxide-induced apoptosis in RAW 264.7 macrophages is mediated by endoplasmic reticulum stress pathway involving ATF6 and CHOP. J Biol Chem. 2002, 277: 12343-12350. 10.1074/jbc.M107988200.CrossRefPubMed
51.
Zurück zum Zitat Shang YY, Wang ZH, Zhang LP, Zhong M, Zhang Y, Deng JT, Zhang W: TRB3, upregulated by ox-LDL, mediates human monocyte-derived macrophage apoptosis. Febs J. 2009, 276: 2752-2761. 10.1111/j.1742-4658.2009.06998.x.CrossRefPubMed Shang YY, Wang ZH, Zhang LP, Zhong M, Zhang Y, Deng JT, Zhang W: TRB3, upregulated by ox-LDL, mediates human monocyte-derived macrophage apoptosis. Febs J. 2009, 276: 2752-2761. 10.1111/j.1742-4658.2009.06998.x.CrossRefPubMed
52.
Zurück zum Zitat Lee S, Lee J, Kim S, Park JY, Lee WH, Mori K, Kim SH, Kim IK, Suk K: A dual role of lipocalin 2 in the apoptosis and deramification of activated microglia. J Immunol. 2007, 179: 3231-3241.CrossRefPubMed Lee S, Lee J, Kim S, Park JY, Lee WH, Mori K, Kim SH, Kim IK, Suk K: A dual role of lipocalin 2 in the apoptosis and deramification of activated microglia. J Immunol. 2007, 179: 3231-3241.CrossRefPubMed
53.
Zurück zum Zitat Granucci F, Petralia F, Urbano M, Citterio S, Di Tota F, Santambrogio L, Ricciardi-Castagnoli P: The scavenger receptor MARCO mediates cytoskeleton rearrangements in dendritic cells and microglia. Blood. 2003, 102: 2940-2947. 10.1182/blood-2002-12-3651.CrossRefPubMed Granucci F, Petralia F, Urbano M, Citterio S, Di Tota F, Santambrogio L, Ricciardi-Castagnoli P: The scavenger receptor MARCO mediates cytoskeleton rearrangements in dendritic cells and microglia. Blood. 2003, 102: 2940-2947. 10.1182/blood-2002-12-3651.CrossRefPubMed
54.
Zurück zum Zitat Kim JS, Lee HJ, Lee MH, Kim J, Jin C, Ryu JH: Luteolin inhibits LPS-stimulated inducible nitric oxide synthase expression in BV-2 microglial cells. Planta Med. 2006, 72: 65-68. 10.1055/s-2005-873145.CrossRefPubMed Kim JS, Lee HJ, Lee MH, Kim J, Jin C, Ryu JH: Luteolin inhibits LPS-stimulated inducible nitric oxide synthase expression in BV-2 microglial cells. Planta Med. 2006, 72: 65-68. 10.1055/s-2005-873145.CrossRefPubMed
55.
Zurück zum Zitat Al-Ubaidi MR, Font RL, Quiambao AB, Keener MJ, Liou GI, Overbeek PA, Baehr W: Bilateral retinal and brain tumors in transgenic mice expressing simian virus 40 large T antigen under control of the human interphotoreceptor retinoid-binding protein promoter. J Cell Biol. 1992, 119: 1681-1687. 10.1083/jcb.119.6.1681.CrossRefPubMed Al-Ubaidi MR, Font RL, Quiambao AB, Keener MJ, Liou GI, Overbeek PA, Baehr W: Bilateral retinal and brain tumors in transgenic mice expressing simian virus 40 large T antigen under control of the human interphotoreceptor retinoid-binding protein promoter. J Cell Biol. 1992, 119: 1681-1687. 10.1083/jcb.119.6.1681.CrossRefPubMed
56.
Zurück zum Zitat Tan E, Ding XQ, Saadi A, Agarwal N, Naash MI, Al-Ubaidi MR: Expression of cone-photoreceptor-specific antigens in a cell line derived from retinal tumors in transgenic mice. Invest Ophthalmol Vis Sci. 2004, 45: 764-768. 10.1167/iovs.03-1114.PubMedCentralCrossRefPubMed Tan E, Ding XQ, Saadi A, Agarwal N, Naash MI, Al-Ubaidi MR: Expression of cone-photoreceptor-specific antigens in a cell line derived from retinal tumors in transgenic mice. Invest Ophthalmol Vis Sci. 2004, 45: 764-768. 10.1167/iovs.03-1114.PubMedCentralCrossRefPubMed
57.
Zurück zum Zitat Ross JA, Kasum CM: Dietary flavonoids: bioavailability, metabolic effects, and safety. Annu Rev Nutr. 2002, 22: 19-34. 10.1146/annurev.nutr.22.111401.144957.CrossRefPubMed Ross JA, Kasum CM: Dietary flavonoids: bioavailability, metabolic effects, and safety. Annu Rev Nutr. 2002, 22: 19-34. 10.1146/annurev.nutr.22.111401.144957.CrossRefPubMed
58.
Zurück zum Zitat Rice-Evans CA, Miller NJ, Paganga G: Structure-antioxidant activity relationships of flavonoids and phenolic acids. Free Radic Biol Med. 1996, 20: 933-956. 10.1016/0891-5849(95)02227-9.CrossRefPubMed Rice-Evans CA, Miller NJ, Paganga G: Structure-antioxidant activity relationships of flavonoids and phenolic acids. Free Radic Biol Med. 1996, 20: 933-956. 10.1016/0891-5849(95)02227-9.CrossRefPubMed
59.
Zurück zum Zitat Seelinger G, Merfort I, Schempp CM: Anti-oxidant, anti-inflammatory and anti-allergic activities of luteolin. Planta Med. 2008, 74: 1667-1677. 10.1055/s-0028-1088314.CrossRefPubMed Seelinger G, Merfort I, Schempp CM: Anti-oxidant, anti-inflammatory and anti-allergic activities of luteolin. Planta Med. 2008, 74: 1667-1677. 10.1055/s-0028-1088314.CrossRefPubMed
60.
Zurück zum Zitat Li N, Venkatesan MI, Miguel A, Kaplan R, Gujuluva C, Alam J, Nel A: Induction of heme oxygenase-1 expression in macrophages by diesel exhaust particle chemicals and quinones via the antioxidant-responsive element. J Immunol. 2000, 165: 3393-3401.CrossRefPubMed Li N, Venkatesan MI, Miguel A, Kaplan R, Gujuluva C, Alam J, Nel A: Induction of heme oxygenase-1 expression in macrophages by diesel exhaust particle chemicals and quinones via the antioxidant-responsive element. J Immunol. 2000, 165: 3393-3401.CrossRefPubMed
61.
Zurück zum Zitat Lim JH, Park HS, Choi JK, Lee IS, Choi HJ: Isoorientin induces Nrf2 pathway-driven antioxidant response through phosphatidylinositol 3-kinase signaling. Arch Pharm Res. 2007, 30: 1590-1598. 10.1007/BF02977329.CrossRefPubMed Lim JH, Park HS, Choi JK, Lee IS, Choi HJ: Isoorientin induces Nrf2 pathway-driven antioxidant response through phosphatidylinositol 3-kinase signaling. Arch Pharm Res. 2007, 30: 1590-1598. 10.1007/BF02977329.CrossRefPubMed
62.
Zurück zum Zitat Singh A, Ling G, Suhasini AN, Zhang P, Yamamoto M, Navas-Acien A, Cosgrove G, Tuder RM, Kensler TW, Watson WH, Biswal S: Nrf2-dependent sulfiredoxin-1 expression protects against cigarette smoke-induced oxidative stress in lungs. Free Radic Biol Med. 2009, 46: 376-386. 10.1016/j.freeradbiomed.2008.10.026.PubMedCentralCrossRefPubMed Singh A, Ling G, Suhasini AN, Zhang P, Yamamoto M, Navas-Acien A, Cosgrove G, Tuder RM, Kensler TW, Watson WH, Biswal S: Nrf2-dependent sulfiredoxin-1 expression protects against cigarette smoke-induced oxidative stress in lungs. Free Radic Biol Med. 2009, 46: 376-386. 10.1016/j.freeradbiomed.2008.10.026.PubMedCentralCrossRefPubMed
63.
Zurück zum Zitat Osburn WO, Wakabayashi N, Misra V, Nilles T, Biswal S, Trush MA, Kensler TW: Nrf2 regulates an adaptive response protecting against oxidative damage following diquat-mediated formation of superoxide anion. Arch Biochem Biophys. 2006, 454: 7-15. 10.1016/j.abb.2006.08.005.PubMedCentralCrossRefPubMed Osburn WO, Wakabayashi N, Misra V, Nilles T, Biswal S, Trush MA, Kensler TW: Nrf2 regulates an adaptive response protecting against oxidative damage following diquat-mediated formation of superoxide anion. Arch Biochem Biophys. 2006, 454: 7-15. 10.1016/j.abb.2006.08.005.PubMedCentralCrossRefPubMed
64.
Zurück zum Zitat de Vries HE, Witte M, Hondius D, Rozemuller AJ, Drukarch B, Hoozemans J, van Horssen J: Nrf2-induced antioxidant protection: a promising target to counteract ROS-mediated damage in neurodegenerative disease?. Free Radic Biol Med. 2008, 45: 1375-1383. 10.1016/j.freeradbiomed.2008.09.001.CrossRefPubMed de Vries HE, Witte M, Hondius D, Rozemuller AJ, Drukarch B, Hoozemans J, van Horssen J: Nrf2-induced antioxidant protection: a promising target to counteract ROS-mediated damage in neurodegenerative disease?. Free Radic Biol Med. 2008, 45: 1375-1383. 10.1016/j.freeradbiomed.2008.09.001.CrossRefPubMed
65.
Zurück zum Zitat Harayama T, Shindou H, Ogasawara R, Suwabe A, Shimizu T: Identification of a novel noninflammatory biosynthetic pathway of platelet-activating factor. J Biol Chem. 2008, 283: 11097-11106. 10.1074/jbc.M708909200.CrossRefPubMed Harayama T, Shindou H, Ogasawara R, Suwabe A, Shimizu T: Identification of a novel noninflammatory biosynthetic pathway of platelet-activating factor. J Biol Chem. 2008, 283: 11097-11106. 10.1074/jbc.M708909200.CrossRefPubMed
66.
Zurück zum Zitat Sheikh AM, Nagai A, Ryu JK, McLarnon JG, Kim SU, Masuda J: Lysophosphatidylcholine induces glial cell activation: role of rho kinase. Glia. 2009, 57: 898-907. 10.1002/glia.20815.CrossRefPubMed Sheikh AM, Nagai A, Ryu JK, McLarnon JG, Kim SU, Masuda J: Lysophosphatidylcholine induces glial cell activation: role of rho kinase. Glia. 2009, 57: 898-907. 10.1002/glia.20815.CrossRefPubMed
67.
Zurück zum Zitat Stuart LM, Bell SA, Stewart CR, Silver JM, Richard J, Goss JL, Tseng AA, Zhang A, El Khoury JB, Moore KJ: CD36 signals to the actin cytoskeleton and regulates microglial migration via a p130Cas complex. J Biol Chem. 2007, 282: 27392-27401. 10.1074/jbc.M702887200.CrossRefPubMed Stuart LM, Bell SA, Stewart CR, Silver JM, Richard J, Goss JL, Tseng AA, Zhang A, El Khoury JB, Moore KJ: CD36 signals to the actin cytoskeleton and regulates microglial migration via a p130Cas complex. J Biol Chem. 2007, 282: 27392-27401. 10.1074/jbc.M702887200.CrossRefPubMed
68.
Zurück zum Zitat Martin CB, Ingersoll SA, Martin BK: Transcriptional control of the C3a receptor gene in glial cells: Dependence upon AP-1 but not Ets. Mol Immunol. 2007, 44: 703-712. 10.1016/j.molimm.2006.04.017.CrossRefPubMed Martin CB, Ingersoll SA, Martin BK: Transcriptional control of the C3a receptor gene in glial cells: Dependence upon AP-1 but not Ets. Mol Immunol. 2007, 44: 703-712. 10.1016/j.molimm.2006.04.017.CrossRefPubMed
69.
Zurück zum Zitat Nguyen H, Teskey L, Lin R, Hiscott J: Identification of the secretory leukocyte protease inhibitor (SLPI) as a target of IRF-1 regulation. Oncogene. 1999, 18: 5455-5463. 10.1038/sj.onc.1202924.CrossRefPubMed Nguyen H, Teskey L, Lin R, Hiscott J: Identification of the secretory leukocyte protease inhibitor (SLPI) as a target of IRF-1 regulation. Oncogene. 1999, 18: 5455-5463. 10.1038/sj.onc.1202924.CrossRefPubMed
70.
Zurück zum Zitat Ramsauer K, Farlik M, Zupkovitz G, Seiser C, Kroger A, Hauser H, Decker T: Distinct modes of action applied by transcription factors STAT1 and IRF1 to initiate transcription of the IFN-gamma-inducible gbp2 gene. Proc Natl Acad Sci USA. 2007, 104: 2849-2854. 10.1073/pnas.0610944104.PubMedCentralCrossRefPubMed Ramsauer K, Farlik M, Zupkovitz G, Seiser C, Kroger A, Hauser H, Decker T: Distinct modes of action applied by transcription factors STAT1 and IRF1 to initiate transcription of the IFN-gamma-inducible gbp2 gene. Proc Natl Acad Sci USA. 2007, 104: 2849-2854. 10.1073/pnas.0610944104.PubMedCentralCrossRefPubMed
71.
Zurück zum Zitat Zauberman A, Lapter S, Zipori D: Smad proteins suppress CCAAT/enhancer-binding protein (C/EBP) beta- and STAT3-mediated transcriptional activation of the haptoglobin promoter. J Biol Chem. 2001, 276: 24719-24725. 10.1074/jbc.M005813200.CrossRefPubMed Zauberman A, Lapter S, Zipori D: Smad proteins suppress CCAAT/enhancer-binding protein (C/EBP) beta- and STAT3-mediated transcriptional activation of the haptoglobin promoter. J Biol Chem. 2001, 276: 24719-24725. 10.1074/jbc.M005813200.CrossRefPubMed
72.
Zurück zum Zitat White CA, McCombe PA, Pender MP: Microglia are more susceptible than macrophages to apoptosis in the central nervous system in experimental autoimmune encephalomyelitis through a mechanism not involving Fas (CD95). Int Immunol. 1998, 10: 935-941. 10.1093/intimm/10.7.935.CrossRefPubMed White CA, McCombe PA, Pender MP: Microglia are more susceptible than macrophages to apoptosis in the central nervous system in experimental autoimmune encephalomyelitis through a mechanism not involving Fas (CD95). Int Immunol. 1998, 10: 935-941. 10.1093/intimm/10.7.935.CrossRefPubMed
73.
Zurück zum Zitat Streit WJ, Braak H, Xue QS, Bechmann I: Dystrophic (senescent) rather than activated microglial cells are associated with tau pathology and likely precede neurodegeneration in Alzheimer's disease. Acta Neuropathol. 2009, 118: 475-485. 10.1007/s00401-009-0556-6.PubMedCentralCrossRefPubMed Streit WJ, Braak H, Xue QS, Bechmann I: Dystrophic (senescent) rather than activated microglial cells are associated with tau pathology and likely precede neurodegeneration in Alzheimer's disease. Acta Neuropathol. 2009, 118: 475-485. 10.1007/s00401-009-0556-6.PubMedCentralCrossRefPubMed
Metadaten
Titel
Luteolin triggers global changes in the microglial transcriptome leading to a unique anti-inflammatory and neuroprotective phenotype
verfasst von
Konstantin Dirscherl
Marcus Karlstetter
Stefanie Ebert
Dominik Kraus
Julia Hlawatsch
Yana Walczak
Christoph Moehle
Rudolf Fuchshofer
Thomas Langmann
Publikationsdatum
01.12.2010
Verlag
BioMed Central
Erschienen in
Journal of Neuroinflammation / Ausgabe 1/2010
Elektronische ISSN: 1742-2094
DOI
https://doi.org/10.1186/1742-2094-7-3

Weitere Artikel der Ausgabe 1/2010

Journal of Neuroinflammation 1/2010 Zur Ausgabe

Neu in den Fachgebieten Neurologie und Psychiatrie

Akuter Schwindel: Wann lohnt sich eine MRT?

28.04.2024 Schwindel Nachrichten

Akuter Schwindel stellt oft eine diagnostische Herausforderung dar. Wie nützlich dabei eine MRT ist, hat eine Studie aus Finnland untersucht. Immerhin einer von sechs Patienten wurde mit akutem ischämischem Schlaganfall diagnostiziert.

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.