Skip to main content
Erschienen in: Journal of Neuroinflammation 1/2015

Open Access 01.12.2015 | Research

β-Lapachone suppresses neuroinflammation by modulating the expression of cytokines and matrix metalloproteinases in activated microglia

verfasst von: Eun-Jung Lee, Hyun-Myung Ko, Yeon-Hui Jeong, Eun-Mi Park, Hee-Sun Kim

Erschienen in: Journal of Neuroinflammation | Ausgabe 1/2015

Abstract

Background

β-Lapachone (β-LAP) is a natural naphthoquinone compound isolated from the lapacho tree (Tabebuia sp.), and it has been used for treatment of rheumatoid arthritis, infection, and cancer. In the present study, we investigated whether β-LAP has anti-inflammatory effects under in vitro and in vivo neuroinflammatory conditions.

Methods

The effects of β-LAP on the expression of inducible nitric oxide synthase (iNOS), cytokines, and matrix metalloproteinases (MMPs) were examined in lipopolysaccharide (LPS)-stimulated BV2 microglial cells and rat primary microglia by ELISA, reverse transcription polymerase chain reaction (RT-PCR), and Western blot analysis. Microglial activation and the expression levels of proinflammatory molecules were measured in the LPS-injected mouse brain by immunohistochemistry and RT-PCR analysis. The detailed molecular mechanism underlying the anti-inflammatory effects of β-LAP was analyzed by electrophoretic mobility shift assay, reporter gene assay, Western blot, and RT-PCR analysis.

Results

β-LAP inhibited the expression of iNOS, proinflammatory cytokines, and MMPs (MMP-3, MMP-8, MMP-9) at mRNA and protein levels in LPS-stimulated microglia. On the other hand, β-LAP upregulated the expressions of anti-inflammatory molecules such as IL-10, heme oxygenase-1 (HO-1), and the tissue inhibitor of metalloproteinase-2 (TIMP-2). The anti-inflammatory effect of β-LAP was confirmed in an LPS-induced systemic inflammation mouse model. Thus, β-LAP inhibited microglial activation and the expressions of iNOS, proinflammatory cytokines, and MMPs in the LPS-injected mouse brain. Further mechanistic studies revealed that β-LAP exerts anti-inflammatory effects by inhibiting MAPKs, PI3K/AKT, and NF-κB/AP-1 signaling pathways in LPS-stimulated microglia. β-LAP also inhibited reactive oxygen species (ROS) production by suppressing the expression and/or phosphorylation of NADPH oxidase subunit proteins, such as p47phox and gp91phox. The anti-oxidant effects of β-LAP appeared to be related with the increase of HO-1 and NQO1 via the Nrf2/anti-oxidant response element (ARE) pathway and/or the PKA pathway.

Conclusions

The strong anti-inflammatory/anti-oxidant effects of β-LAP may provide preventive therapeutic potential for various neuroinflammatory disorders.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s12974-015-0355-z) contains supplementary material, which is available to authorized users.
Eun-Jung Lee and Hyun-Myung Ko contributed equally to this work.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

E-JL and H-MK designed the study and performed the experiments and wrote the manuscript. Y-HJ performed the experiments. E-MP designed the study and analyzed the data. H-SK supervised the design of the study and analyzed the data and wrote the manuscript. All authors read and approved the final manuscript.
Abkürzungen
ARE
Anti-oxidant response element
BBB
Blood–brain barrier
CREB
cAMP response element-binding protein
CM
Conditioned media
EMSA
Electrophoretic mobility shift assay
ERK
Extracellular signal-regulated kinase
HO-1
Heme oxygenase-1
iNOS
Inducible nitric oxide synthase
β-LAP
β-Lapachone
LPS
Lipopolysaccharide
MAPK
Mitogen-activated protein kinase
JNK
c-Jun N-terminal kinase
MMP
Matrix metalloproteinase
NF-κB
Nuclear factor-κB
Nrf
Nuclear factor-E2-related factor
NQO1
NAD(P)H:quinone oxidoreductase 1
ROS
Reactive oxygen species
TIMP
Tissue inhibitor of metalloproteinase

Background

Microglia are innate immune cells of the central nervous system that constantly move through the brain parenchyma and constitute an immune surveillance system [1, 2]. Microglia become activated in response to various stimuli or injury and produce inflammatory mediators such as nitric oxide (NO), cytokines, and matrix metalloproteinases (MMPs). Alternatively, activated microglia produce anti-inflammatory cytokines and lead to matrix deposition and wound healing [3, 4]. Thus, the balance between the inflammatory M1 and the anti-inflammatory M2 phase of microglial activation is important to maintain homeostasis in the brain. However, prolonged and unresolved inflammatory response leads to destructive, chronic inflammation (neuroinflammation) that results in neuronal cell death and ultimately in the onset of neurodegenerative diseases [5, 6]. Therefore, inhibition of exaggerated inflammatory responses by microglia has been suggested as an important strategy to develop therapeutic agents for various neuroinflammatory disorders.
β-Lapachone (3,4-dihydro-2,2-dimethyl-2H-naphtho[1,2-b]pyran-5,6-dione; β-LAP) is a natural compound which was originally isolated from the bark of the South American lapacho tree (Tabebuia avellanedae) [7]. β-LAP has been reported to have a wide variety of pharmacological effects including anti-inflammatory, anti-cancer, anti-bacterial, anti-fungal, anti-platelet, and anti-angiogenic action [810]. In particular, β-LAP exerts anti-neoplastic effects against various human cancer cell lines, and it is now being used in clinical trials for the treatment of various forms of cancer [1113]. β-LAP has topoisomerase inhibitory activity and elevates NQO1 levels, leading to a futile redox cycle and apoptosis of cancer cells [11, 14]. A recent study reports that β-LAP attenuates cisplatin-mediated nephrotoxicity by increasing NAD+ levels with elevated tumoricidal effects of cisplatin [15]. Several studies have reported anti-inflammatory effects of β-LAP. β-LAP suppresses inflammatory responses in activated macrophages and protects from lung edema and high mortality in septic mice [16]. β-LAP alleviates carrageenan-induced rat paw edema by suppressing neutrophil migration and cytokine production [17]. In addition, β-LAP induces anti-inflammatory heme oxygenase-1 (HO-1) via AMPK activation in RAW264.7 macrophages and endothelial cells [18, 19]. A previous study has shown anti-inflammatory effects of β-LAP in activated microglia [20]. It was demonstrated that β-LAP inhibits inducible nitric oxide synthase (iNOS) and cytokine expressions in lipopolysaccharide (LPS)-stimulated BV2 cells. However, the in vivo effects of β-LAP and the detailed molecular mechanism underlying the anti-inflammatory effects of β-LAP have not been fully elucidated.
Therefore, in the present study, we examined the anti-inflammatory effects of β-LAP under both in vitro and in vivo neuroinflammatory conditions and analyzed, in detail, the molecular mechanism. In particular, we investigated the effects of β-LAP on the gene expression and activity of MMPs, because our group recently demonstrated the proinflammatory role of MMPs in activated microglia [2123]. Through this study, we report for the first time that β-LAP inhibits microglial activation and expression of iNOS, cytokines, and several MMPs in the LPS-injected mouse brain. Furthermore, we demonstrated that multiple signaling pathways are involved in the anti-inflammatory mechanism of β-LAP in activated microglia.

Materials and methods

Reagents and antibodies

All reagents for cell culture were purchased from Gibco BRL (Grand Island, NY, USA). β-Lapachone and LPS (Escherichia coli serotype 055:B5) were obtained from Sigma–Aldrich (St. Louis, MO, USA). All reagents and enzymes for reverse transcription polymerase chain reaction (RT-PCR) and oligonucleotides for electrophoretic mobility shift assay (EMSA) were purchased from Promega (Madison, WI, USA). Antibodies against phospho-/total forms of MAPKs, CREB, β-actin, MMPs (MMP-3, MMP-8, MMP-9), and tissue inhibitor of metalloproteinase-2 (TIMP-2) were supplied by Cell Signaling Technology (Beverley, CA, USA), Abcam (Cambridge, UK), or Chemicon (Temecula, CA, USA). Antibodies against HO-1, NQO1, and Iba1 were purchased from Santa Cruz Biotechnology (Santa Cruz, CA, USA) or Novus (Littleton, CO, USA). The antibody for phospho-p47phox (Ser370) was purchased from Assay Biotechnology Company Inc. (Sunnyvale, CA, USA). All other chemicals were obtained from Sigma–Aldrich, unless otherwise stated.

Microglial cell cultures

The immortalized mouse BV2 microglial cell line [24] was grown and maintained in Dulbecco’s modified Eagle’s medium (DMEM), supplemented with 10 % heat-inactivated fetal bovine serum, streptomycin (10 μg/ml), and penicillin (10 U/ml) at 37 °C under 5 % CO2. Primary microglial cells were cultured from the cerebral cortices of 1- to 2-day-old Sprague Dawley rat pups as described previously [21]. The purity of microglial cultures was >95 %, as confirmed by Western blot and immunocytochemistry analyses using an antibody specific to ionized calcium-binding adapter protein-1 (IBA-1) staining (data not shown).

Measurement of cytokines, nitrite, and intracellular ROS levels

Cells (1 × 105 cells per well in a 48-well plate) were pretreated with β-LAP for 1 h and further stimulated with LPS (100 ng/ml) for 16 h. Concentrations of TNF-α, IL-1β, IL-6, and IL-10 in conditioned medium (CM) were measured by ELISA using monoclonal antibodies and procedures recommended by the supplier (PharMingen, San Diego, CA). Accumulated nitrite in CM and intracellular accumulation of reactive oxygen species (ROS) were measured using Griess reagent (Promega) and H2DCF-DA (Invitrogen, La Jolla, USA), respectively, as previously described [25].

Assays for MMP-3, MMP-8, and MMP-9 activity

BV2 cells were stimulated with LPS in the presence or absence of β-LAP for 24 h, and the supernatants were collected to measure MMP activity using the SensoLyte® 520 MMP assay system (AnaSpec, San Jose, CA, USA). MMP activity measurements were performed by continuous detection of peptide cleavage using a fluorescence plate reader (Molecular Devices, Sunnyvale, CA, USA). MMP activity units were expressed as a change in the fluorescence intensity at an excitation wavelength of 490 nm and an emission wavelength of 520 nm.

LPS-induced inflammation and administration of β-LAP

C57BL/6 mice (10–11 weeks old) were purchased from the Orient Co., Ltd. (Seoul, Korea). All animal experiments were approved by the Institutional Animal Care and Use Committee at the School of Medicine, Ewha Womans University. All efforts were made to minimize animal suffering, to reduce the number of animals used, and to utilize alternatives to in vivo techniques, if available. Systemic inflammation was induced by LPS administration (5 mg/kg, i.p.) to male C57BL/6 mice as previously described [26]. β-LAP (10 mg/kg, i.p.), dissolved in vehicle solution (1 % DMSO and normal saline), was given daily for 4 days before the LPS treatment. Samples were obtained 3 or 6 h after LPS treatment.

Immunohistochemistry

Three hours after LPS treatment, the animals were anesthetized with sodium pentobarbital (120 mg/kg i.p.) and perfused transcardially with normal saline containing heparin (5 U/ml), followed by 4 % paraformaldehyde (PFA) in 0.1 M sodium phosphate buffer (PBS), pH 7.2. The brains were removed and incubated overnight in fixatives and stored in a 30 % sucrose solution. Serial coronal brain sections of regions containing the hippocampus (20 μm thick, at 600-μm intervals) were collected using a cryostat. Brain sections were incubated in PBS containing 0.1 % Triton X-100, 5 % normal serum, and 1 % bovine serum albumin for 1 h, and then subsequently incubated with primary antibody. On the next day, sections were incubated in a 1:200 dilution of Alexa Fluor 488-labeled donkey anti-rabbit secondary antibody or Alexa Fluor 594-labeled chicken anti-goat antibody (Molecular Probes Inc., Eugene, OR, USA) for 60 min at room temperature, and then washed with 0.05 % Tween 20 in PBS three times, 5 min each. Sections were then stained with a 0.5-μg/ml DAPI staining solution for 20 min at room temperature and washed. The sections were mounted with Vectashield mounting medium (Vector Laboratories, Burlingame, CA, USA), and fluorescence microcopy images were obtained using confocal microscopy (TSC-SP, Leica, Heidelberg, Germany). Iba1-, MMP-3-, MMP-8-, and MMP-9-positive cells were quantified using the Metamorph program (Carl Zeiss, Jena, Germany). Two serial brain sections from each animal were used for further analysis, and quantification of Iba1-, MMP-3-, MMP-8-, and MMP-9-positive cells was performed in three different areas (500 μm2 in size) in the lateral cortex and dentate gyrus of the right hemisphere per brain section. The mean cell number from six 500-μm2 areas per animal was calculated.

RT-PCR

Total RNA (1 μg) isolated from BV2 or primary microglial cells (4.5 × 105 cells on a 6-cm dish), or from the brain tissue of LPS-injected mice, was reverse transcribed, and synthesized cDNA was used as a template for PCR. RT-PCR was performed on a T100 Thermal Cycler (Bio-Rad) with GoTaq polymerase (Promega). The primer sets shown in Table 1 were used to detect specific PCR products, and their values were calculated as fold change relative to control after normalization to the GAPDH gene.
Table 1
Primers used in RT-PCR reactions
Species
Gene
Forward primer (5′ → 3′)
Reverse primer (5′ → 3′)
Size (bp)
Mouse
TNF-α
CCTATGTCTCAGCCTCTTCT
CCTGGTATGAGATAGCAAAT
354
iNOS
CAAGAGTTTGACCAGAGGACC
TGGAACCACTCGTACTTGGGA
450
IL-1β
GGCAACTGTTCCTGAACTCAACTG
CCATTGAGGTGGAGAGCTTTCAGC
447
IL-6
CCACTTCACAAGTCGGAGGCTT
CCAGCTTATCTGTTAGGAGA
395
IL-10
GCCAGTACAGCCGGGAAGACAATA
GCCTTGTAGACACCTTGGTCTT
409
MMP-3
ATTCAGTCCCTCTATGGA
CTCCAGTATTTGTCCTCTAC
375
MMP-8
CCAAGGAGTGTCCAAGCCAT
CCTGCAGGAAAACTGCATCG
180
MMP-9
GTGATCCCCACTTACTATGGAAAC
GAAGCCATACAGTTTATCCTGGTC
352
TIMP-2
TCTAATTGCAGGAAAGGCAGA
TGCTCTTCTCTGTGACCCAGT
218
HO-1
TGTCACCCTGTGCTTGACCT
ATACCCGCTACCTGGGTGAC
209
NQO1
AGAGGCTCTGAAGAAGAGAGG
CACCCTGAAGAGAGTACATGG
401
p47phox
CGATGGATTGTCCTTTGTGC
ATCACCGGCTATTTCCCATC
256
p67phox
CTTCAACATAGGCTGCGTGA
CTTCATGTTGGTTGCCAATG
334
p22phox
AAAGAGGAAAAAGGGGTCCA
TAGGCTCAATGGGAGTCCAC
239
gp91phox
GTCAAGTGCCCCAAGGTATCCA
TTGTAGCTGAGGAAGTTGGC
453
GAPDH
ATGTACGTAGCCATCCAGGC
AGGAAGGAAGGCTGGAAGAG
420
Rat
TNF-α
AAGTTCCCAAATGGGCTCCCT
TGAAGTGGCAAATCGGCTGAC
306
iNOS
GCAGAATGTGACCATCATGG
ACAACCTTGGTGTTGAAGGC
426
IL-1β
AAATGCCTCGTGCTGTCTGACC
TCCCGACCATTGCTGTTTCCT
377
IL-6
TCATTCTGTCTCGAGCCCAC
GAAGTAGGGAAGGCAGTGGC
345
IL-10
AGGGCTGCCTTCAGTCAAGT
AGAAATCGATGACAGCGTCG
396
MMP-3
GTACCAACCTATTCCTGGTTGC
CCAGAGAGTTAGATTTGGTGGG
231
MMP-8
TACAACCTGTTTCTCGTGGCTGC
TCAACTGTTCTCAGCTGGGGATG
317
MMP-9
AAGTTGAACTCAGCCTTTGAGG
GTCGAATTTCCAGATACGTTCC
225
TIMP-2
CGTAGTGATCAGAGCCAAGC
TCTGCCTTTCCTGCAATTAGA
225
GAPDH
GTGCTGAGTATGTCGTGGAGTCT
ACAGTCTTCTGAGTGGCAGTGA
292

Western blot analysis

Proteins isolated from total cell lysates, and from CM, were separated by SDS-PAGE, transferred to nitrocellulose membranes, and incubated with primary antibodies against MMP-3, MMP-8, and MMP-9; TIMP-2 (1:1000); the phospho- or total form of MAP kinases or CREB; HO-1; NQO1 (1:1000); or p-p47phox [anti-phospho-(Ser345)-p47phox Ab] (1:1000, Assay Biotechnology). After thorough washing with Tris-buffered saline with Tween 20 (TBST), horseradish peroxidase-conjugated secondary antibodies (1:2000 dilution in TBST; New England Biolabs, Beverly, MA, USA) were applied, and the blots were developed using an enhanced chemiluminescence detection kit (Pierce Biotechnology, Rockford, IL, USA). To detect secreted MMPs, MMP proteins in the conditioned media were enriched using an Amicon® centrifugal filter (Millipore Corp., Billerica, MA, USA).

Transient transfection and luciferase assay

BV2 cells plated at 50–60 % confluence (2 × 105 cells per well) in 12-well plates were transfected with 1 μg of plasmid DNA (ARE-luc, CRE-luc) using the Convoy™ Platinum transfection reagent (CellTAGen, Seoul, Korea). After 36 h of transfection, cells were treated with β-LAP and LPS (100 ng/ml), or β-LAP only, for 6 h. The luciferase assay was used to determine the effect of β-LAP on ARE or CRE promoter activity. The ARE-luciferase reporter gene was kindly provided by Dr. Young-Joon Surh (Seoul National University, Seoul, Korea). The sequence of the anti-oxidant response element (ARE) construct is as follows: 5′-CTCAGCCTTCCAAATCG CAGTCACAGTGACTCAGCAGAATC-3′ [27, 28]. The CRE-luc vector, which contains four copies of the cyclic AMP response element (CRE, TGACGTCA), was obtained from Stratagene (La Jolla, CA).

EMSA

Nuclear extracts from treated microglia were prepared as follows. Cells (2 × 107) were treated with 1 ml of lysis buffer (10 mM Tris–HCl, pH 7.9; 10 mM NaCl; 3 mM MgCl2; 1 % NP-40) on ice for 5 min. After 10 min of centrifugation at 3000 rpm, the pellet was resuspended in 50 μl of extraction buffer (20 mM HEPES, pH 7.9; 20 % glycerol; 1.5 mM MgCl2; 0.2 mM EDTA; 300 mM NaCl; 1 mM DTT; 1 mM PMSF) and incubated on ice for 30 min. After centrifugation at 13,200 rpm for 15 min, the supernatant was harvested as a nuclear protein extract and stored at −70 °C. Double-stranded DNA oligonucleotides containing the NF-κB, AP-1, ARE, or CRE consensus sequences were end labeled using T4 polynucleotide kinase (New England Biolabs, Beverly, MA) in the presence of [γ-32P]ATP. Nuclear proteins (5 μg) were incubated with 32P-labeled probe on ice for 30 min, resolved on a 5 % acrylamide gel, and visualized by autoradiography. We purchased double-stranded DNA oligonucleotides containing the NF-κB, AP-1, or CRE consensus sequences from Promega (Madison, WI, USA) and that containing ARE from Santa Cruz Biotechnology (Santa Cruz, CA, USA). The DNA sequences of the probes are as follows: NF-κB (5′-AGTTGAGGGGAC TTTCCCAGGC-3′), AP-1 (5′-CGCTTGATGAGTCAGCCGGAA-3′), ARE (5′-TGG GGAACCTGTGCTGAGTCACTGGAG-3′), and CRE (5′-AGAGATTGCCTGACGTCAGAGAGCTA-3′).

Statistical analysis

Unless otherwise stated, all experiments were performed with triplicate samples and repeated at least three times. Data are presented as mean ± SEM, and statistical comparisons among groups were performed using one-way ANOVA followed by Newman–Keuls post hoc tests or t tests. Statistical significance was accepted for P values <0.05.

Results

β-LAP showed anti-inflammatory effects in LPS-stimulated microglial cells

To investigate the anti-inflammatory effect of β-LAP, BV2 or primary microglial cells were treated with LPS in the presence or absence of β-LAP for 16 h. Consecutively, NO and cytokine levels in CM were measured. We observed that β-LAP significantly inhibited the LPS-induced production of NO and proinflammatory cytokines such as TNF-α, IL-1β, and IL-6, whereas it raised anti-inflammatory IL-10 in BV2 cells and primary microglia (Fig. 1a, b). β-LAP did not have any cytotoxicity in the concentrations used in this study in both the BV2 and primary microglial cells, at least for 48 h (MTT assay, data not shown). Subsequent RT-PCR analysis showed that β-LAP reduced the mRNA expression of iNOS, TNF-α, IL-1β, and IL-6, and increased IL-10 (Fig. 1c, d). The data suggest that β-LAP modulates the expressions of iNOS and cytokines at the transcriptional level.

β-LAP suppressed the expression and activity of MMP-3, MMP-8, and MMP-9, while it enhanced TIMP-2 expression in LPS-stimulated microglia

Our group recently reported that MMP-3, MMP-8, and MMP-9 are important proinflammatory mediators in activated microglia, while TIMP-2 plays an anti-inflammatory role [2123, 29]. To investigate whether β-LAP affects the expression of MMPs and TIMP-2, we performed RT-PCR analysis and Western blotting using cell lysates and CM from LPS + β-LAP-treated microglial cells. We found that β-LAP inhibited LPS-induced expressions of MMP-3, MMP-8, and MMP-9 at the mRNA and protein levels in both BV2 cells and primary microglia (Fig. 2a, b). Moreover, β-LAP reduced the enzymatic activity of MMP-3, MMP-8, and MMP-9 in the CM (Fig. 2d). On the other hand, β-LAP restored the expression of TIMP-2 (Fig. 2ac). The results suggest that the inhibition of MMPs and the upregulation of TIMP-2 are at least partly involved in the anti-inflammatory effects of β-LAP.

β-LAP inhibited microglial activation and the expression of iNOS, cytokines, and MMPs in the brains of LPS-induced systemic inflammation mice

To verify the anti-inflammatory effects of β-LAP in vivo, β-LAP (10 mg/kg) was injected before LPS administration. After 3 h, microglial activation and the expression levels of proinflammatory cytokines and MMPs were measured in the LPS-injected mouse brain. Systemic LPS led to an increase in densely stained Iba1-positive cells in the cortex and dentate gyrus (Fig. 3a). This indicates the presence of activated microglia. Pretreatment of β-LAP, however, reduced their number. The smaller amount of densely stained activated microglia was significant as seen in the subsequent quantification of Iba1-positive cells. However, we did not observe any infiltration of neutrophils or monocytes into the brain at this time point (i.e., 3 h) (data not shown). In addition, we found that β-LAP inhibited the mRNA expression of proinflammatory cytokines (IL-6, IL-lβ, TNF-α), iNOS, and MMPs (MMP-3, MMP-8, MMP-9) that were elevated in the cortex region of the LPS-injected mouse brain (Fig. 3b, c). To determine whether MMPs, upregulated in the cortex of LPS-injected mouse, were co-localized with microglia, immunohistochemistry was performed using antibodies against Iba1 (red) and MMP-3, MMP-8, or MMP-9 (green), 24 h after the LPS challenge. The number of double-immunopositive cells was significantly increased by LPS injection, but it was reduced in the β-LAP-injected groups (Fig. 4). The results indicate that β-LAP inhibits the expression of MMP-3, MMP-8, and MMP-9 in the microglia of the LPS-injected mouse brain.

β-LAP inhibited the phosphorylation of MAPKs and AKT and the DNA binding activity of NF-κB and AP-1 in LPS-stimulated BV2 cells

To further investigate the anti-inflammatory mechanism of β-LAP, we examined the effects of β-LAP on MAPKs and PI3K/AKT signaling, which play a critical role in the process of microglial activation and the production of NO and cytokines [30, 31]. We found that β-LAP significantly inhibited the phosphorylation of three types of MAPKs and AKT in LPS-stimulated BV2 cells (Fig. 5a, b). Furthermore, we found that β-LAP suppressed the DNA binding activity of NF-κB and AP-1, which are key transcription factors modulating cytokine and iNOS gene expression in microglia (Fig. 5c, d).

β-LAP suppressed ROS production and expression of NADPH oxidase subunits, whereas it enhanced anti-oxidant enzyme HO-1/NQO1 expression

Next, we examined the effects of β-LAP on LPS-induced ROS production, which acts as a second messenger in inflammatory reactions and subsequent neuronal cell death [32, 33]. β-LAP significantly inhibited LPS-induced ROS production in the BV2 cells and primary microglia (Fig. 6ac). Since NADPH oxidase is a major enzyme for microglial ROS release, we examined the effect of β-LAP on membrane (gp91phox, p22phox) and cytosolic (p47phox, p67phox) components of NADPH oxidase. β-LAP suppressed the expression and phosphorylation of p47phox (Fig. 6d, e). Moreover, β-LAP inhibited the mRNA expression of gp91phox, without affecting p67phox or p22phox (Fig. 6d). We next examined the effects of β-LAP on HO-1 and NQO1, which mediate anti-inflammatory and anti-oxidant effects in the activated microglia [33, 34]. We observed that β-LAP increased LPS-induced HO-1 and NQO1 expression at mRNA and protein levels (Fig. 7a, b). Interestingly, we found that β-LAP itself also increased HO-1 and NQO1 expression.

β-LAP activated Nrf2/ARE and PKA/CREB pathways

Because Nrf2 binds to ARE on the promoters of phase II anti-oxidant enzyme genes such as HO-1 and NQO1, and controls their expression [27, 34], we examined the effects of β-LAP on the binding of Nrf2 to ARE on HO-1/NQO1 promoters. We observed that LPS induced Nrf2 binding to ARE, which was enhanced by β-LAP (Fig. 7c). Moreover, β-LAP increased ARE-driven luciferase activity in the absence and presence of LPS (Fig. 7d). Previous studies of our own and by other groups reported that the PKA/CREB pathway contributes to the resolution of inflammation and ROS detoxification, and that PKA is an upstream modulator of HO-1 expression in microglia [35, 36]. In the present study, we observed that β-LAP itself increased the phosphorylation of CREB, which is a downstream target of PKA. In addition, β-LAP potentiated the CREB phosphorylation induced by LPS (Fig. 7e). β-LAP also increased the DNA binding and transcriptional activity of CREB in the absence or presence of LPS (Fig. 7f, g). Thus, the data suggest that the Nrf2/ARE and PKA pathways are largely involved in the anti-inflammatory/anti-oxidant mechanism of β-LAP, in association with other signaling pathways such as MAPKs, and PI3K/AKT.

Discussion

Our present study demonstrates the anti-inflammatory properties of β-LAP in brain microglia and their underlying molecular mechanisms. β-LAP inhibited the expressions of iNOS and proinflammatory cytokines in LPS-stimulated microglia. In addition, β-LAP reduced the expression and activity of MMP-3, MMP-8, and MMP-9, which are inflammatory mediators in activated microglia [2123]. By using a systemic inflammation mouse model, we confirmed the anti-inflammatory role of β-LAP. Thus, β-LAP inhibited microglial activation and the expression of proinflammatory molecules in the LPS-injected mouse brain. By mechanistic analysis, we showed that β-LAP inhibited the phosphorylation of MAPKs and AKT and the DNA binding activity of NF-κB/AP-1 induced by LPS. Furthermore, we found that β-LAP exerts anti-oxidant effects by reducing ROS production via suppression of NADPH oxidase subunit activity and/or expression, and upregulation of anti-oxidant enzymes such as HO-1 and NQO1. We showed that β-LAP activated Nrf2/ARE and PKA/CREB pathways, which are involved in the upregulation of HO-1/NQO1 expression. Therefore, β-LAP appears to act as an anti-inflammatory/anti-oxidant agent by modulating multiple signaling pathways (i.e., inhibition of MAPKs and PI3K/AKT, upregulation of Nrf2/ARE and PKA).
A previous study reported that β-LAP inhibits the mRNA expression of TLR4 signaling molecules in experimental autoimmune encephalomyelitis mice [37]. In the present study, however, β-LAP did not affect the mRNA expression of TLR4 or MyD88 in LPS-stimulated microglia (Additional file 1: Figure S1), suggesting that β-LAP exerts anti-inflammatory effects by modulating signaling downstream of TLR4/MyD88 and/or via a MyD88-independent pathway. In addition, we found that β-LAP also inhibited the inflammatory reactions induced by TLR2 or TLR3 agonists. Treatment with β-LAP inhibited NO and TNF-α production in BV2 cells stimulated with lipoteichoic acid (LTA; TLR2 agonist) or polyinosinic–polycytidylic acid (Poly I:C; TLR3 agonist) (Additional file 1: Figure S2). Thus, the data imply that the anti-inflammatory effect of β-LAP is not confined to TLR4 activation. Further studies are necessary to investigate the detailed mechanism underlying the effect of β-LAP on TLR signaling.
MMPs are zinc-dependent endopeptidases which are involved not only in normal brain development but also in various neuropathological conditions such as Alzheimer’s disease, Parkinson’s disease, stroke, and multiple sclerosis [38]. MMPs are aberrantly expressed in neuropathological conditions and cause breakdown of the blood–brain barrier (BBB), infiltration of peripheral immune cells, demyelination, and neuronal cell death [39, 40]. Our group recently reported that MMPs play an important role in various neuroinflammatory conditions [2123]. We showed that MMP-3, MMP-8, and MMP-9 are upregulated in LPS or α-synuclein-stimulated microglia and mediate neuroinflammatory reactions. Thus, the specific inhibition of MMP-3, MMP-8, or MMP-9 suppresses iNOS and cytokine expressions in LPS or α-synuclein-stimulated microglia. We demonstrated that MMPs cleave the N-terminal extracellular domain of protease-activated receptor-1 and activate intracellular inflammatory signaling pathways in α-synuclein-activated microglia [21]. More recently, we showed that MMP-8 plays a pivotal role in neuroinflammation by activating TNF-α processing in microglia [22]. We also reported that TIMP-2, as an endogenous inhibitor of MMPs, has an anti-inflammatory effect by modulating MMP-3, MMP-8, and MMP-9 in activated microglia [29]. Based on these findings, our present study examined the effects of β-LAP on MMPs and TIMP-2 in LPS-stimulated microglia. We observed that β-LAP significantly suppressed the expression and activity of MMP-3, MMP-8, and MMP-9 with enhancement of TIMP-2 under in vitro and/or in vivo neuroinflammatory conditions, which may contribute to the anti-inflammatory properties of β-LAP.
In this study, we found that β-LAP induced phase II anti-oxidant enzymes such as HO-1 and NQO1, which are regulated through the Nrf2/ARE signaling pathway [34]. Under normal conditions, Nrf2 is sequestered by cytosolic Keap1, which serves as an adaptor to link Nrf2 to the ubiquitin ligase Cul3–Rbx1 complex that ubiquitinates and degrades Nrf2. However, upon stimulation by electrophilic agents or ROS, Nrf2 dissociates from its cytosolic docking protein Keap1, translocates into the nucleus, and binds to the ARE site [41]. It has been suggested that Nrf2 phosphorylation is involved in this release process. In the present study, we observed that β-LAP increased Nrf2 binding to ARE, as well as ARE-mediated transcriptional activity. However, we did not further examine the effect of β-LAP on the Nrf2 release mechanism related to Keap1; this would be an interesting study in the future.
It is well-known that β-LAP is a substrate and activator of NQO1, which catalyzes the oxidation of NADH to NAD+. A recent study reported that β-LAP induces HO-1 expression by increasing NQO1 activity and AMPK phosphorylation in RAW264.7 macrophages [18, 19]. As a mechanism to resolve the neurotoxic responses brought about by microglial activation, microglia usually express anti-inflammatory cytokines (i.e., IL-10, TGF-β1), suppressor of cytokine signaling (SOCS)-family proteins (i.e., SOCS1 and SOCS3), and anti-oxidant enzymes (i.e., HO-1, NQO1, SOD) [42, 43]. In the present study, we found that LPS itself increased HO-1 expression, which is associated with ARE-dependent transcription. We previously reported that the activation of PKA/CREB signaling is upstream of HO-1 expression and that the upregulation of the HO-1 and PKA pathway plays a key role in mediating the anti-inflammatory mechanism in LPS-stimulated microglia [35]. Therefore, the potentiation of HO-1/ARE and PKA/CREB by β-LAP may at least partly contribute to the anti-inflammatory and anti-oxidant effects of β-LAP in LPS-stimulated microglia. Interestingly, a recent study demonstrated that HO-1 knockout markedly increases MMP-9 expression in arteriovenous fistulas in mice and that MMP-9 induction reflects the prooxidant and proinflammatory effects recognized in a state of HO-1 deficiency [44]. Therefore, the upregulation of HO-1/NQO1 may at least partly contribute to the anti-inflammatory effects of β-LAP by suppressing proinflammatory molecules such as cytokines and MMPs in LPS-stimulated microglia.
A number of studies have reported therapeutic and health benefits of β-LAP supplementation in experimental animal models and clinical trials. The most extensively studied property of β-LAP is its anti-cancer potential, and β-LAP is currently being evaluated in clinical trials for treatment of cancer [9, 11]. In addition, β-LAP has beneficial effects on metabolic syndromes such as obesity, diabetes, hypertension, arterial restenosis, and salt-induced renal injury [4548]. The anti-inflammatory/cytoprotective effects of β-LAP have also been reported in several disease models. β-LAP attenuates cisplatin-mediated acute kidney injury in mice by suppressing critical mediators for inflammation and ROS [15]. β-LAP has shown therapeutic effects against rheumatoid arthritis by inhibiting synoviocyte proliferation and suppressing MMP expression in chondrocytes [49]. β-LAP also protects against renal ischemia/reperfusion injury in mice by inducing NQO1 activation and subsequent inhibition of ROS [50]. In experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis, the administration of β-LAP ameliorates the development of EAE by inhibiting the production of IL-12 family cytokines [37]. Interestingly, a recent study demonstrated that potentiation of NQO1 activity by feeding β-LAP prevents the age-dependent decline of motor and cognitive function in aged mice [51]. β-LAP also increases memory performance and prevents the loss of synapses in aged mice, suggesting the therapeutic potential of β-LAP for neurodegenerative diseases.
As to the BBB permeability of β-LAP, Huntingdon Life Sciences (UK) has reported that a minimal concentration of β-LAP penetrates into the rat brain, compared with other organs, under normal conditions (unpublished report). Therefore, we suggest two possibilities regarding the mechanism of β-LAP: first, β-LAP suppresses the peripheral inflammation induced by LPS and results in the inhibition of subsequent brain inflammation, and second, in systemic inflammatory conditions, BBB permeability is compromised and thus the penetration of β-LAP into the brain might be enhanced. In the latter case, β-LAP may directly modulate microglial activation. We believe that both of these mechanisms may be simultaneously involved in β-LAP action.

Conclusions

The present study demonstrates that β-LAP inhibits neuroinflammation by modulating various inflammatory molecules and multiple signaling pathways. Considering the high demand for anti-inflammatory agents that can modulate microglial activation, β-LAP may be a promising preventive therapeutic agent against neuroinflammatory disorders such as Alzheimer’s disease and Parkinson’s disease.

Acknowledgements

This research was supported by the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Science, ICT & Future Planning (Grant No. NRF-2012R1A5A2A32671866 and NRF-2015R1A2A2A01005226).
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made.
The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.
The Creative Commons Public Domain Dedication waiver (https://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

E-JL and H-MK designed the study and performed the experiments and wrote the manuscript. Y-HJ performed the experiments. E-MP designed the study and analyzed the data. H-SK supervised the design of the study and analyzed the data and wrote the manuscript. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Gomez-Nicola D, Perry VH. Microglial dynamics and role in the healthy and diseased brain: a paradigm of functional plasticity. Neuroscientist. 2015;21:169–84.PubMedCentralPubMedCrossRef Gomez-Nicola D, Perry VH. Microglial dynamics and role in the healthy and diseased brain: a paradigm of functional plasticity. Neuroscientist. 2015;21:169–84.PubMedCentralPubMedCrossRef
2.
Zurück zum Zitat Tremblay MÈ, Stevens B, Sierra A, Wake H, Bessis A, Nimmerjahn A. The role of microglia in the healthy brain. J Neurosci. 2011;31:16064–9.PubMedCrossRef Tremblay MÈ, Stevens B, Sierra A, Wake H, Bessis A, Nimmerjahn A. The role of microglia in the healthy brain. J Neurosci. 2011;31:16064–9.PubMedCrossRef
3.
Zurück zum Zitat Cherry JD, Olschowka JA, O’Banion MK. Neuroinflammation and M2 microglia: the good, the bad, and the inflamed. J Neuroinflammation. 2014;11:98.PubMedCentralPubMedCrossRef Cherry JD, Olschowka JA, O’Banion MK. Neuroinflammation and M2 microglia: the good, the bad, and the inflamed. J Neuroinflammation. 2014;11:98.PubMedCentralPubMedCrossRef
4.
Zurück zum Zitat Hanisch UK, Kettenmann H. Microglia: active sensor and versatile effector cells in the normal and pathologic brain. Nat Neurosci. 2007;10:1387–94.PubMedCrossRef Hanisch UK, Kettenmann H. Microglia: active sensor and versatile effector cells in the normal and pathologic brain. Nat Neurosci. 2007;10:1387–94.PubMedCrossRef
6.
Zurück zum Zitat Norden DM, Muccigrosso MM, Godbout JP. Microglial priming and enhanced reactivity to secondary insult in aging, and traumatic CNS injury, and neurodegenerative disease. Neuropharmacology. 2014;96:29–41.PubMedCrossRef Norden DM, Muccigrosso MM, Godbout JP. Microglial priming and enhanced reactivity to secondary insult in aging, and traumatic CNS injury, and neurodegenerative disease. Neuropharmacology. 2014;96:29–41.PubMedCrossRef
7.
Zurück zum Zitat Schaffner-Sabba K, Schmidt-Ruppin KH, Wehrli W, Schuerch AR, Wasley JW. beta-Lapachone: synthesis of derivatives and activities in tumor models. J Med Chem. 1984;27:990–4.PubMedCrossRef Schaffner-Sabba K, Schmidt-Ruppin KH, Wehrli W, Schuerch AR, Wasley JW. beta-Lapachone: synthesis of derivatives and activities in tumor models. J Med Chem. 1984;27:990–4.PubMedCrossRef
8.
Zurück zum Zitat de Castro SL, Emery FS, da Silva Junior EN. Synthesis of quinoidal molecules: strategies towards bioactive compounds with an emphasis of lapachones. Eur J Med Chem. 2013;69:678–700.PubMedCrossRef de Castro SL, Emery FS, da Silva Junior EN. Synthesis of quinoidal molecules: strategies towards bioactive compounds with an emphasis of lapachones. Eur J Med Chem. 2013;69:678–700.PubMedCrossRef
9.
Zurück zum Zitat Gomez Castellanos JR, Prieto JM, Heinrich M. Red lapacho (Tabebuia impetiginosa)—a global ethnopharmacological commodity? J Ethnopharmacol. 2009;121:1–13.PubMedCrossRef Gomez Castellanos JR, Prieto JM, Heinrich M. Red lapacho (Tabebuia impetiginosa)—a global ethnopharmacological commodity? J Ethnopharmacol. 2009;121:1–13.PubMedCrossRef
10.
Zurück zum Zitat Pardee AB, Li YZ, Li CJ. Cancer therapy with β-lapachone. Curr Cancer Drug Targets. 2002;2:227–42.PubMedCrossRef Pardee AB, Li YZ, Li CJ. Cancer therapy with β-lapachone. Curr Cancer Drug Targets. 2002;2:227–42.PubMedCrossRef
11.
Zurück zum Zitat Kung HN, Lu KS, Chau YP. The chemotherapeutic effects of lapacho tree extract: β-lapachone. Chemotherapy. 2014;3:2. Kung HN, Lu KS, Chau YP. The chemotherapeutic effects of lapacho tree extract: β-lapachone. Chemotherapy. 2014;3:2.
12.
Zurück zum Zitat Reinicke KE, Bey EA, Bentle MS, Pink JJ, Ingalls ST, Hoppel CL, et al. Development of beta-lapachone prodrugs for therapy against human cancer cells with elevated NAD(P)H:quinone oxidoreductase 1 levels. Clin Cancer Res. 2005;11:3055–64.PubMedCrossRef Reinicke KE, Bey EA, Bentle MS, Pink JJ, Ingalls ST, Hoppel CL, et al. Development of beta-lapachone prodrugs for therapy against human cancer cells with elevated NAD(P)H:quinone oxidoreductase 1 levels. Clin Cancer Res. 2005;11:3055–64.PubMedCrossRef
13.
Zurück zum Zitat Yu HY, Kim SO, Jin GY, Kim GY, Kim WJ, Yoo YH, et al. β-Lapachone-induced apoptosis of human gastric carcinoma AGS cells is caspase-dependent and regulated by the PI3K/Akt pathway. Biomol Ther (Seoul). 2014;22:184–92.CrossRef Yu HY, Kim SO, Jin GY, Kim GY, Kim WJ, Yoo YH, et al. β-Lapachone-induced apoptosis of human gastric carcinoma AGS cells is caspase-dependent and regulated by the PI3K/Akt pathway. Biomol Ther (Seoul). 2014;22:184–92.CrossRef
14.
Zurück zum Zitat Hueber A, Esser P, Heimann K, Kociok N, Wineter S, Weller M. The topoisomerase I inhibitors, camptothecin and β-lapachone, induce apoptosis of human retinal pigment epithelial cells. Exp Eye Res. 1998;67:525–30.PubMedCrossRef Hueber A, Esser P, Heimann K, Kociok N, Wineter S, Weller M. The topoisomerase I inhibitors, camptothecin and β-lapachone, induce apoptosis of human retinal pigment epithelial cells. Exp Eye Res. 1998;67:525–30.PubMedCrossRef
15.
Zurück zum Zitat Oh GS, Kim HJ, Choi JH, Shen A, Choe SK, Karna A, et al. Pharmacological activation of NQO1 increases NAD+ levels and attenuates cisplatin-mediated acute kidney injury in mice. Kidney Int. 2014;85:547–60.PubMedCentralPubMedCrossRef Oh GS, Kim HJ, Choi JH, Shen A, Choe SK, Karna A, et al. Pharmacological activation of NQO1 increases NAD+ levels and attenuates cisplatin-mediated acute kidney injury in mice. Kidney Int. 2014;85:547–60.PubMedCentralPubMedCrossRef
16.
Zurück zum Zitat Tzeng HP, Ho FM, Chao KF, Kuo ML, Lin-Shiau SY, Liu SH. beta-Lapachone reduces endotoxin-induced macrophage activation and lung edema and mortality. Am J Respir Crit Care Med. 2003;168:85–91.PubMedCrossRef Tzeng HP, Ho FM, Chao KF, Kuo ML, Lin-Shiau SY, Liu SH. beta-Lapachone reduces endotoxin-induced macrophage activation and lung edema and mortality. Am J Respir Crit Care Med. 2003;168:85–91.PubMedCrossRef
17.
Zurück zum Zitat Sitônio MM, Carvalho Júnior CH, Campos Ide A, Silva JB, Lima Mdo C, Góes AJ, et al. Anti-inflammatory and anti-arthritic activities of 3,4-dihydro-2,2-dimethyl-2H-naphthol[1,2-b]pyran-5,6-dione(β-lapachone). Inflamm Res. 2013;62:107–13.PubMedCrossRef Sitônio MM, Carvalho Júnior CH, Campos Ide A, Silva JB, Lima Mdo C, Góes AJ, et al. Anti-inflammatory and anti-arthritic activities of 3,4-dihydro-2,2-dimethyl-2H-naphthol[1,2-b]pyran-5,6-dione(β-lapachone). Inflamm Res. 2013;62:107–13.PubMedCrossRef
18.
Zurück zum Zitat Byun SJ, Son Y, Hwan Cho B, Chung HT, Pae HO. β-Lapachone, a substrate of NAD(P)H:quinone oxidoreductase, induces anti-inflammatory heme oxygenase-1 via AMP-activated protein kinase activation in RAW264.7 macrophages. J Clin Biochem Nutr. 2013;52:106–11.PubMedCentralPubMedCrossRef Byun SJ, Son Y, Hwan Cho B, Chung HT, Pae HO. β-Lapachone, a substrate of NAD(P)H:quinone oxidoreductase, induces anti-inflammatory heme oxygenase-1 via AMP-activated protein kinase activation in RAW264.7 macrophages. J Clin Biochem Nutr. 2013;52:106–11.PubMedCentralPubMedCrossRef
19.
Zurück zum Zitat Byun SJ, Son Y, Pae HO. Cytoprotective effect of β-lapachone by inducing heme oxygenase-1 expression and AMP-activated protein kinase activation in human endothelial cells. Eur Rev Med Pharmacol Sci. 2014;18:949–58.PubMed Byun SJ, Son Y, Pae HO. Cytoprotective effect of β-lapachone by inducing heme oxygenase-1 expression and AMP-activated protein kinase activation in human endothelial cells. Eur Rev Med Pharmacol Sci. 2014;18:949–58.PubMed
20.
Zurück zum Zitat Moon DO, Choi YH, Kim ND, Park YM, Kim GY. Anti-inflammatory effects of beta-lapachone in lipopolysaccharide-stimulated BV2 microglia. Int Immunopharmacol. 2007;7:506–14.PubMedCrossRef Moon DO, Choi YH, Kim ND, Park YM, Kim GY. Anti-inflammatory effects of beta-lapachone in lipopolysaccharide-stimulated BV2 microglia. Int Immunopharmacol. 2007;7:506–14.PubMedCrossRef
21.
Zurück zum Zitat Lee EJ, Woo MS, Moon PG, Baek MC, Choi IY, Kim WK, et al. α-Synuclein activates microglia by inducing the expressions of matrix metalloproteases and the subsequent activation of protease-activated receptor-1. J Immunol. 2010;185:615–23.PubMedCrossRef Lee EJ, Woo MS, Moon PG, Baek MC, Choi IY, Kim WK, et al. α-Synuclein activates microglia by inducing the expressions of matrix metalloproteases and the subsequent activation of protease-activated receptor-1. J Immunol. 2010;185:615–23.PubMedCrossRef
22.
Zurück zum Zitat Lee EJ, Han JE, Woo MS, Shin JA, Park EM, Kang JL, et al. Matrix metalloproteinase-8 plays a pivotal role in neuroinflammation by modulating TNF-α activation. J Immunol. 2014;193:2384–93.PubMedCrossRef Lee EJ, Han JE, Woo MS, Shin JA, Park EM, Kang JL, et al. Matrix metalloproteinase-8 plays a pivotal role in neuroinflammation by modulating TNF-α activation. J Immunol. 2014;193:2384–93.PubMedCrossRef
23.
Zurück zum Zitat Woo MS, Park JS, Choi IY, Kim WK, Kim HS. Inhibition of MMP-3 or -9 suppresses lipopolysaccharide-induced expression of proinflammatory cytokines and iNOS in microglia. J Neurochem. 2008;106:770–80.PubMedCrossRef Woo MS, Park JS, Choi IY, Kim WK, Kim HS. Inhibition of MMP-3 or -9 suppresses lipopolysaccharide-induced expression of proinflammatory cytokines and iNOS in microglia. J Neurochem. 2008;106:770–80.PubMedCrossRef
24.
Zurück zum Zitat Bocchini V, Mazzolla R, Barluzzi R, Blasi E, Sick P, Kettenmann H. An immortalized cell line expresses properties of activated microglial cells. J Neurosci Res. 1992;31:616–21.PubMedCrossRef Bocchini V, Mazzolla R, Barluzzi R, Blasi E, Sick P, Kettenmann H. An immortalized cell line expresses properties of activated microglial cells. J Neurosci Res. 1992;31:616–21.PubMedCrossRef
25.
Zurück zum Zitat Lee KM, Kang HS, Yun CH, Kwak HS. Potential in vitro protective effect of quercetin, catechin, caffeic acid and phytic acid against ethanol-induced oxidative stress in SK-Hep-1 cells. Biomol Ther (Seoul). 2012;20:492–8.CrossRef Lee KM, Kang HS, Yun CH, Kwak HS. Potential in vitro protective effect of quercetin, catechin, caffeic acid and phytic acid against ethanol-induced oxidative stress in SK-Hep-1 cells. Biomol Ther (Seoul). 2012;20:492–8.CrossRef
26.
Zurück zum Zitat Jung JS, Shin KO, Lee YM, Shin JA, Park EM, Jeong J, et al. Anti-inflammatory mechanism of exogenous C2 ceramide in lipopolysaccharide-stimulated microglia. Biochim Biophys Acta. 2013;1831:1016–26.PubMedCrossRef Jung JS, Shin KO, Lee YM, Shin JA, Park EM, Jeong J, et al. Anti-inflammatory mechanism of exogenous C2 ceramide in lipopolysaccharide-stimulated microglia. Biochim Biophys Acta. 2013;1831:1016–26.PubMedCrossRef
27.
Zurück zum Zitat Li MH, Cha YN, Surh YJ. Peroxynitrite induces HO-1 expression via PI3K/Akt-dependent activation of NF-E2-related factor 2 in PC12 cells. Free Rad Biol Med. 2006;41:1079–91.PubMedCrossRef Li MH, Cha YN, Surh YJ. Peroxynitrite induces HO-1 expression via PI3K/Akt-dependent activation of NF-E2-related factor 2 in PC12 cells. Free Rad Biol Med. 2006;41:1079–91.PubMedCrossRef
28.
Zurück zum Zitat Lee JM, Moehlenkamp JD, Hanson JM, Johnson JA. Nrf2-dependent activation of the antioxidant response elements by tert-butylhydroquinone is independent of oxidative stress in IMR-32 human neuroblastoma cells. Biochem Biophys Res Commun. 2001;280:286–92.PubMedCrossRef Lee JM, Moehlenkamp JD, Hanson JM, Johnson JA. Nrf2-dependent activation of the antioxidant response elements by tert-butylhydroquinone is independent of oxidative stress in IMR-32 human neuroblastoma cells. Biochem Biophys Res Commun. 2001;280:286–92.PubMedCrossRef
29.
Zurück zum Zitat Lee EJ, Kim HS. The anti-inflammatory role of tissue inhibitor of metalloproteinase-2 in lipopolysaccharide-stimulated microglia. J Neuroinflammation. 2014;11:116.PubMedCentralPubMedCrossRef Lee EJ, Kim HS. The anti-inflammatory role of tissue inhibitor of metalloproteinase-2 in lipopolysaccharide-stimulated microglia. J Neuroinflammation. 2014;11:116.PubMedCentralPubMedCrossRef
30.
Zurück zum Zitat Kim WK, Hwang SY, Oh ES, Piao HZ, Kim KW, Han IO. TGF-beta1 represses activation and resultant death of microglia via inhibition of phosphatidylinositol 3-kinase activity. J Immunol. 2004;172:7015–23.PubMedCrossRef Kim WK, Hwang SY, Oh ES, Piao HZ, Kim KW, Han IO. TGF-beta1 represses activation and resultant death of microglia via inhibition of phosphatidylinositol 3-kinase activity. J Immunol. 2004;172:7015–23.PubMedCrossRef
31.
Zurück zum Zitat Van Eldik LJ, Thompson WL, Ralay Ranaivo H, Behanna HA, Martin Watterson D. Glia proinflammatory cytokine upregulation as a therapeutic target for neurodegenerative diseases: function-based and target-based discovery approaches. Int Rev Neurobiol. 2007;82:277–96.PubMedCrossRef Van Eldik LJ, Thompson WL, Ralay Ranaivo H, Behanna HA, Martin Watterson D. Glia proinflammatory cytokine upregulation as a therapeutic target for neurodegenerative diseases: function-based and target-based discovery approaches. Int Rev Neurobiol. 2007;82:277–96.PubMedCrossRef
32.
Zurück zum Zitat Bedard K, Krause KH. The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology. Physiol Rev. 2007;87:245–313.PubMedCrossRef Bedard K, Krause KH. The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology. Physiol Rev. 2007;87:245–313.PubMedCrossRef
33.
34.
Zurück zum Zitat Keum YS. Regulation of Nrf2-mediated phase II detoxification and anti-oxidant genes. Biomol Ther (Seoul). 2012;20:144–51.PubMedCentralCrossRef Keum YS. Regulation of Nrf2-mediated phase II detoxification and anti-oxidant genes. Biomol Ther (Seoul). 2012;20:144–51.PubMedCentralCrossRef
35.
Zurück zum Zitat Jung JS, Shin JA, Park EM, Lee JE, Kang YS, Min SW, et al. Anti-inflammatory mechanism of ginsenoside Rh1 in lipopolysaccharide-stimulated microglia: critical role of the protein kinase A pathway and hemeoxygenase-1 expression. J Neurochem. 2010;115:1668–80.PubMedCrossRef Jung JS, Shin JA, Park EM, Lee JE, Kang YS, Min SW, et al. Anti-inflammatory mechanism of ginsenoside Rh1 in lipopolysaccharide-stimulated microglia: critical role of the protein kinase A pathway and hemeoxygenase-1 expression. J Neurochem. 2010;115:1668–80.PubMedCrossRef
36.
Zurück zum Zitat Lee B, Cao R, Choi YS, Cho HY, Rhee AD, Hah CK, et al. The CREB/CRE transcriptional pathway: protection against oxidative stress-mediated neuronal cell death. J Neurochem. 2009;108:1251–65.PubMedCentralPubMedCrossRef Lee B, Cao R, Choi YS, Cho HY, Rhee AD, Hah CK, et al. The CREB/CRE transcriptional pathway: protection against oxidative stress-mediated neuronal cell death. J Neurochem. 2009;108:1251–65.PubMedCentralPubMedCrossRef
37.
Zurück zum Zitat Xu J, Wagoner G, Douglas JC, Drew PD. β-Lapachone ameliorization of experimental autoimmune encephalomyelitis. J Neuroimmunol. 2013;254:46–54.PubMedCentralPubMedCrossRef Xu J, Wagoner G, Douglas JC, Drew PD. β-Lapachone ameliorization of experimental autoimmune encephalomyelitis. J Neuroimmunol. 2013;254:46–54.PubMedCentralPubMedCrossRef
38.
Zurück zum Zitat Agrawal SM, Lau L, Yong VW. MMPs in the central nervous system: where the good guys go bad. Semin Cell Dev Biol. 2008;19:42–51.PubMedCrossRef Agrawal SM, Lau L, Yong VW. MMPs in the central nervous system: where the good guys go bad. Semin Cell Dev Biol. 2008;19:42–51.PubMedCrossRef
39.
Zurück zum Zitat Morancho A, Rosell A, García-Bonilla L, Montaner J. Matrix metalloproteinase and stroke infarct size: role for anti-inflammatory treatment. Ann N Y Acad Sci. 2010;1207:123–33.PubMedCrossRef Morancho A, Rosell A, García-Bonilla L, Montaner J. Matrix metalloproteinase and stroke infarct size: role for anti-inflammatory treatment. Ann N Y Acad Sci. 2010;1207:123–33.PubMedCrossRef
40.
Zurück zum Zitat Rosenberg GA. Matrix metalloproteinases and their multiple roles in neurodegenerative diseases. Lancet Neurol. 2009;8:205–16.PubMedCrossRef Rosenberg GA. Matrix metalloproteinases and their multiple roles in neurodegenerative diseases. Lancet Neurol. 2009;8:205–16.PubMedCrossRef
41.
Zurück zum Zitat Kansanen E, Jykkanen HK, Levonen AL. Activation of stress signaling pathways by electrophilic oxidized and nitrated lipids. Free Rad Biol Med. 2012;52:973–82.PubMedCrossRef Kansanen E, Jykkanen HK, Levonen AL. Activation of stress signaling pathways by electrophilic oxidized and nitrated lipids. Free Rad Biol Med. 2012;52:973–82.PubMedCrossRef
42.
Zurück zum Zitat Yang MS, Min KJ, Joe E. Multiple mechanisms that prevent excessive brain inflammation. J Neurosci Res. 2007;85:2298–305.PubMedCrossRef Yang MS, Min KJ, Joe E. Multiple mechanisms that prevent excessive brain inflammation. J Neurosci Res. 2007;85:2298–305.PubMedCrossRef
43.
Zurück zum Zitat Syapin PJ. Regulation of heme oxygenase-1 for treatment of neuroinflammation and brain disorders. Brit J Pharmacol. 2008;155:623–40.CrossRef Syapin PJ. Regulation of heme oxygenase-1 for treatment of neuroinflammation and brain disorders. Brit J Pharmacol. 2008;155:623–40.CrossRef
44.
Zurück zum Zitat Juncos JP, Tracz MJ, Croatt AJ, Grande JP, Ackerman AW, Katusic ZS, et al. Genetic deficiency of heme oxygenase-1 impairs functionality and form of an arteriovenous fistula in the mouse. Kidney Int. 2008;74:47–51.PubMedCentralPubMedCrossRef Juncos JP, Tracz MJ, Croatt AJ, Grande JP, Ackerman AW, Katusic ZS, et al. Genetic deficiency of heme oxygenase-1 impairs functionality and form of an arteriovenous fistula in the mouse. Kidney Int. 2008;74:47–51.PubMedCentralPubMedCrossRef
45.
Zurück zum Zitat Hwang JH, Kim DW, Jo EJ, Kim YK, Jo YS, Park JH, et al. Pharmacological stimulation of NADH oxidation ameliorates obesity and related phenotypes in mice. Diabetes. 2009;58:965–74.PubMedCentralPubMedCrossRef Hwang JH, Kim DW, Jo EJ, Kim YK, Jo YS, Park JH, et al. Pharmacological stimulation of NADH oxidation ameliorates obesity and related phenotypes in mice. Diabetes. 2009;58:965–74.PubMedCentralPubMedCrossRef
46.
Zurück zum Zitat Kim SY, Jeoung NH, Oh CJ, Choi YK, Lee HJ, Kim HJ, et al. Activation of NAD(P)H:quinone oxidoreductase 1 prevents arterial restenosis by suppressing vascular smooth muscle cell proliferation. Circ Res. 2009;104:842–50.PubMedCrossRef Kim SY, Jeoung NH, Oh CJ, Choi YK, Lee HJ, Kim HJ, et al. Activation of NAD(P)H:quinone oxidoreductase 1 prevents arterial restenosis by suppressing vascular smooth muscle cell proliferation. Circ Res. 2009;104:842–50.PubMedCrossRef
47.
Zurück zum Zitat Kim YH, Hwang JH, Noh JR, Gang GT, Tadi S, Yim YH, et al. Prevention of salt-induced renal injury by activation of NAD(P)H:quinone oxidoreductase 1, associated with NADPH oxidase. Free Radic Biol Med. 2012;52:880–8.PubMedCrossRef Kim YH, Hwang JH, Noh JR, Gang GT, Tadi S, Yim YH, et al. Prevention of salt-induced renal injury by activation of NAD(P)H:quinone oxidoreductase 1, associated with NADPH oxidase. Free Radic Biol Med. 2012;52:880–8.PubMedCrossRef
48.
Zurück zum Zitat Kim YH, Hwang JH, Kim KS, Noh JR, Gang GT, Seo Y, et al. NAD(P)H:quinone oxidoreductase 1 activation reduces blood pressure through regulation of endothelial nitric oxide synthase acetylation in spontaneously hypertensive rats. Am J Hypertens. 2015;28:50–7.PubMedCrossRef Kim YH, Hwang JH, Kim KS, Noh JR, Gang GT, Seo Y, et al. NAD(P)H:quinone oxidoreductase 1 activation reduces blood pressure through regulation of endothelial nitric oxide synthase acetylation in spontaneously hypertensive rats. Am J Hypertens. 2015;28:50–7.PubMedCrossRef
49.
Zurück zum Zitat Jackson JK, Higo T, Hunter WL, Burt HM. Topoisomerase inhibitors as anti-arthritic agents. Inflamm Res. 2008;57:126–34.PubMedCrossRef Jackson JK, Higo T, Hunter WL, Burt HM. Topoisomerase inhibitors as anti-arthritic agents. Inflamm Res. 2008;57:126–34.PubMedCrossRef
50.
Zurück zum Zitat Gang GT, Hwang JH, Kim YH, Noh JR, Kim KS, Jeong JY, et al. Protection of NAD(P)H:quinone oxidoreductase 1 against renal ischemia/reperfusion injury in mice. Free Radic Biol Med. 2014;67:139–49.PubMedCrossRef Gang GT, Hwang JH, Kim YH, Noh JR, Kim KS, Jeong JY, et al. Protection of NAD(P)H:quinone oxidoreductase 1 against renal ischemia/reperfusion injury in mice. Free Radic Biol Med. 2014;67:139–49.PubMedCrossRef
51.
Zurück zum Zitat Lee JS, Park AH, Lee SH, Lee SH, Kim JH, Yang SJ, et al. beta-Lapachone, a modulator of NAD metabolism, prevents health declines in aged mice. PLoS ONE. 2012;7:e47122.PubMedCentralPubMedCrossRef Lee JS, Park AH, Lee SH, Lee SH, Kim JH, Yang SJ, et al. beta-Lapachone, a modulator of NAD metabolism, prevents health declines in aged mice. PLoS ONE. 2012;7:e47122.PubMedCentralPubMedCrossRef
Metadaten
Titel
β-Lapachone suppresses neuroinflammation by modulating the expression of cytokines and matrix metalloproteinases in activated microglia
verfasst von
Eun-Jung Lee
Hyun-Myung Ko
Yeon-Hui Jeong
Eun-Mi Park
Hee-Sun Kim
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
Journal of Neuroinflammation / Ausgabe 1/2015
Elektronische ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-015-0355-z

Weitere Artikel der Ausgabe 1/2015

Journal of Neuroinflammation 1/2015 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.