Skip to main content
Erschienen in: Journal of Neuroinflammation 1/2018

Open Access 01.12.2018 | Research

Unveiling anti-oxidative and anti-inflammatory effects of docosahexaenoic acid and its lipid peroxidation product on lipopolysaccharide-stimulated BV-2 microglial cells

verfasst von: Bo Yang, Runting Li, C. Michael Greenlief, Kevin L. Fritsche, Zezong Gu, Jiankun Cui, James C. Lee, David Q. Beversdorf, Grace Y. Sun

Erschienen in: Journal of Neuroinflammation | Ausgabe 1/2018

Abstract

Background

Phospholipids in the central nervous system are enriched in n-3 and n-6 polyunsaturated fatty acids (PUFA), especially docosahexaenoic acid (DHA) and arachidonic acid (ARA). These PUFA can undergo enzymatic reactions to produce lipid mediators, as well as reaction with oxygen free radicals to produce 4-hydroxyhexenal (4-HHE) from DHA and 4-hydroxynonenal (4-HNE) from ARA. Recent studies demonstrated pleiotropic properties of these peroxidation products through interaction with oxidative and anti-oxidant response pathways. In this study, BV-2 microglial cells were used to investigate ability for DHA, 4-HHE, and 4-HNE to stimulate the anti-oxidant stress responses involving the nuclear factor erythroid-2-related factor 2 (Nrf2) pathway and synthesis of heme oxygenase (HO-1), as well as to mitigate lipopolysaccharide (LPS)-induced nitric oxide (NO), reactive oxygen species (ROS), and cytosolic phospholipase A2 (cPLA2). In addition, LC-MS/MS analysis was carried out to examine effects of exogenous DHA and LPS stimulation on endogenous 4-HHE and 4-HNE levels in BV-2 microglial cells.

Methods

Effects of DHA, 4-HHE, and 4-HNE on LPS-induced NO production was determined using the Griess reagent. LPS-induced ROS production was measured using CM-H2DCFDA. Western blots were used to analyze expression of p-cPLA2, Nrf2, and HO-1. Cell viability and cytotoxicity were measured using the WST-1 assay, and cell protein concentrations were measured using the BCA protein assay kit. An ultra-high-performance liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis was used to determine levels of free 4-HHE and 4-HNE in cells.

Results

DHA (12.5–100 μM), 4-HHE (1.25–10 μM), and 4-HNE (1.25–10 μM) dose dependently suppressed LPS-induced production of NO, ROS, and as p-cPLA2 in BV-2 microglial cells. With the same concentrations, these compounds could enhance Nrf2 and HO-1 expression in these cells. Based on the estimated IC50 values, 4-HHE and 4-HNE were five- to tenfold more potent than DHA in inhibiting LPS-induced NO, ROS, and p-cPLA2. LC-MS/MS analysis indicated ability for DHA (10–50 μM) to increase levels of 4-HHE and attenuate levels of 4-HNE in BV-2 microglial cells. Stimulation of cells with LPS caused an increase in 4-HNE which could be abrogated by cPLA2 inhibitor. In contrast, bromoenol lactone (BEL), a specific inhibitor for the Ca2+-independent phospholipase A2 (iPLA2), could only partially suppress levels of 4-HHE induced by DHA or DHA + LPS.

Conclusions

This study demonstrated the ability of DHA and its lipid peroxidation products, namely, 4-HHE and 4-HNE at 1.25–10 μM, to enhance Nrf2/HO-1 and mitigate LPS-induced NO, ROS, and p-cPLA2 in BV-2 microglial cells. In addition, LC-MS/MS analysis of the levels of 4-HHE and 4-HNE in microglial cells demonstrates that increases in production of 4-HHE from DHA and 4-HNE from LPS are mediated by different mechanisms.
Hinweise
Bo Yang and Runting Li contributed equally to this work.
Abkürzungen
4-HHE
4-Hydroxyhexenal
4-HHE-d3
4-Hydroxyhexenal-d3
4-HNE
4-Hydroxynonenal
ARA
Arachidonic acid
ATK
Arachidonyl trifluoromethyl ketone
BCA
Bicinchoninic acid
BEL
Bromoenol lactone
COX-2
Cyclooxygenase-2
cPLA2
Cytosolic phospholipase A2
DHA
Docosahexaenoic acid
DMEM
Dulbecco’s modified Eagle’s medium
DMSO
Dimethyl sulfoxide
FBS
Fetal bovine albumin
HO-1
Heme oxygenase-1
IFNγ
Interferon-γ
iNOS
Inducible nitric oxide synthase
iPLA2
Ca2+-independent phospholipase A2
LC-MS/MS
Liquid chromatography-tandem mass spectrometry
LLOQ
Lower limit of quantification
LOX
Lipoxygenase
LPS
Lipopolysaccharide
MAPKs
Mitogen-activated protein kinases
MRM
Multiple reaction monitoring
NF-κB
Nuclear factor kappa-light-chain-enhancer of activated B cells
NO
Nitric oxide
Nrf2
Nuclear factor E2-related factor 2
PBS
Phosphate-buffered saline
PLA2
Phospholipases A2
PUFAs
Polyunsaturated fatty acids
ROS
Reactive oxygen species
SD
Standard deviation
SDS-PAGE
Sodium dodecyl sulfate-polyacrylamide gel electrophoresis
SEM
Standard error of mean
SPE
Solid-phase extraction
TBS
Tris-buffered saline

Background

The high abundance of n-3 polyunsaturated fatty acids (PUFA) in the phospholipids in the brain and retina has led to immense interest in the search of their functional roles in health and diseases in these organs [1]. A number of studies with animal models have demonstrated the ability of docosahexaenoic acid (DHA) to protect the brain against aging and neurodegenerative diseases, including stroke, spinal cord injury, and traumatic brain injury [26]. Studies with human populations have also demonstrated effects of n-3 PUFA to improve neurologic functions such as depression, cognition, and memory [7]. To date, n-3 PUFA in the form of fish oil are probably one of the most highly consumed supplements by humans. Nevertheless, despite of a possible link between the n-3 PUFA and neural functions, the underlying mechanisms for the beneficial effects remain elusive [8].
Based on the “deacylation-reacylation” hypothesis, the PUFA in membrane phospholipids, in particular, the arachidonic acid (ARA) and DHA, may become metabolically active upon release by phospholipases A2 (PLA2) [9]. Studies have demonstrated the release of ARA through the action of cPLA2, a Ca2+-dependent enzyme with active phosphorylation sites linked to the mitogen-activated protein kinases (MAPKs) [9]. In microglial cells, activation of cPLA2 has been linked to inflammatory responses, increases in reactive oxygen species (ROS), and inducible nitric oxide synthase (iNOS) [10]. In contrast, the release of DHA is mediated by the action of Ca2+-independent PLA2 (iPLA2) [1114]. While ARA is metabolized by cyclooxygenases (COX) and lipoxygenases (LOX) to produce eicosanoids, which are inflammatory mediators, DHA is metabolized by specific LOX to form oxylipins which are generally protective [1].
Besides the enzymatic pathways, these PUFA are also substrates of non-enzymatic action through peroxidation by radiation and ROS generated from extracellular and intracellular sources. Lipid peroxidation products such as 4-hydroxynonenal (4-HNE) from ARA and 4-hydroxyhexenal (4-HHE) from DHA are bioactive molecules capable of forming adducts with DNA, proteins, and lipids [1517]. Increases in lipid peroxidation have been implicated in neuroinflammatory diseases [18, 19]. In fact, 4-HNE protein adducts have been used as a marker for different types of brain injury [16]. Although less is known about adducts with 4-HHE, advances in studies using LC-MS/MS methods have identified these 4-hydroxy alkenals in tissues, plasma, and urine [2024]. Due to their electrophilic properties [25], there is evidence that these 4-hydroxy alkenals can engage in adaptive responses by upregulating the anti-oxidant response pathway involving Nrf2 which leads to increases in synthesis of phase II enzymes, including HO-1 [20, 24, 2628].
Microglia are important immune cells in the brain, and their activation has been implicated in a number of neurodegenerative diseases and different types of brain injury. In recent years, numerous studies have used murine immortalized BV-2 microglial cells as a model to investigate inflammatory and oxidative responses. Our previous studies showed that BV-2 cells exhibit many properties similar to primary microglial cells isolated from 7- to 10-day-old C57BL/6 pup brains using the magnetic activated cell sorting protocol [29]. In this study, primary microglial cells were isolated using a cluster of differentiation molecule CD11b microbeads. Cells were cultured in DMEM supplemented with 10% FBS containing 100 units/mL penicillin and streptomycin, and culture medium was replaced every 3–5 days and cultures were used between 5 and 7 days. Our results showed that both primary microglia and BV-2 cells exhibited active response to LPS and interferon gamma (IFNγ) which induced time-dependent increases in ERK-1/2, p-cPLA2, and iNOS [29]. However, despite the similarities in these responses, subtle differences could be found in the extent and time of the responses (see Fig. 2 in Chuang’s paper). Based on the presence of Toll-like receptors as well as mitogen-activated protein kinase (MAPK)-induced cPLA2 pathways in both primary and BV-2 cells, numerous studies have used BV-2 cells to study the effects of botanical polyphenols and other factors on neuroinflammation [2931].
Despite an yet unknown mechanism, a number of studies demonstrated the ability for DHA and its oxidative products to modulate the oxidant and inflammatory responses in microglial cells [3236]. Studies on the vascular cell system also demonstrated ability for DHA to inhibit inflammation by modulating the cross-talk between the Nrf2/HO-1 and the NF-κB pathways [37]. In endothelial cells, relatively low concentrations of 4-HHE added exogenously could upregulate the Nrf2 pathway and increase synthesis of HO-1, a potent anti-oxidative enzyme [38, 39]. However, a systemic comparison of the effects of DHA, 4-HHE, and 4-HNE on the Nrf2/HO-1 and LPS-induced inflammatory responses in BV-2 microglial cells has not been investigated. In addition, little is known whether the endogenous lipid peroxidation products in these cells can be regulated by exogenous DHA and stimulation with LPS. Here, BV-2 cells were used to examine the effects of exogenous DHA, 4-HHE, and 4-HNE to enhance the Nrf2 and HO-1 pathway, and to mitigate LPS-induced NO, ROS, and cPLA2. Subsequently, using LC-MS/MS analysis to measure intracellular levels of 4-HHE and 4-HNE in the BV-2 cells, studies were performed to evaluate mechanisms for DHA and LPS on these peroxidation products.

Methods

Materials

Dulbecco’s modified Eagle’s medium (DMEM) and penicillin/streptomycin were obtained from GIBCO (Gaithersburg, MD). U0126 inhibitor was purchased from Cell Signaling (Beverly, MA). Arachidonyl trifluoromethyl ketone (ATK, AACOCF3), racemic bromoenol lactone (BEL), 4-hydroxyhexenal (4-HHE, 1 mg in 0.1 mL of ethanol), 4-hydroxynonenal (4-HNE, 1 mg in 0.1 mL of ethanol), 4-hydroxyhexenal-d3 (4-HHE-d3, 100 μg in 0.1 mL of methyl acetate), and docosahexaenoic acid (DHA, 100 mg in 0.4 mL of ethanol) were purchased from Cayman Chemical (Ann Arbor, MI). Fetal bovine albumin (FBS), Greiss reagent (sulfanilamide and N-1-napthylethylenediamine dihydrochloride), 1, 3-cyclohexanedione (CHD, 97%), ammonium acetate (HPLC grade), acetic acid (ACS grade), and formic acid (mass spectrometry grade) were purchased from Sigma-Aldrich (St. Louis, MO). For ROS detection, CM-H2DCFDA was purchased from Invitrogen Inc. (Eugene, OR). WST-1 assay was purchased from Clontech (Mountain View, CA). Antibodies used include the following: Nrf2 and HO-1 from Santa Cruz Biotechnology (Santa Cruz, CA), p-cPLA2 and cPLA2 from Cell Signaling (Beverly, MA), and monoclonal anti-β-actin peroxidase from Sigma-Aldrich (St. Louis, MO). C18 Sep-Pak cartridges (1 mL, 100 mg) were obtained from Waters Corporation (Milford, MA). Phospholipid removal cartridges (Phree™, 1 mL) were purchased from Phenomenex Inc. (Torrance, CA). All solvents (HPLC grade) used for LC and MS analysis were obtained from Thermo Fisher Scientific Inc. (Fair Lawn, NJ).

Cell culture and treatments

BV-2 microglial cells were used between 14 and 25 passages and were prepared as previously described [40]. Initially, cells were cultured in 75-cm2 flasks with DMEM supplemented with 5% FBS containing 100 units/mL penicillin and streptomycin (100 μg/mL), and maintained in a 5% CO2 incubator at 37 °C. For experiments involving assays of NO and ROS, cells were subcultured in 96-well plates; for Western blot analysis, cells were subcultured in 12-well plates, and for LC-MS/MS analysis, cells were subcultured in 60-mm dishes. In all conditions, cells were subcultured to 80–90% confluence and then serum starved for 3 h prior to adding DHA and inhibitors for 1 h, and followed by stimulation with LPS (100 ng/mL). DHA was suspended in DMEM with 2% fatty acid-free BSA (Sigma-Aldrich, St. Louis, MO), and different concentrations were added to cells. Controls received DMEM with BSA without DHA. 4-HHE and 4-HNE were dissolved in dimethyl sulfoxide (DMSO). The following LPS treatment time schedule was used for different assays: 16 h for NO production, 12 h for assay of ROS, 4 h for assay of p-cPLA2/cPLA2, and 6 h for assay of Nrf2 and HO-1. For LC-MS/MS analysis of 4-HHE and 4-HNE, cells were incubated for 6 h.

Assessment of cell viability

Cell viability was determined using the WST-1 protocol (Clontech Laboratories, Inc., Mountain View, CA). Cells were treated with DHA or alkenals for 17 h with or without LPS. WST-1 was added into each well (96-well plates) with medium/WST-1 reagent at 15:1 (v/v). Cells were incubated for 1 h at 37 °C, and after dissolving the formazan dye with DMSO, absorption was read at 450 nm using a Synergy4 Plate Reader (BioTek Instruments, Inc., St. Louis, MO).

Measurement of ROS production

ROS production was measured using the ROS detection reagent CM-H2DCFDA (DCF) (Invitrogen Inc., Eugene, OR) as described previously [41]. Briefly, BV-2 cells were seeded in a 96-well plates for 24 h. Cells were then starved for 3 h in the serum-free DMEM prior to adding DHA (1 h) and followed by addition of LPS for 11 h. DCF (1 μM final concentration) was added to each well for 1 h. The fluorescence intensity of DCF was measured using the Synergy4 Plate Reader with an excitation wavelength of 490 nm and an emission wavelength of 520 nm.

NO determination in culture medium

NO released from cells was converted to nitrite in the culture medium and was determined using Griess reagent [42]. Cells were cultured in phenol red free DMEM. After treatment with LPS for 16 h, aliquots (50 μL) of culture medium were transferred to 96-well plates and incubated with 50 μL of reagent A [1% (w/v) sulfanilamide (Sigma-Aldrich, St. Louis, MO) in 5% phosphoric acid] for 10 min at room temperature in the dark, and followed by incubation with 50 μL of reagent B [0.1%, w/v, N-1-napthylethylenediamine dihydrochloride (Sigma-Aldrich, St. Louis, MO)] for 10 min at room temperature in the dark. The absorbance of samples was measured at a wavelength of 565 nm using the Synergy4 Plate Reader. Sodium nitrite (0–100 μM), diluted in culture media, was used to prepare the nitrite standard curve.

Western blot analyses

Western blot analysis was carried out as described earlier [29]. Briefly, after various treatment protocols, cells were harvested in Laemmli lysis buffer and centrifuged at 10,000 × g for 15 min at 4 °C to remove cell debris. After protein quantification with the BCA protein assay kit (Pierce Biotechnology, Rockford, IL), samples together with Precision Plus Protein standards (Dual color, BioRad, Hercules, CA) were loaded in sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) gels and resolved at 100 V. After electrophoresis, proteins were transferred to 0.45-μm nitrocellulose membranes at 100 V for 1.5 h. Membrane strips were blocked in Tris-buffered saline (TBS), pH 7.4, with 0.1% Tween 20 (TBS-T) containing 5% non-fat milk for 1.5 h at room temperature. The blots were incubated with anti-Nrf2 (1:500 dilution), anti-HO-1 (1:800 dilution), or p-cPLA2 (1:1000 dilution) or cPLA2 (1:1000 dilution) antibodies overnight at 4 °C. After repeated washing with TBS-T, blots were incubated with goat anti-rabbit IgG-horseradish peroxidase (1:6000 dilution) for 1 h at room temperature and then washed three times with TBS-T. Immuno-labeling was detected by SuperSignal chemiluminescent substrates (Thermo Scientific, Rockford, IL). For loading control, blots were incubated with anti-β-actin (1:50,000) and goat anti-mouse IgG-horseradish peroxidase (1:6000). Films were scanned, and the optical density of protein bands was measured using the QuantityOne software program (BioRad, Hercules, CA).

Quantitative analysis of 4-HHE and 4-HNE in microglial cells

Cells were subcultured in 60-mm dishes, and after different treatment conditions, the medium was removed and 0.5 mL of phosphate-buffered saline (PBS)-methanol (1:1, v/v) was added. Cells were harvested by scraping and then transferred into an Eppendorf tube. The cell suspension was mixed, and aliquots were taken for protein determination and for determination of 4-HHE and 4-HNE using LC-MS/MS. For the analysis, 30 μL of analyte was added to equal volume of internal standard (4-HHE-d3, 1000 ng/mL), and acetonitrile (0.5 mL) containing 1% formic acid was added to the mixture to precipitate the proteins. Solid-phase extraction (SPE) was carried out using a Phree™ cartridge to remove the phospholipids [43]. After evaporation by nitrogen to remove the excess solvent, samples were dissolved in 20 μL of methanol, followed by adding 200 μL of freshly prepared acidified 1,3-cyclohexanedione (CHD) reagent [44]. In brief, CHD (25 mg), ammonium acetate (1 g), and acetic acid (0.5 mL) were dissolved in HPLC water to a total volume of 10 mL. The pale yellow solution (pH 5.0) was incubated at 60 °C for 1 h and cooled on ice prior to addition. The derivatization reaction mixture was heated at 60 °C for another 1 h, cooled on ice, and then desalted through a C18 SPE cartridge. The C18 cartridge was pre-conditioned with methanol (0.7 mL) and water (0.7 mL). The reaction mixture (of 220 μL total volume) was loaded onto the cartridge, followed by washing twice with water (0.7 mL) and finalized with 5% acetonitrile in water (0.7 mL). 4-HHE and 4-HNE derivatives were eluted with 100% acetonitrile (0.7 mL) and dried with a steady stream of nitrogen gas. The samples were reconstituted in 40% methanol in water (300 μL) containing 0.1% formic acid and were ready for LC-MS/MS analysis.
A Waters Acquity LC system equipped with a quaternary solvent manager was used in conjunction with a C18 column (Acquity CSH, 1.7 μm, 100 × 2.1 mm2, Waters, Milford, MA, USA). A LC method was developed and optimized to separate the 4-HHE and 4-HNE derivatives with a 7-min LC run time including equilibration. Water containing 0.1% formic acid (solution A) and methanol containing 0.1% formic acid (solution B) were used as the mobile phases. The solvent gradient was 60% A 40% B at 0 min, 60% A 40% B at 0.25 min, 10% A 90% B at 3.55 min, 10% A 90% B at 3.80 min, 60% A 40% B at 3.90 min, and 60% A 40% B at 7.00 min. The flow rate was 0.3 mL/min, and the injection volume was 10 μL in the full-loop mode. The column was heated to 40 °C, and the sample chamber was maintained at 10 °C.
A Waters Xevo TQ-S triple quadrupole mass spectrometer with electrospray ionization, operated in the positive-ionization mode, was used. The multiple reaction monitoring (MRM) scans were conducted by selecting parent and daughter ion pairs of a specific analyte, which were optimized using the IntelliStart™ software. The 4-HNE derivatives exhibited an abundant pseudo parent ion, [M-18+H]+ at m/z 326.3 Da. The dehydrated parent ion generates a major principal daughter ion at m/z 216.1 Da. The MRM transition m/z 326.3 to > 216.1 Da showed higher sensitivity than other transitions and consequently was chosen as the quantifying transition for the 4-HNE derivative. Similarly, the MRM transitions m/z 284.2 to > 216.1 Da and 287.2 to > 216.1 Da were chosen for simultaneous monitoring of 4-HHE and 4-HHE-d3 (internal standard) derivatives, respectively. The desolvation temperature was 350 °C, and the source temperature was 150 °C. A capillary voltage of 3.8 kV, cone voltage of 40 V, and collision energy of 25 eV were used. The nebulizer gas rate was 800 L N2/h. MassLynx software (Version 4.1, Waters) was used for all data acquisition.
Because of the presence of endogenous 4-HHE and 4-HNE in the cell suspension, aqueous solutions were used to prepare the calibration standards. All the stock solutions (50 μg/mL) were prepared in methanol. A working solution containing 2.5 μg/mL of 4-HHE was prepared by diluting the stock solution with methanol. The internal standard working solution (1000 ng/mL) was prepared in methanol. Calibration curves were prepared by spiking the 4-HHE working solution into aqueous solution and preparing serial dilutions that yielded seven calibration standards (15.63, 31.25, 62.50, 125.0, 250.0, 500.0, and 1000 ng/mL). Similar calibration standards with the same spiking concentrations but in cell suspensions for 4-HHE were also prepared and used to assess any difference between standard curves prepared using aqueous solution or cell suspensions. The results showed the same response for 4-HHE in the two solvent systems and that calibration curves prepared in aqueous solution are suitable for analysis of the biological samples. Similarly, seven calibration standards (6.25, 12.5, 25.0, 50.0, 100, 125, and 250 ng/mL) of 4-HNE were prepared as described, and no difference between solvent systems was observed.
A full method for validation was performed, including analysis of limit of quantification, linearity, recovery, matrix effect, and intraday/interday reproducibility as previously described [45]. Briefly, the lower limit of quantification (LLOQ) is defined as a signal-to-noise ratio of 10:1 for a given MRM chromatogram. Recovery of phospholipid removal by the SPE cartridge was determined by comparing the ratio of peak areas of analyte to the internal standard peak area for samples spiked before SPE to the ratio of peak areas of analyte to internal standard spiked after SPE. Recovery of the C18 SPE cartridge was accessed by comparing the ratio of peak areas of analyte to the internal standard for samples prior to SPE to the ratio of peak areas of analyte to internal standard spiked after SPE. Because of the presence of endogenous levels of 4-HHE and 4-HNE in the cell suspension, the potential matrix effects of the cell suspension were analyzed by comparing the peak area of the spiked 4-HHE-d3 in cell suspension to neat standard prepared at the same concentration in methanol. The precision and accuracy of interday and intraday reproducibility were analyzed by performing analysis on three different batches of quality control samples over 3 days.

Statistical analysis

For studies to assess oxidative and inflammatory responses, triplicate analyses were performed on a given sample and at least three independent experiments (with different passages) were performed for each condition. For studies measuring 4-HHE and 4-HNE levels using the LC-MS/MS analysis, cells in each passage were cultured in 60-mm dishes (in triplicate) and assays of alkenals from each extract were carried out in duplicate. Results are expressed as standard error of the mean (SEM) and analyzed by one-way ANOVA followed by Bonferroni post-tests (v7.00; GraphPad Prism Software Inc., San Diego, CA). When two groups were compared, differences were analyzed by a two-tailed Student’s t test. Differences were considered significant at p < 0.05 for all analyses.

Results

Effects of DHA, 4-HHE, and 4-HNE on BV-2 cell viability

In this study, the WST-1 assay was used to test the concentrations of DHA, 4-HHE, and 4-HNE to confer cytotoxic effects in BV-2 microglial cells. Cells were serum starved for 3 h and followed by incubation of test compounds at different concentrations for 16 h. This time point was also used for the NO assay. As shown in Fig. 1a, cytotoxic effects for DHA were observed with concentrations greater than 150 μM. Figure 1b, c shows cell viability decreasing for 4-HHE at concentrations higher than 40 μM and for 4-HNE at concentrations higher than 30 μM. Based on these results, subsequent studies to test the effects of these compounds were carried out below the cytotoxic concentrations.

DHA, 4-HHE, and 4-HNE suppress LPS-induced NO production in microglial cells

In this study, we tested the effects of DHA as well as 4-HHE and 4-HNE on LPS-induced NO production in BV-2 cells. Figure 2a shows that DHA (12.5–100 μM) diminished LPS-induced NO production in a dose-dependent manner with significant decrease (p < 0.05) noted at concentrations > 25 μM and an IC50 of 76.8 μM. Under this condition, incubation of cells with DHA at different concentrations followed by LPS treatment did not alter cell viability as demonstrated by WST-1 assay (Fig. 2d).
Initial studies to test the effects of 4-HHE and 4-HNE on LPS-mediated oxidative and inflammatory responses in microglial cells indicated an effective dose range of 1.25 to 10 μM. As shown in Fig. 2b, c, 4-HHE (1.25–10 μM) and 4-HNE (1.25–10 μM) reduced LPS-induced NO production in a dose-dependent manner with significant reduction (p < 0.05) at concentrations more than 2.5 μM. The IC50 value for 4-HHE to reduce LPS-induced NO was 6.9 μM and that for 4-HNE was 5.9 μM. At these conditions, neither 4-HHE nor 4-HNE showed cytotoxic effects to the cells (Fig. 2e, f).

DHA, 4-HHE, and 4-HNE suppress LPS-induced production of ROS

Using CM-H2DCFDA, we examined the effects of DHA (12.5–100 μM), 4-HHE (1.25–10 μM), and 4-HNE (1.25–10 μM) on ROS production in microglial cells incubated for 12 h with or without LPS (100 ng/mL). As shown in Fig. 3a, DHA suppressed LPS-induced ROS in a dose-dependent manner with significant decrease (p < 0.05) at 50 μM or higher. Estimation of IC50 for DHA indicated a value of 44.7 μM (Fig. 3a). Treatment with 4-HHE (1.25–10 μM) and 4-HNE (1.25–10 μM) also reduced LPS-induced ROS with IC50 of 7.1 and 6.8 μM, respectively (Fig. 3b, c). Treatment with DHA, 4-HHE, or 4-HNE alone without LPS did not alter endogenous ROS levels in the cells (Fig. 3a–c).

DHA, 4-HHE, and 4-HNE suppress LPS-induced p-cPLA2

Results from earlier studies have demonstrated the ability of LPS to activate p-cPLA2 in microglial cells through the p-ERK1/2-dependent pathway [10, 29]. In this study, we tested the ability of DHA, 4-HHE, and 4-HNE to suppress LPS-induced p-cPLA2. Figure 4a–c shows that DHA (12.5–100 μM) as well as 4-HHE (1.25–10 μM) and 4-HNE (1.25–10 μM) suppressed LPS-induced p-cPLA2 expression in a dose-dependent manner. The IC50 values for DHA, 4-HHE, and 4-HNE are 46.3, 6.6, and 4.8 μM, respectively.

DHA, 4-HHE, and 4-HNE upregulate the anti-oxidant response involving Nrf2 and HO-1

In our earlier studies, we observed that some botanical polyphenols, such as quercetin, not only suppressed LPS-stimulated NO and ROS production, but could also enhance the anti-oxidant pathway involving Nrf2 and production of the potent anti-oxidative enzyme HO-1 [46]. In the present study, DHA (12.5–100 μM) as well as 4-HHE (1.25–10 μM) and 4-HNE (1.25–10 μM) showed a dose-dependent increase in the expression of Nrf2 and HO-1 in microglial cells (Fig. 5a–c). A comparison of the effects of 4-HHE and 4-HNE on HO-1 expression showed that 4-HHE was more potent than 4-HNE in producing HO-1 (Fig. 5d).

DHA and LPS regulate 4-HHE and 4-HNE levels in microglial cells

Studies so far have indicated anti-inflammatory and anti-oxidative effects of 4-HHE and 4-HNE added exogenously to microglial cells. Since these two alkenals are derived from two different PUFA with opposing functions, it seems important to examine whether these compounds are produced within the cells and how their production is regulated by LPS and DHA. First, we developed and validated the LC-MS/MS analysis of 4-HHE and 4-HNE. A summary of the validation is shown in Tables 1 and 2. The LC-MS/MS analysis was then used to simultaneous measure levels of 4-HHE and 4-HNE in microglial cells (106 cells plated in 60 mm dish) which were pre-treated with DHA (50 μM) for 1 h and followed by stimulation with LPS (100 ng/mL) for 6 h. Initially, analysis of the culture medium after 6-h incubation indicated no alkenals were detected in the culture medium. Subsequently, after removing the culture medium, cells were harvested from the culture dish by scraping with 0.5 mL of PBS/methanol (1:1, v/v). The cell suspension was then transferred into an Eppendorf tube and was used for both protein determination and for LC-MS/MS analysis.
Table 1
Method validation parameters for detection of 4-HHE in microglia cells
Linear range (ng/mL)
Linearity
Matrix effect
Phree recovery
Sep-Pak recovery
Intraday
Interday
Accuracy (bias, %)
Precision (CV, %)
Accuracy (bias, %)
Precision (CV, %)
15.63–1000
0.9994
92.6%
104.4%
92.4%
0.5%
0.7%
1.5%
0.8%
CV coefficient of variance
Table 2
Method validation parameters for detection of 4-HNE in microglia cells
Linear range (ng/mL)
Linearity
Matrix effect
Phree recovery
Sep-Pak recovery
Intraday
Interday
Accuracy (bias, %)
Precision (CV, %)
Accuracy (bias, %)
Precision (CV, %)
6.25–250
0.9989
92.6%
88.8%
92.1%
2.4%
1.0%
9.9%
1.3%
CV coefficient of variance

Effects of DHA and/or LPS on 4-HHE and 4-HNE in BV-2 microglial cells

Using the LC-MS/MS method, we first tested the effects of different doses of DHA (10–50 μM) on the levels of 4-HHE and 4-HNE in BV-2 microglial cells in the presence and absence of LPS (100 ng/mL). As shown in Fig. 6a, when cells were treated with DHA (10, 25, and 50 μM) for 7 h, there was a dose-dependent increase in levels of 4-HHE with significant increases (p < 0.05) at 25 μM or higher. Under these conditions, treatment with DHA resulted in a dose-dependent decrease in levels of 4-HNE with significant decrease (p < 0.05) at 50 μM (Fig. 6b). We further determined levels of 4-HHE and 4-HNE in cells treated with DHA (50 μM) and/or LPS (100 ng/mL). Figure 6c showed that when LPS is added after DHA, there is a small but no significant further increase in 4-HHE as compared with treatment with DHA alone (Fig. 6c). Cells stimulated with LPS showed a significant increase (p < 0.05) in levels of 4-HNE (Fig. 6d), albeit no change in the levels of 4-HHE (Fig. 6c). Furthermore, when cells were pre-treated with DHA and followed with LPS, the ability of LPS to increase 4-HNE was reduced (Fig. 6d). In this experiment, levels of 4-HHE and 4-HNE were expressed based on the amount of proteins in the cell samples and there was no observable change in protein levels for all the treatment conditions (Fig. 6e).

Regulation of 4-HHE and 4-HNE by cPLA2 and iPLA2 inhibitors

Since our earlier studies provided evidence for LPS to stimulate p-ERK1/2 and subsequently phosphorylation of cPLA2 in primary and BV-2 cells, we surmise that the LPS-induced increase in 4-HNE is associated with activation of the LPS/p-ERK1/2/p-cPLA2 pathway, and in turn, the increase in ARA is subsequently subject to peroxidation to produce 4-HNE [29]. In order to test this hypothesis, cells were pre-treated with U0126 (5 μM), an inhibitor for the MEK1/2 and ERK1/2 pathway, and ATK (AACOCF3) (5 μM), an inhibitor of cPLA2, and followed by stimulation with LPS. As shown in Fig. 7a, both U0126 and ATK abrogated the LPS-induced increases in the 4-HNE levels, whereas the inhibitors alone did not alter basal levels of 4-HNE or 4-HHE.
Since the iPLA2 has been regarded as the responsible enzyme for the release of DHA from membrane phospholipids, we further investigated whether the increase in 4-HHE due to addition of DHA involves iPLA2. As shown in Fig. 7b, addition of BEL (5 μM), a specific inhibitor for iPLA2, significantly (p < 0.001) decreased the basal levels of 4-HHE in the cells, suggesting the involvement of iPLA2 in the metabolism of the endogenous DHA pool. On the other hand, although exogenous DHA resulted in increased levels of 4-HHE, this increase was only partially inhibited by BEL (Fig. 7b). Similarly, treatment of cells with DHA and LPS resulted in a small increase in 4-HHE as compared to treatment with DHA alone, the increase in 4-HHE was only partially inhibited by BEL (Fig. 7b). With a small decrease in 4-HNE due to the presence of DHA, BEL did not produce obvious changes in 4-HNE levels (Fig. 7b).

Discussion

In this study, we provided evidence that depending on concentration, DHA (12.5–100 μM) as well as 4-HNE (1.25–10 μM) and 4-HHE (1.25–10 μM) can suppress LPS-induced increase in production of NO, ROS, and p-cPLA2 in the BV-2 microglial cells. Also, with the same concentration ranges, these compounds can stimulate the Nrf2 anti-oxidant pathway and increased synthesis of HO-1. A scheme in Fig. 8 depicts the signaling pathways for these compounds acting as electrophiles to upregulate the Nrf2/HO-1 pathway and downregulate pathways for LPS-induced NO/ROS/cPLA2. Based on the IC50, the effects of 4-HNE and 4-HHE are five- to tenfold more potent than DHA. In this study, although both 4-HHE and 4-HNE showed similar effects on suppressing the NO/ROS/cPLA2 and upregulating the Nrf2 pathways, subtle differences can be observed. For example, based on IC50 values, 4-HNE is more potent than 4-HHE in suppressing LPS-induced p-cPLA2 (Fig. 4b). In addition, 4-HHE is more potent than 4-HNE in stimulation of HO-1 (Fig. 5d). This latter observation is in line with a report showing higher electrophilic activity for 4-HHE as compared to 4-HNE [6]. Taken together, results from our study with BV-2 microglial cells, as well as studies with smooth muscle and endothelial cells, clearly demonstrate that exogenous 4-HHE and 4-HNE are capable of enhancing the Nrf2 anti-oxidative activity and suppressing the LPS-induced oxidative/inflammatory responses [3739]. The effective concentration ranges for these alkenals to enhance Nrf2/HO-1 and to suppress the LPS pathways are comparable to those observed with quercetin, a botanical polyphenol enriched in berries [31, 46]. In fact, due to the electrophilic properties of these alkenals, their ability to offer adaptive responses can be shown in a number of cell systems [28, 47]. Studies have also shown the ability of these alkenals to alter cell metabolism through forming adducts with proteins, nucleic acids, and phospholipids [16]. Obviously, more studies are needed to determine the physiological function of these alkenals generated within the cells.
Our ability to develop a sensitive LC-MS/MS method for simultaneous determination of 4-HHE and 4-HNE levels has produced interesting results regarding the underlying mechanism(s) for the regulation of these alkenals in microglial cells. In our protocol, the sensitivity of detection was improved by utilizing a SPE strategy to remove phospholipids which are a major source of matrix effects. When compared with traditional antibody-based methods, the analysis of free aldehydes avoids epitopes that are bound to target proteins or nucleic acids [48].
As shown in Fig. 6a, b, the LC-MS/MS methodology showed higher basal levels of 4-HNE as compared to 4-HHE (around 250 pg to 75 pg/10 μg protein, respectively, in 106 cells cultured in the 60-mm dish). The higher endogenous levels of 4-HNE as compared with 4-HHE in these cells is consistent with results of the fatty acid analysis showing higher levels of ARA than DHA in these cells [34]. In the present study, stimulation of cells with LPS led to the increase in 4-HNE levels without altering the 4-HHE levels (Fig. 6c, d). The ability for LPS to induce an increase in 4-HNE is in agreement with previous studies linking LPS to p-ERK1/2 and p-cPLA2, and in turn, releasing ARA for lipid peroxidation [29]. Consequently, U0126 (an inhibitor for MEK1/2 and p-ERK1/2) and ATK (an inhibitor for cPLA2) were shown to abrogate LPS-induced 4-HNE (Fig. 7a). Based on these results, it can be assumed that measurement of 4-HNE can be used as a biomarker for oxidative and inflammatory responses in microglial cells.
In this study, supplementation of cells with DHA was shown to result in a dose-dependent increase in 4-HHE levels whereas this condition resulted in a concomitant decrease in the levels of 4-HNE (Fig. 6a, b). The mechanism for exogenous DHA to inhibit 4-HNE is not well understood but a possible explanation could be due to inhibition of cPLA2 which is responsible for the release of ARA and hence 4-HNE. This phenomenon can also explain the ability for DHA to inhibit the increase in 4-HNE induced by LPS (Fig. 6d). In this study, cells pre-treated with DHA and followed by stimulation with LPS did not greatly alter the levels of 4-HHE (as compared with those with DHA alone) (Fig. 6c). These results are in agreement with the observation that LPS caused the increases in 4-HNE through activation of cPLA2 and ARA and that this pathway is not related to the DHA-induced 4-HHE pathway (Fig. 9).
Since BEL is a specific inhibitor for iPLA2, its ability to decrease the basal levels of 4-HHE suggests that under normal physiological conditions, iPLA2 is active in providing the pool of DHA from membrane phospholipids. However, it is surprising that BEL only showed a small ability to suppress the increase in 4-HHE due to exogenous DHA (Fig. 7b). These results suggest that during this period of exposure (7 h), a large portion of exogenous DHA probably enters the cell without incorporation into the membrane phospholipids, and this pool is directly available for peroxidation (Fig. 9). This form of entry is in agreement with the study by Tremblay et al. who observed the ability of exogenous DHA to form intracellular lipid bodies which subsequently showed interplay with mitochondrial and other intracellular organelles [49]. A study by Ishikado’s group also showed a fourfold increase in 4-HHE levels upon incubating endothelial cells with DHA (75 μM) for 6 h [38]. Taken together, these results show that lipid peroxidation is an on-going process in the microglial cells and production of 4-HNE and 4-HHE is regulated based on the availability of ARA and DHA within the cells.
Similar to results of our studies, Lu et al. demonstrated ability for DHA to suppress IFNγ-induced inflammatory responses (TNFα, IL-6, NO, and COX-2), as well as increases in HO-1 levels in BV-2 microglial cells [36]. However, relatively high levels for DHA are required to exert these effects. It remains to be further tested whether under these conditions, DHA acts as an electrophile to interact with components of the Nrf2 and NF-κB pathways. It is also possible that DHA, being a highly unsaturated fatty acid, may alter cell surface receptors and thus their downstream pathways [35]. In our earlier study, ARA and DHA were shown to alter membrane properties and activity of amyloid precursor protein in neuronal cells [50]. There is also evolving evidence showing protective effects of DHA due to the hormetic and stress response of 4-HHE in stimulating the Nrf2/HO-1 pathway [20, 27, 38, 51, 52]. However, further studies are needed to test whether this phenomenon can be demonstrated in the brain and other organs.

Conclusions

In summary, our study indicated effects of DHA, 4-HHE, and 4-HNE to enhance Nrf2/HO-1 and mitigate LPS-induced NO, ROS, and cPLA2 responses in BV-2 microglial cells. LC-MS/MS analysis indicated measurable endogenous levels of 4-HNE and 4-HHE in microglial cells, and treatment with DHA resulted in the increases in 4-HHE levels, whereas, treatment with LPS resulted in the increases in 4-HNE levels through a cPLA2-dependent pathway. These measurements demonstrate that lipid peroxidation can be mediated through intracellular redox activity. These results provide a cell model to investigate how lipid peroxidation products may be regulated through intracellular signaling pathways associated with neuroinflammatory responses [53].

Acknowledgements

Thanks are due to Dr. Agnes Simonyi for her initial contribution to the project and in statistical assessments. We also thank Dr. Brian Mooney, associate director of the Charles W. Gehrke Proteomics Center at the University of Missouri, for providing assistance with the LC-MS/MS.

Funding

This study is supported by the MU Research Council grant (17-009) from the University of Missouri to DQB.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.
Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Sun GY, Simonyi A, Fritsche KL, Chuang DY, Hannink M, Gu Z, Greenlief CM, Yao JK, Lee JC, Beversdorf DQ. Docosahexaenoic acid (DHA): an essential nutrient and a nutraceutical for brain health and diseases. Prostaglandins Leukot Essent Fatty Acids. 2017. Sun GY, Simonyi A, Fritsche KL, Chuang DY, Hannink M, Gu Z, Greenlief CM, Yao JK, Lee JC, Beversdorf DQ. Docosahexaenoic acid (DHA): an essential nutrient and a nutraceutical for brain health and diseases. Prostaglandins Leukot Essent Fatty Acids. 2017.
2.
Zurück zum Zitat Salem N Jr, Vandal M, Calon F. The benefit of docosahexaenoic acid for the adult brain in aging and dementia. Prostaglandins Leukot Essent Fatty Acids. 2015;92:15–22.CrossRefPubMed Salem N Jr, Vandal M, Calon F. The benefit of docosahexaenoic acid for the adult brain in aging and dementia. Prostaglandins Leukot Essent Fatty Acids. 2015;92:15–22.CrossRefPubMed
3.
Zurück zum Zitat Denis I, Potier B, Vancassel S, Heberden C, Lavialle M. Omega-3 fatty acids and brain resistance to ageing and stress: body of evidence and possible mechanisms. Ageing Res Rev. 2013;12:579–94.CrossRefPubMed Denis I, Potier B, Vancassel S, Heberden C, Lavialle M. Omega-3 fatty acids and brain resistance to ageing and stress: body of evidence and possible mechanisms. Ageing Res Rev. 2013;12:579–94.CrossRefPubMed
4.
Zurück zum Zitat Paterniti I, Impellizzeri D, Di Paola R, Esposito E, Gladman S, Yip P, Priestley JV, Michael-Titus AT, Cuzzocrea S. Docosahexaenoic acid attenuates the early inflammatory response following spinal cord injury in mice: in-vivo and in-vitro studies. J Neuroinflammation. 2014;11:6.CrossRefPubMedPubMedCentral Paterniti I, Impellizzeri D, Di Paola R, Esposito E, Gladman S, Yip P, Priestley JV, Michael-Titus AT, Cuzzocrea S. Docosahexaenoic acid attenuates the early inflammatory response following spinal cord injury in mice: in-vivo and in-vitro studies. J Neuroinflammation. 2014;11:6.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Michael-Titus AT, Priestley JV. Omega-3 fatty acids and traumatic neurological injury: from neuroprotection to neuroplasticity? Trends Neurosci. 2014;37:30–8.CrossRefPubMed Michael-Titus AT, Priestley JV. Omega-3 fatty acids and traumatic neurological injury: from neuroprotection to neuroplasticity? Trends Neurosci. 2014;37:30–8.CrossRefPubMed
6.
Zurück zum Zitat Zhang M, Wang S, Mao L, Leak RK, Shi Y, Zhang W, Hu X, Sun B, Cao G, Gao Y, Xu Y, Chen J, Zhang F. Omega-3 fatty acids protect the brain against ischemic injury by activating Nrf2 and upregulating heme oxygenase 1. J Neurosci. 2014;34:1903–15.CrossRefPubMedPubMedCentral Zhang M, Wang S, Mao L, Leak RK, Shi Y, Zhang W, Hu X, Sun B, Cao G, Gao Y, Xu Y, Chen J, Zhang F. Omega-3 fatty acids protect the brain against ischemic injury by activating Nrf2 and upregulating heme oxygenase 1. J Neurosci. 2014;34:1903–15.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Grosso G, Pajak A, Marventano S, Castellano S, Galvano F, Bucolo C, Drago F, Caraci F. Role of omega-3 fatty acids in the treatment of depressive disorders: a comprehensive meta-analysis of randomized clinical trials. PLoS One. 2014;9:e96905.CrossRefPubMedPubMedCentral Grosso G, Pajak A, Marventano S, Castellano S, Galvano F, Bucolo C, Drago F, Caraci F. Role of omega-3 fatty acids in the treatment of depressive disorders: a comprehensive meta-analysis of randomized clinical trials. PLoS One. 2014;9:e96905.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Trepanier MO, Hopperton KE, Orr SK, Bazinet RP. N-3 polyunsaturated fatty acids in animal models with neuroinflammation: an update. Eur J Pharmacol. 2016;785:187–206.CrossRefPubMed Trepanier MO, Hopperton KE, Orr SK, Bazinet RP. N-3 polyunsaturated fatty acids in animal models with neuroinflammation: an update. Eur J Pharmacol. 2016;785:187–206.CrossRefPubMed
9.
Zurück zum Zitat Sun GY, Chuang DY, Zong Y, Jiang J, Lee JC, Gu Z, Simonyi A. Role of cytosolic phospholipase A2 in oxidative and inflammatory signaling pathways in different cell types in the central nervous system. Mol Neurobiol. 2014;50:6–14.CrossRefPubMedPubMedCentral Sun GY, Chuang DY, Zong Y, Jiang J, Lee JC, Gu Z, Simonyi A. Role of cytosolic phospholipase A2 in oxidative and inflammatory signaling pathways in different cell types in the central nervous system. Mol Neurobiol. 2014;50:6–14.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Szaingurten-Solodkin I, Hadad N, Levy R. Regulatory role of cytosolic phospholipase A2alpha in NADPH oxidase activity and in inducible nitric oxide synthase induction by aggregated Abeta1-42 in microglia. Glia. 2009;57:1727–40.CrossRefPubMed Szaingurten-Solodkin I, Hadad N, Levy R. Regulatory role of cytosolic phospholipase A2alpha in NADPH oxidase activity and in inducible nitric oxide synthase induction by aggregated Abeta1-42 in microglia. Glia. 2009;57:1727–40.CrossRefPubMed
11.
Zurück zum Zitat Strokin M, Sergeeva M, Reiser G. Prostaglandin synthesis in rat brain astrocytes is under the control of the n-3 docosahexaenoic acid, released by group VIB calcium-independent phospholipase A2. J Neurochem. 2007;102:1771–82.CrossRefPubMed Strokin M, Sergeeva M, Reiser G. Prostaglandin synthesis in rat brain astrocytes is under the control of the n-3 docosahexaenoic acid, released by group VIB calcium-independent phospholipase A2. J Neurochem. 2007;102:1771–82.CrossRefPubMed
12.
Zurück zum Zitat Ong WY, Yeo JF, Ling SF, Farooqui AA. Distribution of calcium-independent phospholipase A2 (iPLA 2) in monkey brain. J Neurocytol. 2005;34:447–58.CrossRefPubMed Ong WY, Yeo JF, Ling SF, Farooqui AA. Distribution of calcium-independent phospholipase A2 (iPLA 2) in monkey brain. J Neurocytol. 2005;34:447–58.CrossRefPubMed
13.
Zurück zum Zitat Green JT, Orr SK, Bazinet RP. The emerging role of group VI calcium-independent phospholipase A2 in releasing docosahexaenoic acid from brain phospholipids. J Lipid Res. 2008;49:939–44.CrossRefPubMed Green JT, Orr SK, Bazinet RP. The emerging role of group VI calcium-independent phospholipase A2 in releasing docosahexaenoic acid from brain phospholipids. J Lipid Res. 2008;49:939–44.CrossRefPubMed
14.
Zurück zum Zitat Ramanadham S, Ali T, Ashley JW, Bone RN, Hancock WD, Lei X. Calcium-independent phospholipases A2 and their roles in biological processes and diseases. J Lipid Res. 2015;56:1643–68.CrossRefPubMedPubMedCentral Ramanadham S, Ali T, Ashley JW, Bone RN, Hancock WD, Lei X. Calcium-independent phospholipases A2 and their roles in biological processes and diseases. J Lipid Res. 2015;56:1643–68.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Yang Y, Sharma R, Sharma A, Awasthi S, Awasthi YC. Lipid peroxidation and cell cycle signaling: 4-hydroxynonenal, a key molecule in stress mediated signaling. Acta Biochim Pol. 2003;50:319–36.PubMed Yang Y, Sharma R, Sharma A, Awasthi S, Awasthi YC. Lipid peroxidation and cell cycle signaling: 4-hydroxynonenal, a key molecule in stress mediated signaling. Acta Biochim Pol. 2003;50:319–36.PubMed
16.
Zurück zum Zitat Luczaj W, Gegotek A, Skrzydlewska E. Antioxidants and HNE in redox homeostasis. Free Radic Biol Med. 2017;111:87–101.CrossRefPubMed Luczaj W, Gegotek A, Skrzydlewska E. Antioxidants and HNE in redox homeostasis. Free Radic Biol Med. 2017;111:87–101.CrossRefPubMed
17.
Zurück zum Zitat Csala M, Kardon T, Legeza B, Lizak B, Mandl J, Margittai E, Puskas F, Szaraz P, Szelenyi P, Banhegyi G. On the role of 4-hydroxynonenal in health and disease. Biochim Biophys Acta. 2015;1852:826–38.CrossRefPubMed Csala M, Kardon T, Legeza B, Lizak B, Mandl J, Margittai E, Puskas F, Szaraz P, Szelenyi P, Banhegyi G. On the role of 4-hydroxynonenal in health and disease. Biochim Biophys Acta. 2015;1852:826–38.CrossRefPubMed
18.
Zurück zum Zitat Moniuszko-Malinowska A, Luczaj W, Jarocka-Karpowicz I, Pancewicz S, Zajkowska J, Andrisic L, Zarkovic N, Skrzydlewska E. Lipid peroxidation in the pathogenesis of neuroborreliosis. Free Radic Biol Med. 2016;96:255–63.CrossRefPubMed Moniuszko-Malinowska A, Luczaj W, Jarocka-Karpowicz I, Pancewicz S, Zajkowska J, Andrisic L, Zarkovic N, Skrzydlewska E. Lipid peroxidation in the pathogenesis of neuroborreliosis. Free Radic Biol Med. 2016;96:255–63.CrossRefPubMed
19.
Zurück zum Zitat Luczaj W, Moniuszko A, Jarocka-Karpowicz I, Pancewicz S, Andrisic L, Zarkovic N, Skrzydlewska E. Tick-borne encephalitis—lipid peroxidation and its consequences. Scand J Clin Lab Invest. 2016;76:1–9.CrossRefPubMed Luczaj W, Moniuszko A, Jarocka-Karpowicz I, Pancewicz S, Andrisic L, Zarkovic N, Skrzydlewska E. Tick-borne encephalitis—lipid peroxidation and its consequences. Scand J Clin Lab Invest. 2016;76:1–9.CrossRefPubMed
20.
Zurück zum Zitat Nakagawa F, Morino K, Ugi S, Ishikado A, Kondo K, Sato D, Konno S, Nemoto K, Kusunoki C, Sekine O, Sunagawa A, Kawamura M, Inoue N, Nishio Y, Maegawa H. 4-Hydroxy hexenal derived from dietary n-3 polyunsaturated fatty acids induces anti-oxidative enzyme heme oxygenase-1 in multiple organs. Biochem Biophys Res Commun. 2014;443:991–6.CrossRefPubMed Nakagawa F, Morino K, Ugi S, Ishikado A, Kondo K, Sato D, Konno S, Nemoto K, Kusunoki C, Sekine O, Sunagawa A, Kawamura M, Inoue N, Nishio Y, Maegawa H. 4-Hydroxy hexenal derived from dietary n-3 polyunsaturated fatty acids induces anti-oxidative enzyme heme oxygenase-1 in multiple organs. Biochem Biophys Res Commun. 2014;443:991–6.CrossRefPubMed
21.
Zurück zum Zitat Guichardant M, Bacot S, Moliere P, Lagarde M. Hydroxy-alkenals from the peroxidation of n-3 and n-6 fatty acids and urinary metabolites. Prostaglandins Leukot Essent Fatty Acids. 2006;75:179–82.CrossRefPubMed Guichardant M, Bacot S, Moliere P, Lagarde M. Hydroxy-alkenals from the peroxidation of n-3 and n-6 fatty acids and urinary metabolites. Prostaglandins Leukot Essent Fatty Acids. 2006;75:179–82.CrossRefPubMed
22.
Zurück zum Zitat Wang M, Fang H, Han X. Shotgun lipidomics analysis of 4-hydroxyalkenal species directly from lipid extracts after one-step in situ derivatization. Anal Chem. 2012;84:4580–6.CrossRefPubMedPubMedCentral Wang M, Fang H, Han X. Shotgun lipidomics analysis of 4-hydroxyalkenal species directly from lipid extracts after one-step in situ derivatization. Anal Chem. 2012;84:4580–6.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Tie C, Hu T, Jia ZX, Zhang JL. Derivatization strategy for the comprehensive characterization of endogenous fatty aldehydes using HPLC-multiple reaction monitoring. Anal Chem. 2016;88:7762–8.CrossRefPubMed Tie C, Hu T, Jia ZX, Zhang JL. Derivatization strategy for the comprehensive characterization of endogenous fatty aldehydes using HPLC-multiple reaction monitoring. Anal Chem. 2016;88:7762–8.CrossRefPubMed
24.
Zurück zum Zitat Long EK, Picklo MJ Sr. Trans-4-hydroxy-2-hexenal, a product of n-3 fatty acid peroxidation: make some room HNE. Free Radic Biol Med. 2010;49:1–8.CrossRefPubMed Long EK, Picklo MJ Sr. Trans-4-hydroxy-2-hexenal, a product of n-3 fatty acid peroxidation: make some room HNE. Free Radic Biol Med. 2010;49:1–8.CrossRefPubMed
25.
26.
Zurück zum Zitat Huang Y, Li W, Kong AN. Anti-oxidative stress regulator NF-E2-related factor 2 mediates the adaptive induction of antioxidant and detoxifying enzymes by lipid peroxidation metabolite 4-hydroxynonenal. Cell Biosci. 2012;2:40.CrossRefPubMedPubMedCentral Huang Y, Li W, Kong AN. Anti-oxidative stress regulator NF-E2-related factor 2 mediates the adaptive induction of antioxidant and detoxifying enzymes by lipid peroxidation metabolite 4-hydroxynonenal. Cell Biosci. 2012;2:40.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Chen ZH, Saito Y, Yoshida Y, Sekine A, Noguchi N, Niki E. 4-Hydroxynonenal induces adaptive response and enhances PC12 cell tolerance primarily through induction of thioredoxin reductase 1 via activation of Nrf2. J Biol Chem. 2005;280:41921–7.CrossRefPubMed Chen ZH, Saito Y, Yoshida Y, Sekine A, Noguchi N, Niki E. 4-Hydroxynonenal induces adaptive response and enhances PC12 cell tolerance primarily through induction of thioredoxin reductase 1 via activation of Nrf2. J Biol Chem. 2005;280:41921–7.CrossRefPubMed
28.
Zurück zum Zitat Maulucci G, Daniel B, Cohen O, Avrahami Y, Sasson S. Hormetic and regulatory effects of lipid peroxidation mediators in pancreatic beta cells. Mol Asp Med. 2016;49:49–77.CrossRef Maulucci G, Daniel B, Cohen O, Avrahami Y, Sasson S. Hormetic and regulatory effects of lipid peroxidation mediators in pancreatic beta cells. Mol Asp Med. 2016;49:49–77.CrossRef
29.
Zurück zum Zitat Chuang DY, Simonyi A, Kotzbauer PT, Gu Z, Sun GY. Cytosolic phospholipase A2 plays a crucial role in ROS/NO signaling during microglial activation through the lipoxygenase pathway. J Neuroinflammation. 2015;12:199.CrossRefPubMedPubMedCentral Chuang DY, Simonyi A, Kotzbauer PT, Gu Z, Sun GY. Cytosolic phospholipase A2 plays a crucial role in ROS/NO signaling during microglial activation through the lipoxygenase pathway. J Neuroinflammation. 2015;12:199.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Block ML, Zecca L, Hong JS. Microglia-mediated neurotoxicity: uncovering the molecular mechanisms. Nat Rev Neurosci. 2007;8:57–69.CrossRefPubMed Block ML, Zecca L, Hong JS. Microglia-mediated neurotoxicity: uncovering the molecular mechanisms. Nat Rev Neurosci. 2007;8:57–69.CrossRefPubMed
31.
Zurück zum Zitat Chuang DY, Simonyi A, Cui J, Lubahn DB, Gu Z, Sun GY. Botanical polyphenols mitigate microglial activation and microglia-induced neurotoxicity: role of cytosolic phospholipase A2. NeuroMolecular Med. 2016;18:415–25.CrossRefPubMed Chuang DY, Simonyi A, Cui J, Lubahn DB, Gu Z, Sun GY. Botanical polyphenols mitigate microglial activation and microglia-induced neurotoxicity: role of cytosolic phospholipase A2. NeuroMolecular Med. 2016;18:415–25.CrossRefPubMed
32.
Zurück zum Zitat Antonietta Ajmone-Cat M, Lavinia Salvatori M, De Simone R, Mancini M, Biagioni S, Bernardo A, Cacci E, Minghetti L. Docosahexaenoic acid modulates inflammatory and antineurogenic functions of activated microglial cells. J Neurosci Res. 2012;90:575–87.CrossRefPubMed Antonietta Ajmone-Cat M, Lavinia Salvatori M, De Simone R, Mancini M, Biagioni S, Bernardo A, Cacci E, Minghetti L. Docosahexaenoic acid modulates inflammatory and antineurogenic functions of activated microglial cells. J Neurosci Res. 2012;90:575–87.CrossRefPubMed
33.
Zurück zum Zitat Inoue T, Tanaka M, Masuda S, Ohue-Kitano R, Yamakage H, Muranaka K, Wada H, Kusakabe T, Shimatsu A, Hasegawa K, Satoh-Asahara N. Omega-3 polyunsaturated fatty acids suppress the inflammatory responses of lipopolysaccharide-stimulated mouse microglia by activating SIRT1 pathways. Biochim Biophys Acta. 2017;1862:552–60.CrossRefPubMed Inoue T, Tanaka M, Masuda S, Ohue-Kitano R, Yamakage H, Muranaka K, Wada H, Kusakabe T, Shimatsu A, Hasegawa K, Satoh-Asahara N. Omega-3 polyunsaturated fatty acids suppress the inflammatory responses of lipopolysaccharide-stimulated mouse microglia by activating SIRT1 pathways. Biochim Biophys Acta. 2017;1862:552–60.CrossRefPubMed
34.
Zurück zum Zitat Fourrier C, Remus-Borel J, Greenhalgh AD, Guichardant M, Bernoud-Hubac N, Lagarde M, Joffre C, Laye S. Docosahexaenoic acid-containing choline phospholipid modulates LPS-induced neuroinflammation in vivo and in microglia in vitro. J Neuroinflammation. 2017;14:170.CrossRefPubMedPubMedCentral Fourrier C, Remus-Borel J, Greenhalgh AD, Guichardant M, Bernoud-Hubac N, Lagarde M, Joffre C, Laye S. Docosahexaenoic acid-containing choline phospholipid modulates LPS-induced neuroinflammation in vivo and in microglia in vitro. J Neuroinflammation. 2017;14:170.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat De Smedt-Peyrusse V, Sargueil F, Moranis A, Harizi H, Mongrand S, Laye S. Docosahexaenoic acid prevents lipopolysaccharide-induced cytokine production in microglial cells by inhibiting lipopolysaccharide receptor presentation but not its membrane subdomain localization. J Neurochem. 2008;105:296–307.CrossRefPubMed De Smedt-Peyrusse V, Sargueil F, Moranis A, Harizi H, Mongrand S, Laye S. Docosahexaenoic acid prevents lipopolysaccharide-induced cytokine production in microglial cells by inhibiting lipopolysaccharide receptor presentation but not its membrane subdomain localization. J Neurochem. 2008;105:296–307.CrossRefPubMed
36.
Zurück zum Zitat Lu DY, Tsao YY, Leung YM, Su KP. Docosahexaenoic acid suppresses neuroinflammatory responses and induces heme oxygenase-1 expression in BV-2 microglia: implications of antidepressant effects for omega-3 fatty acids. Neuropsychopharmacology. 2010;35:2238–48.CrossRefPubMedPubMedCentral Lu DY, Tsao YY, Leung YM, Su KP. Docosahexaenoic acid suppresses neuroinflammatory responses and induces heme oxygenase-1 expression in BV-2 microglia: implications of antidepressant effects for omega-3 fatty acids. Neuropsychopharmacology. 2010;35:2238–48.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Yang YC, Lii CK, Wei YL, Li CC, Lu CY, Liu KL, Chen HW. Docosahexaenoic acid inhibition of inflammation is partially via cross-talk between Nrf2/heme oxygenase 1 and IKK/NF-kappaB pathways. J Nutr Biochem. 2013;24:204–12.CrossRefPubMed Yang YC, Lii CK, Wei YL, Li CC, Lu CY, Liu KL, Chen HW. Docosahexaenoic acid inhibition of inflammation is partially via cross-talk between Nrf2/heme oxygenase 1 and IKK/NF-kappaB pathways. J Nutr Biochem. 2013;24:204–12.CrossRefPubMed
38.
Zurück zum Zitat Ishikado A, Morino K, Nishio Y, Nakagawa F, Mukose A, Sono Y, Yoshioka N, Kondo K, Sekine O, Yoshizaki T, Ugi S, Uzu T, Kawai H, Makino T, Okamura T, Yamamoto M, Kashiwagi A, Maegawa H. 4-Hydroxy hexenal derived from docosahexaenoic acid protects endothelial cells via Nrf2 activation. PLoS One. 2013;8:e69415.CrossRefPubMedPubMedCentral Ishikado A, Morino K, Nishio Y, Nakagawa F, Mukose A, Sono Y, Yoshioka N, Kondo K, Sekine O, Yoshizaki T, Ugi S, Uzu T, Kawai H, Makino T, Okamura T, Yamamoto M, Kashiwagi A, Maegawa H. 4-Hydroxy hexenal derived from docosahexaenoic acid protects endothelial cells via Nrf2 activation. PLoS One. 2013;8:e69415.CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Ishikado A, Nishio Y, Morino K, Ugi S, Kondo H, Makino T, Kashiwagi A, Maegawa H. Low concentration of 4-hydroxy hexenal increases heme oxygenase-1 expression through activation of Nrf2 and antioxidative activity in vascular endothelial cells. Biochem Biophys Res Commun. 2010;402:99–104.CrossRefPubMed Ishikado A, Nishio Y, Morino K, Ugi S, Kondo H, Makino T, Kashiwagi A, Maegawa H. Low concentration of 4-hydroxy hexenal increases heme oxygenase-1 expression through activation of Nrf2 and antioxidative activity in vascular endothelial cells. Biochem Biophys Res Commun. 2010;402:99–104.CrossRefPubMed
40.
Zurück zum Zitat Shen S, Yu S, Binek J, Chalimoniuk M, Zhang X, Lo SC, Hannink M, Wu J, Fritsche K, Donato R, Sun GY. Distinct signaling pathways for induction of type II NOS by IFNgamma and LPS in BV-2 microglial cells. Neurochem Int. 2005;47:298–307.CrossRefPubMed Shen S, Yu S, Binek J, Chalimoniuk M, Zhang X, Lo SC, Hannink M, Wu J, Fritsche K, Donato R, Sun GY. Distinct signaling pathways for induction of type II NOS by IFNgamma and LPS in BV-2 microglial cells. Neurochem Int. 2005;47:298–307.CrossRefPubMed
41.
Zurück zum Zitat Chuang DY, Chan MH, Zong Y, Sheng W, He Y, Jiang JH, Simonyi A, Gu Z, Fritsche KL, Cui J, Lee JC, Folk WR, Lubahn DB, Sun AY, Sun GY. Magnolia polyphenols attenuate oxidative and inflammatory responses in neurons and microglial cells. J Neuroinflammation. 2013;10:15.CrossRefPubMedPubMedCentral Chuang DY, Chan MH, Zong Y, Sheng W, He Y, Jiang JH, Simonyi A, Gu Z, Fritsche KL, Cui J, Lee JC, Folk WR, Lubahn DB, Sun AY, Sun GY. Magnolia polyphenols attenuate oxidative and inflammatory responses in neurons and microglial cells. J Neuroinflammation. 2013;10:15.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Sheng W, Zong Y, Mohammad A, Ajit D, Cui J, Han D, Hamilton JL, Simonyi A, Sun AY, Gu Z, Hong JS, Weisman GA, Sun GY. Pro-inflammatory cytokines and lipopolysaccharide induce changes in cell morphology, and upregulation of ERK1/2, iNOS and sPLA (2)-IIA expression in astrocytes and microglia. J Neuroinflammation. 2011;8:121.CrossRefPubMedPubMedCentral Sheng W, Zong Y, Mohammad A, Ajit D, Cui J, Han D, Hamilton JL, Simonyi A, Sun AY, Gu Z, Hong JS, Weisman GA, Sun GY. Pro-inflammatory cytokines and lipopolysaccharide induce changes in cell morphology, and upregulation of ERK1/2, iNOS and sPLA (2)-IIA expression in astrocytes and microglia. J Neuroinflammation. 2011;8:121.CrossRefPubMedPubMedCentral
43.
Zurück zum Zitat Johnson MC, Song H, Cui J, Mossine VV, Gu Z, Greenlief CM. Development of a method and validation for the quantitation of FruArg in mice plasma and brain tissue using UPLC-MS/MS. ACS Omega. 2016;1:663–8.CrossRefPubMedPubMedCentral Johnson MC, Song H, Cui J, Mossine VV, Gu Z, Greenlief CM. Development of a method and validation for the quantitation of FruArg in mice plasma and brain tissue using UPLC-MS/MS. ACS Omega. 2016;1:663–8.CrossRefPubMedPubMedCentral
44.
Zurück zum Zitat O'Brien-Coker IC, Perkins G, Mallet AI. Aldehyde analysis by high performance liquid chromatography/tandem mass spectrometry. Rapid Commun Mass Spectrom. 2001;15:920–8.CrossRefPubMed O'Brien-Coker IC, Perkins G, Mallet AI. Aldehyde analysis by high performance liquid chromatography/tandem mass spectrometry. Rapid Commun Mass Spectrom. 2001;15:920–8.CrossRefPubMed
45.
Zurück zum Zitat Sidhu R, Jiang H, Farhat NY, Carrillo-Carrasco N, Woolery M, Ottinger E, Porter FD, Schaffer JE, Ory DS, Jiang X. A validated LC-MS/MS assay for quantification of 24(S)-hydroxycholesterol in plasma and cerebrospinal fluid. J Lipid Res. 2015;56:1222–33.CrossRefPubMedPubMedCentral Sidhu R, Jiang H, Farhat NY, Carrillo-Carrasco N, Woolery M, Ottinger E, Porter FD, Schaffer JE, Ory DS, Jiang X. A validated LC-MS/MS assay for quantification of 24(S)-hydroxycholesterol in plasma and cerebrospinal fluid. J Lipid Res. 2015;56:1222–33.CrossRefPubMedPubMedCentral
46.
Zurück zum Zitat Sun GY, Chen Z, Jasmer KJ, Chuang DY, Gu Z, Hannink M, Simonyi A. Quercetin attenuates inflammatory responses in BV-2 microglial cells: role of MAPKs on the Nrf2 pathway and induction of heme oxygenase-1. PLoS One. 2015;10:e0141509.CrossRefPubMedPubMedCentral Sun GY, Chen Z, Jasmer KJ, Chuang DY, Gu Z, Hannink M, Simonyi A. Quercetin attenuates inflammatory responses in BV-2 microglial cells: role of MAPKs on the Nrf2 pathway and induction of heme oxygenase-1. PLoS One. 2015;10:e0141509.CrossRefPubMedPubMedCentral
47.
Zurück zum Zitat Chen ZH, Niki E. 4-Hydroxynonenal (4-HNE) has been widely accepted as an inducer of oxidative stress. Is this the whole truth about it or can 4-HNE also exert protective effects? IUBMB Life. 2006;58:372–3.CrossRefPubMed Chen ZH, Niki E. 4-Hydroxynonenal (4-HNE) has been widely accepted as an inducer of oxidative stress. Is this the whole truth about it or can 4-HNE also exert protective effects? IUBMB Life. 2006;58:372–3.CrossRefPubMed
48.
Zurück zum Zitat Sousa BC, Pitt AR, Spickett CM. Chemistry and analysis of HNE and other prominent carbonyl-containing lipid oxidation compounds. Free Radic Biol Med. 2017;111:294–308.CrossRefPubMed Sousa BC, Pitt AR, Spickett CM. Chemistry and analysis of HNE and other prominent carbonyl-containing lipid oxidation compounds. Free Radic Biol Med. 2017;111:294–308.CrossRefPubMed
49.
Zurück zum Zitat Tremblay ME, Zhang I, Bisht K, Savage JC, Lecours C, Parent M, Titorenko V, Maysinger D. Remodeling of lipid bodies by docosahexaenoic acid in activated microglial cells. J Neuroinflammation. 2016;13:116.CrossRefPubMedPubMedCentral Tremblay ME, Zhang I, Bisht K, Savage JC, Lecours C, Parent M, Titorenko V, Maysinger D. Remodeling of lipid bodies by docosahexaenoic acid in activated microglial cells. J Neuroinflammation. 2016;13:116.CrossRefPubMedPubMedCentral
50.
Zurück zum Zitat Yang X, Sun GY, Eckert GP, Lee JC. Cellular membrane fluidity in amyloid precursor protein processing. Mol Neurobiol. 2014;50:119–29.CrossRefPubMed Yang X, Sun GY, Eckert GP, Lee JC. Cellular membrane fluidity in amyloid precursor protein processing. Mol Neurobiol. 2014;50:119–29.CrossRefPubMed
51.
Zurück zum Zitat Nagayama K, Morino K, Sekine O, Nakagawa F, Ishikado A, Iwasaki H, Okada T, Tawa M, Sato D, Imamura T, Nishio Y, Ugi S, Kashiwagi A, Okamura T, Maegawa H. Duality of n-3 polyunsaturated fatty acids on Mcp-1 expression in vascular smooth muscle: a potential role of 4-hydroxy hexenal. Nutrients. 2015;7:8112–26.CrossRefPubMedPubMedCentral Nagayama K, Morino K, Sekine O, Nakagawa F, Ishikado A, Iwasaki H, Okada T, Tawa M, Sato D, Imamura T, Nishio Y, Ugi S, Kashiwagi A, Okamura T, Maegawa H. Duality of n-3 polyunsaturated fatty acids on Mcp-1 expression in vascular smooth muscle: a potential role of 4-hydroxy hexenal. Nutrients. 2015;7:8112–26.CrossRefPubMedPubMedCentral
52.
Zurück zum Zitat Chen ZH, Yoshida Y, Saito Y, Noguchi N, Niki E. Adaptive response induced by lipid peroxidation products in cell cultures. FEBS Lett. 2006;580:479–83.CrossRefPubMed Chen ZH, Yoshida Y, Saito Y, Noguchi N, Niki E. Adaptive response induced by lipid peroxidation products in cell cultures. FEBS Lett. 2006;580:479–83.CrossRefPubMed
53.
Zurück zum Zitat Mancera P, Wappenhans B, Cordobilla B, Virgili N, Pugliese M, Rueda F, Espinosa-Parrilla JF, Domingo JC. Natural docosahexaenoic acid in the triglyceride form attenuates in vitro microglial activation and ameliorates autoimmune encephalomyelitis in mice. Nutrients. 2017;9. Mancera P, Wappenhans B, Cordobilla B, Virgili N, Pugliese M, Rueda F, Espinosa-Parrilla JF, Domingo JC. Natural docosahexaenoic acid in the triglyceride form attenuates in vitro microglial activation and ameliorates autoimmune encephalomyelitis in mice. Nutrients. 2017;9.
Metadaten
Titel
Unveiling anti-oxidative and anti-inflammatory effects of docosahexaenoic acid and its lipid peroxidation product on lipopolysaccharide-stimulated BV-2 microglial cells
verfasst von
Bo Yang
Runting Li
C. Michael Greenlief
Kevin L. Fritsche
Zezong Gu
Jiankun Cui
James C. Lee
David Q. Beversdorf
Grace Y. Sun
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Journal of Neuroinflammation / Ausgabe 1/2018
Elektronische ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-018-1232-3

Weitere Artikel der Ausgabe 1/2018

Journal of Neuroinflammation 1/2018 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.