Skip to main content
Erschienen in: Journal of Neuroinflammation 1/2016

Open Access 01.12.2016 | Research

In vitro evaluation of physiologically relevant concentrations of teriflunomide on activation and proliferation of primary rodent microglia

verfasst von: Tanja Wostradowski, Chittappen Kandiyil Prajeeth, Viktoria Gudi, Jessica Kronenberg, Sina Witte, Marina Brieskorn, Martin Stangel

Erschienen in: Journal of Neuroinflammation | Ausgabe 1/2016

Abstract

Background

Teriflunomide, an inhibitor of dihydroorotate dehydrogenase, is thought to ameliorate multiple sclerosis by reducing activation-induced proliferation of lymphocytes, which is highly dependent on de novo pyrimidine synthesis. Nevertheless, its immunomodulatory effects on resident glial cells in the central nervous system are only poorly understood.

Methods

In this study, we employed physiologically relevant concentrations of teriflunomide and investigated its effects on survival, proliferation, activation, and function of primary rat microglia in vitro.

Results

We demonstrate that teriflunomide had no cytotoxic effect on microglia and had only a minor impact on microglial activation. In a concentration- and time-dependent manner, teriflunomide significantly downregulated surface expression of the co-stimulatory molecule CD86. Furthermore, in the highest concentration applied (5 μM), it slightly increased the expression of interleukin-10 in microglia in response to lipopolysaccharide. Treatment with low concentrations of teriflunomide (0.25–1 μM) did not have any impact on the activation or proliferation of microglia. At 5 μM concentration of teriflunomide, we observed a reduction of approximately 30 % in proliferation of microglia in mixed glial cell cultures.

Conclusions

Taken together, our in vitro findings suggest that at higher concentrations, teriflunomide potentially exerts its effects by reducing microglial proliferation and not by modulating the M1-/M2-like cell differentiation of primary rat microglia. Thus, teriflunomide has no major impact on the plasticity of microglia; however, the anti-proliferative and minimal anti-inflammatory effects might be clinically relevant for immune modulation in the treatment of neuroinflammatory CNS diseases such as multiple sclerosis.
Begleitmaterial
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s12974-016-0715-3) contains supplementary material, which is available to authorized users.
Abkürzungen
APC
Allophycocyanin
Arg
Arginase
BBB
Blood-brain barrier
BrdU
5-Bromo-2′-deoxyuridine
cDNA
Complementary deoxyribonucleic acid
CFSE
Carboxyfluorescein succinimidyl ester
CNS
Central nervous system
DHODH
Dihydroorotate dehydrogenase
DMEM
Dulbecco’s Modified Eagle Medium
DMSO
Dimethyl sulfoxide
DMT
Disease-modifying therapy
ECL
Enhanced chemiluminescence
EDTA
Ethylenediaminetetraacetic acid
FCS
Fetal calf serum
FITC
Fluorescein isothiocyanate
GM-CSF
Granulocyte macrophage-colony stimulating factor
HPRT
Hypoxanthine-guanine phosphoribosyl-transferase
HRP
Horseradish peroxidase
IFN
Interferon
IGF-1
Insulin-like growth factor-1
IkBα
Inhibitor of nuclear factor-kB
IL
Interleukin
IMDM
Iscove’s Modified Dulbecco’s Medium
iNOS
Inducible nitric oxide synthase
LPS
Lipopolysaccharide
MFI
Mean fluorescence intensity
MGP
Mixed glial cell preparation
MS
Multiple sclerosis
NF-kB
Nuclear factor-kB
NH4Cl
Ammonium chloride
PE
Phycoerythrin
RA
Rheumatoid arthritis
RIPA
Radioimmunoprecipitation assay buffer
RRMS
Relapsing-remitting multiple sclerosis
RT-PCR
Reverse transcription polymerase chain reaction
SD
Standard deviation
TNF
Tumor necrosis factor

Background

Teriflunomide is an immunomodulatory disease-modifying therapy (DMT) for relapsing-remitting multiple sclerosis (RRMS). It is thought to act primarily via a specific, non-competitive, and reversible inhibition of the activity of the mitochondrial enzyme dihydroorotate dehydrogenase (DHODH), which is required for de novo pyrimidine synthesis of rapidly dividing cells such as activated lymphocytes [1, 2]. Resting lymphocytes synthesize the nucleotide pyrimidine through the DHODH-independent salvage pathway and therefore are largely unaffected by teriflunomide [3]. Teriflunomide is the active metabolite of leflunomide in vivo which is an effective agent in the treatment of rheumatoid arthritis (RA) [4].
Several studies indicate that teriflunomide rather exerts cytostatic and not cytotoxic effects on actively proliferating lymphocytes in the periphery [5, 6]. This limits the availability of autoreactive T and B cells that can infiltrate into the central nervous system (CNS) in CNS inflammatory diseases such as multiple sclerosis (MS) [7]. Other effects of teriflunomide such as the inhibition of adhesion molecules, cytokines, protein tyrosine kinases, nuclear factor-kB (NF-kB) activation, and cyclooxygenase 2 activity have also been demonstrated in some in vitro studies, suggesting that teriflunomide in addition to its anti-proliferative effects may also impact signal transduction, migration, and inflammatory processes [8, 9].
Pharmacokinetic analysis of patients treated with teriflunomide revealed a concentration of 20–60 mg/l in the serum. Owing to its low permeability through the blood-brain barrier (BBB), only approximately 1–2 % of the serum concentration of drug reaches the CNS, which is approximate to a concentration of 1–3 μM in vivo [10, 11]. Thus, CNS resident cells like microglia are exposed to much lower concentrations of teriflunomide than the immune cells in the periphery. However, these concentrations are still in the range where, e.g., the DHODH is inhibited and thus therapeutic concentrations of teriflunomide may modulate microglia. Microglia are considered to play key regulatory and effector functions during the onset and progression of CNS diseases [12]. Microglial activation in the CNS during neuroinflammation can have both beneficial and detrimental effects. By virtue of their phagocytic ability, they assist in clearing debris which in turn might hasten the repair processes [13]. Furthermore, several inflammatory factors produced by activated microglia are essential for repair processes [14]. On the other hand, activated microglia serve as antigen-presenting cells and are involved in the reactivation of the CNS-infiltrating autoreactive T lymphocytes and thereby promoting neural tissue damage. Additionally, neuroinflammation triggers the release of toxic reactive oxygen species such as nitric oxide by microglia, which is highly toxic to neurons.
In order to explore the potential of teriflunomide to modulate microglia, we applied physiologically relevant concentration of 0.25–5 μM on primary rat microglia and investigated various functions.

Methods

Mixed glial cell cultures

Primary microglia from neonatal Sprague-Dawley rats (Crl:CD) were isolated by the method of Giulian and Baker [15] as previously described [16]. Briefly, brains were freed from meninges and dissociated mechanically and enzymatically with 0.1 % trypsin (Biochrom, Merck Millipore, Darmstadt, Germany) and 0.25 % DNase (Roche Diagnostic GmbH, Mannheim, Germany). Cells were plated at 1–2 brains per poly-l-lysine (Sigma-Aldrich, Munich, Germany) coated culture flasks (25 cm2; 75 cm2; Sarstedt, Nümbrecht, Germany) and cultivated in Dulbecco’s Modified Eagle Medium (DMEM; life technologies, Carlsbad, USA) supplemented with 10 % fetal calf serum (FCS; Biochrom, Merck Millipore, Darmstadt, Germany) and 1 % penicillin/streptomycin (life technologies, Carlsbad, USA; culture medium referred to as: MGP+), where indicated culture flasks were treated with granulocyte macrophage-colony stimulating factor (GM-CSF; 5 ng/ml; Peprotech; Hamburg, Germany). After 7–8 days, loosely attached microglia were harvested from mixed glial cell cultures by shaking for 30–45 min on an orbital shaker-incubator (Edmund Bühler, Hechingen, Germany) at 37 °C. Cells in the supernatant were re-plated at 3–6 × 104 cells per 12-mm glass coverslip in 24-well plates for immunocytochemistry staining, directly resuspended in 500 μl MGP+ for BrdU flow cytometry, 5 × 105 per well in 6-well plates for reverse transcription polymerase chain reaction (RT-PCR), or 2.5 × 105 in 12-well plates (all Nunc, Life Technologies, Paisley, UK) for phagocytosis and flow cytometry analysis. The remaining astrocytes were washed with PBS and harvested with 0.25 % trypsin/0.05 % EDTA. After isolation, the astrocytes were incubated with an APC-conjugated mouse/rat anti-GLAST (ACSA-1) antibody and with FITC-conjugated anti-BrdU antibody.
Microglia were incubated overnight at 37 °C, 5 % CO2, and the following day, cells were treated with 0, 0.25, 0.5, 1, and 5 μM of teriflunomide (stock: 10 mM, dissolved in dimethyl sulfoxide (DMSO); Genzyme, Waltham, MA; USA) with or without indicated stimuli for the designated period of time for each experiment. Cell culture purity and cell viability were analyzed by immunofluorescence stainings with CD11b/c or Iba-1, markers for microglia/macrophages, and DAPI or PI, markers for cell viability. Results were quantified by flow cytometry or fluorescence microscopy.

BrdU flow cytometry analysis of primary mixed glial cell cultures

The incorporation of 5-bromo-2′-deoxyuridine (BrdU) was analyzed in mixed glial cell cultures using flow cytometry and labeling with a conjugate anti-BrdU antibody (FITC BrdU flow kit, BD Pharmingen, CA, USA). Mixed glial cell cultures were incubated with 0.25–5 μM teriflunomide and co-treated with GM-CSF (5 ng/ml; Peprotech, Hamburg, Germany) from day 5 after preparation. The next day, 10 μM BrdU (FITC BrdU flow kit, BD Pharmingen) was added for 16 h.
After microglia isolation, the Fc receptors were blocked with mouse anti-rat CD32 for 30 min on ice (clone: D34-485; BD Pharmingen). Surface staining of microglia was done with allophycocyanin (APC)-conjugated rat anti-CD11b/c antibody for 30 min at 4 °C. Cells were then fixed and permeabilized with fixation/permeabilization buffer (Foxp3 Staining Buffer Set; eBioscience) according to the manufacturer’s instructions. The samples were treated with DNase to expose incorporated BrdU (diluted to 300 μg/ml; BrdU flow kit, BD Pharmingen) for 1 h at 37 °C and finally stained with fluorescein isothiocyanate (FITC)-conjugated anti-BrdU antibody (30 min at RT, 1:100; BrdU flow kit, BD Pharmingen). The analysis was carried out using a FACSCalibur with CellQuest software (BD). Measurements were performed in duplicates per condition and in four independent experiments.

T cell proliferation assay

Single-cell suspensions from spleens of adult Sprague-Dawley rats (Crl:CD) were prepared in complete Iscove’s Modified Dulbecco’s Medium (IMDM) medium. After passage through a 100-μm-mesh-size cell strainer (BD Falcon), red blood cells were lysed by NH4Cl treatment. CD4+ T cells were purified by MACS technology using MagCellect rat CD4+ T Cell Isolation Kit (R&D Systems, Minneapolis, USA) according to the manufacturer’s instructions. The final depleted cell fraction contained the desired highly enriched CD4+ T cells. Purity of the enriched CD4+ T cells was analyzed with a phycoerythrin (PE)-conjugated anti-rat CD4 antibody (W3/25) and a FITC-conjugated anti-rat T cell receptor α/β antibody (R73). Enriched CD4+ T cells were labeled with 2.5 μM carboxyfluorescein succinimidyl ester (CFSE; Invitrogen, Darmstadt, Germany), respectively, for 10 min at 37 °C. Labeling of cells was stopped by the addition of ice-cold complete IMDM medium and by three washing steps. CFSE-labeled and unlabeled CD4+ Rat T cells (1 × 105 cells/well) were seeded in 96-well microtiter plates (Nunc™ 96-Well Polystyrene Round Bottom Microwell Plates, Thermo Fisher Scientific, USA) in 100 μl medium with the addition of different concentrations of teriflunomide and stimulated with 3 μg/ml of plate-bound anti-CD3 mAb/anti-CD28 mAb. The final culture volume was adjusted to 200 μl per well. Cells were maintained at 37 °C in a humidified atmosphere with 5 % CO2. After 65–72 h incubation, cells were washed in PBS, collected and the absolute amount of proliferating rat T cells was determined by flow cytometry.

RNA isolation and reverse transcription polymerase chain reaction

To determine the messenger RNA (mRNA) levels of pro-inflammatory factors such as iNOS, TNF-α, and IL-1β, anti-inflammatory factors such as Arg1 and IL-10, and growth factor IGF-1, or mRNA level of DHODH, real-time PCR analysis was performed (Table 1).
Table 1
Primer used for polymerase chain reaction
Gene
Gene expression assay number
DHODH
Rn01432611_m1
IL-1β
Rn00580432_m1
IL-10
Rn00563409_m1
iNOS
Rn00561646_m1
Arg1
Rn00691090_m1
TNF-α
Rn99999017_m1
IGF-1
Rn00710306_m1
HPRT
Rn01527840_m1
DHODH dihydroorotate dehydrogenase, IL-1β interleukin-1beta, IL-10 interleukin-10, iNOS inducible nitric oxide synthase, Arg1 arginase1, TNF-α tumor necrosis factor-alpha, IGF-1 insulin-like growth factor-1, HPRT hypoxanthine-guanine phosphoribosyl-transferase
For cytokine analysis, total RNA was isolated from microglia pre-treated with different concentrations of teriflunomide (0.25–5 μM) for 12 h and cultured for an additional 12 h with different supplements: (1) recombinant rat interferon (IFN)-γ (50 ng/ml; Peprotech, Hamburg, Germany) plus lipopolysaccharide (LPS; 100 ng/ml; Sigma-Aldrich, Munich, Germany) and (2) recombinant rat IL-4 (20 ng/ml; Peprotech, Hamburg, Germany) to induce a M1- and M2-like phenotype, respectively.
For DHODH expression analysis, we used two experimental designs. Firstly, we isolated total RNA from microglia pre-treated with different concentrations of teriflunomide (0.25–5 μM) for 12 h and cultured for an additional 12 h with medium containing IFN-γ (50 ng/ml; Peprotech, Hamburg, Germany) plus LPS (100 ng/ml; Sigma-Aldrich, Munich, Germany) or MGP+ medium. Secondly, we isolated total RNA from microglia from mixed glial cell cultures co-treated with different concentrations of teriflunomide (0.25–5 μM) and 5 ng/ml GM-CSF or medium for 12 h.
The RNeasy®Micro Kit (Qiagen, Hilden, Germany) was used according to the manufacturer’s instructions. The RNA concentration was measured using a NanoDrop 2000 spectrophotometer (Thermo Fisher Scientific, MA, USA). Complementary deoxyribonucleic acid (cDNA) was synthesized using the High Capacity cDNA Reverse Transcription Kit (Applied Biosystems, Foster City, CA, USA). qPCR analysis was performed using the StepOne™ Real-Time PCR System and appropriate TaqMan assay (Applied Biosystems; see Table 1). The experiments were performed with cDNA from 400 ng total RNA, and all primers were exon-spanning. The ΔΔCT method was used to determine differences in the expression between controls and drug-treated microglia exposed to M1/M2 or GM-CSF stimuli. Changes in the mRNA expression levels were quantified against the housekeeping gene hypoxanthine-guanine phosphoribosyl-transferase (HPRT) 1 compared with control.

Flow cytometry

LPS (100 ng/ml; Sigma-Aldrich, Munich, Germany) stimulated microglia were cultured in the absence or presence of teriflunomide (0.25–5 μM) for 48 or 72 h. After incubation, cells were washed and harvested by 0.05 % trypsin/EDTA treatment and collected in polystyrene tubes. After centrifugation (352×g, 10 min), cells were resuspended in 100 μl of PBS and subsequently incubated with the fluorescent-labeled antibodies for 30 min on ice: APC-conjugated anti-rat CD11b/c mAb (OX-42, 1:100), PE-conjugated anti-rat CD86 mAb (24 F, 1:100), or appropriate isotype control for anti-rat CD86 (mouse IgG1, k, 1:100) (all from BioLegend, California, USA).
After incubation, cells were washed, pelleted by centrifugation (352×g, 10 min), and resuspended in 250 μl of PBS. Cells were immediately analyzed with FACSCalibur (Becton-Dickinson, San Jose, CA, USA) and CellQuest software (BD Biosciences). Dead cells were excluded by propidium iodide staining.

Western blot analysis

For Western blot analysis, microglia were treated with or without 5 μM teriflunomide for 12 h and then stimulated additionally with LPS/IFN-γ (100 ng/ml; Sigma-Aldrich, Munich, Germany; 50 ng/ml; Peprotech, Hamburg, Germany) for 0, 15, 30, or 60 min at 37 °C. Cells were washed with cold PBS and lysed in RIPA buffer supplemented with 1 % protease and phosphatase inhibitor (Cell Signaling Technology; Roche). The protein content was measured using a Pierce BCA Protein Assay Kit (Thermo Fisher Scientific). The lysate (30 μg of protein) was mixed with Laemmli buffer and boiled for 5 min at 95 °C. After the NF-kB activation reaction, Western blot analysis was performed as described [17] using anti-total IkBα (1:1000, rabbit, Cell Signaling Technology) and β-actin (1:3000, mouse, Santa Cruz Biotechnology) followed by horseradish peroxidase (HRP)-coupled goat anti-mouse or goat anti-rabbit secondary antibodies (Santa Cruz Biotechnology).
Proteins were visualized by enhanced chemiluminescence (ECL, Pierce Biotechnology, Illinois, USA; Millipore, Massachusetts, USA) after treatment with the secondary antibody using the ChemoCam system (Intas, Science Imaging Instruments GmbH, Göttingen, Germany) according to the manufacturer’s instructions. Quantification of protein levels by densitometry was conducted on acquired images using LabImage 1D software (Kapelan Bio-Imaging Solutions, Leipzig, Germany).

Phagocytosis assay

Phagocytic activity of microglia was determined by measuring the uptake of fluorescent latex beads on flow cytometry as previously described [18]. After 24 h teriflunomide (±12 h LPS; 100 ng/ml; Sigma-Aldrich, Munich, Germany) treatment, FITC-labeled latex beads (1 μm, Fluoresbrite™ Yellow Green carboxylate microspheres; Polysciences, Warrington, USA) were added at a cell:bead ratio of 1:100 and incubated for 1 h at 37 °C. In parallel, cells incubated with beads on ice (4 °C) served as negative controls. Non-phagocytosed and surface bound beads were removed by washing six times with ice-cold PBS. Adherent microglia were then harvested by 0.05 % trypsin/EDTA treatment, and the uptake of the beads was determined by flow cytometry (FACSCalibur; Becton-Dickinson, San Jose, CA, USA). Dead cells were excluded by propidium iodide staining.
A shift in mean fluorescence intensity (MFI) resulting from the uptake of fluorescent beads and the percentage of gated microglia that phagocytosed latex beads were used as a measure to assess phagocytosis. Active phagocytosis was then calculated by subtracting measured values of cells incubated at 4 °C from the values obtained at 37 °C.

Statistical analysis

All experiments were performed at least four times, and arithmetic means ± standard deviation (SD) was calculated using GraphPad Prism 5.0 software. Comparison of two samples was performed using a one-sample t test or a paired Student’s t test followed by the correction for multiple comparisons by Benjamini and Hochberg [19]. Multiple samples were evaluated using repeated measures ANOVA with Bonferroni’s multiple comparison t tests. P values <0.05 were considered statistically significant (*p < 0.05, **p < 0.01, ***p < 0.001).

Results

Regulation of DHODH-mRNA expression in microglia

Previous reports have revealed that teriflunomide inhibits activation-induced proliferation of T cells by targeting DHODH, which is a crucial enzyme involved in de novo synthesis of pyrimidine nucleotides [1, 2]. Therefore, we initially assessed if DHODH is induced in microglia upon activation. Following the stimulation of microglia with LPS/IFN-γ or GM-CSF, we studied the expression of DHODH-mRNA by RT-PCR. Compared to medium-treated control, we observed a significant upregulation of DHODH in LPS/IFN-γ (Fig. 1a) and GM-CSF-treated microglia (Fig. 1b). Furthermore, we observed that pre-treatment of microglia with teriflunomide prior to activation per se did not influence the mRNA expression, confirming that teriflunomide inhibition of DHODH might be at the protein level and not at the gene expression level. The slight decrease of DHODH-mRNA of approximately 20 % in GM-CSF-treated microglia by 5 μM teriflunomide was not biologically significant (Fig. 1b).

Teriflunomide does not interfere with morphological shaping of microglia

Morphological rearrangement is a major feature during the transition of different functional stages of microglia. Thus, we investigated if teriflunomide affects the shaping of non-activated and activated microglia. After resting overnight, microglia were cultured for 24 h in the presence of teriflunomide with or without LPS/IFN-γ to induce activation in vitro. Using immunocytochemistry, we observed a morphological change of their phenotype in the presence of LPS/IFN-γ. After plating, microglia had an amoeboid appearance that was maintained after activation (Fig. 2). Teriflunomide treatment did not interfere with morphological changes of microglia when compared with non-activated and activated microglia in control medium. Except for cell culture-associated death, we did not observe any cytotoxic effects of teriflunomide (Additional file 1: Figure S2, Additional file 2: Figure S3).

Teriflunomide reduces proliferation of primary microglia

It is known that teriflunomide inhibits active T cell proliferation; therefore, we tested if the physiologically relevant concentrations (0.25–5 μM) chosen for this study were sufficient to inhibit T cell proliferation. For this purpose, CFSE-labeled CD4+ T cells isolated from the spleen of Sprague-Dawley rats were stimulated on anti-CD3/anti-CD28 coated culture plates in the presence or absence of different concentrations of teriflunomide (0.25–5 μM) and CFSE dilution as an indicator of proliferation was measured by flow cytometry. Even at a concentration as low as 0.25 μM, teriflunomide significantly inhibited proliferation of T cells with a concentration-dependent effect with a maximum at 5 μM (Additional file 3: Figure S1). Having confirmed that low concentrations of teriflunomide were sufficient to mediate anti-proliferative effects in the periphery, we next investigated microglial proliferation.
We stimulated mixed glial cell cultures containing astrocytes, microglia, and oligodendrocyte precursor cells with GM-CSF in the presence or absence of teriflunomide and assessed the proliferation and yield of microglia. While the cultures were treated with GM-CSF (5 ng/ml) and 0.25–5 μM teriflunomide on day 5, the controls received media changes only. The proliferation was determined after day 7 as described in the “Methods” section. We observed an increase in microglial proliferation in mixed glial cell cultures after treatment with 5 ng/ml GM-CSF (Fig. 3). We also detected a slight but significant reduction by 29.7 % ± 2.3 % (mean ± SD; p = 0.0242) in the percentage of BrdU+CD11b/c+ glial cells in GM-CSF-treated cultures after the treatment with 5 μM teriflunomide (Fig. 3). Thus, higher concentrations of teriflunomide may have an anti-proliferative effect on GM-CSF-induced proliferation of microglia. We also investigated if the drug teriflunomide could affect the proliferation of astrocytes and in this way would indirectly influence the microglial proliferation. As presented in Additional file 4: Figure S4, teriflunomide did not alter astrocytic proliferation and the expression of chosen astrocyte-secreted factors.

Teriflunomide has no impact on microglial plasticity

Similar to macrophages, microglia can be functionally classified into either pro-inflammatory (M1-like) or anti-inflammatory (M2-like) cells [20]. We thus investigated the effect of teriflunomide on these functions. Primary rat microglia were driven into either M1- or M2-like cells by culturing them in medium containing either LPS/IFN-γ or IL-4, respectively.
As demonstrated in Fig. 4, pro-inflammatory factors such as iNOS, IL-1β, and TNF-α were highly upregulated in microglia treated with LPS/IFN-γ and anti-inflammatory factors such as Arg1 and IGF-1 were significantly upregulated in IL-4-treated cells. Pre-treatment of microglia with teriflunomide for 12 h prior to M1- and M2-like differentiation had no effect on the mRNA expression of any of the abovementioned pro- or anti-inflammatory genes except for a slight but significant increase in IL-10 expression in LPS/IFN-γ-treated microglia pre-treated with 5 μM teriflunomide. In summary, teriflunomide had no major impact on the plasticity of microglia to differentiate into various phenotypes.

Teriflunomide may downregulate LPS-induced CD86 expression

Antigen-presenting properties of microglia are well appreciated. As an efficient antigen-presenting cell, microglia present antigens and provide the crucial co-stimulatory signals required for the reactivation of infiltrating autoreactive T cells. Upon activation, microglia upregulate co-stimulatory molecules such as CD86 on their surface and we tested if teriflunomide interfered with this process. Following the treatment of microglia with LPS in the presence or absence of teriflunomide for 48 and 72 h, cell surface expression of the co-stimulatory molecule CD86 was determined by flow cytometry. As expected, CD86 expression was significantly increased in LPS-treated microglia. No significant effect of different concentrations of teriflunomide on CD86 expression was observed after 48 h treatment (Fig. 5a, c). However, the expression of CD86 was slightly but significantly decreased on LPS-treated microglia cultured in the presence of 5 μM teriflunomide for 72 h (Fig. 5b, d).

Teriflunomide does not inhibit NF-kB activation

NF-kB is an important transcription factor which regulates the expression of pro-inflammatory genes and is an ideal target for immunomodulatory and anti-inflammatory therapies. Previous studies have shown that 5–10 μM of leflunomide completely blocked NF-kB activation in T cells, myeloid, and epithelial cells [21].
We therefore investigated whether teriflunomide displayed similar inhibitory effects on NF-kB activation in primary microglia. NF-kB is found as a complex with IkBα in the cytoplasm of resting cells. Upon activation IkBα is phosphorylated and subsequently degraded to release NF-kB for nuclear translocation [2224]. Hence, we followed the kinetics of IkBα degradation, as a measure of NF-kB activation, in response to LPS/IFN-γ in microglia pre-treated with 5 μM teriflunomide for 12 h.
As demonstrated in Fig. 6a, IkBα degradation was evident within 15 min of LPS/IFN-γ-treatment. However, IkBα degradation was unaffected by teriflunomide treatment suggesting that teriflunomide had no impact on this regulation in activated microglia at the physiologically relevant concentration used here (Fig. 6b).

Phagocytic activity of microglia is unaffected by teriflunomide

Phagocytosis is one of the major functions of microglia. Microglia treated with or without LPS were incubated with different concentrations (0.25–5 μM) of teriflunomide for 24 h. Subsequently, the uptake of latex beads by microglia was measured by flow cytometry. As shown in Fig. 7, teriflunomide did not change the phagocytic activity of both untreated and LPS-treated microglia.

Discussion

In 2012, teriflunomide was approved for the treatment of RRMS in the USA [25]. Teriflunomide selectively inhibits the proliferation of activated T and B lymphocytes. This effect is achieved already at nanomolar concentrations of teriflunomide by blockage of DHODH, an enzyme involved in de novo pyrimidine synthesis [4]. When used at very high concentrations (>50 μM), teriflunomide regulates also other important immune cell functions [26, 27]. For example, treatment with teriflunomide decreased release of pro-inflammatory cytokines in human macrophages or peripheral blood lymphocytes such as IL-6, IL-17, and TNF-α [2830]. However, pharmacokinetic studies have revealed that only 1–2 % of the peripheral teriflunomide might reach the CNS owing to its low BBB permeability. It is thus conceivable that resident CNS cells such as microglia may be exposed to low teriflunomide concentrations around 0.2–0.6 mg/l in vivo, equivalent to approximately 1–3 μM [10, 11]. In order to evaluate further mechanisms of action of teriflunomide, we investigated its effects on microglia in a range of concentrations between 0.25 and 5 μM that may be reached in the CNS by oral administration.
There is only limited knowledge on how teriflunomide influences the phenotype and function of microglia, the resident immune cell in the CNS. Microglia characteristically undergo rapid proliferation in response to an inflammatory insult in the CNS [31]. From our own experiences and from published reports, we know that in mixed glial cell cultures, microglia rapidly proliferate from days 4–5 and that this process is further enhanced by the addition of growth factors such as GM-CSF [32, 33]. Interestingly, we observed that microglia upregulated DHODH gene expression upon activation. Consequently, we expected similar inhibitory effects of teriflunomide on microglial proliferation. We therefore treated mixed glial cell cultures directly with teriflunomide. Teriflunomide treatment significantly lowered the yield of microglia from GM-CSF-treated mixed glial cell cultures. A reduced frequency of BrdU+ microglia indicated decreased proliferation (Fig. 3). However, the exact mechanism how this inhibition is mediated remains to be clarified. We could exclude cytotoxicity influencing our results as we did not detect morphological changes as well as microglial cell death in cells exposed to teriflunomide (Fig. 2; Additional file 1: Figure S2, Additional file 2: Figure S3). Since there was an increased expression of DHODH-mRNA in microglia in response to GM-CSF-treatment (Fig. 1b), we can speculate that the anti-proliferative effect of teriflunomide on activated microglia might be mediated through DHODH inhibition. However, we do not have direct evidence to support this, a DHODH-independent mechanism may also act, as we observed proliferation of microglia in mixed glial cell cultures devoid of GM-CSF is similarly affected by teriflunomide treatment (data not shown), and the concentration required to induce this inhibition is much higher than the concentration that is sufficient to inhibit T cell proliferation (Additional file 3: Figure S1).
Apart from anti-proliferative effects, reports suggest that teriflunomide has significant influence on the inflammatory response of various immune cells. For example, Cutolo et al. showed that leflunomide, the prodrug of teriflunomide, has a concentration-dependent anti-inflammatory effect on cultured synovial macrophages from patients with rheumatoid arthritis [34]. Similar to macrophages, microglia can attain a pro-inflammatory (M1-like) or an anti-inflammatory (M2-1ike) phenotype [35]. This led us to ask if teriflunomide has similar effects on the M1- and M2-like phenotype of microglia in vitro. Interestingly, at 5 μM of teriflunomide, we found a slight but significant enhancement in the mRNA expression of the anti-inflammatory cytokine IL-10 in microglia exposed to LPS/IFN-γ. This is in accordance with the findings of Korn et al. who observed a similar increase in IL-10 expression in LPS-treated microglia that were exposed to teriflunomide [36]. On the other hand, we observed a decreased expression of the co-stimulatory molecule CD86 on LPS-treated microglia that were exposed to 5 μM of teriflunomide. However, no significant effects were detectable on the mRNA expression of pro-inflammatory mediators in M1-microglia. Similarly, the NF-kB pathway, that regulates the expression of pro-inflammatory cytokines, was unaffected in teriflunomide-treated microglia. This stands in contrast to studies in a human T cell line. In this cell line, 5–10 μM of leflunomide were able to block TNF-α-induced NF-kB activation [21, 37]. This may be due to the lower concentrations of teriflunomide used in our studies. Furthermore, microglia seem to be less responsive to teriflunomide as compared to lymphocytes. Taken together, we believe that low concentrations of teriflunomide have no major effects on pro-inflammatory properties of microglia, but higher concentration might possibly enhance their anti-inflammatory properties.
In MS, infiltrating T cells can mediate CNS damage either by directly targeting neurons or by triggering an inflammatory response in microglia. Furthermore microglia respond to CNS damage and remove the damaged cells by phagocytosis. Phagocytosis is one of the main features of microglial activation. In the past, the importance of microglia in the clearance of myelin debris and subsequent repair processes has been demonstrated [38, 39]. However, we did not observe any enhancing or suppressive effects of teriflunomide on phagocytosis in either untreated or LPS-activated microglia.

Conclusions

In conclusion, we demonstrate here that by and large, only the highest concentration of teriflunomide that microglia may be exposed to in vivo in the CNS has an effect on proliferation. Furthermore, only stimulation with the highest concentration of teriflunomide (5 μM) had a significant effect on mRNA expression of IL-10. Lower concentrations were not sufficient to modulate significantly further anti- or pro-inflammatory responses. Thus, we provide evidence that teriflunomide may have an effect on microglial proliferation which may be of importance in the treatment of neuroinflammatory CNS diseases. Further in vivo studies are required to corroborate this effect.

Acknowledgements

The authors thank I. Cierpka-Leja, S. Lang, and A. Niesel for their excellent technical assistance. The authors thank Dr. Christoph Lindemann (University Medical Center Hamburg-Eppendorf) for his assistance with the statistical analysis.

Funding

This study was partly funded by Genzyme Corporation, a Sanofi company, and the authors thank Genzyme for the financial support.

Availability of data and materials

No project-specific resources (cell lines, plasmids, rat strains) were used in this study. All reagents used are commercially available. Raw data for these studies will be made to interested parties available upon request.

Authors’ contributions

TW performed all the experiments and acquisition of data and analysis and interpretation of data. TW and CKP contributed to the scientific design. CKP assisted with the analysis and interpretation of data. VG helped to design the RT-PCR experiments. JK performed the additional experiments for revision. SW and MB helped to prepare the rat mixed glial cell cultures. TW drafted the manuscript. MS were responsible for the conception and scientific design and contributed to the interpretation of data. All authors read and approved the final manuscript.

Competing interests

This research work was supported partly by Genzyme Corporation. The sponsor was not involved in the data collection or analysis, drafting the manuscript, or the decision to publish. Results of this manuscript are part of a doctoral thesis at the University of Veterinary Medicine Hannover (Tanja Wostradowski, December 2015).
Not applicable.
All animal tissue experiments were performed following approval from the Review Board for the Care of Animal Subjects of the district government (Lower Saxony, Germany).
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.
Anhänge

Additional files

Literatur
1.
Zurück zum Zitat Cherwinski HM, Cohn RG, Cheung P, Webster DJ, Xu YZ, Caulfield JP, Young JM, Nakano G, Ransom JT. The immunosuppressant leflunomide inhibits lymphocyte proliferation by inhibiting pyrimidine biosynthesis. J Pharmacol Exp Ther. 1995;275:1043–9.PubMed Cherwinski HM, Cohn RG, Cheung P, Webster DJ, Xu YZ, Caulfield JP, Young JM, Nakano G, Ransom JT. The immunosuppressant leflunomide inhibits lymphocyte proliferation by inhibiting pyrimidine biosynthesis. J Pharmacol Exp Ther. 1995;275:1043–9.PubMed
2.
Zurück zum Zitat Bruneau JM, Yea CM, Spinella-Jaegle S, Fudali C, Woodward K, Robson PA, Sautes C, Westwood R, Kuo EA, Williamson RA, Ruuth E. Purification of human dihydro-orotate dehydrogenase and its inhibition by A77 1726, the active metabolite of leflunomide. Biochem J. 1998;336(Pt 2):299–303.CrossRefPubMedPubMedCentral Bruneau JM, Yea CM, Spinella-Jaegle S, Fudali C, Woodward K, Robson PA, Sautes C, Westwood R, Kuo EA, Williamson RA, Ruuth E. Purification of human dihydro-orotate dehydrogenase and its inhibition by A77 1726, the active metabolite of leflunomide. Biochem J. 1998;336(Pt 2):299–303.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Bar-Or A. Teriflunomide (Aubagio(R)) for the treatment of multiple sclerosis. Exp Neurol. 2014;262 Pt A:57–65.CrossRefPubMed Bar-Or A. Teriflunomide (Aubagio(R)) for the treatment of multiple sclerosis. Exp Neurol. 2014;262 Pt A:57–65.CrossRefPubMed
4.
Zurück zum Zitat Fox RI, Herrmann ML, Frangou CG, Wahl GM, Morris RE, Strand V, Kirschbaum BJ. Mechanism of action for leflunomide in rheumatoid arthritis. Clin Immunol. 1999;93:198–208.CrossRefPubMed Fox RI, Herrmann ML, Frangou CG, Wahl GM, Morris RE, Strand V, Kirschbaum BJ. Mechanism of action for leflunomide in rheumatoid arthritis. Clin Immunol. 1999;93:198–208.CrossRefPubMed
5.
Zurück zum Zitat Kraan MC, Reece RJ, Barg EC, Smeets TJ, Farnell J, Rosenburg R, Veale DJ, Breedveld FC, Emery P, Tak PP. Modulation of inflammation and metalloproteinase expression in synovial tissue by leflunomide and methotrexate in patients with active rheumatoid arthritis. Findings in a prospective, randomized, double-blind, parallel-design clinical trial in thirty-nine patients at two centers. Arthritis Rheum. 2000;43:1820–30.CrossRefPubMed Kraan MC, Reece RJ, Barg EC, Smeets TJ, Farnell J, Rosenburg R, Veale DJ, Breedveld FC, Emery P, Tak PP. Modulation of inflammation and metalloproteinase expression in synovial tissue by leflunomide and methotrexate in patients with active rheumatoid arthritis. Findings in a prospective, randomized, double-blind, parallel-design clinical trial in thirty-nine patients at two centers. Arthritis Rheum. 2000;43:1820–30.CrossRefPubMed
6.
Zurück zum Zitat Ringheim GE, Lee L, Laws-Ricker L, Delohery T, Liu L, Zhang D, Colletti N, Soos TJ, Schroeder K, Fanelli B, et al. Teriflunomide attenuates immunopathological changes in the dark agouti rat model of experimental autoimmune encephalomyelitis. Front Neurol. 2013;4:169.CrossRefPubMedPubMedCentral Ringheim GE, Lee L, Laws-Ricker L, Delohery T, Liu L, Zhang D, Colletti N, Soos TJ, Schroeder K, Fanelli B, et al. Teriflunomide attenuates immunopathological changes in the dark agouti rat model of experimental autoimmune encephalomyelitis. Front Neurol. 2013;4:169.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Gold R, Wolinsky JS. Pathophysiology of multiple sclerosis and the place of teriflunomide. Acta Neurol Scand. 2011;124:75–84.CrossRefPubMed Gold R, Wolinsky JS. Pathophysiology of multiple sclerosis and the place of teriflunomide. Acta Neurol Scand. 2011;124:75–84.CrossRefPubMed
8.
Zurück zum Zitat Hamilton LC, Vojnovic I, Warner TD. A771726, the active metabolite of leflunomide, directly inhibits the activity of cyclo-oxygenase-2 in vitro and in vivo in a substrate-sensitive manner. Br J Pharmacol. 1999;127:1589–96.CrossRefPubMedPubMedCentral Hamilton LC, Vojnovic I, Warner TD. A771726, the active metabolite of leflunomide, directly inhibits the activity of cyclo-oxygenase-2 in vitro and in vivo in a substrate-sensitive manner. Br J Pharmacol. 1999;127:1589–96.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Papadopoulou A, Kappos L, Sprenger T. Teriflunomide for oral therapy in multiple sclerosis. Expert Rev Clin Pharmacol. 2012;5:617–28.CrossRefPubMed Papadopoulou A, Kappos L, Sprenger T. Teriflunomide for oral therapy in multiple sclerosis. Expert Rev Clin Pharmacol. 2012;5:617–28.CrossRefPubMed
10.
Zurück zum Zitat Tallantyre E, Evangelou N, Constantinescu CS. Spotlight on teriflunomide. Int MS J. 2008;15:62–8.PubMed Tallantyre E, Evangelou N, Constantinescu CS. Spotlight on teriflunomide. Int MS J. 2008;15:62–8.PubMed
11.
Zurück zum Zitat Limsakun T, Menguy-Vacheron F. Pharmacokinetics of oral teriflunomide, a novel oral disease-modifying agent under investigation for the treatment of multiple sclerosis. Neurology. 2010;74:P05.032. Limsakun T, Menguy-Vacheron F. Pharmacokinetics of oral teriflunomide, a novel oral disease-modifying agent under investigation for the treatment of multiple sclerosis. Neurology. 2010;74:P05.032.
12.
Zurück zum Zitat Strachan-Whaley M, Rivest S, Yong VW. Interactions between microglia and T cells in multiple sclerosis pathobiology. J Interferon Cytokine Res. 2014;34:615–22.CrossRefPubMed Strachan-Whaley M, Rivest S, Yong VW. Interactions between microglia and T cells in multiple sclerosis pathobiology. J Interferon Cytokine Res. 2014;34:615–22.CrossRefPubMed
13.
Zurück zum Zitat Neumann H, Kotter MR, Franklin RJ. Debris clearance by microglia: an essential link between degeneration and regeneration. Brain. 2009;132:288–95.CrossRefPubMed Neumann H, Kotter MR, Franklin RJ. Debris clearance by microglia: an essential link between degeneration and regeneration. Brain. 2009;132:288–95.CrossRefPubMed
14.
Zurück zum Zitat Franklin RJ, Kotter MR. The biology of CNS remyelination: the key to therapeutic advances. J Neurol. 2008;255 Suppl 1:19–25.CrossRefPubMed Franklin RJ, Kotter MR. The biology of CNS remyelination: the key to therapeutic advances. J Neurol. 2008;255 Suppl 1:19–25.CrossRefPubMed
15.
Zurück zum Zitat Giulian D, Baker TJ. Characterization of ameboid microglia isolated from developing mammalian brain. J Neurosci. 1986;6:2163–78.PubMed Giulian D, Baker TJ. Characterization of ameboid microglia isolated from developing mammalian brain. J Neurosci. 1986;6:2163–78.PubMed
16.
Zurück zum Zitat Singh V, Voss EV, Benardais K, Stangel M. Effects of 2-chlorodeoxyadenosine (Cladribine) on primary rat microglia. J Neuroimmune Pharmacol. 2012;7:939–50.CrossRefPubMed Singh V, Voss EV, Benardais K, Stangel M. Effects of 2-chlorodeoxyadenosine (Cladribine) on primary rat microglia. J Neuroimmune Pharmacol. 2012;7:939–50.CrossRefPubMed
17.
Zurück zum Zitat Wostradowski T, Gudi V, Pul R, Gingele S, Lindquist JA, Stangel M, Lindquist S. Effect of interferon-beta1b on CXCR4-dependent chemotaxis in T cells from multiple sclerosis patients. Clin Exp Immunol. 2015;182:162–72.CrossRefPubMed Wostradowski T, Gudi V, Pul R, Gingele S, Lindquist JA, Stangel M, Lindquist S. Effect of interferon-beta1b on CXCR4-dependent chemotaxis in T cells from multiple sclerosis patients. Clin Exp Immunol. 2015;182:162–72.CrossRefPubMed
18.
Zurück zum Zitat Pul R, Chittappen KP, Stangel M. Quantification of microglial phagocytosis by a flow cytometer-based assay. Methods Mol Biol. 2013;1041:121–7.CrossRefPubMed Pul R, Chittappen KP, Stangel M. Quantification of microglial phagocytosis by a flow cytometer-based assay. Methods Mol Biol. 2013;1041:121–7.CrossRefPubMed
19.
Zurück zum Zitat Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Series B. 1995;57(1):289–300. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Series B. 1995;57(1):289–300.
20.
21.
Zurück zum Zitat Manna SK, Aggarwal BB. Immunosuppressive leflunomide metabolite (A77 1726) blocks TNF-dependent nuclear factor-kappa B activation and gene expression. J Immunol. 1999;162:2095–102.PubMed Manna SK, Aggarwal BB. Immunosuppressive leflunomide metabolite (A77 1726) blocks TNF-dependent nuclear factor-kappa B activation and gene expression. J Immunol. 1999;162:2095–102.PubMed
22.
Zurück zum Zitat Baldwin Jr AS. The NF-kappa B and I kappa B proteins: new discoveries and insights. Annu Rev Immunol. 1996;14:649–83.CrossRefPubMed Baldwin Jr AS. The NF-kappa B and I kappa B proteins: new discoveries and insights. Annu Rev Immunol. 1996;14:649–83.CrossRefPubMed
23.
24.
Zurück zum Zitat Verma IM, Stevenson JK, Schwarz EM, Van Antwerp D, Miyamoto S. Rel/NF-kappa B/I kappa B family: intimate tales of association and dissociation. Genes Dev. 1995;9:2723–35.CrossRefPubMed Verma IM, Stevenson JK, Schwarz EM, Van Antwerp D, Miyamoto S. Rel/NF-kappa B/I kappa B family: intimate tales of association and dissociation. Genes Dev. 1995;9:2723–35.CrossRefPubMed
25.
Zurück zum Zitat Genzyme C. Aubagio Prescribing Information. Cambridge, MA; 2012 Genzyme C. Aubagio Prescribing Information. Cambridge, MA; 2012
26.
Zurück zum Zitat Xu X, Williams JW, Bremer EG, Finnegan A, Chong AS. Inhibition of protein tyrosine phosphorylation in T cells by a novel immunosuppressive agent, leflunomide. J Biol Chem. 1995;270:12398–403.CrossRefPubMed Xu X, Williams JW, Bremer EG, Finnegan A, Chong AS. Inhibition of protein tyrosine phosphorylation in T cells by a novel immunosuppressive agent, leflunomide. J Biol Chem. 1995;270:12398–403.CrossRefPubMed
27.
Zurück zum Zitat Xu X, Williams JW, Gong H, Finnegan A, Chong AS. Two activities of the immunosuppressive metabolite of leflunomide, A77 1726. Inhibition of pyrimidine nucleotide synthesis and protein tyrosine phosphorylation. Biochem Pharmacol. 1996;52:527–34.CrossRefPubMed Xu X, Williams JW, Gong H, Finnegan A, Chong AS. Two activities of the immunosuppressive metabolite of leflunomide, A77 1726. Inhibition of pyrimidine nucleotide synthesis and protein tyrosine phosphorylation. Biochem Pharmacol. 1996;52:527–34.CrossRefPubMed
28.
Zurück zum Zitat González-Alvaro I, Ortiz AM, Dominguez-Jimenez C, Aragon-Bodi A, Diaz Sanchez B, Sanchez-Madrid F. Inhibition of tumour necrosis factor and IL-17 production by leflunomide involves the JAK/STAT pathway. Ann Rheum Dis. 2009;68:1644–50.CrossRefPubMed González-Alvaro I, Ortiz AM, Dominguez-Jimenez C, Aragon-Bodi A, Diaz Sanchez B, Sanchez-Madrid F. Inhibition of tumour necrosis factor and IL-17 production by leflunomide involves the JAK/STAT pathway. Ann Rheum Dis. 2009;68:1644–50.CrossRefPubMed
29.
Zurück zum Zitat Li L, Liu J, Delohery T, Zhang D, Arendt C, Jones C. The effects of teriflunomide on lymphocyte subpopulations in human peripheral blood mononuclear cells in vitro. J Neuroimmunol. 2013;265:82–90.CrossRefPubMed Li L, Liu J, Delohery T, Zhang D, Arendt C, Jones C. The effects of teriflunomide on lymphocyte subpopulations in human peripheral blood mononuclear cells in vitro. J Neuroimmunol. 2013;265:82–90.CrossRefPubMed
30.
Zurück zum Zitat Montagna P, Soldano S, Brizzolara R, Villaggio B, Triolo P, Clerico P, Meroni M, Sulli A, Seriolo B, Molfetta L, Cutolo M. Estrogens interfere with leflunomide modulation of cytokine production by human activated monocytes. Ann N Y Acad Sci. 2010;1193:30–5.CrossRefPubMed Montagna P, Soldano S, Brizzolara R, Villaggio B, Triolo P, Clerico P, Meroni M, Sulli A, Seriolo B, Molfetta L, Cutolo M. Estrogens interfere with leflunomide modulation of cytokine production by human activated monocytes. Ann N Y Acad Sci. 2010;1193:30–5.CrossRefPubMed
31.
Zurück zum Zitat Skripuletz T, Salinas Tejedor L, Prajeeth CK, Hansmann F, Chhatbar C, Kucman V, Zhang N, Raddatz BB, Detje CN, Suhs KW, et al. The antiviral drug ganciclovir does not inhibit microglial proliferation and activation. Sci Rep. 2015;5:14935.CrossRefPubMedPubMedCentral Skripuletz T, Salinas Tejedor L, Prajeeth CK, Hansmann F, Chhatbar C, Kucman V, Zhang N, Raddatz BB, Detje CN, Suhs KW, et al. The antiviral drug ganciclovir does not inhibit microglial proliferation and activation. Sci Rep. 2015;5:14935.CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Esen N, Kielian T. Effects of low dose GM-CSF on microglial inflammatory profiles to diverse pathogen-associated molecular patterns (PAMPs). J Neuroinflammation. 2007;4:10.CrossRefPubMedPubMedCentral Esen N, Kielian T. Effects of low dose GM-CSF on microglial inflammatory profiles to diverse pathogen-associated molecular patterns (PAMPs). J Neuroinflammation. 2007;4:10.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Koshida R, Oishi H, Hamada M, Takahashi S. MafB antagonizes phenotypic alteration induced by GM-CSF in microglia. Biochem Biophys Res Commun. 2015;463:109–15.CrossRefPubMed Koshida R, Oishi H, Hamada M, Takahashi S. MafB antagonizes phenotypic alteration induced by GM-CSF in microglia. Biochem Biophys Res Commun. 2015;463:109–15.CrossRefPubMed
34.
Zurück zum Zitat Cutolo M, Sulli A, Ghiorzo P, Pizzorni C, Craviotto C, Villaggio B. Anti-inflammatory effects of leflunomide on cultured synovial macrophages from patients with rheumatoid arthritis. Ann Rheum Dis. 2003;62:297–302.CrossRefPubMedPubMedCentral Cutolo M, Sulli A, Ghiorzo P, Pizzorni C, Craviotto C, Villaggio B. Anti-inflammatory effects of leflunomide on cultured synovial macrophages from patients with rheumatoid arthritis. Ann Rheum Dis. 2003;62:297–302.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Prajeeth CK, Lohr K, Floess S, Zimmermann J, Ulrich R, Gudi V, Beineke A, Baumgartner W, Muller M, Huehn J, Stangel M. Effector molecules released by Th1 but not Th17 cells drive an M1 response in microglia. Brain Behav Immun. 2014;37:248–59.CrossRefPubMed Prajeeth CK, Lohr K, Floess S, Zimmermann J, Ulrich R, Gudi V, Beineke A, Baumgartner W, Muller M, Huehn J, Stangel M. Effector molecules released by Th1 but not Th17 cells drive an M1 response in microglia. Brain Behav Immun. 2014;37:248–59.CrossRefPubMed
36.
Zurück zum Zitat Korn T, Magnus T, Toyka K, Jung S. Modulation of effector cell functions in experimental autoimmune encephalomyelitis by leflunomide—mechanisms independent of pyrimidine depletion. J Leukoc Biol. 2004;76:950–60.CrossRefPubMed Korn T, Magnus T, Toyka K, Jung S. Modulation of effector cell functions in experimental autoimmune encephalomyelitis by leflunomide—mechanisms independent of pyrimidine depletion. J Leukoc Biol. 2004;76:950–60.CrossRefPubMed
37.
Zurück zum Zitat Manna SK, Mukhopadhyay A, Aggarwal BB. Leflunomide suppresses TNF-induced cellular responses: effects on NF-kappa B, activator protein-1, c-Jun N-terminal protein kinase, and apoptosis. J Immunol. 2000;165:5962–9.CrossRefPubMed Manna SK, Mukhopadhyay A, Aggarwal BB. Leflunomide suppresses TNF-induced cellular responses: effects on NF-kappa B, activator protein-1, c-Jun N-terminal protein kinase, and apoptosis. J Immunol. 2000;165:5962–9.CrossRefPubMed
38.
Zurück zum Zitat Kotter MR, Setzu A, Sim FJ, Van Rooijen N, Franklin RJ. Macrophage depletion impairs oligodendrocyte remyelination following lysolecithin-induced demyelination. Glia. 2001;35:204–12.CrossRefPubMed Kotter MR, Setzu A, Sim FJ, Van Rooijen N, Franklin RJ. Macrophage depletion impairs oligodendrocyte remyelination following lysolecithin-induced demyelination. Glia. 2001;35:204–12.CrossRefPubMed
39.
Zurück zum Zitat Skripuletz T, Hackstette D, Bauer K, Gudi V, Pul R, Voss E, Berger K, Kipp M, Baumgartner W, Stangel M. Astrocytes regulate myelin clearance through recruitment of microglia during cuprizone-induced demyelination. Brain. 2013;136:147–67.CrossRefPubMed Skripuletz T, Hackstette D, Bauer K, Gudi V, Pul R, Voss E, Berger K, Kipp M, Baumgartner W, Stangel M. Astrocytes regulate myelin clearance through recruitment of microglia during cuprizone-induced demyelination. Brain. 2013;136:147–67.CrossRefPubMed
Metadaten
Titel
In vitro evaluation of physiologically relevant concentrations of teriflunomide on activation and proliferation of primary rodent microglia
verfasst von
Tanja Wostradowski
Chittappen Kandiyil Prajeeth
Viktoria Gudi
Jessica Kronenberg
Sina Witte
Marina Brieskorn
Martin Stangel
Publikationsdatum
01.12.2016
Verlag
BioMed Central
Erschienen in
Journal of Neuroinflammation / Ausgabe 1/2016
Elektronische ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-016-0715-3

Weitere Artikel der Ausgabe 1/2016

Journal of Neuroinflammation 1/2016 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.