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

Open Access 01.12.2018 | Research

α1-antitrypsin mitigates NLRP3-inflammasome activation in amyloid β1–42-stimulated murine astrocytes

verfasst von: Taraneh Ebrahimi, Marcus Rust, Sarah Nele Kaiser, Alexander Slowik, Cordian Beyer, Andreas Rembert Koczulla, Jörg B. Schulz, Pardes Habib, Jan Philipp Bach

Erschienen in: Journal of Neuroinflammation | Ausgabe 1/2018

Abstract

Background

Neuroinflammation has an essential impact on the pathogenesis and progression of Alzheimer’s disease (AD). Mostly mediated by microglia and astrocytes, inflammatory processes lead to degeneration of neuronal cells. The NLRP3-inflammasome (NOD-like receptor family, pyrin domain containing 3) is a key component of the innate immune system and its activation results in secretion of the proinflammatory effectors interleukin-1β (IL-1β) and interleukin-18 (IL-18). Under physiological conditions, cytosolic NLRP3-inflammsome is maintained in an inactive form, not able to oligomerize. Amyloid β1–42 (Aβ1–42) triggers activation of NLRP3-inflammasome in microglia and astrocytes, inducing oligomerization and thus recruitment of proinflammatory proteases. NLRP3-inflammasome was found highly expressed in human brains diagnosed with AD. Moreover, NLRP3-deficient mice carrying mutations associated with familial AD were partially protected from deficits associated with AD.
The endogenous protease inhibitor α1-antitrypsin (A1AT) is known for its anti-inflammatory and anti-apoptotic properties and thus could serve as therapeutic agent for NLRP3-inhibition. A1AT protects neurons from glutamate-induced toxicity and reduces Aβ1–42-induced inflammation in microglial cells. In this study, we investigated the effect of Aβ1–42-induced NLRP3-inflammasome upregulation in primary murine astrocytes and its regulation by A1AT.

Methods

Primary cortical astrocytes from BALB/c mice were stimulated with Aβ1–42 and treated with A1AT. Regulation of NLRP3-inflammasome was examined by immunocytochemistry, PCR, western blot and ELISA. Our studies included an inhibitor of NLRP3 to elucidate direct interactions between A1AT and NLRP3-inflammasome components.

Results

Our study revealed that A1AT reduces Aβ1–42-dependent upregulation of NLRP3 at the mRNA and protein levels. Furthermore, A1AT time-dependently mitigated the expression of caspase 1 and its cleavage product IL-1β in Aβ1–42-stimulated astrocytes.

Conclusion

We conclude that Aβ1–42-stimulation results in an upregulation of NLRP3, caspase 1, and its cleavage products in astrocytes. A1AT time-dependently hampers neuroinflammation by downregulation of Aβ1–42-mediated NLRP3-inflammasome expression and thus may serve as a pharmaceutical opportunity for the treatment of Alzheimer’s disease.
Begleitmaterial
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12974-018-1319-x) contains supplementary material, which is available to authorized users.
P Habib and J P Bach contributed equally to this work.
Abkürzungen
A1AT
α1-antitrypsin
AD
Alzheimer’s disease
ASC
Apoptosis-associated speck-like protein containing a CARD
Amyloid β
CARD
Caspase activation and recruitment domain
Casp1
Caspase 1
CNS
Central nervous system
Ctr
Control
DMSO
Dimethylsulfoxid
GAPDH
Glycerinaldehyd-3-phosphate-dehydrogenase
GFAP
Glial fibrillary acidic protein
HPRT
Hypoxanthine phosphoribosyltransferase
Hsp90
Heat shock protein 90
Iba1
Ionized calcium binding adaptor molecule 1
ICC
Immunocytochemistry
IL-18
Interleukin-18
IL-1β
Interleukin-1β
LDH
Lactate dehydrogenase
LPS
Lipopolysaccharide
LRR
Leucine-rich repeat
NACHT
NAIP (NLP family apoptosis inhibitor protein), CIITA (class 2 transcription activator), HET-E (heterokaryon incompatibility), TEP1 (telomerase-associated protein 1)
NLRP
NACHT, LRR, and PYD domains-containing
PCR
Polymerase chain reaction
Pos
Positive control
PYD
Pyrin domain
q-RT-PCR
Quantitative real-time PCR
RFU
Relative fluorescence units
sq-PCR
Semi-quantitative PCR

Background

Alzheimer’s disease (AD) is the most common form of dementia with more than 40 million patients affected worldwide [1]. By 2050, the number is expected to quadruple [2]. Age is the most important risk factor [3], because the incidence of AD doubles every 5 years after the age of 65 years [3]. There is no causal treatment so far. To date, almost all biologicals or secretase inhibitors have failed in clinical trials which emphasize the need for further research into novel therapeutic options. Treatment of patients, even with early symptoms, only starts when the disease pathology has progressed and neural tissue has irreversibly been damaged for years. Therefore, current trials focus on patients with prodromal disease signs [411].
Following the amyloid hypothesis, accumulation of extracellular Aβ1–42-oligomers is one of the earliest and driving factors for pathogenesis of AD [12, 13]. The majority of in vitro studies investigated the effect of Aβ1–42 after an incubation time of 24–72 h [1416]. Current scientific literature reveals less data about a possible damaging effect of Aβ1–42-stimulation on the central nervous system (CNS) after short-term stimulation of only a few hours [14].
Besides Aβ1–42, AD is mainly characterized by hyperphosphorylation of tau and neuroinflammation mediated by microglia and astrocytes, causing neuronal cell death [1721]. A key component of the innate immune system is the NOD-like receptor family, pyrin domain-containing 3 (NLRP 3) [22, 23]. Though ubiquitously expressed in CNS, NLRP3 is found highly expressed in Alzheimer’s patients’ brains [2224]. Under physiological conditions, an inactive form of NLRP3 is located in the cytoplasm [14, 25]. However, in the absence of activating signals, the NLRP3-inflammasome is not able to oligomerize [25]. After NLRP3-receptors recognize danger signals released by damaged cells and pathogens [26], NLRP3, the adaptor protein ASC (apoptosis-associated speck-like protein containing a CARD) and pro-caspase 1 form a subcellular multiprotein complex, known as NLRP3-inflammasome [22, 23, 27]. Subsequently, pro-caspase 1 is activated by autoproteolysis and catalyzes the cleavage of the precursors pro-IL-1β and pro-IL-18 [27]. Mostly induced by microglia and astrocytes, secretion of pro-inflammatory cytokines IL-1β and IL-18 drives inflammatory responses and causes neuronal damaging [2729].
Inflammasomes are linked to neurodegenerative diseases: activated NLRP3 was observed in Parkinson’s disease in the midbrain and cerebrospinal fluid [3033]. Furthermore, in an experimental ischemic stroke model, NLRP3-deficiency was protective against ischemic neuronal damage [34].
In a cellular model of Alzheimer’s disease, Halle et al. 2008 first described that activation of NLRP3-inflammasome is induced by Aβ1–42 in microglia, leading to an overexpression of the pro-inflammatory cytokine IL-1β [14]. Moreover, Aβ1–42 activates the NLRP3-inflammasome in astrocytes [35]. Alike microglia, as a part of the CNS immune response, reactive astrocytes surround amyloid deposits and perform phagocytosis [3537]. Most studies investigated Aβ1–42-mediated inflammatory processes in microglia and little is known about inflammasome activation in Aβ1–42-stimulated astrocytes [14, 15, 24, 3840]. Since inflammation occurs as one of the first cellular and molecular responses after cell stress, short-term effects of Aβ-stimulation in astrocytes need further characterization. Aside from cell culture models, also in human models of AD high expression of NLRP3-inflammasome was found. More precise, an upregulation of NLRP3 expression in peripheral monocytes from individuals with AD was identified [16]. Moreover, in frontal cortex and hippocampus lysates from AD patients increased amounts of cleaved caspase 1 were detected [24]. Interestingly, NLRP3 deficient mice carrying genes associated with familial AD were protected from spatial memory deficits [24].
Therefore, a potent inhibition of the NLRP3-inflammasome could be a new therapeutic approach. The protease inhibitor α1-antitrypsin (A1AT) is known for its anti-inflammatory and anti-apoptotic properties in both hepatic and lung cells [4144]. Conveniently, A1AT is therapeutically used in patients with A1AT-deficiency and therefore well-established as a pharmaceutical agent. Recently, we demonstrated that A1AT also protected neurons from glutamate-induced toxicity [45] and reduced Aβ1–42-induced inflammation in microglial cells [15]. In addition, we found that A1AT inhibited calpain and stabilized calcium-homeostasis [15]. This study investigated the regulation of NLRP3-inflammasome by A1AT in Aβ1–42-stimulated murine astrocytes. In order to elucidate a direct interaction between A1AT and the NLRP3-inflammasome, we have included an inhibitor of NLRP3. MCC950 is a highly potent and specific inhibitor of NLRP3, without affecting AIM2, NLRC4, or NLRP4 [4649]. Recent data revealed that MCC950 stimulated Aβ-phagocytosis in vitro, and reduced Aβ-accumulation in a mouse model of AD, which was associated with improved cognitive function [50].

Methods

Primary cortical murine astrocyte culture

Postnatal (P0 to P2) cortical astrocyte culture preparation from BALB/c mice (Charles River) was performed as previously described by Habib et al. 2014 [51]. Preparation was conducted in accordance with animal welfare policy of University Hospital Aachen and the government of the State of North Rhine-Westphalia, Germany (no. 84.02.04.2015.A292). Briefly, after brain dissection meninges and blood vessels were removed, cortex was isolated, homogenized, and dissolved in Dulbecco’s phosphate-buffered saline (DPBS, Life Technologies, USA) containing 1% (v/v) trypsin and 0.02% (v/v) EDTA. The cell suspension was filtered through a 50 μm nylon mesh. After centrifugation (1400 rpm, 5 min), pellets were re-suspended in Gibco™ Dulbeccos’s modified Eagle medium (DMEM, Life Technologies, USA) and seeded on flasks in DMEM with additional 10% fetal bovine serum (FBS, PAA, Austria) and 0.5% penicillin-streptomycin (Invitrogen, USA). All flasks and plates were coated by poly-L-ornithine (PLO, Sigma-Aldrich, Germany) prior to cell seeding. Cells were kept in a humidified incubator at 37 °C and 5% CO2. After cell confluence was about 80%, flasks were shaken for 2 h (150 rpm, 37 °C) to remove microglia and oligodendrocytes from astrocytes. Additionally, before each subcultivation the 2 h machine shaking was repeated, the contaminating cells were transferred to the medium and then removed.
For subcultivation, cells were trypsinized with 2.5% (v/v) trypsin diluted in PBS/EDTA and seeded on new flasks in a 1:3 ratio. Medium was refreshed every second day. Subcultivation was performed when cells reached a confluence of about 80%. At passage 2, astrocytes were seeded on experimental plates 48 h prior to stimulation. 24 h before stimulation medium was changed to phenol red-free Gibco™ Roswell Park Memorial Institute (RPMI 1640, Life Technologies, USA) with additional 5% FBS and 0.5% penicillin-streptomycin (Fig. 1a).
Astrocyte culture purity was examined by immunocytochemistry (ICC) using anti-GFAP-antibody (glial fibrillary acidic protein), anti-Iba1-antibody (ionized calcium binding adaptor molecule 1), anti-Olig2-antibody (oligodendrocyte transcription factor 2) and Hoechst (33342, Trihydrochloride, Trihydrate, Invitrogen, USA) for nucleus staining. A detailed list of antibodies used for ICC is illustrated in Table 1. The average of astrocyte purity was 95%, less than 5% of the cells were microglia, under 0.5% of the cells remained undefined (Additional file 1: Figure S2B).
Table 1
Antibodies for immunocytochemistry
Antibody
Company
Order no.
Host
Dilution
Anti-goat 488
Life Technologies, USA
A11055
Donkey
1:500
Anti-goat 594
Life Technologies, USA
A11058
Donkey
1:500
Anti-mouse 488
Life Technologies, USA
A21202
Donkey
1:500
Anti-mouse 594
Life Technologies, USA
A21203
Donkey
1:500
Anti-rabbit 488
Life Technologies, USA
A21206
Donkey
1:500
Anti-rabbit 594
Life Technologies, USA
A21207
Donkey
1:500
GFAP
EnCor Biotechnology, USA
RPCA-GFAP
Rabbit
1:1000
Iba 1
Abcam, UK
ab107159
Goat
1:200
Iba 1
Millipore, USA
MABN92
Mouse
1:600
Iba 1
Wako, Japan
019–19,741
Rabbit
1:1000
NLRP3
Adipogen, USA
AG-20B-0014
Mouse
1:333
Olig2
Millipore, USA
MABN50
Mouse
1:1000
Olig2
Millipore, USA
AB9610
Rabbit
1:500
Primary and secondary antibodies used for immunocytochemical staining are listed

Preparation of A1AT, amyloid β1–42, LPS, and MCC950

A1AT originated from Prolastin (Grifols, Barcelona, Spain). 1000 mg of the powder were dissolved in 25 mL ultrapure water to obtain a concentration of 40 mg/mL. The solution was aliquoted and stored at − 80 °C.
To generate Amyloid β1–42 oligomers, we used the procedure described by Kayed et al. [52] and Gold et al. [15]. Briefly, 300 μg Aβ1–42 (Bachem, Bubendorf, Switzerland) were dissolved in 90 μL hexafluoroisopropanol, 210 μL ultrapure water and diluted with 900 μL 100 mM NaCl, 50 mM Tris (pH 7.4). The solution was stirred for 48 h on a magnetic stirrer at room temperature. Next, the tube was weighed again, and weight difference was adjusted with 100 mM NaCl 50 mM Tris (pH 7.4). The Aβ1–42 concentration of this solution was 56 mM. After centrifugation (16,000×g, 10 min), the supernatant was used for cell culture experiments. A negative control containing all ingredients but Aβ1–42 was established to evaluate possible solvent effects on astrocytes. Lipopolysaccharides (LPS) from Escherichia coli (Sigma-Aldrich, Germany) were used at a concentration of 1 μg/mL, as an extra stimulus for maximum cell stimulation. Stimulation time of all reagents was 3 h and 6 h. In order to reveal the short-term inflammasome regulation after Aβ1–42 and to understand the mechanism of early inflammation in AD, we decided for short term stimulation of cells.
MCC950 (Adipogen, USA) was diluted in DMSO (dimethylsulfoxid, Sigma-Aldrich, Germany) and used in a concentration of 1 μM. MCC950 incubated 1 h before further treatment, according to previous studies by Coll et al. [46]. Then, treatment with A1AT, Amyloid β1–42, and LPS was performed for 3 h and 6 h.
Cells were kept in the incubator at 37 °C and 5% CO2.

Dose-dependency studies

A dose-dependency study with increasing concentrations of A1AT [1, 2, 4, 8, 10, and 12 mg/mL] and Aβ1–42 [1, 2, 4, 8, 10, and 12 μM] was performed to determine the sublethal concentration for primary astrocytes. Lactate dehydrogenase (LDH) and Cell Titer-Blue (CTB) assay were used to assess cell viability after 3 h stimulation.

Cell viability

LDH-release

CytoTox 96® Non-Radioactive Cytotoxicity Assay (Promega, USA) was performed according to the manufacturer’s protocol to measure release of lactate dehydrogenase (LDH) as a marker of cellular viability. Astrocytes were seeded on a 96-well plate 48 h prior to stimulation and were finally stimulated with Aβ1–42 or LPS, and treated with A1AT. After 3 h/6 h incubation time, 50 μL of each well was transferred to a fresh 96-well plate. In addition to a no-treatment-cell control, a no-cell control, one positive control containing LDH and a control containing astrocytes lysed with Triton X-100 were used. CytoTox 96® Reagent (Promega, USA) was added to each well, and the absorbance was recorded at 490 nm by Infinite® M200 (Tecan, Switzerland). Data are presented as percentage of maximum LDH release (100%), which was determined by astrocytes lysed with 1% Triton X-100.

CTB assay

CellTiter-Blue® Cell Viability Assay (Promega, USA) was performed according to manufacturer’s protocol to assess metabolic activity of the cells. Astrocytes were seeded on a 96-well plate 48 h prior to stimulation and finally stimulated with Aβ1–42 or LPS and treated with A1AT. After 3 h and 6 h incubation time, CellTiter-Blue® Reagent (Promega, USA) was added to each well. After 2.5–3 h, a color switch (reduction of resazurin) was observed and fluorescence was recorded at 560Ex/590Em by Infinite® M200 (Tecan, Switzerland).

Semi-quantitative and quantitative real-time PCR

After 3 h/6 h of stimulation, medium was removed and peqGOLD TriFast™ (Peqlab, Germany) was added to cells. RNA was isolated using phenol-chloroform extraction method as previously described [53]. Afterwards, RNA concentration was measured by NanoDrop® ND-1000 (Thermo Fisher Scientific, USA). RNA purity was examined using 260/280 ratio, which was at 2.0 ± 0.1. Samples were diluted with ultrapure water to attain same RNA concentration in each sample. For DNA transcription, samples were transcribed by moloney murine leukemia virus (M-MLV) reverse transcriptase (Invitrogen™, USA) using random primer (Invitrogen™, USA). Semi-quantitative PCR with 30–32 cycles was performed to assess cDNA transcription success, starting with reference genes primer HPRT (hypoxanthine phosphoribosyltransferase 1), GAPDH (glycerinaldehyd-3-phosphate-dehydrogenase), and Hsp90 (heat shock protein 90). Table 2 contains all primers used for PCR. Positive control contained mouse cortex and negative control contained ultrapure water. A Thermocycler Mastercycler ep gradient S (Eppendorf, Germany) was used with the following settings: 3 min at 95 °C, 40 s at 95 °C, 40 s at respective annealing temperature (Table 2), and 45 s at 72 °C, 45 s at 72 °C. Nucleic acids were detected after application on 3% agarose gel containing Midori Green Advance (Biozym, Germany) for DNA staining and electrophoresis (25 min, 125 V constant, 400 mA). Gels were then photographed in E-box VX2 (Peqlab, Germany).
Table 2
Primers for PCR
Primer
Sequence
Bp
AT
ASC
Forward: CTTGTCAGGGGATGAACTCAAAA
Reverse: GCCATACGACTCCAGATAGTAGC
154
60
Casp1
Forward: CCGTGGAGAGAAACAAGGAGT
Reverse: CCCCTGACAGGATGTCTCCA
180
62
GAPDH
Forward: TGTGTCCGTCGTGGATCTGA
Reverse: CCTGCTTCACCACCTTCTTGA
77
65
HPRT
Forward: GCTGGTGAAAAGGACCTCT
Reverse: CACAGGACTAGAACACCTGC
249
61
Hsp90
Forward: TACTACTACTCGGCTTTCCCGT
Reverse: TCGAATCTTGTCCAGGGCATC
192
64
IL-1β
Forward: CAGCTCATATGGGTCCGACA
Reverse: CTGTGTCTTTCCCGTGGACC
251
61
IL-18
Forward: SGCCTGTGTTCGAGGATATGACT
Reverse: CCTTCACAGAGAGGGTCACAG
122
62
NLRP3
Forward: CCTGGGGGACTTTGGAATCA
Reverse: GATCCTGACAACACGCGGA
113
65
List of primers, respective sequences, base pairs (bp) and annealing temperature in °C (AT) used for sq-PCR and q-RT-PCR
For quantitative real-time PCR, a dilution series containing all samples was established, starting from 100% with dilution factor 2. Then, samples were diluted 1:10 with ultrapure water. Master mixture included SensiMix™ SYBR and Fluorescein (Bioline, USA), ultrapure water and primer (Table 2). CFX Connect™ Real-Time PCR Detection System (Bio-Rad, USA) was used. The following settings were adjusted: 10 min at 95 °C, 15 s at 95 °C, 30 s at respective annealing temperature (40 cycles), 30 s at 72 °C, and 5 s at 72 °C. The software quantified DNA products by melting curve analysis. An addition gel electrophoresis was performed to control the size of the amplified DNA products. First, the expression of reference genes was measured. All following target gene expressions were normalized to reference genes HPRT, GAPDH, and Hsp90. Using the qbase+ software (Biogazelle, Belgium), the relative quantification was calculated by the ∆∆Ct-method and data were expressed as relative amount of the three housekeeping genes, respectively, by using the multiple reference gene normalization method. Untreated cell controls were set to 1.

Immunocytochemistry

Immunocytochemistry (ICC) was performed as previously described by Habib et al. 2014 [51]. Astrocytes were seeded on cover slips on a 24-well plate. After stimulation, cells were fixed with 3.7% paraformaldehyde, lysed with Triton X-100, blocked with blocking buffer, and incubated with primary antibody. A negative control was established by incubating the cover slip only with blocking buffer without primary antibody. On the next day, the secondary antibody was applied and incubated for 2 h. After washing the cover slips, nuclei were stained by Hoechst (33342, Trihydrochloride, Trihydrate, Invitrogen, USA). A detailed list of antibodies used for ICC can be found in Table 1. Fluorescence images were taken with Leica DM6000 B (Leica Microsystems, Germany). For each experiment, the identical microscope settings were selected. Fluorescence intensity was measured using ImageJ (USA),

Western blot

Samples were generated from cell lysate and supernatant. Pierce™ BCA Protein Assay Kit (Thermo Fisher Scientific, USA) was used according to manufacturer’s protocol to measure protein concentration. The absorbance was recorded at 562 nm by Tecan Infinite® M200 reader (Tecan, Switzerland). Western blot was performed as previously described by Dang et al. 2011 [54, 55]. Briefly, after astrocytes were stimulated for 3 h/ 6 h, the lysis and extraction buffer as well as protease inhibitors were added. The buffer consists of 10 mM HEPES (PromoCell, Germany), 1.5 mM MgCl2 (Sigma-Aldrich, Germany), 10 mM KCl, 0.5 mM DTT, and 0.05% NP-40 (pH = 7.9.).
Samples were heated for 5 min at 95 °C, loaded on gels, and electrophoresis was performed (10 min at 80 V, 1 h at 140 V). PVDF membranes (Trans-Blot® Turbo™ RTA Mini PVDF Transfer Kit, Bio-Rad, USA) were activated with methanol, and then blotting was performed by Trans-Blot® Turbo™ Transfer System (Bio-Rad, USA) (22 min, 14 V). Blotting success was verified by incubating the membrane with methanol and Ponceau S. The membrane was incubated with the primary antibody overnight; on the next day, the secondary antibody was added after washing the membrane. Table 3 reveals the antibodies used. Chemiluminescence detection system was performed using Pierce™ ECL Western Blotting Substrate (Thermo Fisher Scientific, USA). Densitometric analysis was performed using ImageJ Software (USA).
Table 3
Antibodies for western blotting
Antibody
Company
Order no.
Host
Dilution
Anti-mouse
Sigma-Aldrich, Germany
A4416
Goat
1:4000
Anti-rabbit
Bio-Rad, USA
170–6515
Goat
1:5000
ASC (N15)-R
Santa Cruz, USA
sc-22514-R
Rabbit
1:1000
Caspase 1
Adipogen
20B0042C100
Mouse
1:1000
Caspase 1 p10
Santa Cruz, USA
sc-514
Rabbit
1:1000
Caspase 1 p20
Bioss
bs-6368R
Rabbit
1:500
IL-1β
Abcam, UK
ab9722
Rabbit
1:1000
IL-1β
Cell Signaling Technology, USA
12242S
Mouse
1:1000
IL-18
Abcam, UK
Ab71495
Rabbit
1:1000
IL-18
Santa Cruz, USA
sc-7954
Rabbit
1:1000
NLRP3
Bioss, USA
bs-10021R
Rabbit
1:1000
β-actin
Santa Cruz, USA
sc-47,778
Mouse
1:5000
List of primary and secondary antibodies used for western blotting. Antibodies were diluted in 5% milk

ELISA

Samples were generated from supernatant. Pierce™ BCA Protein Assay Kit (Thermo Fisher Scientific, USA) was used according to manufacturer’s protocol to measure protein concentration. The absorbance was recorded at 562 nm by Tecan Infinite® M200 reader (Tecan, Switzerland). Murine IL-1β Standard ABTS ELISA (PeproTech, USA) was performed according to manufacturer’s protocol. Capture antibody was incubated on a 96-well plate overnight. Wells were blocked for 1 h then incubated overnight with standard and samples in triplicate. Next, detection antibody was incubated for 2 h. Avidin-HRP conjugate was incubated for 30 min, afterwards ABTS was added. The color development was recorded at 405 nm by Tecan Infinite® M200 reader (Tecan, Switzerland).

Caspase 1 assay

FAM-FLICA® Caspase-1 Assay Kit (ImmunoChemistry Technologies, USA) was performed according to the manufacturer’s protocol to detect caspase 1 activity after 3 h and 6 h stimulation time. FLICA was incubated for 1 h at 37 °C. Hoechst 33342 (1:10000) was used for nuclear staining, cells then were fixed with 3.7% paraformaldehyde. Fluorescence images were taken with Leica DM6000 B (Leica Microsystems, Germany); for each experiment, the exact same microscope settings were adjusted. The number of caspase 1 active cells was counted and set in relation to the total amount of counted astrocytes per well. For each treatment group, 100 cells/well in 5 wells were counted.

Data analysis

All experiments were performed at least three times in triplicate. All data are presented as arithmetic mean ± standard deviation of the mean. Prior to the analysis the residuals of data were tested for normal distribution with the Shapiro-Wilk normality test using JMP® (Version 10, SAS Institute Inc., Cary, NC, USA, 1989–2007). Secondly, equal variance was tested with the Bartlett test. In case that one of these tests was significant, a Box-Cox transformation was performed, and the test for normality and equal variance were repeated with the new calculated values. Finally, a one-way ANOVA was applied followed by the Tukey-HSD post-hoc test for intergroup differences. When transformation of data failed to convert non-normal into normal distributed data, rank data were calculated and used for one-way ANOVA analysis, which results in the Kruskal-Wallis non-parametric analysis followed by the Tukey-HSD post-hoc test. Statistical significance was set at p value ≤ 0.05 (*/a ≤ 0.05, **/aa ≤ 0.01, ***/aaa ≤ 0.001).

Results

After primary astrocytes were stimulated according to work flow (Fig. 1a), culture purity of 95% astrocytes on average was examined by ICC, less than 5% of the remaining cells were microglia (Fig. 1b, Additional file 2: Figure S1 and Additional file 1: Figure S2).

Amyloid β1–42 had a dose-dependent cytotoxic effect on astrocytes

First, we incubated astrocytes with Aβ1–42 for 3 h to assess the short-term effect on cell viability. A concentration range of frequently used doses between 1 μM and 12 μM was selected [14, 15]. Aβ1–42 induced a concentration-depended release of LDH into the medium, reaching 50% release and a significant difference compared to the control condition at a concentration of 12 μM (Additional file 3: Figure S3A). In comparison, the stimulation with LPS (1 μg/mL) led to a LDH-release of about 60%. The metabolic activity of Aβ1–42-stimulated primary astrocytes (Cell Titer-Blue-assay) revealed no significant dose-dependent differences (Additional file 4: Figure S4A). To rule out cytotoxic effects, the further studies were performed with sublethal doses of Aβ1–42 at 4 μM and 10 μM.
To examine a possible cytotoxic effect of A1AT, dose-dependency studies were performed. 3 h incubation of astrocytes with increasing concentrations of 1 mg/mL to 12 mg/mL of A1AT did not affect LDH release (Additional file 3: Figure S3B), also CTB assay showed no influence on metabolic activity of astrocytes (Additional file 4: Figure S4B).
Moreover, co-exposure of astrocytes with Aβ1–42 (4 μM and 10 μM) and A1AT (1 mg/mL) had no impact on LDH release (Fig. 2a, c, Additional file 3: Figure S3C) or cell metabolism (Fig. 2b, d, Additional file 4: Figure S4C). In contrast, A1AT exposure of LPS-stimulated astrocytes significantly reduced LDH release (Additional file 3: Figure S3C).

A1AT prevented Aβ1–42-induced upregulation of NLRP3 mRNA and protein

We next evaluated the expression of NLRP3 in the given experimental settings. Stimulation of astrocytes with Aβ1–42-oligomers significantly increased NLRP3 mRNA-expression by three-fold (Aβ1–42, 4 μM) or four-fold (Aβ1–42, 10 μM) in comparison to untreated controls (Fig. 3a, Fig. 4c, d). Co-treatment with 4 mg/mL A1AT almost completely blocked this increase (Fig. 3a, Fig. 4c, d). Western blot analysis revealed a significant higher protein expression of NLRP3 in Aβ1–42-stimulated astrocytes compared to untreated controls (Fig. 3b, c, Fig. 4a, b). Co-treatment with A1AT significantly prevented this increase in NLRP3 protein expression (Fig. 3c, Fig. 4a, b). These results were confirmed by ICC. Staining with GFAP-, NLRP3-antibody and Hoechst revealed significantly higher fluorescence intensity of NLRP3-stained astrocytes stimulated with 10 μM of Aβ1–42 (Fig. 3d, e, microscope settings were identical for each experiment). Fluorescence intensity of Aβ1–42-stimulated cells significantly declined with co-treatment by A1AT (Fig. 3e).
To exclude that NLRP3-upregulation was due to microglia contamination, ICC-staining using Iba1- and NLRP3-antibody was performed (Additional file 5: Figure S5). Indeed, NLRP3 was expressed by the few present microglia. But as Additional file 1: Figure S2 and Additional file 5: Figure S5 show, the amount of microglia was so low, that their impact on NLRP3-expression is negligible.

1–42-stimulation and A1AT-treatment did not regulate ASC expression

The NLRP3-inflammasome consists of active NLRP3 (LRR, NACHT, PYD, CARD) as well as the adaptor protein ASC and pro-caspase 1. In primary astrocytes, treatment with 4 μM or 10 μM Aβ1–42 or 4 mg/mL A1AT did not result in changes of ASC mRNA or protein expression at 3 h and 6 h stimulation time (Fig. 5).

A1AT abrogated Aβ1–42-induced upregulation of caspase 1 and the pro-inflammatory cytokine IL-1β

Next, we analyzed caspase 1, IL-1β and IL-18 expression. Whereas 3 h treatment of primary astrocytes with 4 μM of Aβ1–42 did not result in an increase of caspase 1 mRNA expression, treatment with 10 μM of Aβ1–42 led to a significant increase of caspase 1 mRNA expression (Fig. 6a, Additional file 6: Figure S6A). Co-treatment with 4 mg/mL A1AT blocked the increase of caspase 1 expression significantly (Additional file 6: Figure S6A). Repeating this experiment, this time performing a 6-h stimulation, revealed that Aβ1–42 significantly increased mRNA levels of caspase 1 in astrocytes (Fig. 6b). Co-treatment with A1AT blocked this increase in caspase 1 mRNA (Fig. 6b).
FAM-FLICA® Caspase-1 Assay, measuring active caspase-1 enzyme, showed that treatment with Aβ1–42 significantly induced the number of caspase 1 positive cells (Fig. 7). Co-treatment with 4 mg/mL of A1AT significantly blocked this increase of caspase 1-positive cells (Fig. 7).
In the next step, pro-inflammatory cytokines cleaved by caspase 1 were assessed. Stimulation of astrocytes with 4 μM and 10 μM of Aβ1–42 increased mRNA expression of IL-1β (Fig. 8a, b, Additional file 6: Figure S6B). Co-treatment with A1AT significantly reduced mRNA expression of IL-1β (Fig. 8a, b, Additional file 6: Fig. 6b). In contrast, IL-18 gene expression was not affected by either of the treatments at 3 h stimulation time (Fig. 8c, Additional file 6: Figure S6C). However, 6 h stimulation with Aβ1–42 significantly increased mRNA levels of IL-18 in astrocytes, but A1AT did not block this effect (Fig. 8d).
Furthermore, western blot revealed a significant upregulation of the IL-1β-precursor in Aβ1–42-treated cells (Fig. 9a, b). Co-treatment with A1AT blocked the effect (Fig. 9a, b). Analysis of IL-1β protein by ELISA revealed a similar effect of increased levels after Aβ1–42-stimulation, which was significant at 6 h, but not at 3 h (9C-D). A1AT-treatment significantly prevented this increase time-dependently after 6 h stimulation (Fig. 9d).

MCC950 reduced caspase 1 activity and IL-1β protein expression

In order to elucidate a direct interaction between A1AT and the NLRP3-inflammasome, all studies were repeated including MCC950—a selective inhibitor of NLRP3. MCC950 had no impact on cell viability (Fig. 2a, c), but led to an increase of cell metabolism in each treatment group at 3 h and 6 h, though not significant (Fig. 2b, d). In western blot, MCC950 did not change protein levels of NLRP3 (Fig. 4a, b). Treatment with MCC950 did not alter gene expression of NLRP3, ASC, caspase 1, IL-1β, and IL-18 (Figs. 4c, d, 5E–F, 6a, b, 8a, d).
MCC950 significantly mitigated caspase 1 activity (Fig. 7) and significantly decreased IL-1β in Aβ-stimulated astrocytes examined by ELISA (Fig. 9c, d). Further, there is a trend towards a decrease of IL-1β-protein levels in the presence of MCC950 in nearly all treatment groups, though not significant (Fig. 9c, d). MCC950 did not affect Aβ-induced expression of IL-1β-precursor protein in western blot (Fig. 9a, b).

Co-treatment with A1AT and Aβ in the presence of MCC950 did not alter expression of NLRP3-inflammasome components

Examined by caspase 1 assay (Fig. 7), IL-1β western blot (Fig. 9a, b) and IL-1β-ELISA (Fig. 9c, d) co-treatment with A1AT and Aβ in the presence of MCC950 did not alter expression of inflammasome components compared to the same stimulation group in absence of MCC950. Therefore, we conclude that A1AT mitigated IL-1β mainly by inhibiting NLRP3-inflammasome.

Discussion

Activation of glia cells and overexpression of pro-inflammatory cytokines are regarded early events in Alzheimer’s disease [56]. In recent years, astrocytes have come into focus in neurodegenerative disorders such as AD and are seen in a new way. Astrocytes express a plethora of receptors and modulate cells and their function in their surroundings [57]. In brief, they are involved in excitotoxic glutamate release, secretion of pro-inflammatory cytokines, growth factor production, stabilization, and organization of the blood-brain barrier and Aβ1–42 production [58]. In microglia cells, the activation of NLRP3 appears to be an essential step during AD. Following activation of NLRP3, inflammation is triggered by the activation of caspase 1 and generation of IL-1β. This hampers the phagocytic capability of microglia cells [39]. The NLRP3-inflammasome is important for the initiation and processing of neuroinflammatory processes, and especially in AD, NLRP3 is associated with age-related inflammation [59]. NLRP3 is known to be activated by Aβ1–42-aggregates [14]. NLRP3 knock-out in transgenic animals carrying mutations associated with AD prevents AD pathology [24].
Our group has recently presented data in acute and chronic neurodegenerative disease models that different components of the NLRP3-inflammasome are allocated to astrocytes [6064]. With respect to AD, Couturier and co-workers were able to show that astrocytes produce and release IL-1β following Aβ1–42-stimulation [18]. In this animal model, the downregulation of NLRP3-inflammasome activation leads to decreased amyloid plaques and a better memory performance [35]. Our data now show that stimulation of primary astrocytes with Aβ1–42 induces a dose-dependent upregulation of NLRP3. This in turn is known to stimulate the activation of caspase 1 and IL-1β [39]. Our work further demonstrates that such an effect also occurs after short-term stimulation with Aβ1–42. In contrast, the co-treatment of astrocytes with Aβ1–42 and A1AT blocks the induction of NLRP3. The regulation of NLRP3 is complex and usually requires a two-step activation process [22]. Currently, it is well accepted that the first step includes a priming signal, usually provided by NF-ΚB signaling or secretion of endogenous cytokines such as IL-1a [65]. NLRP3 is then activated by a variety of cellular signals, amongst them are misfolded proteins. The large variety of possible regulatory signals and pathways suggest that the activation is rather the consequence of a disturbance of cellular equilibrium [22]. In 2012, Lee and co-workers have identified calcium signaling as essential in NLRP3 activation [66]. In this study, increased intracellular calcium levels are associated with NLRP3 activation. In astrocytes, Aβ1–42 potentiates calcium signaling which is triggered by mGlu, α7nAChR, and purinergic substances [57].
Yet, there is little evidence on the regulation of the NLRP3-inflammasome by A1AT. Toldo et al. 2011 stated that A1AT inhibits caspase-1 [67]. Aggarwal et al. 2016 found that—by the presence of polyunsaturated fatty acids—A1AT downregulates NLRP3 and caspase 1 [68]. For astrocytes, no data exist with respect to mechanisms of action of A1AT. In previous studies, we have presented data which show that A1AT reduces inflammation in microglia cells mainly by controlling calcium signaling pathways [15]. A1AT has no effect on classic signaling pathways such as MAPK p38, p44/42, JNK, and cAMP-coupled mechanisms [15]. Using a fluorescent calcium dye, we have shown that A1AT reduces intracellular calcium concentrations in a microglial cell line [15]. A1AT had no direct effect on Aβ1–42-oligomerization [15]. We therefore hypothesize that A1AT effects on NLRP3 upregulation in primary astrocytes are mainly triggered by an inhibition of calcium and calpain. This hypothesis needs further evaluation, since other reports also demonstrate that A1AT is able to reduce glutamate-induced toxicity in murine primary neurons [45]. Since astrocytes release glutamate in response to Aβ1–42-stimulation, this could represent another way how A1AT prevents deleterious Aβ1–42-induced inflammatory cascades in microglia, neurons, and astrocytes. However, our current study does not include research on how A1AT could regulate the NLRP3-inflammasome complex. Further, it must be remarked that microglia might have partially contributed to the observed results due to the slight contamination of approximately 5%.
Our data demonstrate that NLRP3-inflammasome components are upregulated time-dependently following Aβ1–42-stimulation. This can be blocked by A1AT application. NLRP3 is the sensor protein of the NLRP3-inflammasome complex, which explains its upregulation after Aβ1–42-stimulation. ASC, in contrast, has the caspase activating and recruitment domain. In our experiments, ASC concentration on the mRNA and protein level was unchanged, indicating that Aβ1–42 and A1AT mainly regulate NLRP3, but not ASC. We assume the total amount of ASC to be sufficient to lead to the NLRP3-ASC-complex formation and caspase 1 binding.
To further investigate the direct effect of A1AT on inflammasome-dependent IL-1β maturation, we used a specific NLRP3-inhibitor called MCC950. As previously shown by Coll et al., pre-treatment with MCC950 prevents complex formation of apoptosis-associated speck-like protein containing a CARD (ASC) and blocks the release of IL-1β in immunological active cells, without affecting priming of NLRP3 [46]. Furthermore, MCC950 stimulated Aβ phagocytosis in vitro, and it reduced Aβ accumulation in a mouse model of AD, which was associated with improved cognitive function [50]. In our studies, MCC950-pretreatment in cells co-treated with A1AT and Aβ did not further drop IL-1β protein expression. Thus, A1AT had no effect on protein expression, when NLRP3 was selectively blocked. We therefore conclude that A1AT reduces IL-1β by inhibiting NLRP3-inflammasome.
These observations not only clearly highlight the importance of astroglial-related pro-inflammatory processes in the brain and in particular during AD, but also point at A1AT as a potent antagonist in astrocyte-dependent inflammatory signaling.

Conclusion

We demonstrate that Aβ1–42-stimulation results in an upregulation of NLRP3, caspase 1, and its cleavage products in astrocytes. A1AT time-dependently hampers Aβ1–42-triggered neuroinflammation by attenuating NLRP3-inflammasome expression. This suggests that A1AT offers a therapeutic opportunity for AD treatment.

Funding

This work was supported by an internal grant from the Medical Clinic of the RWTH Aachen University (START grant, P. Habib). The funding body had no influence in the design of the study and was neither involved in collection, analysis, and interpretation of results nor in writing the manuscript.

Availability of data and materials

All data generated or analyzed during this study are included in this published article and its supplementary information files.

Ethics approval

No human tissue was involved in this study. Postnatal (P0 to P2) cortical astrocyte culture preparation from BALB/c mice (Charles River) was performed as previously described by Habib et al. 2014 [51]. Preparation was conducted in accordance with animal welfare policy of University Hospital Aachen and the government of the State of North Rhine-Westphalia, Germany (no. 84.02.04.2015.A292).
Not applicable

Competing interests

All 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.
Anhänge

Additional files

Literatur
1.
Zurück zum Zitat Prince M, Bryce R, Albanese E, Wimo A, Ribeiro W, Ferri CP. The global prevalence of dementia: a systematic review and metaanalysis. Alzheimers Dement. 2013;9(1):63–75.e62.CrossRef Prince M, Bryce R, Albanese E, Wimo A, Ribeiro W, Ferri CP. The global prevalence of dementia: a systematic review and metaanalysis. Alzheimers Dement. 2013;9(1):63–75.e62.CrossRef
2.
Zurück zum Zitat Brookmeyer R, Johnson E, Ziegler-Graham K, Arrighi HM. Forecasting the global burden of Alzheimer's disease. Alzheimers Dement. 2007;3(3):186–91.CrossRef Brookmeyer R, Johnson E, Ziegler-Graham K, Arrighi HM. Forecasting the global burden of Alzheimer's disease. Alzheimers Dement. 2007;3(3):186–91.CrossRef
3.
Zurück zum Zitat Hirtz D, Thurman DJ, Gwinn-Hardy K, Mohamed M, Chaudhuri AR, Zalutsky R. How common are the “common” neurologic disorders? Neurology. 2007;68(5):326–37.CrossRef Hirtz D, Thurman DJ, Gwinn-Hardy K, Mohamed M, Chaudhuri AR, Zalutsky R. How common are the “common” neurologic disorders? Neurology. 2007;68(5):326–37.CrossRef
4.
Zurück zum Zitat Rosenberg PB, Lyketsos C. Mild cognitive impairment: searching for the prodrome of Alzheimer's disease. World Psychiatry. 2008;7(2):72–8.CrossRef Rosenberg PB, Lyketsos C. Mild cognitive impairment: searching for the prodrome of Alzheimer's disease. World Psychiatry. 2008;7(2):72–8.CrossRef
5.
Zurück zum Zitat Wilson RS, Leurgans SE, Boyle PA, Bennett DA. Cognitive decline in prodromal Alzheimer disease and mild cognitive impairment. Arch Neurol. 2011;68(3):351–6.CrossRef Wilson RS, Leurgans SE, Boyle PA, Bennett DA. Cognitive decline in prodromal Alzheimer disease and mild cognitive impairment. Arch Neurol. 2011;68(3):351–6.CrossRef
6.
Zurück zum Zitat Ringman JM, Liang LJ, Zhou Y, Vangala S, Teng E, Kremen S, Wharton D, Goate A, Marcus DS, Farlow M, et al. Early behavioural changes in familial Alzheimer’s disease in the dominantly inherited Alzheimer network. Brain. 2015;138(Pt 4):1036–45.CrossRef Ringman JM, Liang LJ, Zhou Y, Vangala S, Teng E, Kremen S, Wharton D, Goate A, Marcus DS, Farlow M, et al. Early behavioural changes in familial Alzheimer’s disease in the dominantly inherited Alzheimer network. Brain. 2015;138(Pt 4):1036–45.CrossRef
7.
Zurück zum Zitat Bature F, Guinn BA, Pang D, Pappas Y. Signs and symptoms preceding the diagnosis of Alzheimer's disease: a systematic scoping review of literature from 1937 to 2016. BMJ Open. 2017;7(8):e015746.CrossRef Bature F, Guinn BA, Pang D, Pappas Y. Signs and symptoms preceding the diagnosis of Alzheimer's disease: a systematic scoping review of literature from 1937 to 2016. BMJ Open. 2017;7(8):e015746.CrossRef
8.
Zurück zum Zitat Hsu D, Marshall GA. Primary and secondary prevention trials in Alzheimer disease: looking back, Moving Forward. Curr Alzheimer Res. 2017;14(4):426–40.PubMedPubMedCentral Hsu D, Marshall GA. Primary and secondary prevention trials in Alzheimer disease: looking back, Moving Forward. Curr Alzheimer Res. 2017;14(4):426–40.PubMedPubMedCentral
9.
Zurück zum Zitat Karakaya T, Fusser F, Schroder J, Pantel J. Pharmacological treatment of mild cognitive impairment as a prodromal syndrome of Alzheimer’s disease. Curr Neuropharmacol. 2013;11(1):102–8.PubMedPubMedCentral Karakaya T, Fusser F, Schroder J, Pantel J. Pharmacological treatment of mild cognitive impairment as a prodromal syndrome of Alzheimer’s disease. Curr Neuropharmacol. 2013;11(1):102–8.PubMedPubMedCentral
10.
Zurück zum Zitat Dubois B, Zaim M, Touchon J, Vellas B, Robert P, Murphy MF, Pujadas-Navines F, Rainer M, Soininen H, Riordan HJ, et al. Effect of six months of treatment with V0191 in patients with suspected prodromal Alzheimer’s disease. J Alzheimers Dis. 2012;29(3):527–35.CrossRef Dubois B, Zaim M, Touchon J, Vellas B, Robert P, Murphy MF, Pujadas-Navines F, Rainer M, Soininen H, Riordan HJ, et al. Effect of six months of treatment with V0191 in patients with suspected prodromal Alzheimer’s disease. J Alzheimers Dis. 2012;29(3):527–35.CrossRef
11.
Zurück zum Zitat Caldwell CC, Yao J, Brinton RD. Targeting the prodromal stage of Alzheimer’s disease: bioenergetic and mitochondrial opportunities. Neurotherapeutics. 2015;12(1):66–80.CrossRef Caldwell CC, Yao J, Brinton RD. Targeting the prodromal stage of Alzheimer’s disease: bioenergetic and mitochondrial opportunities. Neurotherapeutics. 2015;12(1):66–80.CrossRef
12.
Zurück zum Zitat Haass C, Selkoe DJ. Soluble protein oligomers in neurodegeneration: lessons from the Alzheimer's amyloid beta-peptide. Nat Rev Mol Cell Biol. 2007;8(2):101–12.CrossRef Haass C, Selkoe DJ. Soluble protein oligomers in neurodegeneration: lessons from the Alzheimer's amyloid beta-peptide. Nat Rev Mol Cell Biol. 2007;8(2):101–12.CrossRef
13.
Zurück zum Zitat Baglioni S, Casamenti F, Bucciantini M, Luheshi LM, Taddei N, Chiti F, Dobson CM, Stefani M. Prefibrillar amyloid aggregates could be generic toxins in higher organisms. J Neurosci. 2006;26(31):8160–7.CrossRef Baglioni S, Casamenti F, Bucciantini M, Luheshi LM, Taddei N, Chiti F, Dobson CM, Stefani M. Prefibrillar amyloid aggregates could be generic toxins in higher organisms. J Neurosci. 2006;26(31):8160–7.CrossRef
14.
Zurück zum Zitat Halle A, Hornung V, Petzold GC, Stewart CR, Monks BG, Reinheckel T, Fitzgerald KA, Latz E, Moore KJ, Golenbock DT. The NALP3 inflammasome is involved in the innate immune response to amyloid-beta. Nat Immunol. 2008;9(8):857–65.CrossRef Halle A, Hornung V, Petzold GC, Stewart CR, Monks BG, Reinheckel T, Fitzgerald KA, Latz E, Moore KJ, Golenbock DT. The NALP3 inflammasome is involved in the innate immune response to amyloid-beta. Nat Immunol. 2008;9(8):857–65.CrossRef
15.
Zurück zum Zitat Gold M, Dolga AM, Koepke J, Mengel D, Culmsee C, Dodel R, Koczulla AR, Bach JP. alpha1-antitrypsin modulates microglial-mediated neuroinflammation and protects microglial cells from amyloid-beta-induced toxicity. J Neuroinflammation. 2014;11:165.CrossRef Gold M, Dolga AM, Koepke J, Mengel D, Culmsee C, Dodel R, Koczulla AR, Bach JP. alpha1-antitrypsin modulates microglial-mediated neuroinflammation and protects microglial cells from amyloid-beta-induced toxicity. J Neuroinflammation. 2014;11:165.CrossRef
16.
Zurück zum Zitat Saresella M, La Rosa F, Piancone F, Zoppis M, Marventano I, Calabrese E, Rainone V, Nemni R, Mancuso R, Clerici M. The NLRP3 and NLRP1 inflammasomes are activated in Alzheimer’s disease. Mol Neurodegener. 2016;11:23.CrossRef Saresella M, La Rosa F, Piancone F, Zoppis M, Marventano I, Calabrese E, Rainone V, Nemni R, Mancuso R, Clerici M. The NLRP3 and NLRP1 inflammasomes are activated in Alzheimer’s disease. Mol Neurodegener. 2016;11:23.CrossRef
17.
Zurück zum Zitat Glass CK, Saijo K, Winner B, Marchetto MC, Gage FH. Mechanisms underlying inflammation in neurodegeneration. Cell. 2010;140(6):918–34.CrossRef Glass CK, Saijo K, Winner B, Marchetto MC, Gage FH. Mechanisms underlying inflammation in neurodegeneration. Cell. 2010;140(6):918–34.CrossRef
18.
Zurück zum Zitat Kraft AD, Harry GJ. Features of microglia and neuroinflammation relevant to environmental exposure and neurotoxicity. Int J Environ Res Public Health. 2011;8(7):2980–3018.CrossRef Kraft AD, Harry GJ. Features of microglia and neuroinflammation relevant to environmental exposure and neurotoxicity. Int J Environ Res Public Health. 2011;8(7):2980–3018.CrossRef
19.
Zurück zum Zitat Rama Rao KV, Kielian T. Neuron-astrocyte interactions in neurodegenerative diseases: role of neuroinflammation. Clin Exp Neuroimmunol. 2015;6(3):245–63.CrossRef Rama Rao KV, Kielian T. Neuron-astrocyte interactions in neurodegenerative diseases: role of neuroinflammation. Clin Exp Neuroimmunol. 2015;6(3):245–63.CrossRef
20.
Zurück zum Zitat Goedert M, Wischik CM, Crowther RA, Walker JE, Klug A. Cloning and sequencing of the cDNA encoding a core protein of the paired helical filament of Alzheimer disease: identification as the microtubule-associated protein tau. Proc Natl Acad Sci U S A. 1988;85(11):4051–5.CrossRef Goedert M, Wischik CM, Crowther RA, Walker JE, Klug A. Cloning and sequencing of the cDNA encoding a core protein of the paired helical filament of Alzheimer disease: identification as the microtubule-associated protein tau. Proc Natl Acad Sci U S A. 1988;85(11):4051–5.CrossRef
21.
Zurück zum Zitat Goedert M, Spillantini MG, Jakes R, Rutherford D, Crowther RA. Multiple isoforms of human microtubule-associated protein tau: sequences and localization in neurofibrillary tangles of Alzheimer’s disease. Neuron. 1989;3(4):519–26.CrossRef Goedert M, Spillantini MG, Jakes R, Rutherford D, Crowther RA. Multiple isoforms of human microtubule-associated protein tau: sequences and localization in neurofibrillary tangles of Alzheimer’s disease. Neuron. 1989;3(4):519–26.CrossRef
22.
Zurück zum Zitat Song L, Pei L, Yao S, Wu Y, Shang Y. NLRP3 Inflammasome in neurological diseases, from functions to therapies. Front Cell Neurosci. 2017;11:63.PubMedPubMedCentral Song L, Pei L, Yao S, Wu Y, Shang Y. NLRP3 Inflammasome in neurological diseases, from functions to therapies. Front Cell Neurosci. 2017;11:63.PubMedPubMedCentral
23.
Zurück zum Zitat Agostini L, Martinon F, Burns K, McDermott MF, Hawkins PN, Tschopp J. NALP3 forms an IL-1beta-processing inflammasome with increased activity in Muckle-Wells autoinflammatory disorder. Immunity. 2004;20(3):319–25.CrossRef Agostini L, Martinon F, Burns K, McDermott MF, Hawkins PN, Tschopp J. NALP3 forms an IL-1beta-processing inflammasome with increased activity in Muckle-Wells autoinflammatory disorder. Immunity. 2004;20(3):319–25.CrossRef
24.
Zurück zum Zitat Heneka MT, Kummer MP, Stutz A, Delekate A, Schwartz S, Vieira-Saecker A, Griep A, Axt D, Remus A, Tzeng TC, et al. NLRP3 is activated in Alzheimer's disease and contributes to pathology in APP/PS1 mice. Nature. 2013;493(7434):674–8.CrossRef Heneka MT, Kummer MP, Stutz A, Delekate A, Schwartz S, Vieira-Saecker A, Griep A, Axt D, Remus A, Tzeng TC, et al. NLRP3 is activated in Alzheimer's disease and contributes to pathology in APP/PS1 mice. Nature. 2013;493(7434):674–8.CrossRef
25.
Zurück zum Zitat Sutterwala FS, Haasken S, Cassel SL. Mechanism of NLRP3 inflammasome activation. Ann N Y Acad Sci. 2014;1319:82–95.CrossRef Sutterwala FS, Haasken S, Cassel SL. Mechanism of NLRP3 inflammasome activation. Ann N Y Acad Sci. 2014;1319:82–95.CrossRef
26.
Zurück zum Zitat Martinon F. Detection of immune danger signals by NALP3. J Leukoc Biol. 2008;83(3):507–11.CrossRef Martinon F. Detection of immune danger signals by NALP3. J Leukoc Biol. 2008;83(3):507–11.CrossRef
27.
Zurück zum Zitat Martinon F, Burns K, Tschopp J. The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol Cell. 2002;10(2):417–26.CrossRef Martinon F, Burns K, Tschopp J. The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol Cell. 2002;10(2):417–26.CrossRef
28.
Zurück zum Zitat Yatsiv I, Morganti-Kossmann MC, Perez D, Dinarello CA, Novick D, Rubinstein M, Otto VI, Rancan M, Kossmann T, Redaelli CA, et al. Elevated intracranial IL-18 in humans and mice after traumatic brain injury and evidence of neuroprotective effects of IL-18-binding protein after experimental closed head injury. J Cereb Blood Flow Metab. 2002;22(8):971–8.CrossRef Yatsiv I, Morganti-Kossmann MC, Perez D, Dinarello CA, Novick D, Rubinstein M, Otto VI, Rancan M, Kossmann T, Redaelli CA, et al. Elevated intracranial IL-18 in humans and mice after traumatic brain injury and evidence of neuroprotective effects of IL-18-binding protein after experimental closed head injury. J Cereb Blood Flow Metab. 2002;22(8):971–8.CrossRef
29.
Zurück zum Zitat Merrill JE, Benveniste EN. Cytokines in inflammatory brain lesions: helpful and harmful. Trends Neurosci. 1996;19(8):331–8.CrossRef Merrill JE, Benveniste EN. Cytokines in inflammatory brain lesions: helpful and harmful. Trends Neurosci. 1996;19(8):331–8.CrossRef
30.
Zurück zum Zitat Codolo G, Plotegher N, Pozzobon T, Brucale M, Tessari I, Bubacco L, de Bernard M. Triggering of inflammasome by aggregated alpha-synuclein, an inflammatory response in synucleinopathies. PLoS One. 2013;8(1):e55375.CrossRef Codolo G, Plotegher N, Pozzobon T, Brucale M, Tessari I, Bubacco L, de Bernard M. Triggering of inflammasome by aggregated alpha-synuclein, an inflammatory response in synucleinopathies. PLoS One. 2013;8(1):e55375.CrossRef
31.
Zurück zum Zitat Mao Z, Liu C, Ji S, Yang Q, Ye H, Han H, Xue Z. The NLRP3 Inflammasome is involved in the pathogenesis of Parkinson’s disease in rats. Neurochem Res. 2017;42(4):1104–15.CrossRef Mao Z, Liu C, Ji S, Yang Q, Ye H, Han H, Xue Z. The NLRP3 Inflammasome is involved in the pathogenesis of Parkinson’s disease in rats. Neurochem Res. 2017;42(4):1104–15.CrossRef
32.
Zurück zum Zitat Zhou Y, Lu M, Du RH, Qiao C, Jiang CY, Zhang KZ, Ding JH, Hu G. MicroRNA-7 targets nod-like receptor protein 3 inflammasome to modulate neuroinflammation in the pathogenesis of Parkinson's disease. Mol Neurodegener. 2016;11:28.CrossRef Zhou Y, Lu M, Du RH, Qiao C, Jiang CY, Zhang KZ, Ding JH, Hu G. MicroRNA-7 targets nod-like receptor protein 3 inflammasome to modulate neuroinflammation in the pathogenesis of Parkinson's disease. Mol Neurodegener. 2016;11:28.CrossRef
33.
Zurück zum Zitat Zhang P, Shao XY, Qi GJ, Chen Q, Bu LL, Chen LJ, Shi J, Ming J, Tian B. Cdk5-dependent activation of neuronal Inflammasomes in Parkinson's disease. Mov Disord. 2016;31(3):366–76.CrossRef Zhang P, Shao XY, Qi GJ, Chen Q, Bu LL, Chen LJ, Shi J, Ming J, Tian B. Cdk5-dependent activation of neuronal Inflammasomes in Parkinson's disease. Mov Disord. 2016;31(3):366–76.CrossRef
34.
Zurück zum Zitat Yang F, Wang Z, Wei X, Han H, Meng X, Zhang Y, Shi W, Li F, Xin T, Pang Q, et al. NLRP3 deficiency ameliorates neurovascular damage in experimental ischemic stroke. J Cereb Blood Flow Metab. 2014;34(4):660–7.CrossRef Yang F, Wang Z, Wei X, Han H, Meng X, Zhang Y, Shi W, Li F, Xin T, Pang Q, et al. NLRP3 deficiency ameliorates neurovascular damage in experimental ischemic stroke. J Cereb Blood Flow Metab. 2014;34(4):660–7.CrossRef
35.
Zurück zum Zitat Couturier J, Stancu IC, Schakman O, Pierrot N, Huaux F, Kienlen-Campard P, Dewachter I, Octave JN. Activation of phagocytic activity in astrocytes by reduced expression of the inflammasome component ASC and its implication in a mouse model of Alzheimer disease. J Neuroinflammation. 2016;13:20.CrossRef Couturier J, Stancu IC, Schakman O, Pierrot N, Huaux F, Kienlen-Campard P, Dewachter I, Octave JN. Activation of phagocytic activity in astrocytes by reduced expression of the inflammasome component ASC and its implication in a mouse model of Alzheimer disease. J Neuroinflammation. 2016;13:20.CrossRef
36.
Zurück zum Zitat Dong Y, Benveniste EN. Immune function of astrocytes. Glia. 2001;36(2):180–90.CrossRef Dong Y, Benveniste EN. Immune function of astrocytes. Glia. 2001;36(2):180–90.CrossRef
37.
Zurück zum Zitat Akiyama H, Arai T, Kondo H, Tanno E, Haga C, Ikeda K. Cell mediators of inflammation in the Alzheimer disease brain. Alzheimer Dis Assoc Disord. 2000;14(Suppl 1):S47–53.CrossRef Akiyama H, Arai T, Kondo H, Tanno E, Haga C, Ikeda K. Cell mediators of inflammation in the Alzheimer disease brain. Alzheimer Dis Assoc Disord. 2000;14(Suppl 1):S47–53.CrossRef
38.
Zurück zum Zitat Salminen A, Ojala J, Suuronen T, Kaarniranta K, Kauppinen A. Amyloid-beta oligomers set fire to inflammasomes and induce Alzheimer’s pathology. J Cell Mol Med. 2008;12(6A):2255–62.CrossRef Salminen A, Ojala J, Suuronen T, Kaarniranta K, Kauppinen A. Amyloid-beta oligomers set fire to inflammasomes and induce Alzheimer’s pathology. J Cell Mol Med. 2008;12(6A):2255–62.CrossRef
39.
Zurück zum Zitat Gold M, El Khoury J. beta-amyloid, microglia, and the inflammasome in Alzheimer's disease. Semin Immunopathol. 2015;37(6):607–11.CrossRef Gold M, El Khoury J. beta-amyloid, microglia, and the inflammasome in Alzheimer's disease. Semin Immunopathol. 2015;37(6):607–11.CrossRef
40.
Zurück zum Zitat Freeman L, Guo H, David CN, Brickey WJ, Jha S, Ting JP. NLR members NLRC4 and NLRP3 mediate sterile inflammasome activation in microglia and astrocytes. J Exp Med. 2017;214(5):1351–70.CrossRef Freeman L, Guo H, David CN, Brickey WJ, Jha S, Ting JP. NLR members NLRC4 and NLRP3 mediate sterile inflammasome activation in microglia and astrocytes. J Exp Med. 2017;214(5):1351–70.CrossRef
41.
Zurück zum Zitat Tumen J, Meyrick B, Berry L Jr, Brigham KL. Antiproteinases protect cultured lung endothelial cells from endotoxin injury. J Appl Physiol (1985). 1988;65(2):835–43.CrossRef Tumen J, Meyrick B, Berry L Jr, Brigham KL. Antiproteinases protect cultured lung endothelial cells from endotoxin injury. J Appl Physiol (1985). 1988;65(2):835–43.CrossRef
42.
Zurück zum Zitat Libert C, Van Molle W, Brouckaert P, Fiers W. alpha1-Antitrypsin inhibits the lethal response to TNF in mice. J Immunol. 1996;157(11):5126–9.PubMed Libert C, Van Molle W, Brouckaert P, Fiers W. alpha1-Antitrypsin inhibits the lethal response to TNF in mice. J Immunol. 1996;157(11):5126–9.PubMed
43.
Zurück zum Zitat Van Molle W, Libert C, Fiers W, Brouckaert P. Alpha 1-acid glycoprotein and alpha 1-antitrypsin inhibit TNF-induced but not anti-Fas-induced apoptosis of hepatocytes in mice. J Immunol. 1997;159(7):3555–64.PubMed Van Molle W, Libert C, Fiers W, Brouckaert P. Alpha 1-acid glycoprotein and alpha 1-antitrypsin inhibit TNF-induced but not anti-Fas-induced apoptosis of hepatocytes in mice. J Immunol. 1997;159(7):3555–64.PubMed
44.
Zurück zum Zitat Janciauskiene S, Larsson S, Larsson P, Virtala R, Jansson L, Stevens T. Inhibition of lipopolysaccharide-mediated human monocyte activation, in vitro, by alpha1-antitrypsin. Biochem Biophys Res Commun. 2004;321(3):592–600.CrossRef Janciauskiene S, Larsson S, Larsson P, Virtala R, Jansson L, Stevens T. Inhibition of lipopolysaccharide-mediated human monocyte activation, in vitro, by alpha1-antitrypsin. Biochem Biophys Res Commun. 2004;321(3):592–600.CrossRef
45.
Zurück zum Zitat Gold M, Koczulla AR, Mengel D, Koepke J, Dodel R, Dontcheva G, Habib P, Bach JP. Reduction of glutamate-induced excitotoxicity in murine primary neurons involving calpain inhibition. J Neurol Sci. 2015;359(1–2):356–62.CrossRef Gold M, Koczulla AR, Mengel D, Koepke J, Dodel R, Dontcheva G, Habib P, Bach JP. Reduction of glutamate-induced excitotoxicity in murine primary neurons involving calpain inhibition. J Neurol Sci. 2015;359(1–2):356–62.CrossRef
46.
Zurück zum Zitat Coll RC, Robertson AA, Chae JJ, Higgins SC, Munoz-Planillo R, Inserra MC, Vetter I, Dungan LS, Monks BG, Stutz A, et al. A small-molecule inhibitor of the NLRP3 inflammasome for the treatment of inflammatory diseases. Nat Med. 2015;21(3):248–55.CrossRef Coll RC, Robertson AA, Chae JJ, Higgins SC, Munoz-Planillo R, Inserra MC, Vetter I, Dungan LS, Monks BG, Stutz A, et al. A small-molecule inhibitor of the NLRP3 inflammasome for the treatment of inflammatory diseases. Nat Med. 2015;21(3):248–55.CrossRef
47.
Zurück zum Zitat Levy M, Thaiss CA, Elinav E. Taming the inflammasome. Nat Med. 2015;21(3):213–5.CrossRef Levy M, Thaiss CA, Elinav E. Taming the inflammasome. Nat Med. 2015;21(3):213–5.CrossRef
48.
Zurück zum Zitat Ismael S, Nasoohi S, Ishrat T. MCC950, the selective inhibitor of nucleotide oligomerization domain-like receptor Protein-3 inflammasome, protects mice against traumatic brain injury. J Neurotrauma. 2018;35(11):1294–303.CrossRef Ismael S, Nasoohi S, Ishrat T. MCC950, the selective inhibitor of nucleotide oligomerization domain-like receptor Protein-3 inflammasome, protects mice against traumatic brain injury. J Neurotrauma. 2018;35(11):1294–303.CrossRef
49.
Zurück zum Zitat Perera AP, Fernando R, Shinde T, Gundamaraju R, Southam B, Sohal SS, Robertson AAB, Schroder K, Kunde D, Eri R. MCC950, a specific small molecule inhibitor of NLRP3 inflammasome attenuates colonic inflammation in spontaneous colitis mice. Sci Rep. 2018;8(1):8618.CrossRef Perera AP, Fernando R, Shinde T, Gundamaraju R, Southam B, Sohal SS, Robertson AAB, Schroder K, Kunde D, Eri R. MCC950, a specific small molecule inhibitor of NLRP3 inflammasome attenuates colonic inflammation in spontaneous colitis mice. Sci Rep. 2018;8(1):8618.CrossRef
50.
Zurück zum Zitat Dempsey C, Rubio Araiz A, Bryson KJ, Finucane O, Larkin C, Mills EL, Robertson AAB, Cooper MA, O'Neill LAJ, Lynch MA. Inhibiting the NLRP3 inflammasome with MCC950 promotes non-phlogistic clearance of amyloid-beta and cognitive function in APP/PS1 mice. Brain Behav Immun. 2017;61:306–16.CrossRef Dempsey C, Rubio Araiz A, Bryson KJ, Finucane O, Larkin C, Mills EL, Robertson AAB, Cooper MA, O'Neill LAJ, Lynch MA. Inhibiting the NLRP3 inflammasome with MCC950 promotes non-phlogistic clearance of amyloid-beta and cognitive function in APP/PS1 mice. Brain Behav Immun. 2017;61:306–16.CrossRef
51.
Zurück zum Zitat Habib P, Dang J, Slowik A, Victor M, Beyer C. Hypoxia-induced gene expression of aquaporin-4, cyclooxygenase-2 and hypoxia-inducible factor 1alpha in rat cortical astroglia is inhibited by 17beta-estradiol and progesterone. Neuroendocrinology. 2014;99(3–4):156–67.CrossRef Habib P, Dang J, Slowik A, Victor M, Beyer C. Hypoxia-induced gene expression of aquaporin-4, cyclooxygenase-2 and hypoxia-inducible factor 1alpha in rat cortical astroglia is inhibited by 17beta-estradiol and progesterone. Neuroendocrinology. 2014;99(3–4):156–67.CrossRef
52.
Zurück zum Zitat Kayed R, Head E, Thompson JL, McIntire TM, Milton SC, Cotman CW, Glabe CG. Common structure of soluble amyloid oligomers implies common mechanism of pathogenesis. Science. 2003;300(5618):486–9.CrossRef Kayed R, Head E, Thompson JL, McIntire TM, Milton SC, Cotman CW, Glabe CG. Common structure of soluble amyloid oligomers implies common mechanism of pathogenesis. Science. 2003;300(5618):486–9.CrossRef
53.
Zurück zum Zitat Habib P, Dreymueller D, Ludwig A, Beyer C, Dang J. Sex steroid hormone-mediated functional regulation of microglia-like BV-2 cells during hypoxia. J Steroid Biochem Mol Biol. 2013;138:195–205.CrossRef Habib P, Dreymueller D, Ludwig A, Beyer C, Dang J. Sex steroid hormone-mediated functional regulation of microglia-like BV-2 cells during hypoxia. J Steroid Biochem Mol Biol. 2013;138:195–205.CrossRef
54.
Zurück zum Zitat Dang J, Mitkari B, Kipp M, Beyer C. Gonadal steroids prevent cell damage and stimulate behavioral recovery after transient middle cerebral artery occlusion in male and female rats. Brain Behav Immun. 2011;25(4):715–26.CrossRef Dang J, Mitkari B, Kipp M, Beyer C. Gonadal steroids prevent cell damage and stimulate behavioral recovery after transient middle cerebral artery occlusion in male and female rats. Brain Behav Immun. 2011;25(4):715–26.CrossRef
55.
Zurück zum Zitat Johann S, Dahm M, Kipp M, Zahn U, Beyer C. Regulation of choline acetyltransferase expression by 17 beta-oestradiol in NSC-34 cells and in the spinal cord. J Neuroendocrinol. 2011;23(9):839–48.CrossRef Johann S, Dahm M, Kipp M, Zahn U, Beyer C. Regulation of choline acetyltransferase expression by 17 beta-oestradiol in NSC-34 cells and in the spinal cord. J Neuroendocrinol. 2011;23(9):839–48.CrossRef
56.
Zurück zum Zitat Heneka MT, Golenbock DT, Latz E. Innate immunity in Alzheimer’s disease. Nat Immunol. 2015;16(3):229–36.CrossRef Heneka MT, Golenbock DT, Latz E. Innate immunity in Alzheimer’s disease. Nat Immunol. 2015;16(3):229–36.CrossRef
57.
Zurück zum Zitat Acosta C, Anderson HD, Anderson CM. Astrocyte dysfunction in Alzheimer disease. J Neurosci Res. 2017;95(12):2430–47.CrossRef Acosta C, Anderson HD, Anderson CM. Astrocyte dysfunction in Alzheimer disease. J Neurosci Res. 2017;95(12):2430–47.CrossRef
58.
Zurück zum Zitat Sofroniew MV. Molecular dissection of reactive astrogliosis and glial scar formation. Trends Neurosci. 2009;32(12):638–47.CrossRef Sofroniew MV. Molecular dissection of reactive astrogliosis and glial scar formation. Trends Neurosci. 2009;32(12):638–47.CrossRef
59.
Zurück zum Zitat Youm YH, Grant RW, McCabe LR, Albarado DC, Nguyen KY, Ravussin A, Pistell P, Newman S, Carter R, Laque A, et al. Canonical Nlrp3 inflammasome links systemic low-grade inflammation to functional decline in aging. Cell Metab. 2013;18(4):519–32.CrossRef Youm YH, Grant RW, McCabe LR, Albarado DC, Nguyen KY, Ravussin A, Pistell P, Newman S, Carter R, Laque A, et al. Canonical Nlrp3 inflammasome links systemic low-grade inflammation to functional decline in aging. Cell Metab. 2013;18(4):519–32.CrossRef
60.
Zurück zum Zitat Johann S, Heitzer M, Kanagaratnam M, Goswami A, Rizo T, Weis J, Troost D, Beyer C. NLRP3 inflammasome is expressed by astrocytes in the SOD1 mouse model of ALS and in human sporadic ALS patients. Glia. 2015;63(12):2260–73.CrossRef Johann S, Heitzer M, Kanagaratnam M, Goswami A, Rizo T, Weis J, Troost D, Beyer C. NLRP3 inflammasome is expressed by astrocytes in the SOD1 mouse model of ALS and in human sporadic ALS patients. Glia. 2015;63(12):2260–73.CrossRef
61.
Zurück zum Zitat Heitzer M, Kaiser S, Kanagaratnam M, Zendedel A, Hartmann P, Beyer C, Johann S. Administration of 17beta-estradiol improves motoneuron survival and Down-regulates Inflammasome activation in male SOD1(G93A) ALS mice. Mol Neurobiol. 2017;54(10):8429–43.CrossRef Heitzer M, Kaiser S, Kanagaratnam M, Zendedel A, Hartmann P, Beyer C, Johann S. Administration of 17beta-estradiol improves motoneuron survival and Down-regulates Inflammasome activation in male SOD1(G93A) ALS mice. Mol Neurobiol. 2017;54(10):8429–43.CrossRef
62.
Zurück zum Zitat Mortezaee K, Khanlarkhani N, Beyer C, Zendedel A. Inflammasome: its role in traumatic brain and spinal cord injury. J Cell Physiol. 2017;233(7):5160–9.CrossRef Mortezaee K, Khanlarkhani N, Beyer C, Zendedel A. Inflammasome: its role in traumatic brain and spinal cord injury. J Cell Physiol. 2017;233(7):5160–9.CrossRef
63.
Zurück zum Zitat Zendedel A, Monnink F, Hassanzadeh G, Zaminy A, Ansar MM, Habib P, Slowik A, Kipp M, Beyer C. Estrogen attenuates local Inflammasome expression and activation after spinal cord injury. Mol Neurobiol. 2017;55(2):1364–75.CrossRef Zendedel A, Monnink F, Hassanzadeh G, Zaminy A, Ansar MM, Habib P, Slowik A, Kipp M, Beyer C. Estrogen attenuates local Inflammasome expression and activation after spinal cord injury. Mol Neurobiol. 2017;55(2):1364–75.CrossRef
64.
Zurück zum Zitat Debye B, Schmulling L, Zhou L, Rune G, Beyer C, Johann S. Neurodegeneration and NLRP3 inflammasome expression in the anterior thalamus of SOD1(G93A) ALS mice. Brain Pathol. 2016;28(1):14–27.CrossRef Debye B, Schmulling L, Zhou L, Rune G, Beyer C, Johann S. Neurodegeneration and NLRP3 inflammasome expression in the anterior thalamus of SOD1(G93A) ALS mice. Brain Pathol. 2016;28(1):14–27.CrossRef
65.
Zurück zum Zitat Bauernfeind FG, Horvath G, Stutz A, Alnemri ES, MacDonald K, Speert D, Fernandes-Alnemri T, Wu J, Monks BG, Fitzgerald KA, et al. Cutting edge: NF-kappaB activating pattern recognition and cytokine receptors license NLRP3 inflammasome activation by regulating NLRP3 expression. J Immunol. 2009;183(2):787–91.CrossRef Bauernfeind FG, Horvath G, Stutz A, Alnemri ES, MacDonald K, Speert D, Fernandes-Alnemri T, Wu J, Monks BG, Fitzgerald KA, et al. Cutting edge: NF-kappaB activating pattern recognition and cytokine receptors license NLRP3 inflammasome activation by regulating NLRP3 expression. J Immunol. 2009;183(2):787–91.CrossRef
66.
Zurück zum Zitat Lee GS, Subramanian N, Kim AI, Aksentijevich I, Goldbach-Mansky R, Sacks DB, Germain RN, Kastner DL, Chae JJ. The calcium-sensing receptor regulates the NLRP3 inflammasome through Ca2+ and cAMP. Nature. 2012;492(7427):123–7.CrossRef Lee GS, Subramanian N, Kim AI, Aksentijevich I, Goldbach-Mansky R, Sacks DB, Germain RN, Kastner DL, Chae JJ. The calcium-sensing receptor regulates the NLRP3 inflammasome through Ca2+ and cAMP. Nature. 2012;492(7427):123–7.CrossRef
67.
Zurück zum Zitat Toldo S, Seropian IM, Mezzaroma E, Van Tassell BW, Salloum FN, Lewis EC, Voelkel N, Dinarello CA, Abbate A. Alpha-1 antitrypsin inhibits caspase-1 and protects from acute myocardial ischemia-reperfusion injury. J Mol Cell Cardiol. 2011;51(2):244–51.CrossRef Toldo S, Seropian IM, Mezzaroma E, Van Tassell BW, Salloum FN, Lewis EC, Voelkel N, Dinarello CA, Abbate A. Alpha-1 antitrypsin inhibits caspase-1 and protects from acute myocardial ischemia-reperfusion injury. J Mol Cell Cardiol. 2011;51(2):244–51.CrossRef
68.
Zurück zum Zitat Aggarwal N, Korenbaum E, Mahadeva R, Immenschuh S, Grau V, Dinarello CA, Welte T, Janciauskiene S. alpha-Linoleic acid enhances the capacity of alpha-1 antitrypsin to inhibit lipopolysaccharide-induced IL-1beta in human blood neutrophils. Mol Med. 2016;22:680–93. Aggarwal N, Korenbaum E, Mahadeva R, Immenschuh S, Grau V, Dinarello CA, Welte T, Janciauskiene S. alpha-Linoleic acid enhances the capacity of alpha-1 antitrypsin to inhibit lipopolysaccharide-induced IL-1beta in human blood neutrophils. Mol Med. 2016;22:680–93.
Metadaten
Titel
α1-antitrypsin mitigates NLRP3-inflammasome activation in amyloid β1–42-stimulated murine astrocytes
verfasst von
Taraneh Ebrahimi
Marcus Rust
Sarah Nele Kaiser
Alexander Slowik
Cordian Beyer
Andreas Rembert Koczulla
Jörg B. Schulz
Pardes Habib
Jan Philipp Bach
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/s12974-018-1319-x

Weitere Artikel der Ausgabe 1/2018

Journal of Neuroinflammation 1/2018 Zur Ausgabe