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

Open Access 01.12.2015 | Research

P2X7R blockade prevents NLRP3 inflammasome activation and brain injury in a rat model of intracerebral hemorrhage: involvement of peroxynitrite

verfasst von: Liang Feng, Yizhao Chen, Rui Ding, Zhenghao Fu, Shuo Yang, Xinqing Deng, Jun Zeng

Erschienen in: Journal of Neuroinflammation | Ausgabe 1/2015

Abstract

Background

The NLR family, pyrin domain-containing 3 (NLRP3) inflammasome plays a key role in intracerebral hemorrhage (ICH)-induced inflammatory injury, and the purinergic 2X7 receptor (P2X7R) is upstream of NLRP3 activation. This study aimed to investigate how P2X7R functions in ICH-induced inflammatory injury and how the receptor interacts with the NLRP3 inflammasome.

Methods

Rats were treated with P2X7R small interfering RNA (siRNA) 24 h before undergoing collagenase-induced ICH. A selective P2X7R inhibitor (blue brilliant G, BBG) or a peroxynitrite (ONOO) decomposition catalyst (5,10,15,20-tetrakis(4-sulfonatophenyl)porphyrinato iron(III) [FeTPPS]) was injected 30 min after ICH. Brain water content, hemorrhagic lesion volume, and neurological deficits were evaluated, and western blot, immunofluorescence, and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) were carried out.

Results

Striatal P2X7R and NLRP3 inflammasomes were activated after ICH. Gene silencing of P2X7R suppressed NLRP3 inflammasome activation and interleukin (IL)-1β/IL-18 release and significantly ameliorated brain edema and neurological deficits. Additionally, enhanced NADPH oxidase 2 (NOX2, gp91phox) and inducible nitric oxide synthase (iNOS), as well as their cytotoxic product (ONOO) were markedly attenuated by BBG treatment following ICH. This was accompanied by downregulations of the inflammasome components, IL-1β/IL-18 and myeloperoxidase (MPO, a neutrophil marker). Most importantly, inflammasome activation and IL-1β/IL-18 release were significantly inhibited by ONOO decomposition with FeTPPS.

Conclusions

Our findings implicate that P2X7R exacerbated inflammatory progression and brain damage in ICH rats possibly via NLRP3 inflammasome-dependent IL-1β/IL-18 release and neutrophil infiltration. ONOO, a potential downstream signaling molecule of P2X7R, may play a critical role in triggering NLRP3 inflammasome activation.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s12974-015-0409-2) contains supplementary material, which is available to authorized users.
An erratum to this article can be found at http://​dx.​doi.​org/​10.​1186/​s12974-016-0627-2.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

Conceived and designed the experiments: YZC and LF. Performed the experiments: LF. Analyzed the data: YZC and LF. Wrote the paper: LF. Paper revision: YZC. RD, ZHF, SY, XQD, and JZ provided experimental technical support and assisted in completing the study at different stages. All authors read and approved the final manuscript.
Abkürzungen
3-NT
3-nitrotyrosine
ASC
apoptosis-associated speck-like protein containing a CARD
ATP
adenosine triphosphate
BBG
brilliant blue G
CNS
central nervous system
ICH
intracerebral hemorrhage
IL
interleukin
iNOS
inducible nitric oxide synthase
LPS
lipopolysaccharide
mNSS
modified Neurological Severity Score
MPO
myeloperoxidase
mtDNA
mitochondrial DNA
NLRP3
NLR family, pyrin domain-containing 3
NO
nitric oxide
NOX2
NADPH oxidase 2
O2
superoxide anion
ONOO
peroxynitrite
P2X7R
purinergic 2X7 receptor
RNS
reactive nitrogen species
ROS
reactive oxygen species
SD
Sprague–Dawley
Sham
sham-operated animals
siRNA
small interfering RNA
TUNEL
terminal deoxynucleotidyl transferase-mediated dUTP nick 3′-end labeling

Background

Spontaneous intracerebral hemorrhage (ICH) is a devastating stroke subtype, with high morbidity and mortality [1]. Unfortunately, no satisfactory pharmacologic treatments have been found for clinical practice, mainly due to a lack of knowledge underlying the mechanisms of post-ICH brain damage. There is an urgent need to clarify the pathophysiology of this disease to identify effective therapies.
Accumulating evidence suggests that innate immunity and inflammatory responses are involved in ICH-induced secondary brain injury [13]. The intracellular Nod-like receptors have recently been shown to play a critical role in the process of innate immunity and inflammatory responses [4]. The NLR family, pyrin domain-containing 3 (NLRP3) inflammasome, the best characterized member of Nod-like receptor family, is a multiprotein complex that contains the adaptor protein apoptosis-associated speck-like protein containing a CARD (ASC) and the effector caspase-1. Once activated, caspase-1 can cleave the pro forms of interleukin (IL)-1β and IL-18 into their mature and active forms, which leads to the recruitment and activation of other immune cells, such as neutrophils [5]. In this regard, evidence indicates that the NLRP3 inflammasome plays a pivotal role in ICH [2] and other central nervous system (CNS) conditions [611], but the precise mechanisms associated with inflammasome activation continue to be debated.
The role of ATP-gated transmembrane cation channel P2X7R in the signaling cascade has received particular attention due to its widespread involvement as a key regulatory element of NLRP3 inflammasome activation [12]. A growing number of studies have demonstrated the important pathophysiological functions of P2X7R in CNS disorders, including ischemic stroke, subarachnoid hemorrhage, neurotrauma, epilepsy, neuropathic pain, and neurodegenerative illnesses [13, 14]. However, the specific role of P2X7R in ICH has not yet been established, and the interaction between P2X7R and the NLRP3 inflammasome in the development of ICH-induced brain injury remains unclear.
In the case of ICH, both NADPH oxidase 2 (NOX2) and inducible nitric oxide synthase (iNOS) have been reported to contribute to brain injury; knockout mice exhibit less brain edema and cell death than wild-type controls following ICH [15, 16]. Superoxide anion (O2 ) and nitric oxide (NO), released through NOX and iNOS in activated microglia, act as devastating pro-inflammatory mediators in CNS diseases [17]. More importantly, peroxynitrite (ONOO), the product of a diffusion-controlled reaction of NO with O2 , is a more potent oxidant species and is involved in the pathologies of ischemic stroke, neurotrauma, and neurodegenerative diseases [1820]. We previously demonstrated that abundant ONOO formed in a hemoglobin (Hb)-induced ICH rat model [21], but the exact mechanisms of ONOO in brain injury after ICH have not been fully characterized. Besides its ability to oxidize or nitrate proteins, lipids, and DNA, ONOO can also lead to destructive pathological consequences by triggering the activation of several biochemical pathways engaged in the development of neuroinflammation and IL-1β production [2224]. However, the precise link between ONOO formation and IL-1β secretion in ICH is unclear.
The P2X7R acts as an upstream molecule of NOX2 activation signaling in many in vivo and in vitro disease models [2527]. NOX2-mediated oxidative stress was recently proposed to be responsible for activation of the NLRP3 inflammasome and subsequent neurovascular damage in ischemic stroke [7]. Notably, P2X7R-dependent NADPH oxidase activation and ONOO formation play key roles in caspase-1 and IL-1β processing in endotoxin-primed human monocytes [28]. In an animal model of lipopolysaccharide (LPS)-induced striatum injury, activated P2X7R in microglia was associated with increased iNOS and 3-nitrotyrosine (3-NT, a reliable marker of ONOO), and this was reversed by the P2X7R antagonist oxidized ATP (oxATP) [29]. Despite this knowledge, the potential roles of P2X7R and NLRP3 inflammasomes and NOX2/iNOS-dependent ONOO formation in the development of ICH-induced brain damage remain to be clarified.
We hypothesized that ONOO, formed from NOX2-derived O2 and iNOS-derived NO, may be involved in transducing P2X7R-mediated IL-1β/IL-18 production and brain injury via NLRP3 inflammasome activation after ICH. We first investigated the expression profiles of P2X7R and the NLRP3 inflammasome components. Next, a mixed small interfering (si) RNA was applied to knock down P2X7R in vivo, and alterations in NLRP3 inflammasome components and functional outcomes were measured. We then explored the therapeutic effect of the selective P2X7R antagonist, blue brilliant G (BBG). Additionally, we observed iNOS- and NOX2-dependent formation of ONOO and their alterations in ICH rats following BBG treatment. Finally, to determine whether ONOO is involved in P2X7R-mediated NLRP3 inflammasome activation, we used an ONOO decomposition catalyst in vivo and measured the expression levels of P2X7R and NLRP3 inflammasome components.

Materials and methods

Animals

Sprague–Dawley (SD) male rats weighing 280–320 g were purchased from the Animal Experiment Center of Southern Medical University (Guangzhou, China). All experimental procedures and animal care were approved by the Southern Medical University Ethics Committee.

ICH model

After anesthetization (0.3 ml/100 g, 10 % chloral hydrate, Sigma-Aldrich, St. Louis, MO, USA), an incision was made on the skin along the sagittal midline to expose the skull. A burr hole (1 mm) was drilled 3 mm lateral and 1 mm anterior to the bregma, then a 30-gauge needle was inserted through the burr hole into the striatum (6 mm ventral from the skull surface), and ICH was induced by stereotaxic infusion of bacterial collagenase VII-S (0.25U in 1.0 μl sterile saline, Sigma-Aldrich) over a 10-min period. In the Sham group, rats were subjected to only a needle insertion as described above. The needle was kept in situ for another 10 min to prevent backflow and then slowly removed. The craniotomies were sealed with bone wax. Rats were allowed to recover in separate cages with free access to food and water.

Experimental protocol

Four separate experiments were conducted as shown in Additional file 1.

Experiment 1

Thirty-six rats were divided into six groups (Sham, and 6, 12, 24, 48, and 72 h after ICH). The expression levels of P2X7R, NLRP3, ASC, and caspase-1 were detected by western blot. The tissue for immunofluorescence (IF) was collected 24 h after ICH induction.

Experiment 2

Eighty-eight rats were randomized into four groups: Sham, Vehicle (ICH + saline, intracerebroventricular injection), Scramble small interfering RNA (siRNA) (1000 pmol, 2 μl, ICH + scramble siRNA), and P2X7R siRNA (1000 pmol, 2 μl, ICH + P2X7R siRNA). siRNA silencing efficacy was assessed by western blot. Brain water content and modified Neurological Severity Score (mNSS) were also measured.

Experiment 3

One hundred and thirty-two rats were randomized into four groups: Sham, Vehicle (ICH + saline, intraperitoneal injection), BBG (50 mg/kg), and BBG (100 mg/kg). For the 72-h study, BBG was administered daily by intraperitoneal injection. Western blot, hematoxylin and eosin (H&E) staining, immunofluorescence (IF), and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) were measured 24 h after ICH induction; mNSS and brain water content were detected at both 24 and 72 h.

Experiment 4

Thirty-three rats were randomized into three groups: Sham, Vehicle (ICH + saline, intraperitoneal injection), and FeTPPS (30 mg/kg). Western blotting was performed 24 h after ICH induction.

siRNA and drug delivery

BBG (Sigma-Aldrich) was diluted at 50 and 100 mg/kg in Vehicle (saline) solution. Rats were treated intraperitoneally with either BBG or Vehicle immediately after ICH induction and at 12, 36, and 60 h.
For in vivo siRNA administration, P2X7R siRNA or non-silencing RNA (Sigma-Aldrich) was applied 24 h before ICH by intracerebroventricular injection as previously described [2]. A cranial burr hole (1 mm) was drilled, and a 30-gauge needle was inserted stereotaxically into the right lateral ventricle. To improve the gene silence efficiency, two different sequences targeting P2X7R siRNA were combined (a)sense:5′-CAGUGAAUGAGUACUACUA-3′; antisense:5′-UAGUAGUACUCAUUCACUG-3′; (b)sense:5′-CUCUUGAGGAGCGCCGAAA-3′;antisense:5′-UUUCGGCGCUCCUCAAGAG-3′. siRNA was dissolved in RNA free water. Scrambled control siRNA (1000 pmol, 2 μl), P2X7R siRNA (1000 pmol, 2 μl), or RNA free water (2 μl) was delivered intracerebroventricularly for 2 min. The needle was left in place for an additional 10 min after injection and then slowly withdrawn.
FeTPPS (Millipore, Billerica, MA, USA) was diluted to 30 mg/kg in Vehicle (saline) solution. Rats were treated intraperitoneally with either FeTPPS or Vehicle immediately and at 12 h after ICH induction. The FeTPPS dose was selected based on our previous reports that showed that it is effectively protected against injury [30].

RT-PCR

Rats were anesthetized and decapitated. Lesioned tissues (about 40 mg) were obtained, and total RNA was extracted from the tissue with GeneJET™ RNA Purification Kit (Thermo Fisher Scientific Inc., Waltham, MA, USA). RNA (1 μg) was reverse-transcribed to cDNA with high capacity (Life Technologies, Carlsbad, CA, USA). RT-PCR was performed in an ABI Prism 7500 sequence detection system (Applied Biosystems, Foster City, CA, USA) using specific primers designed from known sequences. GAPDH was used as an endogenous control gene. Sequence-specific primers for P2X7R, NLRP3, and GADPH were as follows:
P2X7R, 5′-CTACTCTTCGGTGGGGGCTT-3′
(forward primer),
P2X7R, 5′-CTCTGGATCCGGGTGACTTT-3′
(reverse primer);
NLRP3, 5′-CTGCATGCCGTATCTGGTTG-3′
(forward primer),
NLRP3, 5′-GCTGAGCAAGCTAAAGGCTTC-3′
(reverse primer);
GAPDH, 5′-AGACAGCCGCATCTTCTTGT-3′
(forward primer),
GAPDH, 5′- TGATGGCAACAATGTCCACT-3’
(reverse primer);

Western blot

Western blotting was performed as described previously [30]. The following primary antibodies were used: rabbit polyclonal anti-P2X7R (1:1000, Alomone Labs, Jerusalem, Israel), rabbit polyclonal anti-NLRP3 antibody (1:1000, Santa Cruz, Biotechnology, Santa Cruz, CA, USA), rabbit polyclonal anti-ASC antibody (1:500, Abclonal, Cambridge, MA, USA), mouse monoclonal anti-caspase-1 p20 antibody (1:1000, Santa Cruz Biotechnology), rabbit polyclonal anti-IL-1β antibody (1:1000, Millipore), rabbit monoclonal anti-IL-18 antibody (1:1000, Abcam, Cambridge, UK), mouse monoclonal anti-nitrotyrosine antibody (1:1000, Abcam), mouse monoclonal anti-iNOS antibody (1:200, Santa Cruz Biotechnology), mouse monoclonal anti-gp91phox antibody (1:2000, BD Transduction Laboratories, San Jose, CA, USA), and rabbit polyclonal anti-myeloperoxidase antibody (MPO, 1:500, Abcam). GAPDH (1:1000, Cell Signaling Technology, Danvers, MA, USA) was employed as the loading control. Blot bands were quantified by densitometry with ImageJ software (National Institutes of Health, Baltimore, MD, USA).

Paraffin section preparations

The sections were processed as previously described [30] with minor modifications. After anesthetization, rats were transcardially perfused with 200 ml saline followed by 400 ml 4 % paraformaldehyde solution. Brain tissues were then removed and fixed by immersion in the same solution at 4 °C for 24 h. After dehydration and vitrification, they were embedded in paraffin, and 3-μm sections were prepared. Sections were dewaxed, rehydrated, and then processed for IF and TUNEL.

Histological examination

Coronal sections (1 mm apart) [31] were prepared accordingly and then stained with H&E. Hemorrhagic volumes were calculated using Image Pro Plus 6.0 software (Media Cybernetics, USA) to span the entire hematoma [32].

IF

Antigen retrieval was performed by heat treatment in a microwave oven for 21 min in Tris–EDTA buffer solution (0.05 mol/L Tris, 0.001 mol/L EDTA; pH 8.5). Sections were incubated for 30 min in 5 % bovine serum albumin (BSA) and then incubated at 4 °C overnight with primary antibodies (rabbit polyclonal anti-P2X7R, 1:500, Alomone labs; mouse monoclonal anti-3-Nitrotyrosine, 1:400, Abcam; rabbit polyclonal anti-nitrotyrosine antibody, 1:200, Millipore; rabbit polyclonal anti-MPO antibody, 1:50, Abcam; rabbit polyclonal anti-iNOS antibody, 1:40, Santa Cruz Biotechnology; rabbit polyclonal anti-Iba-1 antibody, 1:600, WAKO, Osaka, Japan; goat polyclonal anti-Iba-1 antibody, 1:300, Abcam; mouse monoclonal gp91phox antibody, 1:400, BD Transduction Laboratories). For double-staining experiments, primary antibodies were separately incubated overnight at 4 °C. After they were washed with phosphate-buffered saline (PBS), sections were then incubated with secondary antibodies. Images were obtained using confocal microscopes (FV10i-W, Olympus, Tokyo, Japan; LSM780, Zeiss, Oberkochen, Germany).

TUNEL

At 24 h after ICH, TUNEL staining was performed with an in situ apoptosis detection kit (Roche, Basel, Switzerland) according to the manufacturer’s instruction. For NeuN and TUNEL co-staining, the sections were first labeled with a NeuN antibody (1:400, Abcam), followed by TUNEL. The slides were analyzed using a fluorescence microscope (Bx51, Olympus).

Brain water content measurement

Brain edema was evaluated by a common wet/dry method as previously described [33]. Briefly, at 24 or 72 h post-ICH, rats were anesthetized and decapitated. The brains were removed and immediately separated into contralateral and ipsilateral hemispheres and the cerebellum and wet weighed. The cerebellum was used as an internal control. Brain specimens were dried in an oven at 100 °C for 24 h to obtain the dry weight. The water content was expressed as a percentage of the wet weight: ([wet weight] – [dry weight]) / (wet weight) × 100 %.

Behavioral testing

Behavioral tests were assessed with mNSS at 24 and 72 h after ICH by an investigator who was blinded to the experimental groups [34].

Statistical analysis

Data are shown as mean ± SD. Statistical analysis was performed using SPSS 13.0 (SPSS Inc., Chicago, IL, USA). Comparison between groups was determined by Student’s t tests or one-way analysis of variance (ANOVA) followed by least significant difference (LSD) tests with multiple comparisons. The statistically significant level was P < 0.05.

Results

P2X7R was increased and mainly expressed in microglia cells following ICH

Protein content was analyzed at different time points after injection to investigate whether P2X7R would respond to collagenase-induced ICH. As shown by western blot (Fig. 1a, b), P2X7R levels were significantly elevated at 6 h after ICH (P < 0.05 vs. Sham) and peaked at around 24 h (P < 0.01 vs. Sham) when P2X7R levels were nearly 4.5 times more than those in the Sham group. Following the peak, P2X7R levels decreased, returning close to baseline levels at 72 h.
Double immunolabeling was performed to identify the cell type that expresses P2X7R. The results showed that P2X7R was predominantly expressed in microglia cells (Fig. 1c) and not in other cell types, such as astrocytes or neurons (Additional file 2).

NLRP3, ASC, and caspase-1 were upregulated after ICH

NLRP3 inflammasome has been proposed to be downstream of P2X7R [12]. We evaluated the expressions of NLRP3 inflammasome components by western blot (Fig. 2a). NLRP3 (Fig. 2b), ASC (Fig. 2c), and cleaved caspase-1 (Fig. 2d) were significantly upregulated at 6 h (P < 0.05 vs. Sham) and reached their peak at 24 h post-ICH (P < 0.01 vs. Sham). Following this peak, levels of all three proteins declined but still remained higher than those in the Sham group at 48 h (P < 0.05 vs. Sham) and 72 h (P < 0.05 vs. Sham).

P2X7R RNA interference reduced brain water content and improved neurological outcomes

We next explored whether P2X7R is involved in brain injury following ICH. Two P2X7R siRNA mixtures were applied 24 h before ICH induction. Silencing efficacy by RT-PCR demonstrated a significant inhibitory effect of P2X7R siRNA on its mRNA levels (P < 0.01) (Fig. 3a). Western blot (Fig. 3b) showed 41.3 and 40.7 % reductions of P2X7R in the P2X7R siRNA group compared with the Vehicle and Scramble siRNA groups, respectively (both P < 0.05), at 24 h after ICH. Brain water content in the ipsilateral hemisphere was significantly increased in the Vehicle (82.56 ± 0.72 % vs. Sham, 79.40 ± 0.44 %, P < 0.01) and Scramble siRNA (82.44 ± 0.75 % vs. Sham, 79.40 ± 0.44 %, P < 0.01) groups at 24 h post-ICH, while that in P2X7R siRNA group was decreased to 81.39 ± 0.58 % (P < 0.05 vs. Vehicle or Scramble siRNA) (Fig. 3c). Consistent with the brain edema results, P2X7R siRNA administration significantly ameliorated neurological deficits at 24 h (8.66 ± 1.15 vs. Vehicle, 10.00 ± 1.95, P < 0.05; vs. Scramble siRNA, 10.50 ± 1.26, P < 0.05) and 72 h (6.33 ± 0.81 vs. Vehicle, 8.10 ± 1.96, P < 0.05; vs. Scramble siRNA, 8.16 ± 1.16, P < 0.05) post-ICH (Fig. 3d).

P2X7R RNA interference inhibited NLRP3 inflammasome activation and subsequent IL-1β/IL-18 release

NLRP3 inflammasome is actively involved in brain injury after ICH [2]. We further clarify the role of P2X7R in NLRP3/ASC/caspase-1 activation and the subsequent processing of IL-1β/IL-18. P2X7R siRNA treatment significantly reduced NLRP3 mRNA expression (P < 0.01) (Fig. 4a). The protein levels of NLRP3 inflammasome component and IL-1β/IL-18 production were evidently elevated in the Vehicle and Scramble siRNA groups at 24 h after ICH (P < 0.01). P2X7R siRNA treatment significantly suppressed caspase-1 activation and the subsequent secretion of mature IL-1β/IL-18 (P < 0.05) (Fig. 4bf).

BBG deceased post-ICH neurological deficits, brain water content, and neuronal apoptosis

Next, we investigated the effects of the selective P2X7R inhibitor, BBG. Both doses (50 and 100 mg/kg) significantly attenuated brain water content at 24 h (50 mg/kg, 81.76 ± 0.32 % vs. Vehicle, 82.54 ± 0.66 %, P < 0.05; 100 mg/kg, 81.67 ± 0.43 % vs. Vehicle, 82.54 ± 0.66 %, P < 0.05) and 72 h (50 mg/kg, 81.59 ± 1.15 % vs. Vehicle, 82.94 ± 1.00 %, P < 0.05; 100 mg/kg, 81.74 ± 1.12 % vs. Vehicle, 82.94 ± 1.00 %, P < 0.05) after ICH (Fig. 5d, h). The hemorrhagic lesion volume at 24 h post-ICH for the Vehicle and BBG groups were 91.17 ± 23.54 and 82.77 ± 21.31, respectively (P > 0.05) (Fig. 5a, b), indicating that BBG did not affect bleeding. However, the hemorrhagic lesion volume for the BBG group (37.79 + 15.56) was significantly decreased compared with the Vehicle (73.03 + 19.34) group at 72 h post-ICH (P < 0.01) (Fig. 5e, f), indicating that BBG could promote tissue reconstruction. Meanwhile, the tissue damage around the lesion site was evidently mitigated by BBG treatment at both 24 and 72 h (Fig. 5c, d, g, h). Consistently, neurological deficits (Fig. 6c) at both 24 h (50 mg/kg, 8.83 ± 1.64 vs. Vehicle, 10.00 ± 1.95, P < 0.05; 100 mg/kg, 8.66 ± 1.55 vs. Vehicle, 10.00 ± 1.95, P < 0.05) and 72 h (50 mg/kg, 6.40 ± 1.64 vs. Vehicle, 8.10 ± 1.96, P < 0.05; 100 mg/kg, 7.00 ± 1.78 vs. Vehicle, 8.10 ± 1.96, P < 0.05) after ICH were improved by BBG treatment. However, there was no difference between animals that received 50 and 100 mg/kg doses of BBG treatment with regard to mNSS scores or brain water contents. Thus, the 50 mg/kg dose was applied in further studies.
The number of apoptotic neurons was significantly increased at 24 h after ICH compared with the Sham group (P < 0.01), and BBG treatment significantly reduced the number of apoptotic neurons (P < 0.01) relative to the Vehicle group (Fig. 6a, b).

BBG decreased P2X7R expression, NLRP3/ASC/caspase-1 activation, and subsequent IL-1β/IL-18 production following ICH

BBG treatment significantly reduced NLRP3 mRNA levels (Fig. 7a). Western blot analysis revealed that BBG (50 mg/kg) treatment attenuated the expressions of P2X7R (P < 0.05 vs. Vehicle), NLRP3 (P < 0.05 vs. Vehicle), ASC (P < 0.01 vs. Vehicle), and cleaved caspase-1 (P < 0.05 vs. Vehicle). Furthermore, the levels of mature IL-1β (P < 0.05 vs. Vehicle) and IL-18 (P < 0.05 vs. Vehicle) were distinctly reduced after BBG treatment (Fig. 7b–h).

BBG reduced neutrophils infiltration after ICH.

We detected MPO levels in brain tissue by IF (Fig. 8a) and western blot (Fig. 8b, c) at 24 h following ICH to determine the effect of P2X7R/NLRP3 inflammasome axis activation on neutrophil infiltration. Striatal MPO levels were evidently increased following ICH compared with the Sham group (P < 0.01 vs. Sham). BBG (50 mg/kg) significantly suppressed MPO expression compared to the Vehicle group (P < 0.05 vs. Vehicle).

BBG suppressed ICH-induced iNOS expression

iNOS is upregulated in both blood infusion and collagenase-induced ICH rat models [31, 35]. The role of P2X7R in iNOS signaling was next investigated using IF and western blot analysis. iNOS expression was weak in Sham-operated rats but was dramatically elevated 24 h after ICH induction (P < 0.01 vs. Sham). To further trace the source of iNOS, double IF was performed and found that iNOS was mainly expressed in Iba-1-positive microglia (Fig. 9a). Western blots and IF showed that the enhanced iNOS levels were markedly attenuated in BBG-treated rats (P < 0.05 vs. Vehicle) (Fig. 9bd).

BBG reduced ICH-induced NOX2 expression

NOX2, a primary source of O2 , is actively involved in ICH-induced brain injury [15]. We studied the expression of gp91phox, a membrane subunit of NOX2, by IF and western blotting. Double IF showed that gp91phox was also mainly expressed in Iba-1-positive areas (Fig. 10a) and most overlapped with iNOS expression (Fig. 10b), implying a close connection between them. Consistent with iNOS, gp91phox was significantly increased after ICH in ipsilateral hemisphere brain tissues as compared with the Sham group (P < 0.01 vs. Sham) at 24 h after ICH. BBG treatment significantly downregulated gp91phox overexpression (P < 0.05 vs. Vehicle) (Fig. 10ce).

BBG attenuated peroxynitrite formation following ICH

The enhancement of iNOS and NOX2 after ICH induction prompted us to examine the involvement of P2X7R in ONOO formation. Double IF demonstrated a high degree of colocalization with Iba-1 (Fig. 11a). Moreover, 3-NT and gp91phox expressions were almost completely overlapped (Fig. 11b), implying that peroxynitrite production is NOX2 dependent. However, BBG treatment significantly downregulated 3-NT overexpression (P < 0.05 vs. Vehicle) (Fig. 11c–e).

The ONOO decomposition catalyst FeTPPS inhibited NLRP3/ASC/Caspase-1 activation and subsequent production of mature IL-1β/IL-18 following ICH

Our findings suggested a pivotal role for microglia-expressed P2X7R in mediating ONOO formation in an iNOS and NOX2-dependent way, which further prompted us to explore whether ONOO served as the key bridge between P2X7R and NLRP3 inflammasome activation. To answer this question, the ONOO decomposition catalyst FeTPPS was applied in vivo. Firstly, FeTPPS significantly reduced 3-NT levels (P < 0.01 vs. Vehicle) on western blot (Fig. 12a, b). Thereafter, the expression levels of P2X7R and NLRP3 inflammasome components were measured by western blot at 24 h following ICH. The results indicated that FeTPPS significantly downregulated the enhanced levels of NLRP3 (P < 0.05 vs. Vehicle), ASC (P < 0.05 vs. Vehicle), and cleaved caspase-1 (P < 0.05 vs. Vehicle) after ICH. Moreover, the upregulation of IL-1β/IL-18 was also attenuated by FeTPPS (P < 0.05 vs. Vehicle) (Fig. 12ch). However, FeTPPS had no influence on P2X7R expressions (Additional file 3). FeTPPS treatment significantly reduced NLRP3 mRNA expression. Together, these results reveal that P2X7R-dependent synthesis of ONOO may be a key activator of the NLRP3 inflammasome.

Discussion

Innate immune and inflammatory responses are increasingly recognized as important factors in the pathophysiology of secondary brain injury following ICH. The NLRP3 inflammasome, the most characterized pattern recognition receptor (PRR) in innate immune response initiation, is strongly involved in ICH-induced inflammation and produces pro-inflammatory factors such as IL-1β [2].
Several distinct mechanisms have been proposed to account for NLRP3 activation, including potassium (K+) efflux, intracellular calcium alteration, ubiquitination, and reactive oxygen species (ROS) generation [36]. It was recently reported that extracellular ATP-induced P2X7R activation could directly mediate K+ efflux through the hemichannel pannexin 1 to activate the NLRP3 inflammasome [5, 28, 36]. However, pannexin 1-deficient mice do not show diminished caspase-1 activation [37], implying that pannexin-1 is dispensable for the assembly of caspase-1 activating inflammasome complexes. This raises the hypothesis that other signaling pathways may be involved in P2X7R-mediated NLRP3 inflammasome activation or that something besides pannexin 1 is necessary for P2X7R-dependent K+ efflux. NOX2-derived ROS after ATP release-mediated P2X7R activation is well established in a growing body of in vitro and in vivo disease models [2325]. Importantly, there are a number of reports supporting that high ROS levels, particularly NOX2-derived ROS, are critical for NLRP3 inflammasome activation [36]. RNS also play an important role as evidenced by the fact that both an ONOO scavenger and NOX2 inhibitor suppressed nigericin-induced caspase-1 activation and IL-1β secretion in human monocytes [28]. Additionally, K+ efflux can be positively regulated by ROS [14] and RNS (e.g., ONOO) [38]. In this regard, we propose that NOX2-mediated ONOO formation may link P2X7R and NLRP3 inflammasome activation.
P2X7R activation in the ICH brain is a novel finding of this study. ATP is released in large quantities following any kind of cell injury [39], and cell death occurs 3 to 6 h after collagenase- induced ICH [40]. Elevated extracellular ATP is necessary for P2X7R activation [13]. In the present study, increased P2X7R was observed as early as 6 h after ICH in the perihematomal tissue along with expression of the microglia marker Iba-1, suggesting that P2X7R may act as a signal in the process of microglia activation as previously reported [41].
Microglia are believed to be one of the first non-neuronal cells to respond to brain injury [42], much earlier than neutrophil invasion [43]. Activated microglia then release pro-inflammatory cytokines, such as IL-1β [44] and IL-18 [45], which recruit leukocytes especially neutrophils. The infiltrated neutrophils amplify neuroinflammation by releasing and expressing neurotoxic factors or even by stimulating microglia to secrete neurotoxic factors, creating a vicious cycle [46]. In the present study, upregulated P2X7R was closely associated with increased NLRP3/ASC/caspase-1 inflammasome expression; siRNA interference or pharmacological blockage of P2X7R impaired inflammasome activation and IL-1β/IL-18 secretion, which had a pronounced neuroprotective effect. Moreover, infiltrated neutrophils were also diminished in the BBG group compared with the Vehicle group. These results indicate that P2X7R may be responsible for ICH-induced inflammation, possibly by NLRP3 inflammasome-dependent IL-1β/IL-18 release and subsequent neutrophil infiltration signals. In accordance with our results, Ma and colleagues [2] found that NLRP3 mainly colocalized with microglia but not with other cell types, providing further support for a tight relationship between P2X7R and NLRP3.
In response to microglia activation, large amounts O2 and NO are released through activated NADPH oxidase and iNOS [17], both of which play pivotal roles in ICH-induced brain damage [15, 16]. In our study, enhanced gp91phox in microglia largely colocalized with iNOS, prompting us to consider ONOO. Double IF revealed that increased 3-NT was also expressed in microglia and mostly overlapped with gp91phox. Consistently with this, Mander and Brown [47] reported that activation of NOX or iNOS alone was relatively harmless, but their simultaneous activation was lethal because it spurred ONOO production. Inhibition of either iNOS [48] or NOX2 [49] significantly reduced ONOO production in CNS disease models. Collectively, we can infer that NOX2-derived O2 and iNOS-derived NO may contribute to ONOO formation upon microglia activation after ICH.
We also found that ICH-induced upregulations of NOX2, iNOS, and ONOO were diminished in BBG treated ICH rats, indicating that they may be downstream of P2X7R as previously reported [26, 29]. We next explored the role of ONOO in NLRP3 inflammasome activation. As a highly active and relatively specific ONOO decomposition catalyst, FeTPPS exerts neuroprotective effects in many CNS disease models [20, 50] because it catalyzes peroxynitrite to become a harmless nitrate [51]. Our results show that overexpression of NLRP3 inflammasome components and mature IL-1β/IL-18 was reduced by FeTPPS. These findings suggest that ONOO may be involved in inflammasome activation following ICH. Consistent with our results, an early report found that FeTPPS inhibited nigericin-induced caspase-1 activation and IL-1β secretion in human monocytes [28].
Although we did not attempt to explore the exact mechanisms of ONOO in modulating NLRP3 inflammasome activation, ONOO may act as a key mediator for inflammasome activation through several mechanisms. Firstly, direct oxidation of mitochondria and the release of mitochondrial DNA (mtDNA) activate the NLRP3 inflammasome; a recent study showed that oxidized mtDNA released into the cytosol from injured mitochondria could bind to and activate the NLRP3 inflammasome [52]. ONOO is a potent ROS with strong abilities to oxidize and nitrate proteins, lipids, and DNA. Mitochondria are a primary target for peroxynitrite [18]. Secondly, nitration can inactivate thioredoxin (Trx), leading to the dissociation of thioredoxin interaction protein (TXNIP) from Trx. Zhou et al. [53] reported that TXNIP could dissociate from Trx in an ROS-sensitive way, allowing it to bind and activate NLRP3. Thirdly, ONOO may mediate K+ efflux and then activate NLRP3. K+ efflux is a well-characterized activator for NLRP3 inflammasome, and ONOO is a positive modulator for K+ efflux [38]. Therefore, ONOO may be responsible for NLRP3 inflammasome activation.
Of translational significance, we investigated the effect of BBG in an ICH rat model. Previous studies have demonstrated a neuroprotective effect of BBG in many acute CNS diseases such as ischemic stroke [54], subarachnoid hemorrhage [6], traumatic brain injury [55], and spinal cord injury [56]. BBG is a derivative of the widely used and Food and Drug Administration-approved food additive FD&C Blue number 1 [57]. With its low toxicity and high selectivity, BBG is considered to be an attractive drug for CNS diseases [56]. We found that BBG treatment inhibited the inflammatory response via the P2X7R/NLRP3 axis following ICH, and this was associated with significantly improved neurological function and less brain edema.
Several potential limitations deserve mention. Firstly, although we employed the most commonly used ICH model and injected sterile-filtered collagenase, the extra-inflammatory responses induced by the collagenase itself seemed unavoidable. Secondly, our observation period was 72 h but it appears that ASC and caspase-1 activation may undergo a second peak at 72 h; we speculate that other signaling pathways may be involved in ASC and caspase-1 activation in a later phase. Finally, although it was significant, the effect of P2X7R siRNA and BBG on changes in brain water content and neurobehavioral scores was relatively small. Thus, further experiments are necessary to address these problems.

Conclusion

In summary, our results indicate that P2X7R contributes to NLRP3 inflammasome activation and subsequent IL-1β/IL-18 release to drive brain inflammation and neuronal damage in an ICH rat model. NOX2/iNOS-dependent ONOO formation, a potential downstream signaling component of P2X7R, may be a key trigger of NLRP3 inflammasome activation. Thus, inhibition of P2X7R or ONOO could be a potential therapeutic target for secondary brain injury accompanying ICH.

Acknowledgements

This study was supported by the National Natural Science Foundation of China (Nos. 81271314 and 30500526), Natural Science Foundation of Guangdong (No. 5300468), Special Project on the Integration of Industry, Education and Research of Guangdong Province and Ministry of Education (No. 2012B091100154), Natural Science Foundation of Guangdong (No.2014A030313346), and The Guangdong Provincial Clinical Medical Centre For Neurosurgery (No. 2013B020400005).
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.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

Conceived and designed the experiments: YZC and LF. Performed the experiments: LF. Analyzed the data: YZC and LF. Wrote the paper: LF. Paper revision: YZC. RD, ZHF, SY, XQD, and JZ provided experimental technical support and assisted in completing the study at different stages. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Keep RF, Hua Y, Xi G. Intracerebral haemorrhage: mechanisms of injury and therapeutic targets. Lancet Neurol. 2012;11:720–31.CrossRefPubMed Keep RF, Hua Y, Xi G. Intracerebral haemorrhage: mechanisms of injury and therapeutic targets. Lancet Neurol. 2012;11:720–31.CrossRefPubMed
2.
Zurück zum Zitat Ma Q, Chen S, Hu Q, Feng H, Zhang JH, Tang J. NLRP3 inflammasome contributes to inflammation after intracerebral hemorrhage. Ann Neurol. 2014;75:209–19.CrossRefPubMedPubMedCentral Ma Q, Chen S, Hu Q, Feng H, Zhang JH, Tang J. NLRP3 inflammasome contributes to inflammation after intracerebral hemorrhage. Ann Neurol. 2014;75:209–19.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Wang YC, Zhou Y, Fang H, Lin S, Wang PF, Xiong RP, et al. Toll-like receptor 2/4 heterodimer mediates inflammatory injury in intracerebral hemorrhage. Ann Neurol. 2014;75:876–89.CrossRefPubMed Wang YC, Zhou Y, Fang H, Lin S, Wang PF, Xiong RP, et al. Toll-like receptor 2/4 heterodimer mediates inflammatory injury in intracerebral hemorrhage. Ann Neurol. 2014;75:876–89.CrossRefPubMed
5.
Zurück zum Zitat Latz E, Xiao TS, Stutz A. Activation and regulation of the inflammasomes. Nat Rev Immunol. 2013;13:397–411.CrossRefPubMed Latz E, Xiao TS, Stutz A. Activation and regulation of the inflammasomes. Nat Rev Immunol. 2013;13:397–411.CrossRefPubMed
6.
Zurück zum Zitat Chen S, Ma Q, Krafft PR, Hu Q, Rolland 2nd W, Sherchan P, et al. P2X7R/cryopyrin inflammasome axis inhibition reduces neuroinflammation after SAH. Neurobiol Dis. 2013;58:296–307.CrossRefPubMedPubMedCentral Chen S, Ma Q, Krafft PR, Hu Q, Rolland 2nd W, Sherchan P, et al. P2X7R/cryopyrin inflammasome axis inhibition reduces neuroinflammation after SAH. Neurobiol Dis. 2013;58:296–307.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Yang F, Wang Z, Wei X, Han H, Meng X, Zhang Y, et al. NLRP3 deficiency ameliorates neurovascular damage in experimental ischemic stroke. J Cereb Blood Flow Metab. 2014;34:660–7.CrossRefPubMedPubMedCentral Yang F, Wang Z, Wei X, Han H, Meng X, Zhang Y, et al. NLRP3 deficiency ameliorates neurovascular damage in experimental ischemic stroke. J Cereb Blood Flow Metab. 2014;34:660–7.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Liu HD, Li W, Chen ZR, Hu YC, Zhang DD, Shen W, et al. Expression of the NLRP3 inflammasome in cerebral cortex after traumatic brain injury in a rat model. Neurochem Res. 2013;38:2072–83.CrossRefPubMed Liu HD, Li W, Chen ZR, Hu YC, Zhang DD, Shen W, et al. Expression of the NLRP3 inflammasome in cerebral cortex after traumatic brain injury in a rat model. Neurochem Res. 2013;38:2072–83.CrossRefPubMed
9.
Zurück zum Zitat Bigford GE, Bracchi-Ricard VC, Keane RW, Nash MS, Bethea JR. Neuroendocrine and cardiac metabolic dysfunction and NLRP3 inflammasome activation in adipose tissue and pancreas following chronic spinal cord injury in the mouse. ASN Neuro. 2013;5:243–55.CrossRefPubMed Bigford GE, Bracchi-Ricard VC, Keane RW, Nash MS, Bethea JR. Neuroendocrine and cardiac metabolic dysfunction and NLRP3 inflammasome activation in adipose tissue and pancreas following chronic spinal cord injury in the mouse. ASN Neuro. 2013;5:243–55.CrossRefPubMed
10.
Zurück zum Zitat Meng XF, Tan L, Tan MS, Jiang T, Tan CC, Li MM, et al. Inhibition of the NLRP3 inflammasome provides neuroprotection in rats following amygdala kindling-induced status epilepticus. J Neuroinflammation. 2014;11:212.CrossRefPubMedPubMedCentral Meng XF, Tan L, Tan MS, Jiang T, Tan CC, Li MM, et al. Inhibition of the NLRP3 inflammasome provides neuroprotection in rats following amygdala kindling-induced status epilepticus. J Neuroinflammation. 2014;11:212.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Parajuli B, Sonobe Y, Horiuchi H, Takeuchi H, Mizuno T, Suzumura A. Oligomeric amyloid beta induces IL-1beta processing via production of ROS: implication in Alzheimer’s disease. Cell Death Dis. 2013;4:e975.CrossRefPubMedPubMedCentral Parajuli B, Sonobe Y, Horiuchi H, Takeuchi H, Mizuno T, Suzumura A. Oligomeric amyloid beta induces IL-1beta processing via production of ROS: implication in Alzheimer’s disease. Cell Death Dis. 2013;4:e975.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Di Virgilio F. Liaisons dangereuses: P2X(7) and the inflammasome. Trends Pharmacol Sci. 2007;28:465–72.CrossRefPubMed Di Virgilio F. Liaisons dangereuses: P2X(7) and the inflammasome. Trends Pharmacol Sci. 2007;28:465–72.CrossRefPubMed
13.
Zurück zum Zitat Sperlagh B, Illes P. P2X7 receptor: an emerging target in central nervous system diseases. Trends Pharmacol Sci. 2014;35:537–47.CrossRefPubMed Sperlagh B, Illes P. P2X7 receptor: an emerging target in central nervous system diseases. Trends Pharmacol Sci. 2014;35:537–47.CrossRefPubMed
14.
Zurück zum Zitat Bartlett R, Stokes L, Sluyter R. The P2X7 receptor channel: recent developments and the use of P2X7 antagonists in models of disease. Pharmacol Rev. 2014;66:638–75.CrossRefPubMed Bartlett R, Stokes L, Sluyter R. The P2X7 receptor channel: recent developments and the use of P2X7 antagonists in models of disease. Pharmacol Rev. 2014;66:638–75.CrossRefPubMed
15.
Zurück zum Zitat Tang J, Liu J, Zhou C, Ostanin D, Grisham MB, Neil Granger D, et al. Role of NADPH oxidase in the brain injury of intracerebral hemorrhage. J Neurochem. 2005;94:1342–50.CrossRefPubMed Tang J, Liu J, Zhou C, Ostanin D, Grisham MB, Neil Granger D, et al. Role of NADPH oxidase in the brain injury of intracerebral hemorrhage. J Neurochem. 2005;94:1342–50.CrossRefPubMed
16.
Zurück zum Zitat Kim DW, Im SH, Kim JY, Kim DE, Oh GT, Jeong SW. Decreased brain edema after collagenase-induced intracerebral hemorrhage in mice lacking the inducible nitric oxide synthase gene. Laboratory investigation. J Neurosurg. 2009;111:995–1000.CrossRefPubMed Kim DW, Im SH, Kim JY, Kim DE, Oh GT, Jeong SW. Decreased brain edema after collagenase-induced intracerebral hemorrhage in mice lacking the inducible nitric oxide synthase gene. Laboratory investigation. J Neurosurg. 2009;111:995–1000.CrossRefPubMed
17.
Zurück zum Zitat Heneka MT, Kummer MP, Latz E. Innate immune activation in neurodegenerative disease. Nat Rev Immunol. 2014;14:463–77.CrossRefPubMed Heneka MT, Kummer MP, Latz E. Innate immune activation in neurodegenerative disease. Nat Rev Immunol. 2014;14:463–77.CrossRefPubMed
18.
Zurück zum Zitat Szabo C, Ischiropoulos H, Radi R. Peroxynitrite: biochemistry, pathophysiology and development of therapeutics. Nat Rev Drug Discov. 2007;6:662–80.CrossRefPubMed Szabo C, Ischiropoulos H, Radi R. Peroxynitrite: biochemistry, pathophysiology and development of therapeutics. Nat Rev Drug Discov. 2007;6:662–80.CrossRefPubMed
19.
Zurück zum Zitat Deng-Bryant Y, Singh IN, Carrico KM, Hall ED. Neuroprotective effects of tempol, a catalytic scavenger of peroxynitrite-derived free radicals, in a mouse traumatic brain injury model. J Cereb Blood Flow Metab. 2008;28:1114–26.CrossRefPubMed Deng-Bryant Y, Singh IN, Carrico KM, Hall ED. Neuroprotective effects of tempol, a catalytic scavenger of peroxynitrite-derived free radicals, in a mouse traumatic brain injury model. J Cereb Blood Flow Metab. 2008;28:1114–26.CrossRefPubMed
20.
Zurück zum Zitat Genovese T, Mazzon E, Esposito E, Muia C, Di Paola R, Bramanti P, et al. Beneficial effects of FeTSPP, a peroxynitrite decomposition catalyst, in a mouse model of spinal cord injury. Free Radic Biol Med. 2007;43:763–80.CrossRefPubMed Genovese T, Mazzon E, Esposito E, Muia C, Di Paola R, Bramanti P, et al. Beneficial effects of FeTSPP, a peroxynitrite decomposition catalyst, in a mouse model of spinal cord injury. Free Radic Biol Med. 2007;43:763–80.CrossRefPubMed
21.
Zurück zum Zitat Ding R, Chen Y, Yang S, Deng X, Fu Z, Feng L, et al. Blood-brain barrier disruption induced by hemoglobin in vivo: involvement of up-regulation of nitric oxide synthase and peroxynitrite formation. Brain Res. 2014;1571:25–38.CrossRefPubMed Ding R, Chen Y, Yang S, Deng X, Fu Z, Feng L, et al. Blood-brain barrier disruption induced by hemoglobin in vivo: involvement of up-regulation of nitric oxide synthase and peroxynitrite formation. Brain Res. 2014;1571:25–38.CrossRefPubMed
22.
Zurück zum Zitat Little JW, Cuzzocrea S, Bryant L, Esposito E, Doyle T, Rausaria S, et al. Spinal mitochondrial-derived peroxynitrite enhances neuroimmune activation during morphine hyperalgesia and antinociceptive tolerance. Pain. 2013;154:978–86.CrossRefPubMedPubMedCentral Little JW, Cuzzocrea S, Bryant L, Esposito E, Doyle T, Rausaria S, et al. Spinal mitochondrial-derived peroxynitrite enhances neuroimmune activation during morphine hyperalgesia and antinociceptive tolerance. Pain. 2013;154:978–86.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Doyle T, Chen Z, Muscoli C, Bryant L, Esposito E, Cuzzocrea S, et al. Targeting the overproduction of peroxynitrite for the prevention and reversal of paclitaxel-induced neuropathic pain. J Neurosci. 2012;32:6149–60.CrossRefPubMedPubMedCentral Doyle T, Chen Z, Muscoli C, Bryant L, Esposito E, Cuzzocrea S, et al. Targeting the overproduction of peroxynitrite for the prevention and reversal of paclitaxel-induced neuropathic pain. J Neurosci. 2012;32:6149–60.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Apolloni S, Amadio S, Parisi C, Matteucci A, Potenza RL, Armida M, et al. Spinal cord pathology is ameliorated by P2X7 antagonism in a SOD1-mutant mouse model of amyotrophic lateral sclerosis. Dis Model Mech. 2014;7:1101–9.CrossRefPubMedPubMedCentral Apolloni S, Amadio S, Parisi C, Matteucci A, Potenza RL, Armida M, et al. Spinal cord pathology is ameliorated by P2X7 antagonism in a SOD1-mutant mouse model of amyotrophic lateral sclerosis. Dis Model Mech. 2014;7:1101–9.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Chatterjee S, Rana R, Corbett J, Kadiiska MB, Goldstein J, Mason RP. P2X7 receptor-NADPH oxidase axis mediates protein radical formation and Kupffer cell activation in carbon tetrachloride-mediated steatohepatitis in obese mice. Free Radic Biol Med. 2012;52:1666–79.CrossRefPubMedPubMedCentral Chatterjee S, Rana R, Corbett J, Kadiiska MB, Goldstein J, Mason RP. P2X7 receptor-NADPH oxidase axis mediates protein radical formation and Kupffer cell activation in carbon tetrachloride-mediated steatohepatitis in obese mice. Free Radic Biol Med. 2012;52:1666–79.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Parvathenani LK, Tertyshnikova S, Greco CR, Roberts SB, Robertson B, Posmantur R. P2X7 mediates superoxide production in primary microglia and is up-regulated in a transgenic mouse model of Alzheimer’s disease. J Biol Chem. 2003;278:13309–17.CrossRefPubMed Parvathenani LK, Tertyshnikova S, Greco CR, Roberts SB, Robertson B, Posmantur R. P2X7 mediates superoxide production in primary microglia and is up-regulated in a transgenic mouse model of Alzheimer’s disease. J Biol Chem. 2003;278:13309–17.CrossRefPubMed
28.
Zurück zum Zitat Hewinson J, Moore SF, Glover C, Watts AG, MacKenzie AB. A key role for redox signaling in rapid P2X7 receptor-induced IL-1 beta processing in human monocytes. J Immunol. 2008;180:8410–20.CrossRefPubMed Hewinson J, Moore SF, Glover C, Watts AG, MacKenzie AB. A key role for redox signaling in rapid P2X7 receptor-induced IL-1 beta processing in human monocytes. J Immunol. 2008;180:8410–20.CrossRefPubMed
29.
Zurück zum Zitat Choi HB, Ryu JK, Kim SU, McLarnon JG. Modulation of the purinergic P2X7 receptor attenuates lipopolysaccharide-mediated microglial activation and neuronal damage in inflamed brain. J Neurosci. 2007;27:4957–68.CrossRefPubMed Choi HB, Ryu JK, Kim SU, McLarnon JG. Modulation of the purinergic P2X7 receptor attenuates lipopolysaccharide-mediated microglial activation and neuronal damage in inflamed brain. J Neurosci. 2007;27:4957–68.CrossRefPubMed
30.
Zurück zum Zitat Ding R, Feng L, He L, Chen Y, Wen P, Fu Z, et al. Peroxynitrite decomposition catalyst prevents matrix metalloproteinase-9 activation and neurovascular injury after hemoglobin injection into the caudate nucleus of rats. Neuroscience. 2015;297:182–93.CrossRefPubMed Ding R, Feng L, He L, Chen Y, Wen P, Fu Z, et al. Peroxynitrite decomposition catalyst prevents matrix metalloproteinase-9 activation and neurovascular injury after hemoglobin injection into the caudate nucleus of rats. Neuroscience. 2015;297:182–93.CrossRefPubMed
31.
Zurück zum Zitat Jung KH, Chu K, Jeong SW, Han SY, Lee ST, Kim JY, et al. HMG-CoA reductase inhibitor, atorvastatin, promotes sensorimotor recovery, suppressing acute inflammatory reaction after experimental intracerebral hemorrhage. Stroke. 2004;35:1744–9.CrossRefPubMed Jung KH, Chu K, Jeong SW, Han SY, Lee ST, Kim JY, et al. HMG-CoA reductase inhibitor, atorvastatin, promotes sensorimotor recovery, suppressing acute inflammatory reaction after experimental intracerebral hemorrhage. Stroke. 2004;35:1744–9.CrossRefPubMed
32.
Zurück zum Zitat Rynkowski MA, Kim GH, Komotar RJ, Otten ML, Ducruet AF, Zacharia BE, et al. A mouse model of intracerebral hemorrhage using autologous blood infusion. Nat Protoc. 2008;3:122–8.CrossRefPubMed Rynkowski MA, Kim GH, Komotar RJ, Otten ML, Ducruet AF, Zacharia BE, et al. A mouse model of intracerebral hemorrhage using autologous blood infusion. Nat Protoc. 2008;3:122–8.CrossRefPubMed
33.
Zurück zum Zitat Chu K, Jeong SW, Jung KH, Han SY, Lee ST, Kim M, et al. Celecoxib induces functional recovery after intracerebral hemorrhage with reduction of brain edema and perihematomal cell death. J Cereb Blood Flow Metab. 2004;24:926–33.CrossRefPubMed Chu K, Jeong SW, Jung KH, Han SY, Lee ST, Kim M, et al. Celecoxib induces functional recovery after intracerebral hemorrhage with reduction of brain edema and perihematomal cell death. J Cereb Blood Flow Metab. 2004;24:926–33.CrossRefPubMed
34.
Zurück zum Zitat Chen J, Sanberg PR, Li Y, Wang L, Lu M, Willing AE, et al. Intravenous administration of human umbilical cord blood reduces behavioral deficits after stroke in rats. Stroke. 2001;32:2682–8.CrossRefPubMed Chen J, Sanberg PR, Li Y, Wang L, Lu M, Willing AE, et al. Intravenous administration of human umbilical cord blood reduces behavioral deficits after stroke in rats. Stroke. 2001;32:2682–8.CrossRefPubMed
35.
Zurück zum Zitat Zhao X, Zhang Y, Strong R, Zhang J, Grotta JC, Aronowski J. Distinct patterns of intracerebral hemorrhage-induced alterations in NF-kappaB subunit, iNOS, and COX-2 expression. J Neurochem. 2007;101:652–63.CrossRefPubMed Zhao X, Zhang Y, Strong R, Zhang J, Grotta JC, Aronowski J. Distinct patterns of intracerebral hemorrhage-induced alterations in NF-kappaB subunit, iNOS, and COX-2 expression. J Neurochem. 2007;101:652–63.CrossRefPubMed
36.
Zurück zum Zitat Abais JM, Xia M, Zhang Y, Boini KM, Li PL. Redox regulation of NLRP3 inflammasomes: ROS as trigger or effector? Antioxid Redox Signal. 2015;22:1111–29. Abais JM, Xia M, Zhang Y, Boini KM, Li PL. Redox regulation of NLRP3 inflammasomes: ROS as trigger or effector? Antioxid Redox Signal. 2015;22:1111–29.
37.
Zurück zum Zitat Qu Y, Misaghi S, Newton K, Gilmour LL, Louie S, Cupp JE, et al. Pannexin-1 is required for ATP release during apoptosis but not for inflammasome activation. J Immunol. 2011;186:6553–61.CrossRefPubMed Qu Y, Misaghi S, Newton K, Gilmour LL, Louie S, Cupp JE, et al. Pannexin-1 is required for ATP release during apoptosis but not for inflammasome activation. J Immunol. 2011;186:6553–61.CrossRefPubMed
38.
Zurück zum Zitat Bossy-Wetzel E, Talantova MV, Lee WD, Scholzke MN, Harrop A, Mathews E, et al. Crosstalk between nitric oxide and zinc pathways to neuronal cell death involving mitochondrial dysfunction and p38-activated K+ channels. Neuron. 2004;41:351–65.CrossRefPubMed Bossy-Wetzel E, Talantova MV, Lee WD, Scholzke MN, Harrop A, Mathews E, et al. Crosstalk between nitric oxide and zinc pathways to neuronal cell death involving mitochondrial dysfunction and p38-activated K+ channels. Neuron. 2004;41:351–65.CrossRefPubMed
40.
Zurück zum Zitat Zhu X, Tao L, Tejima-Mandeville E, Qiu J, Park J, Garber K, et al. Plasmalemma permeability and necrotic cell death phenotypes after intracerebral hemorrhage in mice. Stroke. 2012;43:524–31.CrossRefPubMed Zhu X, Tao L, Tejima-Mandeville E, Qiu J, Park J, Garber K, et al. Plasmalemma permeability and necrotic cell death phenotypes after intracerebral hemorrhage in mice. Stroke. 2012;43:524–31.CrossRefPubMed
41.
Zurück zum Zitat Benarroch EE. Microglia: multiple roles in surveillance, circuit shaping, and response to injury. Neurology. 2013;81:1079–88.CrossRefPubMed Benarroch EE. Microglia: multiple roles in surveillance, circuit shaping, and response to injury. Neurology. 2013;81:1079–88.CrossRefPubMed
42.
Zurück zum Zitat Wang J, Dore S. Inflammation after intracerebral hemorrhage. J Cereb Blood Flow Metab. 2007;27:894–908.CrossRefPubMed Wang J, Dore S. Inflammation after intracerebral hemorrhage. J Cereb Blood Flow Metab. 2007;27:894–908.CrossRefPubMed
44.
Zurück zum Zitat Lu A, Tang Y, Ran R, Ardizzone TL, Wagner KR, Sharp FR. Brain genomics of intracerebral hemorrhage. J Cereb Blood Flow Metab. 2006;26:230–52.CrossRefPubMed Lu A, Tang Y, Ran R, Ardizzone TL, Wagner KR, Sharp FR. Brain genomics of intracerebral hemorrhage. J Cereb Blood Flow Metab. 2006;26:230–52.CrossRefPubMed
45.
Zurück zum Zitat Felderhoff-Mueser U, Schmidt OI, Oberholzer A, Buhrer C, Stahel PF. IL-18: a key player in neuroinflammation and neurodegeneration? Trends Neurosci. 2005;28:487–93.CrossRefPubMed Felderhoff-Mueser U, Schmidt OI, Oberholzer A, Buhrer C, Stahel PF. IL-18: a key player in neuroinflammation and neurodegeneration? Trends Neurosci. 2005;28:487–93.CrossRefPubMed
46.
Zurück zum Zitat Ziai WC. Hematology and inflammatory signaling of intracerebral hemorrhage. Stroke. 2013;44:S74–8.CrossRefPubMed Ziai WC. Hematology and inflammatory signaling of intracerebral hemorrhage. Stroke. 2013;44:S74–8.CrossRefPubMed
47.
Zurück zum Zitat Mander P, Brown GC. Activation of microglial NADPH oxidase is synergistic with glial iNOS expression in inducing neuronal death: a dual-key mechanism of inflammatory neurodegeneration. J Neuroinflammation. 2005;2:20.CrossRefPubMedPubMedCentral Mander P, Brown GC. Activation of microglial NADPH oxidase is synergistic with glial iNOS expression in inducing neuronal death: a dual-key mechanism of inflammatory neurodegeneration. J Neuroinflammation. 2005;2:20.CrossRefPubMedPubMedCentral
48.
Zurück zum Zitat Ryu JK, McLarnon JG. Minocycline or iNOS inhibition block 3-nitrotyrosine increases and blood-brain barrier leakiness in amyloid beta-peptide-injected rat hippocampus. Exp Neurol. 2006;198:552–7.CrossRefPubMed Ryu JK, McLarnon JG. Minocycline or iNOS inhibition block 3-nitrotyrosine increases and blood-brain barrier leakiness in amyloid beta-peptide-injected rat hippocampus. Exp Neurol. 2006;198:552–7.CrossRefPubMed
49.
Zurück zum Zitat Dohi K, Ohtaki H, Nakamachi T, Yofu S, Satoh K, Miyamoto K, et al. Gp91phox (NOX2) in classically activated microglia exacerbates traumatic brain injury. J Neuroinflammation. 2010;7:41.CrossRefPubMedPubMedCentral Dohi K, Ohtaki H, Nakamachi T, Yofu S, Satoh K, Miyamoto K, et al. Gp91phox (NOX2) in classically activated microglia exacerbates traumatic brain injury. J Neuroinflammation. 2010;7:41.CrossRefPubMedPubMedCentral
50.
Zurück zum Zitat Thiyagarajan M, Kaul CL, Sharma SS. Neuroprotective efficacy and therapeutic time window of peroxynitrite decomposition catalysts in focal cerebral ischemia in rats. Br J Pharmacol. 2004;142:899–911.CrossRefPubMedPubMedCentral Thiyagarajan M, Kaul CL, Sharma SS. Neuroprotective efficacy and therapeutic time window of peroxynitrite decomposition catalysts in focal cerebral ischemia in rats. Br J Pharmacol. 2004;142:899–911.CrossRefPubMedPubMedCentral
51.
Zurück zum Zitat Salvemini D, Wang ZQ, Stern MK, Currie MG, Misko TP. Peroxynitrite decomposition catalysts: therapeutics for peroxynitrite-mediated pathology. Proc Natl Acad Sci U S A. 1998;95:2659–63.CrossRefPubMedPubMedCentral Salvemini D, Wang ZQ, Stern MK, Currie MG, Misko TP. Peroxynitrite decomposition catalysts: therapeutics for peroxynitrite-mediated pathology. Proc Natl Acad Sci U S A. 1998;95:2659–63.CrossRefPubMedPubMedCentral
52.
Zurück zum Zitat Shimada K, Crother TR, Karlin J, Dagvadorj J, Chiba N, Chen S, et al. Oxidized mitochondrial DNA activates the NLRP3 inflammasome during apoptosis. Immunity. 2012;36:401–14.CrossRefPubMedPubMedCentral Shimada K, Crother TR, Karlin J, Dagvadorj J, Chiba N, Chen S, et al. Oxidized mitochondrial DNA activates the NLRP3 inflammasome during apoptosis. Immunity. 2012;36:401–14.CrossRefPubMedPubMedCentral
53.
Zurück zum Zitat Zhou R, Tardivel A, Thorens B, Choi I, Tschopp J. Thioredoxin-interacting protein links oxidative stress to inflammasome activation. Nat Immunol. 2010;11:136–40.CrossRefPubMed Zhou R, Tardivel A, Thorens B, Choi I, Tschopp J. Thioredoxin-interacting protein links oxidative stress to inflammasome activation. Nat Immunol. 2010;11:136–40.CrossRefPubMed
54.
Zurück zum Zitat Arbeloa J, Perez-Samartin A, Gottlieb M, Matute C. P2X7 receptor blockade prevents ATP excitotoxicity in neurons and reduces brain damage after ischemia. Neurobiol Dis. 2012;45:954–61.CrossRefPubMed Arbeloa J, Perez-Samartin A, Gottlieb M, Matute C. P2X7 receptor blockade prevents ATP excitotoxicity in neurons and reduces brain damage after ischemia. Neurobiol Dis. 2012;45:954–61.CrossRefPubMed
55.
Zurück zum Zitat Kimbler DE, Shields J, Yanasak N, Vender JR, Dhandapani KM. Activation of P2X7 promotes cerebral edema and neurological injury after traumatic brain injury in mice. PLoS ONE. 2012;7:e41229.CrossRefPubMedPubMedCentral Kimbler DE, Shields J, Yanasak N, Vender JR, Dhandapani KM. Activation of P2X7 promotes cerebral edema and neurological injury after traumatic brain injury in mice. PLoS ONE. 2012;7:e41229.CrossRefPubMedPubMedCentral
56.
Zurück zum Zitat Peng W, Cotrina ML, Han X, Yu H, Bekar L, Blum L, et al. Systemic administration of an antagonist of the ATP-sensitive receptor P2X7 improves recovery after spinal cord injury. Proc Natl Acad Sci U S A. 2009;106:12489–93.CrossRefPubMedPubMedCentral Peng W, Cotrina ML, Han X, Yu H, Bekar L, Blum L, et al. Systemic administration of an antagonist of the ATP-sensitive receptor P2X7 improves recovery after spinal cord injury. Proc Natl Acad Sci U S A. 2009;106:12489–93.CrossRefPubMedPubMedCentral
57.
Zurück zum Zitat Wang J, Jackson DG, Dahl G. The food dye FD&C Blue No. 1 is a selective inhibitor of the ATP release channel Panx1. J Gen Physiol. 2013;141:649–56.CrossRefPubMedPubMedCentral Wang J, Jackson DG, Dahl G. The food dye FD&C Blue No. 1 is a selective inhibitor of the ATP release channel Panx1. J Gen Physiol. 2013;141:649–56.CrossRefPubMedPubMedCentral
Metadaten
Titel
P2X7R blockade prevents NLRP3 inflammasome activation and brain injury in a rat model of intracerebral hemorrhage: involvement of peroxynitrite
verfasst von
Liang Feng
Yizhao Chen
Rui Ding
Zhenghao Fu
Shuo Yang
Xinqing Deng
Jun Zeng
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
Journal of Neuroinflammation / Ausgabe 1/2015
Elektronische ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-015-0409-2

Weitere Artikel der Ausgabe 1/2015

Journal of Neuroinflammation 1/2015 Zur Ausgabe