Skip to main content
Erschienen in: Journal of Neuroinflammation 1/2017

Open Access 01.12.2017 | Research

Thioredoxin-interacting protein links endoplasmic reticulum stress to inflammatory brain injury and apoptosis after subarachnoid haemorrhage

verfasst von: Qing Zhao, Xudong Che, Hongxia Zhang, Pianpian Fan, Guanping Tan, Liu Liu, Dengzhi Jiang, Jun Zhao, Xiang Xiang, Yidan Liang, Xiaochuan Sun, Zhaohui He

Erschienen in: Journal of Neuroinflammation | Ausgabe 1/2017

Abstract

Background

Early brain injury (EBI) is considered a major contributor to the high morbidity and mortality associated with subarachnoid haemorrhage (SAH). Both of sterile inflammation and apoptosis are considered the important causes of EBI. Recently, it was confirmed that thioredoxin-interacting protein (TXNIP) not only participates in inflammatory amplification but also stimulates the apoptosis signalling cascade pathway. However, whether the effects of TXNIP influence the pathogenesis of SAH remains unclear. Here, we hypothesize that TXNIP activity induced by endoplasmic reticulum stress (ER stress) may contribute to the pathogenesis of EBI through pro-inflammatory and pro-apoptotic mechanisms.

Methods

A total of 299 male Sprague–Dawley rats were used to create SAH models. Resveratrol (RES, 60 mg/kg) and two TXNIP small interfering RNA (siRNA) were used to inhibit TXNIP expression. The specific inhibitors of ER stress sensors were used to disrupt the link between TXNIP and ER stress. SAH grade, neurological deficits, brain water content and blood–brain barrier (BBB) permeability were evaluated simultaneously as prognostic indicators. Fluorescent double-labelling was employed to detect the location of TXNIP in cerebral cells. Western blot and TUNEL were performed to study the mechanisms of TXNIP and EBI.

Results

We found that TXNIP expression significantly increased after SAH, peaking at 48 h (0.48 ± 0.04, up to 3.2-fold) and decreasing at 72 h after surgery. This process was accompanied by the generation of inflammation-associated factors. TXNIP was expressed in the cytoplasm of neurons and was widely co-localized with TUNEL-positive cells in both the hippocampus and the cortex of SAH rats. We discovered for the first time that TXNIP was co-localized in neural immunocytes (microglia and astrocytes). After administration of RES, TXNIP siRNA and ER stress inhibitors, TXNIP expression was significantly reduced and the crosstalk between TXNIP and ER stress was disrupted; this was accompanied by a reduction in inflammatory and apoptotic factors, as well as attenuation of the prognostic indices.

Conclusions

These results may represent the critical evidence to support the pro-inflammatory and pro-apoptotic effects of TXNIP after SAH. Our data suggest that TXNIP participates in EBI after SAH by mediating inflammation and apoptosis; these pathways may represent a potential therapeutic strategy for SAH treatment.
Begleitmaterial
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s12974-017-0878-6) contains supplementary material, which is available to authorized users.
Abkürzungen
EBI
Early brain injury
SAH
Subarachnoid haemorrhage
TXNIP
Thioredoxin-interacting protein
ER stress
Endoplasmic reticulum stress
siRNA
Small interfering RNA
BBB
Blood–brain barrier
IL-1β
Interleukin-1β
TRX
Thioredoxin
NLRP3 inflammasome
Nod-like receptor protein 3 inflammasome
PERK
Protein kinase RNA-like ER kinase
IRE1α
Inositol-requiring enzyme-1α
ATF-6
Activating transcription factor-6
RES
Resveratrol
eIF2α
Eukaryotic translation initiation factor-2α
TUNEL
Terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick end labelling
ATF-5
Activating transcription factor 5
ChREBP
Carbohydrate response element-binding protein
ASK-1
Apoptosis signal-regulating kinase-1
XBP1
X-box binding protein-1

Background

Subarachnoid haemorrhage (SAH) is a serious neurological emergency associated with high morbidity and mortality [1]. Current studies show that early brain injury (EBI) is the overriding factor and that both neuroinflammation and apoptosis have been confirmed to contribute to EBI [2, 3]. EBI refers to the direct brain damage occurring within 72 h after SAH, including increased intracranial pressure, cerebral perfusion pressure disorder, blood–brain barrier (BBB) destruction and brain edema [2, 3].
Neuroinflammation is associated with various acute and chronic neurodegenerative diseases, including Alzheimer’s disease, Parkinson’s disease and SAH [3, 4]. Increased levels of cytokines have been found in the cerebrospinal fluid of SAH patients and are correlated with poor neurological outcomes [5]. Once bleeding occurs, neural immunocytes (microglia and astrocytes) rapidly react to the extravascular blood components that enter the cerebral parenchyma [6, 7]. The activated immunocytes then release various inflammation mediators such as Toll-like receptor 4 and interleukin-1β (IL-1β), thereby aggravating the secondary brain injury [810]. Various pro-apoptotic mechanisms are also reported to contribute to EBI after SAH and involve not only the activation of pro-apoptotic proteins but also the inhibition of anti-apoptotic factors [2, 3, 11]. Although some anti-inflammatory and anti-apoptotic strategies have been tested in preclinical and clinical trials, the mortality and disability burdens of SAH remain high, reminding us that the identification of appropriate and effective targets is still a major obstacle.
Thioredoxin-interacting protein (TXNIP), also known as thioredoxin-binding protein-2 or vitamin D3-upregulated protein 1, is a natural antagonist of thioredoxin (TRX) in vivo. Recent research has confirmed that TXNIP is the critical link between nod-like receptor protein 3 (NLRP3) inflammasome activation and inflammatory amplification [12]. Moreover, TXNIP can directly bind to and inhibit TRX function, leading to the activation of downstream apoptosis signalling pathways [13]. However, whether the effects of TXNIP influence the pathogenesis of SAH remains unclear. Previously, our work revealed that downstream factors in endoplasmic reticulum stress (ER stress) can induce neuronal apoptosis after SAH [14]. Intriguingly, recent studies have demonstrated that TXNIP expression can be significantly induced by ER stress at the transcriptional and post-transcriptional levels [15, 16]. The ER is the major site of protein folding, post-translational modification and assembly [17]. Although ER stress is primarily a self-protective signal transduction pathway, high-level ER stress could culminate in cell death via the activation of inflammation and apoptosis [18, 19]. Moreover, persistent and maladaptive ER stress is implicated in various human neurodegenerative diseases [20]. However, whether the induction of TXNIP by ER stress is involved in EBI after SAH remains unknown. Based on these findings, we utilized a TXNIP inhibitor, small interfering RNA (siRNA) and specific inhibitors of ER stress sensors to suppress TXNIP expression and ER stress–TXNIP signalling in order to assess the role of TXNIP in the progression of EBI after SAH.

Methods

Animals

A total of 299 adult male Sprague–Dawley rats (280–350 g) were obtained from the Animal Center of Chongqing Medical University. Animals were maintained in a specific pathogen-free laboratory with regular 12/12 h light/dark cycles under controlled temperature and humidity conditions. The Animal Ethics and Use Committee of Chongqing Medical University approved all the experimental procedures. All surgical procedures were performed under deep anaesthesia to minimize suffering, in accordance with the recommendation guidelines in the Care and Use of Laboratory Animals of the National Institutes of Health. The experiments are designed and performed as in Fig. 1. Rats were divided into different groups as follows: sham (n = 33), SAH (n = 64), SAH + normal saline (NS, as a vehicle, n = 24), SAH + resveratrol (RES, n = 22), SAH + control siRNA (control, n = 23), SAH + TXNIP siRNA (n = 21), SAH + dimethylsulfoxide (DMSO, as a vehicle, n = 31), SAH + GSK2656157 (GSK, n = 42) and SAH + STF083010 (STF, n = 39).

Endovascular perforation model of SAH

SAH animal models were created through endovascular perforation as described before [21]. Rats were anaesthetized with sodium pentobarbital (50 mg/kg) through intraperitoneal injection. Additional single doses (5 mg/kg) of pentobarbital were given to maintain anaesthesia when necessary. Sham-operated rats underwent identical procedures without the vessel puncture. After perforation, rats were kept in heated cages until recovery from anaesthesia. The electric heating pads were used to keep body temperature at 37 °C during and after the perforation.

Resveratrol and TXNIP siRNA injection

Resveratrol (trans-3, 4′, 5-trihydroxystilbene, RES) was obtained from Sigma-Aldrich, St. Louis, MO (R5010, USA), and administered to the rats by intraperitoneal injection 1 h after puncture in a single dose of 60 mg/kg [22]. RES has been reported to significantly suppress TXNIP mRNA and protein expression and is capable of penetrating the BBB and reaching brain tissue rapidly [23, 24]. RES was dissolved in 50% ethanol and diluted with physiological saline (1 mL). Normal saline (1 mL) with 50% ethanol was used as the control.
Rats received TXNIP siRNA and control siRNA at 24 h before surgery via intracerebroventricular infusion as we described previously [14]. Two different TXNIP siRNA (Table 1) were designed as reported before [25, 26] and obtained from Guangzhou Ribo Biotechnology Co., Ltd. (Guangzhou, China). Briefly, 5 nmol of siRNA per rat in 6 μL sterile phosphate-buffered saline was inserted into the left lateral ventricles through a burr hole located at 1.5 mm posterior, 1.0 mm lateral and 3.2 mm under the bregma horizontal plane; the injection was performed with a sterile 10-μL Hamilton syringe and at a rate of 0.5 μL/min. Sham-treated and SAH animals also received a burr hole, but no siRNA injection was performed. After 10 min, the needle was removed and the burr hole was plugged carefully with bone wax.
Table 1
The sequences of two different TXNIP siRNA
Name
Sequences
siRNA 1 [25]
Sense: 5′-GCUGG AUAGACCUAAACAUTT-3′
Antisense: 5′-AUGUUUAGGUCUAUCCAGCTT-3′
siRNA 2 [26]
Sense: 5′-UGGUCACGUCGAAAUGAAUTT-3′
Antisense: 5′-TTACCAGUGCAGCUUUACUUA-3′
Control siRNA
Sense: 5′-UUCUCCGAACGUGUCACGUTT-3′
Antisense: 5′-ACGUGACACGUUCGGAGAATT-3′

ER stress sensor inhibitor injection

Various stimuli could result in the activation of stress sensors in the ER transmembrane domain, including protein kinase RNA-like ER kinase (PERK) and inositol-requiring enzyme-1α (IRE1α) [27]. GSK2656157 (MedChem Express, HY-13820, China) is an adenosine triphosphate (ATP) competitive inhibitor that has been reported to effectively suppress PERK autophosphorylation via interaction with the PERK kinase domain [28]. However, the use of GSK2656157 in animal brain has not yet been reported before. Meanwhile, to avoid the BBB effect and to improve local drug concentration, SAH rats received GSK2656157 through intracerebroventricular injection at the same coordinate as TXNIP siRNA injection (1.5 mm posterior, 1.0 mm lateral and 3.2 mm below the horizontal plane of bregma). According to the previous study, GSK2656157 completely inhibits PERK autophosphorylation at 8 h after treatment [28]. Therefore, in this experiment, GSK2656157 was administered twice through two symmetrical bilateral burr holes, at 8 h before and after perforation. Ten microlitres of 1% DMSO in saline containing 30, 60, 120, 180, 240 or 300 μg of GSK2656157 was injected under anaesthesia at a rate of 0.5 μL/min by using a sterile 10-μL Hamilton syringe. Ten minutes later, the needle was removed and the burr hole was plugged with bone wax. The control group rats received 10 μL of 1% DMSO in saline without the inhibitor. The rats were quickly decapitated under deep anaesthesia at 24 h after surgery for Western blot.
STF083010, an IRE1α-specific inhibitor and cell-permeable molecule, was obtained from Sigma-Aldrich, St. Louis, MO (SML0409, USA). STF083010 has been reported to significantly inhibit IRE1α endoribonuclease activity, IRE1α-mediated X-box binding protein-1 (XBP1) mRNA splicing and TXNIP expression, without affecting IRE1α kinase in vitro and in vivo [15, 29]. A previous study found that the inhibition of IRE1α-mediated XBP1 splicing occurred at 6 h after STF083010 treatment [29]. STF083010 was also dissolved in DMSO at different concentrations: a total of 10 μL DMSO containing 10, 20, 30, 40 and 50 μg of STF083010 was administered twice a day, 6 h before and after perforation.

Neurological scores, SAH grade and mortality assessment

Neurological scores were evaluated at 24 and 72 h in a blinded fashion using previously reported scoring methods [30]. An 18-point scoring system consisting of six subsets (spontaneous activity, spontaneous movements of four limbs, forelimb outstretching, wire cage wall climb, trunk touch reaction and vibrissae touch response) was used. After neurologic evaluation, the rats were sacrificed to quantify the severity of SAH according to a previously described grading scale [30]. Animals with a score below 5 points were excluded due to low SAH grade. Mortality was recorded after the operation.

Blood–brain barrier (BBB) permeability and brain water content detection

BBB permeability assessment was based on the Evans blue extravasation [31]. At 72 h after perforation, Evans blue dye 4% (2.5 ml/kg) was injected into the rats’ left femoral vein under deep anaesthesia and allowed to circulate for 60 min. Spectrophotometry was used to quantify Evans blue dye at an excitation wavelength of 620 nm, an emission wavelength of 680 nm and a bandwidth of 10 nm. For brain water content measure, the brains were quickly divided into different portions under deep anaesthesia, then weighed immediately (wet weight) and kept in the oven for 72 h at 105 °C (dry weight). The brain water content percentage was recorded as the equation: (wet weight to dry weight)/wet weight × 100% [14].

Immunofluorescence

Animals were sacrificed at 24 h after perforation for immunofluorescence analysis in accordance with methods reported in our previous work [14]. Ten-micrometre-thick sections of SAH rat brains were incubated with the following primary antibodies: TXNIP (1:50, Abcam, Cambridge, MA), NEUN (1:100, Merck Millipore), GFAP (1:50, Cell Signalling Technology, CST) and IBA1 (1:25, Abcam) overnight at 4 °C. Appropriate fluorescent secondary antibodies (Abbkine, Redlands, CA, USA) were used. Fluorescence microscopy (FV1200, Olympus, Japan) was used to detect TXNIP localization in neurons, microglia and astrocytes. For negative controls, the same staining procedures were conducted without primary antibodies.

TUNEL staining

Fluorescent Terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick end labelling (TUNEL, Roche Inc, Mannheim, Germany) was performed according to the manufacturer’s protocol and previously reported methods [21]. TUNEL staining was used to detect TXNIP localization within apoptotic neurons in the hippocampus and cortex of SAH rats using fluorescence microscopy (Olympus, Japan). TUNEL-positive cells in five different fields per animal in the ipsilateral basal cortex (left) were counted under Olympus microscopy at ×400 magnification in a blind manner. The number of TUNEL-positive cells per square millimetre predicted the severity of brain injury. Negative controls were performed using a labelling solution without TUNEL reagent.

Western blot

Rats were quickly decapitated under deep anaesthesia, and the left hemispheres were isolated for Western blot as described before [32]. The polyvinylidene fluoride (PVDF) membranes were incubated with primary antibodies as follows: TXNIP (1:500; Abcam, Cambridge, Mass), TRX1 (1:2000; Abcam), NLRP3 (1:1000; Abcam), cleaved Caspase-1 (1:500; Abcam), cleaved IL-1β (1:500; Abcam), cleaved Caspase-3 (1:1000; CST, Danvers, MA), BCL-2 (1:1000; CST), sXBP1 (1:100; Santa Cruz), phosphorylated-PERK (p-PERK, 1:200; Santa Cruz), eukaryotic translation initiation factor-2α (eIF2α) (1:100; Santa Cruz), phosphorylated-eIF2α (p-eIF2α, 1:500; Abcam), carbohydrate response element-binding protein (ChREBP) (1:100; Santa Cruz), activating transcription factor 5 (ATF-5) (1:2000; Abcam) and β-actin (NeoBioscience Technology Co., Ltd, Beijing, China). Membranes were incubated with the appropriate horseradish peroxidase-conjugated secondary antibodies, visualized with the enhanced chemiluminescent reagent kit (ECL, Engreen Biosystem Co., Ltd. Beijing, China) and analysed using the Fusion system (Fusion fx 7 Spectra, Vilber, France). Results are expressed as a percentage of the values for β-actin.

Statistical analysis

Statistical analysis was performed using SPSS17.0 software. Mortality data was analysed using Fisher’s exact test. All of the other data were expressed as the mean ± standard error of the mean, and comparisons between multiple groups were performed by one-way analysis of variance (ANOVA). The Dunnett t test was used for comparisons between the control group and treatment group, and Student–Newman–Keuls was used for comparisons between pairs of treatment groups receiving different interventions. p < 0.05 was considered to be statistically significant.

Results

TXNIP expression increased after SAH

First, we created SAH models and performed Western blots to detect the expression of TXNIP and various inflammatory factors, including TXNIP, TRX1, NLRP3, cleaved Caspase-1 (CC1) and cleaved IL-1β, at different time intervals. Our results showed that TXNIP expression significantly increased after SAH compared with that of the sham group (0.15 ± 0.05), reached a peak at 48 h (0.48 ± 0.04, up to 3.2-fold, p = 0.007, Fig. 2a, b) and was still in a high level at 72 h (0.46 ± 0.06) after the surgery. Compared with the sham group, we also found that the expression of NLRP3 (p = 0.017), CC1 (p = 0.006) and cleaved IL-1β (p = 0.002) was elevated to varying degrees (Fig. 2a, c, e, f) and was accompanied by the downregulation of TRX1 (p = 0.023, Fig. 2d). These results indicated that the significantly elevated TXNIP and inflammatory cytokine expression were clearly induced by SAH.

TXNIP is widely expressed in neurons and co-localizes with microglia and astrocytes after SAH

Immunofluorescence was used to detect TXNIP localization in SAH rat brains. We found that TXNIP was widely expressed in cytoplasm of neurons after SAH (n = 3, Fig. 2g). Both microglia and astrocytes play a crucial role in promoting neuroinflammation and secondary brain damage [33, 34]. However, there is no published literature to describe the histological localization of TXNIP in microglia and astrocytes after SAH. Therefore, we used specific markers of microglia and astrocytes to support these hypotheses. Importantly, we discovered for the first time that TXNIP was co-localized with microglia and astrocytes the cortex of SAH rats (n = 3, Fig. 2h, i).

Downregulation of TXNIP by resveratrol (RES) and siRNA

To provide further evidence of TXNIP’s pro-inflammatory role after SAH, both RES and TXNIP siRNA were used to suppress TXNIP expression. Our results showed that TXNIP blockage by RES decreased the generation of inflammatory factors (Fig. 3). These effects were associated with a reduction of TXNIP (p = 0.001), NLRP3 (p = 0.006), CC1 (p = 0.019) and cleaved IL-1β (p = 0.000) and improved TRX1 (p = 0.000) at 24 h after SAH (Fig. 3a–c and Additional file 1: Figure S1).
TXNIP is considered a promising therapeutic target. Although specific inhibitors and agonists of TXNIP would provide more convincing experimental evidences, such compounds are still lacking. Moreover, we cannot exclude the possibility that some other mechanisms may also mediate the observed protective effects. Previous studies have reported that RES may attenuate acute inflammatory brain injury after SAH by inhibiting other inflammatory mediator-dependent pathways [35]. We therefore used two reported siRNA mixtures to knock down TXNIP expression [25, 26]. We found that siRNA injection significantly reduced TXNIP expression (p = 0.003), as well as the expression of NLRP3 (p = 0.015), CC1 (p = 0.005) and cleaved IL-1β (p = 0.015), whereas TRX1 (p = 0.001) expression was enhanced (Fig. 3d–f and Additional file 2: Figure S2). Although we did not detect changes in TXNIP mRNA and immunofluorescence after RES and siRNA treatment, these results could indicate that TXNIP can aggravate EBI by triggering downstream inflammatory signalling pathways.

TXNIP has a pro-apoptotic effect after SAH

A recent study reported that TXNIP exerts a pro-apoptotic effect by directly binding to TRX1, leading to the activation of apoptosis [13]. In order to confirm that TXNIP also has a pro-apoptotic after SAH, we labelled cells with TUNEL, a marker for cell death. TUNEL analysis revealed that TXNIP was widely co-localized with TUNEL-positive cells in both the hippocampus and the cortex (n = 3, Fig. 4a, b). Furthermore, TXNIP inhibition by RES and siRNAs treatment significantly downregulated cleaved Caspase-3 (CC3, p = 0.000 and p = 0.001) expression and elevated B cell lymphoma (BCL)-2 (BCL-2, p = 0.001 and p = 0.032) generation (Fig. 4c–h), while reducing the number of TUNEL-positive cells (Fig. 4i–j, p < 0.05). Although we did not detect the other apoptosis-related factors of TXNIP downstream signalling pathways and some researchers have previously reported that RES may reduce cell apoptosis through the activation of some anti-apoptotic pathways [22], these results indicated that TXNIP may aggravate apoptosis after SAH by triggering downstream apoptosis signalling pathways.

Downregulation of TXNIP by PERK and IRE1α inhibition

Previous studies have confirmed that ER stress may be a principal response against neurodegeneration in various neurodegenerative diseases [20]. Our previous work also has found that the silencing of PERK downstream proteins could also attenuate EBI [14]. PERK, IRE1α and activating transcription factor-6 (ATF-6) are three important proteins in the ER transmembrane domain with multiple biological functions [27]. Recently, two research groups have found that irremediable ER stress could promote TXNIP expression through PERK at the transcriptional level and through IRE1α at the post-transcriptional level [15, 16]. However, ATF-6 has no effect on TXNIP expression [16]. Based on these findings, we speculated that TXNIP induced by ER stress may also occur after SAH. Therefore, we used a specific inhibitor of PERK and IRE1α to disrupt the links between TXNIP and ER stress.
As we know, PERK is a type 1 transmembrane protein in the ER transmembrane domain that undergoes trans-autophosphorylation upon ER stress and further phosphorylates the eukaryotic translation initiation factor-2α (eIF-2α). Interestingly, recent studies have confirmed that TXNIP transcription is significantly regulated by the PERK–eIF2α pathway during ER stress; two signalling factors downstream of PERK–eIF2α, ATF-5 and ChREBP play an especially important role [15, 16]. In this study, we found that PERK phosphorylation was suppressed by GSK2656157 in a dose-dependent manner after SAH; the inhibition effect was apparent at a dose of 120 μg and pronounced at 180 μg to 300 μg (Fig. 5a, b, p = 0.000). The levels of eIF2α phosphorylation (p = 0.001), ChREBP (p = 0.000) and ATF-5 (p = 0.001) were also reduced (Fig. 5a, c and Additional file 3: Figure S3). We used 300 μg of GSK2656157 as the final dosage and found that TXNIP expression was significantly suppressed by GSK2656157 treatment (300 μg, p = 0.020, Fig. 5d–f and Additional file 4: Figure S4). Meanwhile, GSK2656157 treatment produced positive effect on protecting BBB permeability (Fig. 7c, p < 0.05). These results indirectly indicated that the protective effects of PERK inhibition were mediated by TXNIP downregulation, although we could not exclude the existence of other contributory mechanisms. Yan and colleagues recently found that PERK inhibition also exerts a neuroprotective effect through the activation of some protection signal factors [36]. That may explain why PERK inhibition showed no significant mortality decrease or improvement of neurological deficits in our work.
IRE1α is also a type 1 ER transmembrane protein with kinase and endoribonuclease activities that can induce XBP1 mRNA splicing through its endoribonuclease activity [37]. Interestingly, Lerner and colleagues recently reported that IRE1α can regulate TXNIP expression at the post-transcriptional level [15]. Thus, we speculated that IRE1α inhibition may also suppress TXNIP expression after SAH. STF083010 is an IRE1α-specific inhibitor that can completely inhibit IRE1α endoribonuclease-mediated XBP1 mRNA splicing [15, 29]. We found that STF-083010 inhibits IRE1α in a dose-dependent manner, at a minimal dose of 10 μg and with significant effect at 30 to 50 μg; the effect was reflected by XBP1 splicing (p = 0.000, Fig. 5g, h), an indirect measure, due to the difficulty of detecting IRE1α endoribonuclease activity [29]. Compared with the control group, animals treated with STF-083010 (50 μg) showed significantly inhibited TXNIP expression (p = 0.021) after SAH (Fig. 5i–j).
Above all, these results demonstrated that the inhibition of ER stress sensors may downregulate TXNIP expression after SAH. Therefore, these findings might indirectly suggest a potential link between ER stress, TXNIP and SAH.

The analogue signalling pathway of TXNIP mediated by ER stress after SAH

The signalling pathway of TXNIP mediated by ER stress may be a novel therapeutic target in SAH-related EBI. ER stress occurs after SAH, activating PERK and IRE1α that are both ER stress sensors, as well as the downstream coherent factors. TXNIP expression is closely mediated by PERK and IRE1α. Firstly, ER stress activates the PERK-eIF2α pathway through activating the sensor PERK and its downstream transcription factors, ATF-5 and ChREBP, which significantly promote the transcription of TXNIP. In addition, hyperactivated IRE1α improves TXNIP mRNA stability through selective degradation of TXNIP-destabilizing microRNA-17 by its endoribonuclease activity. TXNIP is ubiquitously present in vivo and is a natural antagonist of TRX, while TRX plays an important role in redox homeostasis regulation [38]. Under various stresses, TXNIP can directly bind to TRX and disturb the redox homeostasis and TRX/apoptosis signal-regulating kinase-1 (ASK-1) inhibitory complex, leading to the reactive oxygen species (ROS) release and autophosphorylation of ASK-1 which is responsible for apoptosis. At the same time, TXNIP has been confirmed as a critical link for NLRP3 inflammasome activation, activating downstream Caspase-1 and IL-1β. Through the above mechanisms, TXNIP participates in EBI after SAH by mediating neuroinflammation and apoptosis.

SAH severity and mortality

Because SAH grade and mortality are prognostic indicators, they were recorded after surgery and treatment. SAH grading scores showed no significant difference between the surgery and treatment groups (Fig. 6a, p = 0.987), indicating that the extent of haemorrhage after SAH was similar to both control and treatment groups of rats. Mortality occurred mainly within 24 h after surgery in our study. A total of 22 rats died during or immediately after surgery due to severe SAH. Twenty rats were excluded because of their low SAH grade. No death was observed in the sham group. After surgery, 59 rats died within 24 h, with mortality rates which were calculated as follows: sham = 0% (0 of 33), SAH = 25.0% (16 of 64), NS = 25.0% (6 of 24), RES = 18.1% (4 of 22), control siRNA = 21.7% (5 of 23), TXNIP siRNA = 14.3% (3 of 21), DMSO = 22.6% (7 of 31), GSK2656157 = 21.4% (9 of 42) and STF083010 = 23.1% (9 of 39). However, there was no statistically significant difference in mortality between treatment groups (TXNIP siRNA vs. control siRNA, p = 0.701; RES vs. NS, p = 0.725; GSK2656157 vs. DMSO, p = 0.772; STF083010 vs. DMSO, p = 0.743) based on Fisher’s two-sided exact test.

Neurological deficits

SAH significantly induced neurological dysfunction after surgery. Compared with the scores of the sham group (18.00 ± 0.00), the neurological scores of the SAH group were significantly lower at 24 and 72 h (24 h = 10.75 ± 0.37 and 72 h = 12.58 ± 0.36, n = 12, Fig. 7b, p < 0.05). However, there was no significant difference between groups (SAH, control siRNA, NS and DMSO groups, p > 0.05). Compared with the control group (24 h = 10.58 ± 0.36 and 72 h = 12.50 ± 0.38), neurological scores were improved after TXNIP siRNA treatment (24 h = 12.67 ± 0.40, p = 0.001 and 72 h = 15.00 ± 0.33, p = 0.000, Fig. 7b). The neurological scores of the RES treatment group (24 h = 12.50 ± 0.36, p = 0.016 and 72 h = 14.83 ± 0.39, p = 0.002) were higher than those of the control group (NS, 24 h = 11.16 ± 0.37 and 72 h = 12.83 ± 0.44, Fig. 7b). In contrast, there was no significant difference with GSK2656157 (24 h = 12.58 ± 0.36, p = 0.133 and 72 h = 15.58 ± 0.31, p = 0.068) or STF083010 (24 h = 11.91 ± 0.10 and 72 h = 14.42 ± 0.13, p = 0.160) compared with the control group (DMSO, 24 h = 11.18 ± 0.32 and 72 h = 14.58 ± 0.42, Fig. 7b). These results suggested that TXNIP inhibition could attenuate the neurological deficits of SAH rats, but PERK and IRE1α inhibition showed no significant improvement.

BBB permeability

Evans blue dye extravasation was used to measure BBB permeability. SAH caused remarkable BBB disruption in the cerebral hemispheres, the cerebellum (CB) and the brain stem (BS) at 72 h post-surgery (Fig. 7c, p < 0.05). Compared with that observed in the control group, TXNIP siRNA injection significantly prevented BBB disruption, as reflected by the reduced levels of Evans blue dye in the left hemisphere (LH), right hemisphere (RH) and CB at 72 h (Fig. 7c, p < 0.05). RES treatment also attenuated BBB disruption in three areas (LH, RH and CB, Fig. 7c, p < 0.05). Both GSK2656157 and STF083010 had a protective effect on BBB permeability compared with the disruption observed in the DMSO control group (Fig. 7c, p < 0.05). However, there was no significant difference between groups in the BS (Fig. 7c, p > 0.05). These results showed that the inhibition of TXNIP, PERK and IRE1α could prevent BBB disruption in different regions after SAH.

Brain water content

We found that SAH caused remarkably diffused brain swelling after surgery, especially at 72 h. Compared with that in the sham group, the brain water content was significantly worse in the LH, RH and CB at 72 h (Fig. 7d and Additional file 5: Table S1, p < 0.05). However, there is no significant difference observed in the BS between the control and treatment groups (Fig. 7d, p > 0.05). TXNIP siRNA injection significantly decreased the water content in the LH, RH and CB, when compared with rats in the control group (Fig. 7d and Additional file 5: Table S1, p < 0.05). SAH rats treated with RES showed a decrease in brain water content in the LH, RH and CB (Fig. 7d and Additional file 5: Table S1, p < 0.05). Brain water content was also significantly reduced by GSK2656157 and STF083010 treatment when compared with that of the control group (Fig. 7d and Additional file 5: Table S1, p < 0.05). These results demonstrated that TXNIP, PERK and IRE1α inhibition could reduce cerebral edema after SAH.

Discussion

The present study proves that both pharmacological inhibition and gene knockdown of TXNIP significantly attenuated brain injury and improved early prognosis after SAH. We found that these effects were closely associated with TXNIP pro-inflammatory and pro-apoptotic signalling pathways. TXNIP was extensively co-localized with TUNEL-positive cells in both the rat hippocampus and subcortex after SAH. We also found for the first time that TXNIP is localized in microglia and astrocytes. These results represent critical evidence to support the pro-inflammatory and pro-apoptotic effects of TXNIP after SAH. PERK and IRE1α inhibition resulted in significant TXNIP suppression and attenuation of some prognostic indicators. More generally, we once again showed the important role of ER stress in EBI after SAH, highlighting the tight relationships between SAH, TXNIP and ER stress.
SAH results in severe neurological deficits, and there are few therapeutic drug targets available. Inflammation and apoptosis are regarded as the main causes in EBI after SAH. Although suppressing inflammation is generally neuroprotective in animal experiments, little success has been seen in clinical trials to date. Numerous apoptotic proteins are increased during this process, and many anti-apoptotic strategies have been developed for EBI treatment. Nevertheless, the mortality and disability of SAH remain unacceptably high, reminding us that there are some unknown mechanisms involved. Several studies recently reported that TXNIP plays dual pro-inflammatory and pro-apoptotic roles in ischaemic diseases [23, 39], but the relationship between TXNIP and SAH has not been elucidated. Kaya and colleagues reported that SAH induced TXNIP mRNA expression [40]. Therefore, we speculated that TXNIP may also participate in the pathogenesis of EBI. We found that TXNIP protein levels were significantly elevated and expressed in microglia and astrocytes during the initial phases of our SAH experiments. As described above, the neural immunocytes (microglia and astrocytes) play a major role in promoting neuroinflammation and secondary brain damage [33, 34]. Excessive microglial activation can aggravate cerebral haemorrhage-induced brain injury by inflammatory amplification [41, 42]. Inflammatory mediators could further activate astrocytes to induce secondary inflammatory responses [43]. In combination with that NLRP3 inflammasome is reported to be expressed in microglia cells and participates in the inflammatory brain injury of SAH [44, 45], our results provide a supplementary histological evidence to support that TXNIP may play a possible effect on EBI after SAH through NLRP3 inflammasome activation and inflammatory amplification.
Recent studies have found that TXNIP can interact closely with the leucine-rich repeats (LRRs) of the NLRP3 inflammasome, further inducing Caspase-1 and IL-1β activation under stress conditions and eventually promoting inflammatory amplification [12]. The NLRP3 inflammasome, as the best characterized inflammasome, consists of an NLRP3 scaffold, apoptosis-associated speck-like protein containing a CARD (ASC) and pro-Caspase-1 [46]. Under stress conditions, NLRP3 recruits ASC and pro-Caspase-1, further causing Caspase-1 autocatalytic activation and the downstream transformation of pro-IL-1β and pro-IL-18 into their cleaved and secreted forms [46]. In addition, TXNIP can directly bind to the active cysteine residue of TRX and further disturb the TRX/ASK-1 inhibitory complex, leading to ASK-1 release and pro-apoptotic effects [47]. TRX1 is a TRX protein that interacts closely with the N-terminal portion of ASK-1 and inhibits ASK-1-dependent apoptosis [48]. Researchers have found that TRX inhibition can induce neuronal apoptosis after cerebral ischaemia [38]. Likewise, we found that TRX1 expression was inhibited after SAH, which may be due to the co-localization of TXNIP with TRX1 and inhibition of TRX1 transcription [38]. We also found that TXNIP was widely expressed in SAH rat brain neurons. More importantly, TUNEL staining revealed that TXNIP was widely co-localized with apoptotic cells in the hippocampus and cortex after SAH. Brain cell apoptosis was reduced, and prognostic indicators were attenuated with inhibition of TXNIP. These results could support our speculation that TXNIP participates in EBI after SAH.
Recently, Oslowski and colleagues found that PERK significantly promotes TXNIP expression via the ATF-5 and ChREBP transcription factors under ER stress [16]. It has also been shown that hyperactivated IRE1α improves TXNIP mRNA stability through selective degradation of TXNIP-destabilizing microRNA via IRE1α endoribonuclease activity, eventually promoting TXNIP expression at the post-transcriptional level. When ER stress reaches a certain threshold, IRE1α selectively degrades four pre-microRNAs, including four microRNAs (miR-17, miR-34a, miR-96 and miR-125b) [49]. Among these, miR-17 has been confirmed to function as a TXNIP mRNA-destabilizing microRNA [15]. There are also some other mechanisms involved in regulating TXNIP expression, such as the p38 MAPK and forkhead box O1 transcriptional factor (FOXO1) pathways [50]. Of course, more research is still needed to identify additional potential mechanisms of TXNIP and ER stress in SAH.
Meanwhile, there are also some limitations of this current study and were listed as follows: (1) firstly, we did not detect a dose-dependent change of the mRNA, protein and immunofluorescence levels of TXNIP and downstream-associated factors after treatment with TXNIP siRNA, RES, GSK2656157 and STF083010; (2) secondly, we only tested the PERK phosphorylation levels. And a more thorough method is to detect the total-PERK levels and then calculate the ratios between the phosphorylation of PERK and total PERK, which will provide more favourable evidence to support our results; (3) in addition, ER stress can also occur in other pro-inflammatory immune or supporting cell types of the neurovascular unit, such as oligodendrocytes, vascular smooth muscle and endothelial cells. It is also possible that ER stress might induce pro-inflammatory activation of other cell types, which then can mediate indirect activation of TXNIP in neuronal cells, microglia and astrocytes in a paracrine matter; and (4) our Western blot experiments showed that TXNIP expression was the most obvious at 48 h. However, the immunolocalization experiment in Fig. 2 and the follow-up Western blot study for TXNIP was not done at the same 48 h time point used. Due to these, we should choose 48 h as the best testing point, while, combined with our found clinical and experimental observations, clinical symptoms appear and are significant at 24 h in patients and animals after intracranial aneurysm bleeding and SAH. So at the beginning of the experiment design, we aimed to observe the immunofluorescence and protein detection at 24 h. At the same time, TXNIP expression at 24 h was also significant when compared with that in the sham group. Therefore, the results of immunofluorescence and protein detection at 24 h may also be powerful to support the conclusions. These are limitations of our study design, and more work needs to be done in the future to address them.

Conclusions

Our findings demonstrate that TXNIP participates in EBI after SAH by mediating neuroinflammation and apoptosis. The inhibition of TXNIP induced by ER stress alleviated SAH-induced inflammatory amplification and cell apoptosis and resulted in attenuation of some prognostic indicators, suggesting that TXNIP inhibition may represent a potential therapeutic strategy for SAH treatment. We speculate that the crosstalk between ER stress, apoptosis, inflammation and SAH will play an important role in SAH treatment and that TXNIP may be the critical link in these relationships. Of course, there are many questions that need to be addressed in our future work.

Acknowledgements

None.

Funding

The study was supported by research grants from the National Natural Science Foundation of China (no. 81371309) and Natural Science Foundation Project of Chongqing Science and Technology Commission (no. cstc2012jjA10109).

Availability of data and materials

The datasets generated and/or analysed during the current study are available from the corresponding author on reasonable request.

Authors’ contributions

QZ performed the study and wrote the paper. QZ, XC, HZ, JZ and XX participated in making the SAH model. QZ, HZ, GT, LL, DJ, JZ, XX and YL performed the Western blot, TUNEL and immunofluorescence staining. QZ and PF analysed the samples and data. QZ and ZH conceived and designed the study. ZH, XS and PF reviewed and edited the manuscript. All authors approved the final version of the manuscript.

Competing interests

The authors declare that they have no competing interests.
Not applicable.

Ethics approval

All animal experiments were conducted according to a protocol approved by the Animal Ethics and Use Committee of Chongqing Medical University (permit no. SCXK (Chongqing) 2007–0001).

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.
2.
Zurück zum Zitat Cahill J, Calvert JW, Zhang JH. Mechanisms of early brain injury after subarachnoid hemorrhage. J Cereb Blood Flow Metab. 2006;26:1341–53.CrossRefPubMed Cahill J, Calvert JW, Zhang JH. Mechanisms of early brain injury after subarachnoid hemorrhage. J Cereb Blood Flow Metab. 2006;26:1341–53.CrossRefPubMed
3.
5.
Zurück zum Zitat Ma C, Zhou W, Yan Z, Qu M, Bu X. Toll-like receptor 4 (TLR4) is correlated with delayed cerebral ischemia (DCI) and poor prognosis in aneurysmal subarachnoid hemorrhage. J Neurol Sci. 2015;359:67–71.CrossRefPubMed Ma C, Zhou W, Yan Z, Qu M, Bu X. Toll-like receptor 4 (TLR4) is correlated with delayed cerebral ischemia (DCI) and poor prognosis in aneurysmal subarachnoid hemorrhage. J Neurol Sci. 2015;359:67–71.CrossRefPubMed
6.
Zurück zum Zitat Rolland WB, Lekic T, Krafft PR, Hasegawa Y, Altay O, Hartman R, Ostrowski R, Manaenko A, Tang J, Zhang JH. Fingolimod reduces cerebral lymphocyte infiltration in experimental models of rodent intracerebral hemorrhage. Exp Neurol. 2013;241:45–55.CrossRefPubMed Rolland WB, Lekic T, Krafft PR, Hasegawa Y, Altay O, Hartman R, Ostrowski R, Manaenko A, Tang J, Zhang JH. Fingolimod reduces cerebral lymphocyte infiltration in experimental models of rodent intracerebral hemorrhage. Exp Neurol. 2013;241:45–55.CrossRefPubMed
8.
Zurück zum Zitat Wilms H, Sievers J, Rickert U, Rostami-Yazdi M, Mrowietz U, Lucius R. Dimethylfumarate inhibits microglial and astrocytic inflammation by suppressing the synthesis of nitric oxide, IL-1beta, TNF-alpha and IL-6 in an in-vitro model of brain inflammation. J Neuroinflammation. 2010;7:30.CrossRefPubMedPubMedCentral Wilms H, Sievers J, Rickert U, Rostami-Yazdi M, Mrowietz U, Lucius R. Dimethylfumarate inhibits microglial and astrocytic inflammation by suppressing the synthesis of nitric oxide, IL-1beta, TNF-alpha and IL-6 in an in-vitro model of brain inflammation. J Neuroinflammation. 2010;7:30.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Fang H, Wang PF, Zhou Y, Wang YC, Yang QW. Toll-like receptor 4 signaling in intracerebral hemorrhage-induced inflammation and injury. J Neuroinflammation. 2013;10:27.CrossRefPubMedPubMedCentral Fang H, Wang PF, Zhou Y, Wang YC, Yang QW. Toll-like receptor 4 signaling in intracerebral hemorrhage-induced inflammation and injury. J Neuroinflammation. 2013;10:27.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Brough D, Tyrrell PJ, Allan SM. Regulation of interleukin-1 in acute brain injury. Trends Pharmacol Sci. 2011;32:617–22.CrossRefPubMed Brough D, Tyrrell PJ, Allan SM. Regulation of interleukin-1 in acute brain injury. Trends Pharmacol Sci. 2011;32:617–22.CrossRefPubMed
11.
Zurück zum Zitat Broughton BR, Reutens DC, Sobey CG. Apoptotic mechanisms after cerebral ischemia. Stroke. 2009;40:e331–9.CrossRefPubMed Broughton BR, Reutens DC, Sobey CG. Apoptotic mechanisms after cerebral ischemia. Stroke. 2009;40:e331–9.CrossRefPubMed
12.
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
13.
Zurück zum Zitat Hwang J, Suh HW, Jeon YH, Hwang E, Nguyen LT, Yeom J, Lee SG, Lee C, Kim KJ, Kang BS, et al. The structural basis for the negative regulation of thioredoxin by thioredoxin-interacting protein. Nat Commun. 2014;5:2958.PubMedPubMedCentral Hwang J, Suh HW, Jeon YH, Hwang E, Nguyen LT, Yeom J, Lee SG, Lee C, Kim KJ, Kang BS, et al. The structural basis for the negative regulation of thioredoxin by thioredoxin-interacting protein. Nat Commun. 2014;5:2958.PubMedPubMedCentral
14.
Zurück zum Zitat He Z, Ostrowski RP, Sun X, Ma Q, Huang B, Zhan Y, Zhang JH. CHOP silencing reduces acute brain injury in the rat model of subarachnoid hemorrhage. Stroke. 2012;43:484–90.CrossRefPubMed He Z, Ostrowski RP, Sun X, Ma Q, Huang B, Zhan Y, Zhang JH. CHOP silencing reduces acute brain injury in the rat model of subarachnoid hemorrhage. Stroke. 2012;43:484–90.CrossRefPubMed
15.
Zurück zum Zitat Lerner AG, Upton J-P, Praveen P, Ghosh R, Nakagawa Y, Igbaria A, Shen S, Nguyen V, Backes BJ, Heiman M. IRE1α induces thioredoxin-interacting protein to activate the NLRP3 inflammasome and promote programmed cell death under irremediable ER stress. Cell Metab. 2012;16:250–64.CrossRefPubMedPubMedCentral Lerner AG, Upton J-P, Praveen P, Ghosh R, Nakagawa Y, Igbaria A, Shen S, Nguyen V, Backes BJ, Heiman M. IRE1α induces thioredoxin-interacting protein to activate the NLRP3 inflammasome and promote programmed cell death under irremediable ER stress. Cell Metab. 2012;16:250–64.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Oslowski CM, Hara T, O’Sullivan-Murphy B, Kanekura K, Lu S, Hara M, Ishigaki S, Zhu LJ, Hayashi E, Hui ST. Thioredoxin-interacting protein mediates ER stress-induced β cell death through initiation of the inflammasome. Cell Metab. 2012;16:265–73.CrossRefPubMedPubMedCentral Oslowski CM, Hara T, O’Sullivan-Murphy B, Kanekura K, Lu S, Hara M, Ishigaki S, Zhu LJ, Hayashi E, Hui ST. Thioredoxin-interacting protein mediates ER stress-induced β cell death through initiation of the inflammasome. Cell Metab. 2012;16:265–73.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Roussel BD, Kruppa AJ, Miranda E, Crowther DC, Lomas DA, Marciniak SJ. Endoplasmic reticulum dysfunction in neurological disease. Lancet Neurol. 2013;12:105–18.CrossRefPubMed Roussel BD, Kruppa AJ, Miranda E, Crowther DC, Lomas DA, Marciniak SJ. Endoplasmic reticulum dysfunction in neurological disease. Lancet Neurol. 2013;12:105–18.CrossRefPubMed
20.
Zurück zum Zitat Lindholm D, Wootz H, Korhonen L. ER stress and neurodegenerative diseases. Cell Death Differ. 2006;13:385–92.CrossRefPubMed Lindholm D, Wootz H, Korhonen L. ER stress and neurodegenerative diseases. Cell Death Differ. 2006;13:385–92.CrossRefPubMed
21.
Zurück zum Zitat Duris K, Manaenko A, Suzuki H, Rolland WB, Krafft PR, Zhang JH. α7 nicotinic acetylcholine receptor agonist PNU-282987 attenuates early brain injury in a perforation model of subarachnoid hemorrhage in rats. Stroke. 2011;42:3530–6.CrossRefPubMedPubMedCentral Duris K, Manaenko A, Suzuki H, Rolland WB, Krafft PR, Zhang JH. α7 nicotinic acetylcholine receptor agonist PNU-282987 attenuates early brain injury in a perforation model of subarachnoid hemorrhage in rats. Stroke. 2011;42:3530–6.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Zhou XM, Zhou ML, Zhang XS, Zhuang Z, Li T, Shi JX, Zhang X. Resveratrol prevents neuronal apoptosis in an early brain injury model. J Surg Res. 2014;189:159–65.CrossRefPubMed Zhou XM, Zhou ML, Zhang XS, Zhuang Z, Li T, Shi JX, Zhang X. Resveratrol prevents neuronal apoptosis in an early brain injury model. J Surg Res. 2014;189:159–65.CrossRefPubMed
23.
Zurück zum Zitat Ishrat T, Mohamed IN, Pillai B, Soliman S, Fouda AY, Ergul A, El-Remessy AB, Fagan SC. Thioredoxin-interacting protein: a novel target for neuroprotection in experimental thromboembolic stroke in mice. Mol Neurobiol. 2015;51:766–78.CrossRefPubMed Ishrat T, Mohamed IN, Pillai B, Soliman S, Fouda AY, Ergul A, El-Remessy AB, Fagan SC. Thioredoxin-interacting protein: a novel target for neuroprotection in experimental thromboembolic stroke in mice. Mol Neurobiol. 2015;51:766–78.CrossRefPubMed
24.
Zurück zum Zitat Bedarida T, Baron S, Vibert F, Ayer A, Henrion D, Thioulouse E, Marchiol C, Beaudeux JL, Cottart CH, Nivet-Antoine V. Resveratrol decreases TXNIP mRNA and protein nuclear expressions with an arterial function improvement in old mice. J Gerontol A Biol Sci Med Sci. 2016;71:720–9.CrossRefPubMed Bedarida T, Baron S, Vibert F, Ayer A, Henrion D, Thioulouse E, Marchiol C, Beaudeux JL, Cottart CH, Nivet-Antoine V. Resveratrol decreases TXNIP mRNA and protein nuclear expressions with an arterial function improvement in old mice. J Gerontol A Biol Sci Med Sci. 2016;71:720–9.CrossRefPubMed
25.
Zurück zum Zitat Xiao J, Zhu Y, Liu Y, Tipoe GL, Xing F, So K-F. Lycium barbarum polysaccharide attenuates alcoholic cellular injury through TXNIP-NLRP3 inflammasome pathway. Int J Biol Macromol. 2014;69:73–8.CrossRefPubMed Xiao J, Zhu Y, Liu Y, Tipoe GL, Xing F, So K-F. Lycium barbarum polysaccharide attenuates alcoholic cellular injury through TXNIP-NLRP3 inflammasome pathway. Int J Biol Macromol. 2014;69:73–8.CrossRefPubMed
26.
Zurück zum Zitat Luo B, Li B, Wang W, Liu X, Xia Y, Zhang C, Zhang M, Zhang Y, An F. NLRP3 gene silencing ameliorates diabetic cardiomyopathy in a type 2 diabetes rat model. PLoS One. 2014;9, e104771.CrossRefPubMedPubMedCentral Luo B, Li B, Wang W, Liu X, Xia Y, Zhang C, Zhang M, Zhang Y, An F. NLRP3 gene silencing ameliorates diabetic cardiomyopathy in a type 2 diabetes rat model. PLoS One. 2014;9, e104771.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Ron D, Walter P. Signal integration in the endoplasmic reticulum unfolded protein response. Nat Rev Mol Cell Biol. 2007;8:519–29.CrossRefPubMed Ron D, Walter P. Signal integration in the endoplasmic reticulum unfolded protein response. Nat Rev Mol Cell Biol. 2007;8:519–29.CrossRefPubMed
28.
Zurück zum Zitat Atkins C, Liu Q, Minthorn E, Zhang SY, Figueroa DJ, Moss K, Stanley TB, Sanders B, Goetz A, Gaul N, et al. Characterization of a novel PERK kinase inhibitor with antitumor and antiangiogenic activity. Cancer Res. 2013;73:1993–2002.CrossRefPubMed Atkins C, Liu Q, Minthorn E, Zhang SY, Figueroa DJ, Moss K, Stanley TB, Sanders B, Goetz A, Gaul N, et al. Characterization of a novel PERK kinase inhibitor with antitumor and antiangiogenic activity. Cancer Res. 2013;73:1993–2002.CrossRefPubMed
29.
Zurück zum Zitat Papandreou I, Denko NC, Olson M, Van Melckebeke H, Lust S, Tam A, Solow-Cordero DE, Bouley DM, Offner F, Niwa M. Identification of an Ire1alpha endonuclease specific inhibitor with cytotoxic activity against human multiple myeloma. Blood. 2011;117:1311–4.CrossRefPubMedPubMedCentral Papandreou I, Denko NC, Olson M, Van Melckebeke H, Lust S, Tam A, Solow-Cordero DE, Bouley DM, Offner F, Niwa M. Identification of an Ire1alpha endonuclease specific inhibitor with cytotoxic activity against human multiple myeloma. Blood. 2011;117:1311–4.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Sugawara T, Ayer R, Jadhav V, Zhang JH. A new grading system evaluating bleeding scale in filament perforation subarachnoid hemorrhage rat model. J Neurosci Methods. 2008;167:327–34.CrossRefPubMed Sugawara T, Ayer R, Jadhav V, Zhang JH. A new grading system evaluating bleeding scale in filament perforation subarachnoid hemorrhage rat model. J Neurosci Methods. 2008;167:327–34.CrossRefPubMed
31.
Zurück zum Zitat Tsubokawa T, Solaroglu I, Yatsushige H, Cahill J, Yata K, Zhang JH. Cathepsin and calpain inhibitor E64d attenuates matrix metalloproteinase-9 activity after focal cerebral ischemia in rats. Stroke. 2006;37:1888–94.CrossRefPubMed Tsubokawa T, Solaroglu I, Yatsushige H, Cahill J, Yata K, Zhang JH. Cathepsin and calpain inhibitor E64d attenuates matrix metalloproteinase-9 activity after focal cerebral ischemia in rats. Stroke. 2006;37:1888–94.CrossRefPubMed
32.
Zurück zum Zitat Suzuki H, Hasegawa Y, Kanamaru K, Zhang JH. Mechanisms of osteopontin-induced stabilization of blood-brain barrier disruption after subarachnoid hemorrhage in rats. Stroke. 2010;41:1783–90.CrossRefPubMedPubMedCentral Suzuki H, Hasegawa Y, Kanamaru K, Zhang JH. Mechanisms of osteopontin-induced stabilization of blood-brain barrier disruption after subarachnoid hemorrhage in rats. Stroke. 2010;41:1783–90.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Kreutzberg GW. Microglia: a sensor for pathological events in the CNS. Trends Neurosci. 1996;19:312–8.CrossRefPubMed Kreutzberg GW. Microglia: a sensor for pathological events in the CNS. Trends Neurosci. 1996;19:312–8.CrossRefPubMed
34.
Zurück zum Zitat Farina C, Aloisi F, Meinl E. Astrocytes are active players in cerebral innate immunity. Trends Immunol. 2007;28:138–45.CrossRefPubMed Farina C, Aloisi F, Meinl E. Astrocytes are active players in cerebral innate immunity. Trends Immunol. 2007;28:138–45.CrossRefPubMed
35.
Zurück zum Zitat Zhang XS, Li W, Wu Q, Wu LY, Ye ZN, Liu JP, Zhuang Z, Zhou ML, Zhang X, Hang CH. Resveratrol attenuates acute inflammatory injury in experimental subarachnoid hemorrhage in rats via inhibition of TLR4 pathway. Int J Mol Sci. 2016;17. Zhang XS, Li W, Wu Q, Wu LY, Ye ZN, Liu JP, Zhuang Z, Zhou ML, Zhang X, Hang CH. Resveratrol attenuates acute inflammatory injury in experimental subarachnoid hemorrhage in rats via inhibition of TLR4 pathway. Int J Mol Sci. 2016;17.
36.
Zurück zum Zitat Yan F, Cao S, Li J, Dixon B, Yu X, Chen J, Gu C, Lin W, Chen G. Pharmacological inhibition of PERK attenuates early brain injury after subarachnoid hemorrhage in rats through the activation of Akt. Mol Neurobiol. 2016. Yan F, Cao S, Li J, Dixon B, Yu X, Chen J, Gu C, Lin W, Chen G. Pharmacological inhibition of PERK attenuates early brain injury after subarachnoid hemorrhage in rats through the activation of Akt. Mol Neurobiol. 2016.
37.
Zurück zum Zitat Calfon M, Zeng H, Urano F, Till JH, Hubbard SR, Harding HP, Clark SG, Ron D. IRE1 couples endoplasmic reticulum load to secretory capacity by processing the XBP-1 mRNA. Nature. 2002;415:92–6.CrossRefPubMed Calfon M, Zeng H, Urano F, Till JH, Hubbard SR, Harding HP, Clark SG, Ron D. IRE1 couples endoplasmic reticulum load to secretory capacity by processing the XBP-1 mRNA. Nature. 2002;415:92–6.CrossRefPubMed
38.
Zurück zum Zitat Yoshihara E, Masaki S, Matsuo Y, Chen Z, Tian H, Yodoi J. Thioredoxin/Txnip: redoxisome, as a redox switch for the pathogenesis of diseases. Front Immunol. 2014;4:514.CrossRefPubMedPubMedCentral Yoshihara E, Masaki S, Matsuo Y, Chen Z, Tian H, Yodoi J. Thioredoxin/Txnip: redoxisome, as a redox switch for the pathogenesis of diseases. Front Immunol. 2014;4:514.CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Kim GS, Jung JE, Narasimhan P, Sakata H, Chan PH. Induction of thioredoxin-interacting protein is mediated by oxidative stress, calcium, and glucose after brain injury in mice. Neurobiol Dis. 2012;46:440–9.CrossRefPubMedPubMedCentral Kim GS, Jung JE, Narasimhan P, Sakata H, Chan PH. Induction of thioredoxin-interacting protein is mediated by oxidative stress, calcium, and glucose after brain injury in mice. Neurobiol Dis. 2012;46:440–9.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Kaya B, Erdi F, Kilinc I, Keskin F, Feyzioglu B, Esen H, Karatas Y, Uyar M, Kalkan E. Alterations of the thioredoxin system during subarachnoid hemorrhage-induced cerebral vasospasm. Acta Neurochir (Wien). 2015;157:793–9. discussion 799–800.CrossRef Kaya B, Erdi F, Kilinc I, Keskin F, Feyzioglu B, Esen H, Karatas Y, Uyar M, Kalkan E. Alterations of the thioredoxin system during subarachnoid hemorrhage-induced cerebral vasospasm. Acta Neurochir (Wien). 2015;157:793–9. discussion 799–800.CrossRef
41.
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
42.
Zurück zum Zitat Gao Z, Wang J, Thiex R, Rogove AD, Heppner FL, Tsirka SE. Microglial activation and intracerebral hemorrhage. Acta Neurochir Suppl. 2008;105:51–3.CrossRefPubMed Gao Z, Wang J, Thiex R, Rogove AD, Heppner FL, Tsirka SE. Microglial activation and intracerebral hemorrhage. Acta Neurochir Suppl. 2008;105:51–3.CrossRefPubMed
43.
Zurück zum Zitat Saijo K, Winner B, Carson CT, Collier JG, Boyer L, Rosenfeld MG, Gage FH, Glass CK. A Nurr1/CoREST pathway in microglia and astrocytes protects dopaminergic neurons from inflammation-induced death. Cell. 2009;137:47–59.CrossRefPubMedPubMedCentral Saijo K, Winner B, Carson CT, Collier JG, Boyer L, Rosenfeld MG, Gage FH, Glass CK. A Nurr1/CoREST pathway in microglia and astrocytes protects dopaminergic neurons from inflammation-induced death. Cell. 2009;137:47–59.CrossRefPubMedPubMedCentral
44.
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
45.
Zurück zum Zitat Shao A, Wu H, Hong Y, Tu S, Sun X, Wu Q, Zhao Q, Zhang J, Sheng J. Hydrogen-rich saline attenuated subarachnoid hemorrhage-induced early brain injury in rats by suppressing inflammatory response: possible involvement of NF-kappaB pathway and NLRP3 inflammasome. Mol Neurobiol. 2016;53:3462–76.CrossRefPubMed Shao A, Wu H, Hong Y, Tu S, Sun X, Wu Q, Zhao Q, Zhang J, Sheng J. Hydrogen-rich saline attenuated subarachnoid hemorrhage-induced early brain injury in rats by suppressing inflammatory response: possible involvement of NF-kappaB pathway and NLRP3 inflammasome. Mol Neurobiol. 2016;53:3462–76.CrossRefPubMed
47.
Zurück zum Zitat Saxena G, Chen J, Shalev A. Intracellular shuttling and mitochondrial function of thioredoxin-interacting protein. J Biol Chem. 2010;285:3997–4005.CrossRefPubMed Saxena G, Chen J, Shalev A. Intracellular shuttling and mitochondrial function of thioredoxin-interacting protein. J Biol Chem. 2010;285:3997–4005.CrossRefPubMed
48.
Zurück zum Zitat Saitoh M, Nishitoh H, Fujii M, Takeda K, Tobiume K, Sawada Y, Kawabata M, Miyazono K, Ichijo H. Mammalian thioredoxin is a direct inhibitor of apoptosis signal-regulating kinase (ASK) 1. Embo j. 1998;17:2596–606.CrossRefPubMedPubMedCentral Saitoh M, Nishitoh H, Fujii M, Takeda K, Tobiume K, Sawada Y, Kawabata M, Miyazono K, Ichijo H. Mammalian thioredoxin is a direct inhibitor of apoptosis signal-regulating kinase (ASK) 1. Embo j. 1998;17:2596–606.CrossRefPubMedPubMedCentral
49.
Zurück zum Zitat Upton J-P, Wang L, Han D, Wang ES, Huskey NE, Lim L, Truitt M, McManus MT, Ruggero D, Goga A. IRE1α cleaves select microRNAs during ER stress to derepress translation of proapoptotic Caspase-2. Science. 2012;338:818–22.CrossRefPubMedPubMedCentral Upton J-P, Wang L, Han D, Wang ES, Huskey NE, Lim L, Truitt M, McManus MT, Ruggero D, Goga A. IRE1α cleaves select microRNAs during ER stress to derepress translation of proapoptotic Caspase-2. Science. 2012;338:818–22.CrossRefPubMedPubMedCentral
50.
Zurück zum Zitat Li X, Rong Y, Zhang M, Wang XL, LeMaire SA, Coselli JS, Zhang Y, Shen YH. Up-regulation of thioredoxin interacting protein (Txnip) by p38 MAPK and FOXO1 contributes to the impaired thioredoxin activity and increased ROS in glucose-treated endothelial cells. Biochem Biophys Res Commun. 2009;381:660–5.CrossRefPubMedPubMedCentral Li X, Rong Y, Zhang M, Wang XL, LeMaire SA, Coselli JS, Zhang Y, Shen YH. Up-regulation of thioredoxin interacting protein (Txnip) by p38 MAPK and FOXO1 contributes to the impaired thioredoxin activity and increased ROS in glucose-treated endothelial cells. Biochem Biophys Res Commun. 2009;381:660–5.CrossRefPubMedPubMedCentral
Metadaten
Titel
Thioredoxin-interacting protein links endoplasmic reticulum stress to inflammatory brain injury and apoptosis after subarachnoid haemorrhage
verfasst von
Qing Zhao
Xudong Che
Hongxia Zhang
Pianpian Fan
Guanping Tan
Liu Liu
Dengzhi Jiang
Jun Zhao
Xiang Xiang
Yidan Liang
Xiaochuan Sun
Zhaohui He
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
Journal of Neuroinflammation / Ausgabe 1/2017
Elektronische ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-017-0878-6

Weitere Artikel der Ausgabe 1/2017

Journal of Neuroinflammation 1/2017 Zur Ausgabe

Neu in den Fachgebieten Neurologie und Psychiatrie