Animals and surgery
Male Sprague-Dawley rats (230‒270 g) were purchased from the Experimental Animal Center of Nantong University (Nantong, China) and housed in an animal room under controlled temperature (23 ± 2 °C) and a 12-h/12-h light/dark cycle with free access to water and food. All experimental procedures were approved by the Experimental Animal Ethics Committee of Nantong University. Traumatic brain injury was induced by controlled cortical impact (CCI) using air pressure [19, 20]. Briefly, rats were anesthetized with 10% chloral hydrate, fixed in a stereotactic frame, and subjected to right parietal craniotomy with a dental drill (3 mm posterior to bregma, 3 mm from the midline, diameter of 6 mm) to expose the underlying dura mater. Injury was induced using the TBI-0310 impactor device (Precision Systems and Instrumentation, USA) with the following impact parameters: speed 4 m/s, depth 3 mm, and impact time 150 ms. After injury, the skull skin was sterilized with iodophor. The rat was then placed next to a heater until it regained consciousness before return to the home cage. A sham operation group underwent the same surgical procedure but did not receive experimental TBI.
Culture of primary cortical astrocytes
Primary astrocyte cultures were prepared from the cerebral cortices of neonatal rats [11, 21, 22] purchased from the Experimental Animal Center of Nantong University. Bilateral cerebral cortices were isolated and transferred to ice-cold D-Hank’s buffer. The meninges were carefully removed and the remaining tissues dissociated, filtered through nylon mesh (pore size of 100 µm), and collected by centrifugation at ~3000 × g for 5 min. The cell pellet was dispersed with a pipette and resuspended in medium containing in low-glucose Dulbecco's Modified Eagle's Medium (DMEM, Life Technologies, Gibco BRL Division, Grand Island, NY, USA) supplemented with 10% fetal bovine serum (FBS). The cells were then triturated using glass pipettes, filtered through a screen with 10-µm pores, seeded on 6-well plates at a density of 2.5 × 105 cells/cm2, and cultured for approximately 10 days with medium replacement every 3 days. Dibutyryl cAMP sodium salt (0.15 mM; Sigma-Aldrich, St. Louis, MO, USA) was added to induce morphological and functional differentiation. The cells were used for experiments 3 days after reaching 90% confluence. Prior to experiments, the medium was replaced with Opti-MEM (Life Technologies, Inc-Gibco BRL Division). The cells were incubated with LPS (1 μg/mL; Santa Cruz Biotechnology, Dallas, TX, USA) for 1 to 6 h and expression levels of inflammatory signaling factors measured by ELISA and western blotting as described below.
Adeno-associated virus injection
We constructed an adeno-associated virus serotype 9 (AAV9) vector expressing TRAF6-specific RNAi (AAV9-TRAF6-RNAi) under control of astrocyte (1.36E+13 v.g./mL) and neuronal promoter (1.22E+13 v.g./mL), and an empty AAV9 vector (AAV-NC) (1.64E+13 v.g/mL) as a negative control according to the manufacturer's protocol (Jikai Gene, Shanghai, China). Male Sprague-Dawley rats (60‒70 g) were randomly divided into 4 groups: (1) sham, (2) TBI, (3) adeno-associated virus negative control (AAV9-NC) with TB, and (4) interference adeno-associated virus (AAV9-TRAF6-RNAi) with TBI. Rats were anesthetized by intraperitoneal injection of 10% chloral hydrate, shaved, sterilized, and fixed on a brain stereotaxic device. The forehead skin was cut along the midline and 3 holes drilled on the right side of the parietal bone with a dental drill (1.5 mm posterior to bregma, 1.5 mm lateral to the midline; 1.5 mm posterior to bregma, 3 mm lateral to the midline; 3 mm posterior to bregma, 1.5 mm lateral to the midline) through which 2 μL of viral solution was injected per hole using Hamilton microliter syringes. Injection depth was 1.3 mm and injection speed was 0.2 μL/min. The needle was kept in place for 8 minutes to insure full vector delivery. The TBI model was created as described 4 weeks after virus injection.
Drugs and Administration
Selective inhibitors of NF-κB, ERK, JNK, and p38 were purchased from Calbiochem (Merck, Darmstadt, Germany) and dissolved in dimethyl sulfoxide (DMSO). One hour after TBI, the injured area was treated with NF-κB inhibitor (BAY117082), ERK inhibitor (PD98059), JNK inhibitor (SP600125), or p38 inhibitor (SB203580) at 25 mg/10 mL (high dose) or 2.5 mg/10 mL (low dose) as indicated. Alternatively, corresponding control (vehicle) subgroups received 10 ml PBS+DMSO. All injections were performed while the rat was fixed to a stereotaxic frame using 10 mL Hamilton microliter syringes. Drugs and solvent were injected at 2 mL/min (requiring about 5 min) and the needle was kept in place for 5 minutes. Rats received the indicated injections for 3 consecutive days. After TBI, the surrounding damaged cerebral cortex tissue was collected for analysis of inflammatory factor expression levels by enzyme-linked immunosorbent assay (ELISA) or real-time quantitative PCR (RT-qPCR).
Astrocytes were transfected with a small interfering (si)RNA against TRAF6 (Guangzhou RiboBio Co., Ltd., Guangzhou, China) or negative control (NC) siRNA for 72 h and then stimulated with LPS. Afterward, the cells were collected for analysis by ELISA or western blotting.
Immunofluorescence
Rats were anesthetized by intraperitoneal injection of 10% chloral hydrate, perfused through the heart with normal saline until the liver became white, and then perfused with 4% paraformaldehyde. The whole brain was post-fixed overnight in 4% paraformaldehyde, dehydrated in 20% and 30% sucrose solutions, and then coronally cryo-sectioned to 20-µm slices. The slices were first blocked with 1% bovine serum albumin (BSA) for 2 h at room temperature and incubated overnight at 4 °C with mouse anti-TRAF6 monoclonal antibody (sc-8409, 1:200, Santa Cruz Biotechnology) plus the astrocyte marker rabbit anti-GFAP monoclonal antibody (ab7260, 1:500, abcam), the microglial marker goat anti-IBA-1 monoclonal antibody (ab5076,1:500, abcam), or the neuronal marker rabbit anti-NeuN monoclonal antibody (ab177487, 1:500, abcam). Slices were then incubated at room temperature with Cy3-conjugated and Alexa 488-conjugated secondary antibodies (1:1000, Jackson ImmunoResearch, West Grove, PA) for 2 h, and examined under a fluorescence microscope (Nikon).
Morris water maze test
Three days before TBI modeling, the rats received successive adaptive training trials in the Morris water maze to eliminate the influences of vision and motor function on performance. Place navigation learning was tested starting on the 3rd day after TBI and the spatial probe trial for spatial memory was conducted 24 hours later [10, 23]. Escape latency to the platform was measured on each of four daily learning trials and averaged. We also recorded swim paths during each trial. If the rat did not reach the platform within 120 s, it was guided to the platform and allowed to stay for 30 s. The escape latency in such cases was recorded as 120 s. The probe trial examined spatial memory for the original platform location by recording the number of crossings over the former platform location after release from the quadrant opposite the target quadrant. Two probe trials were conducted and the average recorded for analysis.
TUNEL
Terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) staining was performed to detect DNA fragmentation as an index of programmed cell death in brain tissues using an apoptosis detection kit (A113, Vazyme, Nanjing, China) according to the manufacturer's protocol. The red fluorescence of fluorescein-12-dUTP was detected against the blue background of 4′,6-diamidino-2-phenylindole (DAPI) under a fluorescence microscope at 20× magnification. The TUNEL-positive cells in the injured cortex were identified and counted in nine 20-μm thick slices per brain.
Real-time fluorescence quantitative PCR
Rats were anesthetized with 10% chloral hydrate and then perfused via the heart with PBS until the liver turned white. The cerebral cortex around the injured area was excised and total RNA extracted using Trizol. The RNAs were then reversed transcribed to cDNA. Real-time PCR was performed in a Step One Plus real-time PCR instrument using the primer sequences shown in Table 1 and the following PCR amplification conditions: pre-denaturation at 95 °C for 3 min; 40 cycles of 95 °C for 10 s and 60 °C for 30 s; dissolution at 95 °C for 15 s, 60 °C for 60 s, and 95 °C for 15 s. Expression of GAPDH served as the internal control and gene expression level was determined using the 2-ΔΔCT method.
Table 1 Primer sequences
Genes
|
Primers
|
Sequences
|
GAPDH
|
Forward
|
5′-TCCTACCCCCAATGTATCCG-3′
|
|
Reverse
|
5′-CCTTTAGTGGGCCCTCGG-3′
|
CCL2
|
Forward
|
5′-TGCTGCTACTCATTCACTGGC-3′
|
|
Reverse
|
5′-CCTTATTGGGGTCAGCACAG-3′
|
CCR2
|
Forward
|
5′-TGCTACTCAGGAATCCTCCACAC-3′
|
|
Reverse
|
5′-GGCCTGGTCTAAGTGCATGTCAAC-3′
|
CXCL1
|
Forward
|
5′-GCACCCAAACCGAAGTCATA-3′
|
|
Reverse
|
5′-GGGGACACCCTTTAGCATCT-3′
|
CXCR2
|
Forward
|
5′-TGGTCCTCGTCTTCCTGCTCTG-3′
|
|
Reverse
|
5′-CGTTCTGGCGTTCACAGGTCTC-3′
|
Western blot analysis
Rats were perfused as described for RT-qPCR analysis. 250ul RIPA lysate was added to brain tissues and the tissue samples were homogenized on ice using an electric homogenizer. After homogenization, they were allowed to stand on ice for 30 minutes and centrifuged at 4°C, 15000 rpm for 20 minutes, and the protein supernatant was taken. Total protein concentrations measured by the BCA Protein Assay (Pierce, Rockford, IL). Proteins (30 μg per gel lane) were separated by electrophoresis and transferred to PVDF membranes. Membranes were blocked with 5% skim-milk and incubated overnight at 4 °C with mouse anti-TRAF6 polyclonal antibody (sc-8409,1:500, Santa Cruz), rabbit anti-p-NF-κB polyclonal antibody (3033, 1:1000, Cell Signaling Technology, Danvers, MA, USA), rabbit anti-p-ERK polyclonal antibody (9101, 1:1000, Cell Signaling Technology), rabbit anti-p-JNK polyclonal antibody (4688,1:1000, Cell Signaling Technology), rabbit anti-p-p38 polyclonal antibody (9211,1:1000, Cell Signaling Technology), and (or) mouse anti-GAPDH polyclonal antibody (MAB374,1:10000, Millipore, Billerica, MA). The next day, membranes were incubated with IRDye 800CW secondary antibodies for 2 h at room temperature and immunolabeling captured using an Odyssey Imaging System (LI-COR Bioscience, Lincoln, NE). The average optical density of each protein band was measured using ImageJ (NIH, Bethesda, MD). Target protein expression is normalized to GAPDH expression as the gel loading control.
ELISA
Cell proteins were prepared as described for western blotting and added at 100 μg/well to 96-well plates prepared for ELISA according to the kit manufacturers’ instructions (below). The absorbance of each well at 450 nm was measured and target protein concentrations calculated according to standard curves prepared by the dilution of standards. A rat CXCL1 ELISA kit was purchased from Hangzhou MultiSciences (Lianke) Biotech (EK396/2-96, Hangzhou, Zhejiang, China), a rat CCL2 ELISA kit from R&D Systems (MJE00, Minneapolis, MN, USA), a rat CCR2 ELISA kit from CUSABIO TECHNOLOGY (CSB-EL004841RA, Wuhan, Hubei, China), and a rat CXCR2 ELISA kit from Cloud-Clone Corp (SEC006Ra, Katy, TX, USA).
Statistical methods
All measurement data are expressed as mean ± SEM. Multiple group means were compared by one-way ANOVA with post hoc Bonferroni correction. A P<0.05 (two-tailed) was considered significant for all tests. All statistical analyses were conducted using GraphPad Prism 5.0 (San Diego, CA, USA).