Proteins Active proteins (IKKα, IKKβ and Aurora kinase C; Signalchem, Vanier Place Richmond, BC,Canada), kinase buffer I and ATP stock solution were purchased from signalchem. Halo-Tag plasmidvector (pFN18A), Restriction enzyme blend (Sgf + Pme ), KRX expression cell, L-rhamnose monohydrate and sequencing-grade modified trypsin were purchased from Promega (Madison, WI). Quikchange lightning site-directed mutagenesis kit was purchased from Agilent technologies. D- glucose and phosphoric acid (PA) were purchased from Sigma-Aldrich. 2,5-Dihydroxybenzoic acid (2,5-DHB) was purchased from Bruker Daltonics. Acetonitrile (ACN, ultrapure ACS reagent grade and methyl alcohol (high purity HPLC reagent) were purchased from USB corporation and samchun pure chemical. All other chemicals used were of ACS or HPLC grade. Primary antibody (anti-IκBα monoclonal antibody) and anti-mouse secondary antibody were purchased from Santa Cruz Biotechnology and Cell signaling Technology. Primary antibody (anti-phospho-IκBα serine32; Cell Signaling Technology, CA, USA, serine63 and serine262 monoclonal antibody; Custom made antibody) and anti-rabbit secondary antibody were purchased from abcam and Cell signaling Technology. Primer and DNA sequencing data were purchased cosmo genetech (Seoul, Korea).
Cloning and mutagenesis of IκBα IκBα constructs were cloned into the Halo-Tag vector pFN18A (Promega) for E.coli overexpression between the restriction sites Sgf and Pme . Mutagenesis reactions of the IκBα single and double point mutant were performed with the Quikchange lightning site-directed mutagenesis kit (Agilent technologies). The following oligonucleotides were used as sense primers:S32A, 5’-CACGACGCCGGCCTG-3’; S36A, 5’-CTGGACGCCATGAAA-3’; S32/36A, 5’-CACGACGCCGGCCTGGACGCCATGAA-3’. The C-terminal deletion mutants were by insertion ofnewGAAGCAGCAGCTCACCGAGTGAGTTTAAACGAATTCGGGCT-3’;CGCACCTCCACTCCATCTGAGTTTAAACGAATTCGGGCT-3’; IκBα stop codon with following PCR antisense primers: IκBα 1-72, 5’-1-175,1-277,CTGACACTAGAAAACCTTTGAGTTTAAACGAATTCGGGCT-3’. The construction of the ankyrinrepeat domain deleted IκBα 17 was used overlap PCR method with following PCR antisence and sensepromers: IκBα 1-72/278-317, 5’-GAAGCAGCAGCTCACCGAGCAGATGCTGCCAGAGAGTG-3’, 5’-GAAGCAGCAGCTCACCGAGCAGATGCTGCCAGAGAGTG-3’. The mutated PCR fragments also were cloned into E.coli overexpression vector pFN18A (Promega).
Overexpression of IκBα in KRX cell Full-length human IκBα was cloned into a pFN18A vector (Promega). The plasmid is transformed into KRX cell (Promega) grown in LB media (add to ampicillin) supplemented with glucose and rhamnose to induce expression without isopropyl-β-D- thiogalactoside (IPTG). After growing the cells to OD 600 of 0.5-0.6 at 37 °C, the temperature was reduced to 20 °C for overnight expression.
Purification of IκBα For IκBα purification, Cell pellets were resuspended in 5 ㎖ of HaloTag purification buffer (50 mM HEPES (Sigma-aldrich), pH 7.5 and 150 mM NaCl) supplemented with 1x Protease Inhibitor (Thermo scientific), and sonicated on ice using a vibra cell sonicator (5 min total time on; 1 min on / 1 min off; amplitude = 40-60, pulse = 4). Lysates were centrifuged at 10,000 ×g for 30 min at 4 °C and the supernatants were directly applied onto pre-equilibrated HaloLink resin (Promega) following manufacturer recommendations. Binding to the resin was conducted at room temperature for 1 hr with constant end-over-end rotation, followed by three washes, each for 5 min with 10 ㎖ purification buffer. Target proteins were released from the resin by proteolytic cleavage using a ratio of HaloTEV to settled resin in HaloTag purification buffer for 1 hr at room temperature. The supernatants containing the released protein of interest and TEV protease were carefully removed into new tube. For TEV protease removal, add of HisLink resin into the tube. Binding to the resin was conducted at room temperature for 20 min with constant end-over-end rotation. Spin at 1,000 ×g for 5 min, and transfer supernatant to another tube. protein quantification was conducted by the Bradford method.[10] Bovine serum albumin was used for the calibration of sample protein quantity.
Cell culture. MCF-7(KCLB60104, Korean Cell line Bank) and MDA-MB 231 (KCLB88064, Korean Cell line Bank) cells were cultured at 37°C with 5% CO2 in Dulbecco’s Modified Eagle Medium (DMEM; Gibco, Grand Island, NY) supplemented with 10% fetal bovine serum (FBS; Gibco, CA, USA), 4.5 g/L glucose, L-glutamine, and 1% penicillin/streptomycin.
Staining or Western Blot analysis For in-gel trypsin digestion, the gels were stained by Coomassie brilliant Blue-R250 (CBB, 3M co.). For deletion mutant of IκBα size visualization, the gels were stained by silver staining (GE Healthcare, uppsala, Sweden). For Western blotting, the protein separated by electrophoresis were transferred to 0.2 ㎛ Polyvinylidene fluoride (PVDF; Bio-Rad Laboratories Inc.)membranes in Transfer buffer (25 mM Tris, 192mM Glycine, 20% (v/v) methanol) at 48 mA for 12 hr using Mini Trans-Blot Cell system (Bio-Rad Laboratories Inc.). in order to confirm the protein expression level as aging is processing, the transferred membrane was incubated in 5% (W/V) skim milk with 50 mM Tris-buffered saline with Tween 20 (TBST, Intron biotechnology, 24.7 mM Tris, pH 7.8, 2.7 mM KCl, 137 mM NaCl and 0.05% Tween-20) for blocking, and exposed to primary antibody (RIP3, MLKL, Caspase-3, Caspase-8, beta-actin: Cell Signaling Technology, CA, USA) overnight at 4 ℃. After washing step (three times for 15 min with fresh TBST), the membrane was exposed to secondary antibody for 2 hr at room temperature. After washing step, the proteins were visualized using ECL solution and detected by ChemiDoc XRS+ system (Bio-Rad Laboratories Inc.). The transcription activity of p65 was measured by enzyme-linked immunosorbent assay (ELISA; R&D Systems, Mineapolis, MN, USA), performed according to the manufacturer’s instructions. In the NF-κB -p65 assay, a double stranded DNA sequence containing the NF-κB response element was immobilized onto the bottom of the wells of a 96-well plate. NF-κB contained in a nuclear extract binds to the NF-κB response element, and then it was detected using an anti NF-κB p65 antibody. A secondary antibody conjugated to HRP was added to provide a colorimetric readout at 450 nm.
In vitro kinase assay Recombinant active AURK (100 ng) (Signalchem, BC, Canada) was incubated with 10 μM ATP (Signalchem, BC, Canada), 20 μl kinase buffer (25 mM HEPES, 25 mM β- glycerophosphate, 25 mM MgCl2, 2mM dithiotreitol, and 0.1 mM NaVO3) and purified inactivated IκBα. Reactions were incubated at 37°C for 30 min and terminated by addition of Laemmli SDS sample dilution buffer (Bio-rad, CA, USA). Proteins were separated by SDS-PAGE, and phosphorylation was detected specific antibody.
Cell Growth Assay. Cell Growth Assay. Breast cancer cells growth rates were measured using the WST-1 (2-(2-methoxy-4-nitrophenyl)-3-(4-nitrophenyl)-5-(2,4-disulfophenyl)-2H-tetrazolium (WST-1, Sigma-Aldrich, CA, USA) method. First, the cells were seeded in a 96-well plate at 5 x 103 cells/well, incubated at 37°C for 24 hours, transferred to serum-free medium, and transfected with S63A and S262A as described above. Then, after cultivation under 21% O2 for 24 hours, the cells were transferred to a culture medium containing 10% FBS. Finally, WST-1 was added to the wells, and the plates were incubated at 37°C for 3~4 hours for efficient cell dyeing, and analyzed for its absorbance at 460 nm using a spectrophotometer (Molecular Devices, USA). And, cell viability was measured by tryphan blue staining. It is based on the principle that live cells possess intact cell membranes that exclude certain dyes, such as trypan blue. When a cell suspension is simply mixed with the dye, add equal parts of 0.4% trypan blue dye to the cell suspension to obtain a 1 to 2 dilution, and incubate mixture for less than three minutes at room temperature. Place the hemacytometer with 1:1 tryphan blue staining cells on the stage of a light microscope (Olympus Optical Co., Tokyo, Japan) and calculated the staining cells.
Cell death assa-FACs FACs Cells were stained with FITC-labeled annexin V and propidium iodide, and tumor cell death was assessed by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL; Millipore, Billerica, MA, USA) assay and flow cytometry (BD- Flow JO).
In gel trypsin digestion The in-gel digestion of phosphorylated IκBα was excised from the polyacrylamide gel, washed with water and 30% methanol, and destained with 50% acetonitrile/10 mM ammonium bicarbonate (ABC) at room temperature until the CBB color was removed. The gel pieces were dehydrated with 100% (v/v) acetonitrile. After drying the organic solvent using Speed vacuum concentrator. Disulfide bonds were reduced with DTT (10 mM, 56 ℃, 1 hr), and the free sulfhydryl groups were alkylated with iodoacetamide (55 mM, room temperature, 40 minutes in the dark). The gels were vortexed and completely dried in Speed vacuum concentrator. Gel pieces were washed with water, and dehydrated with 100% acetonitrile. After drying with a Speed vacuum concentrator, the gel was rehydrated using 100 ng/㎕ trypsin (50 mM ABC, pH 8.3) and the digestion was carried out at 37 ℃ for the maximum 20 hr for full digestion. The tryptic peptides were collected in extraction steps using 50% (v/v) acetonitrile containing 0.1% (v/v) formic acid. The tryptic peptide extracts were pooled and dried in Speed vacuum concentrator. The peptides were redissolved in 1% phosphoric acid for mass spectrometric analysis.
Mass spectrometry Phosphotylation of IκBα were analyzed by both matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) and nanospray-liquid chromatography hybrid quadrupole-Orbitrap mass spectrometer (nanoLC-Q Exactive Plus). For MALDI MS, 0.5 ㎕ of sample and 0.5 ㎕ of matrix solution were mixed followed by sample mixture was spotted onto a stainless-steel MALDI plate. The matrix solution was prepared by supernatant from a saturated solution of 2,5-dihydroxybenzoic acid (2,5-DHB) with 50% acetonitrile/1% phosphoric acid.[12] The spots were analyzed with the ultrafleXtreme™ MALDI TOF-TOF mass spectrometer (Bruker Daltonics). MS spectra were collected in reflectron mode over the set mass range of m/z = 700-3500. Instruments were calibrated using Bruker peptide standard II. For LC-MS/MS, tryptic peptides were loaded on a trapping column with 75 μm inner diameter, packed with 5 μm C18 particles (Acclaim PepMap100, Thermo Scientific) and analyzed using a 15 cm analytical column packed with 2 μm C18 particles (Acclaim PepMap RSLC, Thermo Scientific). Reversed phase chromatography was performed using an Ultimate 3000 RSLC nano system (Thermo Scientific) with a binary solvent consisting of 0.1% formic acid (buffer A) and 80% ACN in 0.1% formic acid (buffer B). The peptides were separated by a linear gradient of buffer B from 5% up to 95% for 180 min with a flow rate of 300 nl/min. The LC was coupled to a Q Exactive hybrid quadrupole-Orbitrap mass spectrometer (Q Exactive Plus, Thermo Scientific). The Q Exactive Plus was operated in data-dependent mode with MS scans acquired at a resolution of 70,000, an ion target value of 1e6, and maximum ion injection time for the MS scan was set to 250 ms. Up to 15 most abundant isotope patterns with charge ≥ 2 from the survey scan were selected for MS/MS. An isolation window of 2.0 m/z and higher energy collisional dissociation (HCD) with normalized collision energies of 27% was applied. The maximum ion injection time for the MS/MS scans was set to 100 ms and the ion target value to 1e5. Repeated sequencing of peptides was kept to a minimum by dynamic exclusion of 40 s.[13-14]
Data analysis Measured MS data were processed using FlexAnalysis (Bruker Daltonice). MS/MS data acquired with the Q Exactive was searched the human IκBα FASTA using Mascot. Search parameters included two missed cleavages from trypsin proteolysis and fixed modification of cysteine residues. Variable modifications included oxidation (M), and phosphorylation (S and T). The ion-score cutoff was less than zero or 10-5 at significance threshold p < 0.05.
Statistical analysis Results were expressed as means ± standard error of the mean (SEM) or frequency (%). An independent t-test was performed to compare the difference of the means between control and experimental groups. All statistical analysis was done using SPSS version 12.0 (SPSS, Inc., Chicago, IL). A p value of less than 0.05 was considered statistically significant.