Cell lines and culture conditions
All media and supplements were purchased from Gibco unless otherwise stated. OC cell line SKOV3, OVCA429, CH1 and NSCLC cell line H1299 and A549 were maintained using Dulbecco’s Modified Eagle Medium (DMEM) media supplemented with 10% (v/v) fetal bovine serum (FBS). OC cell lines HeyA8, PEO1, OVTOKO and RMG5 were maintained using Roswell Park Memorial Institute 1640 media (RPMI) supplemented with 10% (v/v) FBS. Penicillin-Streptomycin, L-glutamine and sodium pyruvate were supplemented where necessary. All cells were grown in a humidified incubator at 37oC with 5% CO2 and sub-cultured when 80–90% confluence was reached using 0.25% trypin in 1 mM EDTA pH 7.4.
Antibodies
Primary antibodies used were anti-AXL (CS8661; C44E1), anti-ATM (92356S), anti-pATM (13050S), anti-ATR (13934S), anti-pATR (58014S), anti CHK1 (2360S), anti-pCHK1 (2348S), anti-CHK2 (6334S) and anti-pCHK2 (2197S) from Cell signalling; anti-SAM68 (07-415) and anti-γH2AX (3BW301) from Millipore; antiRPA2 [p Ser33] (NB100-544) from Novus Biologicals; anti-SAM68 (ab76471), anti-SREBP2 (30682), anti-HMGCR (242315) from Abcam; anti-squalene synthase (sc-271602), anti-MVK (sc-390669), anti-OSC (sc-514507), anti-pADPr (sc-56198), anti-GAPDH (sc-47724), anti-ß-actin (sc-47778) from Santa Cruz. Secondary antibodies used were HRP-conjugated goat anti-mouse and goat anti-rabbit from Cell signalling. All primary antibody dilutions were at 1:1,000. Secondary antibody dilutions were at 1:5,000.
Chemicals
All compounds were reconstituted to 10 mM stock using DMSO (Sigma Aldrich) and stored in -20oC. AXL inhibitor R428 (BGB324) (#S2841), ATR inhibitor BAY1895344 (#S8666), ATR inhibitor VE-821 (#S8007), PARP1 inhibitor Olaparib (#S1060), PARP1 inhibitor Niraparib (#S2741) were purchased from Selleckchem.
shRNA knockdown, sgRNA CRISPR-Cas9 mediated knockout and lentiviral transduction
shRNA and sgRNA sequences were obtained from the broad institute genetic perturbation platform (Tables 1 & 2). shRNA was cloned into pLKO.1 lentiviral plasmid, which was a gift from Bob Weinberg (Addgene plasmid #8453; http://n2t.net/addgene:8453; RRID: Addgene_8453), while sgRNA were cloned into the lentiCRISPR v2 lentiviral plasmid which was a gift from Feng Zhang (Addgene plasmid #52961; http://n2t.net/addgene:52961; RRID: Addgene_52961). SKOV3 and HeyA8 were infected with pLKO.1 and/or lentiCRISPR v2 lentiviral plasmid and selected using puromycin (1 µg/ml) or sorted for GFP expression. Single clones were generated for CRISPR knockout lines.
Table 1
Target Gene
|
shRNA ID
|
Target sequences
|
Luciferase
|
shLUC
|
CGCTGAGTACTTCGAAATGTC
|
AXL
|
shAXL#40
|
CGAAATCCTCTATGTCAACAT
|
SREBF2
|
shSREBF2#1
|
CCTGAGTTTCTCTCTCCTGAA
|
SREBF2
|
shSREBF2#2
|
GCAACAACAGACGGTAATGAT
|
HMGCR
|
shHMGCR#1
|
ATCAACTTGTGTACTGATAAA
|
HMGCR
|
shHMGCR#2
|
CAAGTTATTACCCTAAGTTTA
|
Table 2
List of CRISPR knockout sgRNA sequences
Gene
|
sgRNA ID
|
sgRNA sequence
|
AXL Exon 1
|
1.1.2
|
GCGTGGCGGTGCCCCAGGAT
|
AXL Exon 2
|
2.1.22 / 2.1.1
|
GCTGAAGAAAGTCCCTTCGT
|
SAM68 Exon 5
|
S7
|
TGAACCCTCTCGTGGACGTG
|
SAM68 Exon 6
|
S8
|
TGGTGTTGGACCACCTCGGG
|
Cell fractionation
Cell fractionation was carried out using PARIS™ Kit (Thermo Fisher Scientific). Briefly, cells were scraped and washed with PBS. Cell pellets were lysed using ice cold cell fractionation buffer and incubated for 10 mins. Cytoplasmic fractionation was obtained by pelleting nuclear fraction at 500 x g. Nuclear fraction pellet was lysed using cell disruption buffer for 10 mins. Both fractions were clarified at 13,500 rpm for 15 mins at 4oC
Co-Immunoprecipitation (Co-IP)
Cells were lysed in ice cold co-immunoprecipitation lysis buffer containing 10 mM Tris-HCL (pH7.5), 150 mM NaCl, 0.5% NP40, 0.25% Sodium deoxycholate, 0.5 mM EDTA (pH8.0) supplemented with 100X protease and phosphatase inhibitor (Thermo Fisher Scientific), 50 units/mL benzonase (Millipore) and 2 mM MgCl2 for 45 mins at 4oC. Supernatant were obtained by pelleting cell debris at 13,500 rpm for 15 mins at 4oC. 500 µg of protein was incubated with 1 µg of antibody and rotated at 4oC overnight. Following, samples were incubated with 1% (v/v) bovine serum albumin (BSA) (Sigma Aldrich) blocked Pierce™ Protein A/G Agarose (25 L packed bead volume) (Thermo Fisher Scientific) for 45 mins at 4oC. Beads were washed with co-immunoprecipitation buffer and bound proteins were released by boiling in 2X gel loading buffer.
Immunoblotting
Cells were lysed in RIPA lysis and extraction buffer (Thermo Fisher Scientific) supplemented with 100X protease and phosphatase inhibitor (Thermo Fisher Scientific) and sonicated. Samples were resolved using 4–12% SDS-polyacrylamide gel electrophoresis (SDS-PAGE) (Invitrogen) and transferred onto polyvinylidene fluoride (PVDF) membrane (Bio-Rad) using Trans-Blot Turbo Transfer System (Bio-Rad) at a constant voltage. Membrane was blocked using either 5% (v/v) BSA in Tris-buffered saline with 0.1% Tween-20 (TBST) or 5% Blotting Grade Blocker Non-Fat Dry Milk (Bio-Rad) prior to incubation with antibody of interest. Proteins were visualised using SuperSignal® West Dura Extended Duration Substrate (Thermo Fisher Scientific) and imaged using ChemiDoc MP Imaging System (Bio-Rad).
Immunofluorescence staining
Cells lines were grown on coverslips, fixed with 4% paraformaldehyde (PFA) and blocked in 5% BSA with 0.1% Triton-X-100. Samples were incubated with primary antibody overnight and subsequently with Alexa Fluor 594 donkey anti-mouse (A21203) or/and Alexa Fluor 647 goat anti-rabbit (A21245) (Life Technologies). Cover slips were mounted using Glycergel mounting medium, visualized using Leica light sheet microscope (Leica Microsystems), and analysed using ImageJ. Corrected total cell fluorescence (CTCF) was calculated based on the following formula: CTCF = Cell Area x Cell Mean fluorescence – (Area of selected cell X Mean fluorescence of background readings). Cholesterol was stained using 50µg/ml filipin III (Sigma-Aldrich) for 1 hour and counterstained with propidium iodide.
Flow cytometry / Edu-labelling
Cells were treated accordingly and EdU labelling was carried out using Click-iT™ EdU Alexa Fluor™ 647 flow cytometry assay kit as per written in manufacturer’s protocol (Thermo Fisher Scientific). Cells were then analysed using LSRII Cell Analyzer (BD). Results were analysed with FlowJo v10.
Dose-response curve / Drug treatment
Dose response curve of AXL inhibitor (R428) was carried out using varying concentration for 72 hours with DMSO as a negative control. Cell viability was quantified using CellTiter-Glo Luminescent Cell Viability Assay system (Promega) and plotted using GraphPad Prism and IC50 was calculated. For combination treatment with AXL inhibitor, a fixed IC20 of AXL inhibitor with varying concentration of selected inhibitor was used.
Drug combination and dose reduction index analysis
Combination Index (CI) was calculated using the Chou–Talalay equation [22], (CI) = (D)1 /(Dx)1 + (D)2/(Dx)2 where (Dx)1 and (Dx)2 represents each drug alone exerting x% inhibition, while (D)1 and (D)2 were concentrations of drugs in combination to elicit the same effect. CI < 1 indicates synergism; CI = 1 indicates an additive effect; and CI > 1 indicates antagonism. The dose-reduction index (DRI) measures the extent or folds of dose reduction when in a combination, at a given effect of inhibition, compared to each drug alone. (DRI) = (Dx)1/(D)1. CI and DRI plot were plotted against Fa, the fraction of cells affected cells or cells killed.
Cell Proliferation Assay
Cells were seeded onto 96-well Greiner flat-bottomed white plate. Growth rate was quantified in 48 hrs intervals by measuring ATP levels using CellTiter-Glo Luminescent Cell Viability Assay (Promega) over a duration of 7 days. Proliferation curve was plotted with GraphPad Prism.
RNA extraction, reverse transcription, quantitative polymerase chain reaction
RNA extraction was done using TRIzol® Reagent (#15596026), chloroform and RNeasy Mini Kit (Qiagen). Reverse transcription was carried out using High-Capacity cDNA Reverse Transcription Kit (Thermo Fisher Scientific). Following, qPCR was performed using BlitzAmp qPCR system (MiRXES). All reactions were carried out according to manufacturer’s instructions. The cDNA was diluted to 100 ng before use. mRNA levels were measured with gene-specific primers as listed in Table 3 and reactions were carried out using QuantStudio™ 5 Real-Time PCR System (Applied Biosystems). Samples were assayed in triplicates with GAPDH as the internal normalization controls. Relative mRNA expression was determined using the ΔΔCT method.
Table 3
Sequence of primers used for qPCR
Gene
|
Forward Primer
|
Reverse Primer
|
GAPDH
|
ACAACTTTGGTATCGTGGAAGG
|
GCCATCACGCCACAGTTTC
|
AXL
|
GTGGGCAACCCAGGGAATATC
|
GTACTGTCCCGTGTCGGAAAG
|
SERBF1
|
ACAGTGACTTCCCTGGCCTAT
|
GCATGGACGGGTACATCTTCAA
|
SERBF2
|
CCTGGGAGACATCGACGAGAT
|
TGAATGACCGTTGCACTGAAG
|
ACAT2
|
GCGGACCATCATAGGTTCCTT
|
ACTGGCTTGTCTAACAGGATTCT
|
HMGCS1
|
CTCTTGGGATGGACGGTATGC
|
GCTCCAACTCCACCTGTAGG
|
HMGCR
|
TGATTGACCTTTCCAGAGCAAG
|
CTAAAATTGCCATTCCACGAGC
|
FDFT1
|
CCACCCCGAAGAGTTCTACAA
|
TGCGACTGGTCTGATTGAGATA
|
FASN
|
ACAGCGGGGAATGGGTACT
|
GACTGGTACAACGAGCGGAT
|
CYP51A1
|
GAAACGCAGACAGTCTCAAGA
|
ACGCCCATCCTTGTATGTAGC
|
KHDRBS1
|
TTGGTACGTGGTACACCAGTA
|
AGGCAAAGGTATCCTCTGGATG
|
RNA sequencing
Total RNA was extracted, and RNA integrity and concentration were assessed on 2100 Bioanalyzer Instrument (Agilent). Library preparation was performed using TruSeq RNA Library Prep Kit (Illumina) and sequencing was performed on the Illumina HiSeq 4000 System. Relative gene expression changes were filtered based on log2 fold-change > / < 1.5 with a p-value < 0.05.
Chick CAM model
Specific Pathogen Free fertilized chicken eggs were purchased from JD-SPF Biotech Co. Ltd., washed, and incubated at 37.5 ℃ with 70% humidity on embryonic day 0 (ED0). Preparation of the CAM was operated within a biosafety cabinet. A small hole was pierced through the apex of the shell using an 18-gauge needle, and 3–4 ml albumin was removed on ED3. A 1.5 cm2 window of shell was removed to expose the CAM. The window was then covered and sealed with a 3M™ Tegaderm™ transparent film. On ED7, 0.75 x 106 SKOV3 cells suspended in 50 µl Matrigel (#354234, Corning) were engrafted onto blood vessel area of CAM. The targeted area of vessel was gently bruised using a glass rod before inoculation. IC50 concentrations of R428, BAY1895344, or a combination or both were administered using a piece of filter paper on ED12 and ED13. Tumour volume was measured using ultrasound (FUJIFILM Sonosite SII using HSL25x transducer) on ED11 and ED14. All CAM experiments were performed in accordance with relevant guidelines and regulations and approved by National Taiwan University.
Stable isotope labelling by amino acids in cell culture (SILAC) nuclear co-immunoprecipitation (Co-IP) mass spectrometry analysis
For SILAC labelling, cells were incubated in DMEM or RPMI (-Arg, -Lys) medium containing 10% dialyzed fetal bovine serum supplemented with 42 mg/l 13C615N4 L-arginine and 73 mg/l 13C615N2 L-lysine (Cambridge Isotope) for DMEM, 84 mg/l 13C615N4 L-arginine and 50 mg/l 13C615N2 L-lysine (Cambridge Isotope) or the corresponding non-labeled amino acids, respectively. Successful SILAC incorporation was verified by in-gel trypsin digestion and MS analysis of heavy input samples to ensure an incorporation rate of > 98%.
SILAC Mass Spectrometry analysis
Labelled cell lines were fractionated to obtain the nuclear fraction and Co-IP was performed prior to mass spectrometry analysis. Samples were boiled at 95°C prior to separation on a 12% NuPAGE Bis-Tris precast gel (Thermo Fisher Scientific) for 10 min at 170 V in 1x MOPS buffer, followed by gel fixation using the Colloidal Blue Staining Kit (Thermo Fisher Scientific). For in-gel digestion, samples were destained in destaining buffer (25 mM ammonium bicarbonate; 50% ethanol), reduced in 10 mM DTT for 1h at 56°C followed by alkylation with 55mM iodoacetamide (Sigma) for 45 min in the dark. Tryptic digest was performed in 50 mM ammonium bicarbonate buffer with 2 µg trypsin (Promega) at 37°C overnight. Peptides were desalted on StageTips and analysed by nanoflow liquid chromatography on an EASY-nLC 1200 system coupled to a Q Exactive HF mass spectrometer (Thermo Fisher Scientific). Peptides were separated on a C18-reversed phase column (25 cm long, 75 µm inner diameter) packed in-house with ReproSil-Pur C18-AQ 1.9 µm resin (Dr Maisch). The column was mounted on an Easy Flex Nano Source and temperature controlled by a column oven (Sonation) at 40°C. A 105-min gradient from 2 to 40% acetonitrile in 0.5% formic acid at a flow of 225 nl/min was used. Spray voltage was set to 2.2 kV. The Q Exactive HF was operated with a TOP20 MS/MS spectra acquisition method per MS full scan. MS scans were conducted with 60,000 at a maximum injection time of 20 ms and MS/MS scans with 15,000 resolution at a maximum injection time of 50 ms. The raw files were processed with MaxQuant [63] version 1.5.2.8 and searched against the human Uniprot database (95,934 entries) with preset standard settings for SILAC labeled samples and the re-quantify option was activated. Carbamidomethylation was set as fixed modification while methionine oxidation and protein N-acetylation were considered as variable modifications. Search results were filtered with a false discovery rate of 0.01. Known contaminants, proteins groups only identified by site, and reverse hits of the MaxQuant results were removed and only proteins were kept that were quantified by SILAC ratios in both ‘forward’ and ‘reverse’ samples.
Cholesterol-related LC-MS analyses
The following reagents and materials were purchased from the indicated sources: Methanol, isopropanol and ammonium formate, Fisher Chemical; Ultra-high quality water from an Atrium® Pro lab water system, Sartorius; Tricine, acetonitrile and chloroform, Merck; Formic acid and ammonia solution (25%), VWR Chemical.
Sample preparation for cholesterol-related analyses
Cells were quenched using ice-cold 150 mM NaCl, collected using a cell scraper and pelleted down. Double lipid extraction was performed for detection of cholesterol and cholesteryl ester. Briefly, methanol, chloroform and 3.8 mM tricine solution (approximately 1:1:0.5 vol/vol) was used to separate polar metabolites (aqueous fraction) from lipid species (organic fraction). Lipid metabolites in the lipid layer were collected and re-extracted by addition of chloroform. Lipid layer collected were pooled together and purged with nitrogen gas before storing prior to mass spectrometry run. Whole extraction process was done either on ice or under 4oC conditions.
LC-MS profiling of cholesterol
The lipid extracts were analysed in triplicate using LC-MS analysis of cholesterol was performed using a ACQUITY ultra performance liquid chromatography (UPLC) system (Waters) interfaced with a high resolution triple-quadrupole time-of-flight mass spectrometer (TripleTOF®6600, SCIEX) equipped with an electrospray ionization (ESI) source, in the positive mode. To ensure mass accuracy of the system throughout batch acquisition, calibrant delivery system (CDS) was used to introduce calibration solution for automated mass calibration of the mass spectrometer. The calibration compound was Reserpine (m/z 609.28066) for positive mode. The MS acquisition parameters were optimized and set accordingly: ion spray voltage 5500 V, nebulizer gases (GS1 and GS2) 50 psi, curtain gas (CUR) 40 psi, source temperature (TEM) 450oC, declustering potential (DP) 80 V and collision energy spread (CES) 20 V. Liquid chromatography separation was conducted using an Acquity CSH (C18) column (2.1 mm × 50.0 mm, 1.7 µm, Waters) in gradient elution mode at a flow rate of 0.1 mL/min. The composition of the LC mobile phase is as follows – solvent A comprises of methanol: water mixture (4:1) with 5 mM ammonium formate, while solvent B comprises of methanol with 5 mM ammonium formate. Cholesterol was separated using the following gradient condition: 0–1 mins at 1% B, ramping from 1 to 82.5% B for 1–10 mins, holding at 99% B from 10 to 15 mins before equilibration at 1% B from 15-17.2 mins. The injection volume of each sample was 2µL.
LC-MS profiling of cholesteryl esters
The lipid extracts were analysed in triplicate using an ACQUITY UPLC system (Waters) in tandem with a SYNAPT G2-Si High Definition Mass Spectrometry (Waters,). A C18 UPLC column (Acquity UPLC CSH column, 1.0 × 50 mm, 1.7µm, Waters) was used for separation and the mobile phase comprised of two solvents: ‘A’ comprising of acetonitrile, methanol and water (2:2:1) with 0.1% acetic acid (Merck) and 0.025% ammonia solution (VWR), and ‘B’ comprising of isopropanol (Thermo Fisher Scientific) with 0.1% acetic acid and 0.025% ammonia solution. The UPLC program was performed as follows: the gradient was increased from 50% B to 95% B over 10 mins (flow rate of 0.1 mL/min) before B was further increased to 99% for a 5 mins wash at a flow rate of 0.15 mL/min. The column was re-equilibrated for 2.2 mins at 1% B (flow rate of 0.1 mL/min). Column temperature was maintained at 45°C and eluent from the LC system was directed into the MS. Next, high-resolution mass spectrometry was performed in positive ESI mode with a mass range of 100 to 1,800 m/z and a resolution of ≥ 10,000. Cone and desolvation gas flows was set at 40.0 and 600.0 (L/hour) respectively, with a desolvation temperature of 200oC. The ESI capillary voltage was 2.0 kV for ionization. Mass calibration was performed using sodium formate prior to injection of the samples. Quality control (QC) sample comprising of equal aliquots of each sample was run at regular intervals during the batch LC-MS runs. The lipid extracts were dried under nitrogen gas and reconstituted with Solvent ‘B’ before LC–MS analysis. The injection volume of each sample was 2 µL.
LC-MS data processing and analysis
The raw LC-MS data obtained from the lipid extracts was processed using a XCMS-based peak finding algorithm [64]. The QC samples were used to adjust for instrumental drift and total ion count normalisation was performed. Detected mass peaks were assigned putative metabolite identities by matching the respective masses (< 10 ppm error) with the KEGG [65] and Human Metabolome Database (HMDB) [66]. Subsequently, metabolite identities were confirmed based on mass spectral comparison with available metabolite standards or with online mass spectral libraries mzCloud.
Statistical Analyses
All statistical analyses were performed using GraphPad Prism and data were presented as mean with standard error of the mean, unless otherwise stated. Two-tailed unpaired student’s t-test with Welch’s correction was performed to compare differences between two individual groups. A p-value of less than 0.05 was defined to be statistically significant and are indicated as follows – ns: not significant, *p < 0.05, **p < 0.01, ***p < 0.001. The p-value significance values are indicated in the relevant figures and figure legends.
Data availability
The data that support the findings of this study are available from the corresponding author, R.Y.H, upon reasonable request.