Cell lines, antibodies and reagents
Human pancreatic cancer cell lines, BxPC3, Capan-1, Panc-1, Panc10.05 and 293T cells were obtained from ATCC. CRABP-II knockout (CIIKO) and CRISPR negative control lines were established using Panc-1 as the parental line as described [9]. Gemcitabine resistant lines GR2000 and GR4000 were derived from gemcitabine sensitive line BxPC3 by treating the cells with a stepwise increase of gemcitabine. Panc-1, 293T, CIIKO and negative control cells were maintained in Dulbecco's modified Eagle's medium (DMEM) supplemented with 4.5 g/L glucose, 4.5 g/L L-glutamine, 10% FBS (Gibco) and 100 IU/mL penicillin/streptomycin. BxPC3, GR2000 and GR4000 cells were maintained in RPMI1640 medium supplemented with 4.5 g/L L-glutamine, 1 mM sodium pyruvate, 10% FBS and 100 IU/mL penicillin/streptomycin. All transfections were performed by using lipofectamine 3000 (Invitrogen).
Antibodies used in this study include: CRABP-II mouse mAbs (Millipore, MAB5488), CRABP-II rabbit polyclonal antibody (Proteintech, 10225-1-AP), HuR (3A2, Santa Cruz, sc-5261), Flotilin-2 (Santa Cruz, sc-28320), GAPDH (Santa Cruz, sc-365062), and Actin (Santa Cruz, sc-1615), anti-Flag M2 mAb (Sigma, F9291), anti-Flag agarose beads (Clontech, #635686), Ki67 (SP6, ThermoFisher, RM-9106-S0), ADRP (Novus, NB110-40877), Caspas3 (Cell Signaling, #9662), PARP (Cell Signaling, #9542), AKT (Cell Signaling, #4691), mTOR (Cell Signaling, #2983), S6 (Cell Signaling, #2217), pAKT (S473, Cell Signaling, #9018), pmTOR (Cell Signaling, #5536), pS6 (Cell Signaling, #4858), and pGSK3β (Cell Signaling, #5558).
SNIPER-11 and MV1 were synthesized by Mekoo Biosciences (Morrisville, NC). Other reagents include: retinoic acid, cholesterol, actinomycin D and gemcitabine hydrochloride (USP standard) (Sigma); insulin (ThermoFisher); protein A/G agarose beads (Millipore); BODIPY dyes (ThermoFisher).
RNA isolation and real time Q-PCR
Total RNA was isolated using Trizol (Invitrogen). 500 ng of RNA was reverse transcribed to cDNA by using iScript Reserve Transcription Supermix (Bio-Rad). Real time PCR were performed with SYBR green on C1000 CFX96 real time system (Bio-Rad). The primers used are listed in Table S1.
MTT assay, Annexin V staining and BODIPY staining
MTT assay was performed to assess the cell viability upon gemcitabine treatment. BxPC3 cells and gemcitabine resistant GR2000/GR4000 cells were counted by trypan blue staining and seeded into 96-well plates at 5000 cell/well in triplicates. 6 hrs after seeding, the adhered cells were treated with gemcitabine for 72 hrs in a series of increasing concentrations from 0 µM to 100 µM. Cell viability was assessed by using Celltiter96 AQueous One Solution Reagent (Promega). For gemcitabine sensitivity comparison between Panc-1 cells and CIIKO cells, all parental Panc-1, CIIKO and CRABP-II re-expressing (CIIOE) cells were counted and seeded into 96-well plates at 3000 cell/well in triplicates. After adhesion, cells were treated with gemcitabine in hormone-depleted medium (Gibco) for 96 hrs and cell viability was assessed.
To evaluate the early stage cell apoptosis induced by gemcitabine, Annexin V staining was performed following the manufactory description (BD Pharmingen). Briefly, cells were treated with 50 µM of gemcitabine for 24 hrs. The suspended cells were collected while the adherent cells were shortly trypsinized. Both parts of cells were combined and washed with cold PBS and resuspended in binding buffer at about 1 x 106 cells/mL. 100 µL of cell suspension was stained with 5 µL FITC-Annexin solution and 2 µL Propidium Iodide (PI) for 15 min at 25ºC before FACS analysis on FACSAria™ II flow cytometer (BD).
For neutral lipid content detection, CIIKO and wild type control cells were rinsed with PBS then incubated with 2 µM BODIPY staining solution in PBS at 37ºC for 15 min. After a quick PBS wash, cells were trypsinized to single cell suspension and submitted to FACS analysis.
Immunoprecipitation (IP) and RNA-immunoprecipitation (RIP)
GR4000 cells were cultured in hormone-depleted media for 48 hrs and grew to 70-80% confluence. After cold PBS washes, cells were lysed in EBC buffer (50 mM Tris, pH8.0, 120 mM NaCl and 0.05% IGEPAL) supplemented with cocktail proteinase inhibitor (Roche). 1-2 mg of total proteins in cell lysate were mixed with 10 μg of rabbit IgG isotype control or anti-CRABP-II polyclonal antibody (Proteintech) and incubated at 4°C overnight. Protein A agarose beads were added and incubated for 1 hr. Washed beads were boiled in 30 μL of 2x SDS loading buffer and protein samples were loaded on SDS-PAGE. Separated proteins on PVDF membrane were blotted with anti-HuR antibody.
For RNA immunoprecipitation (RIP), flag-CRABP-II expressing CIIKO cells and empty vector transfected cells were grown in hormone-depleted media for 48 hrs and followed by ice-cold PBS wash for twice. Cells were collected and lysed in polysome lysis buffer (10 mM HEPES, pH7.0, 100 mM KCl, 5 mM MgCl2, 25 mM EDTA, 0.5% IGEPAL) containing 2 mM DTT, RNase OUT (Invitrogen), proteinase inhibitors and 0.2 mg/mL Heparin. After centrifuge, 20 mg cell extract was removed to new 15ml tubes and diluted in 10x volumes of NT2 buffer (50 mM Tris, pH7.5, 150 mM NaCl, 1 mM MgCl2 and 0.05% IGEPAL). Anti-flag beads were washed, suspended in NT2 buffer with 5% BSA, 0.02 mg/ml heparin, and rotated with diluted cell extract at 4°C for 4 hrs. After washes with ice-cold NT2 buffer, the beads and 100 µL of original cell extract (used as input) were mixed with Trizol reagents and centrifuged. RNA in aqueous phase was precipitated by adding isopropanol and RNAase free glycogen (Invitrogen). SREBP-1c and actin messages were assessed by Q-PCR.
Lipid raft isolation and cholesterol quantification
Sucrose gradient ultracentrifugation was used for membrane fractionation as described before [10]. To quantify the total cholesterol in lipid rafts, the raft enriched fractions were mixed with equal volume of methanol: 12 N HCl (10:1) and 2 volumes of chloroform, centrifuged at 13,000 xg for 10 min at 4ºC. The aqueous phases were removed and the chloroform phases were re-extracted with 1 volume of methanol: 1 N HCl (1:1). After removing aqueous phases, the chloroform was evaporated at 50ºC for 3 hrs and the dried lipids were dissolved in cholesterol assay buffer by sonication. The total cholesterol was quantified using the Cholesterol Quantitation Kit (Sigma) according to the manufacturer’s instruction.
Human PDAC collection and immunohistochemistry
Paraffin-embedded blocks of 12 surgically resected, primary PDACs and their relapsing biopsies were collected from the Pathology Archives. Tissue slides were processed using a BenchMark Ultra automated immunostainer (Ventana Medical Systems, Tucson AZ). Slides were deparaffinized, antigen retrieved, and incubated at 37ºC with the primary anti-CRABP-II, or anti-ADRP. Histologic images were obtained with the ScanScope XT digital scanning system (Aperio Technologies, Vista, CA). Cytoplasmic and nuclear immunoreactivity was considered as a positive expression. Immunoreactivity was scored by 2 investigators independently based on the percentage and intensity of positive epithelium cells (intensity: 0, undetectable; 1, weak; 2, moderate; and 3, strong). Score 0 was considered as negative; score 1 or above, as positive.
PDAC PDX mouse models and SNIPER-11 treatment
Xenografts were established in 6-8 weeks NOD-SCID-gamma (NSG) mice. Briefly, the limited passaged (2-3 passages) PDX tumors were resected from mice and rinsed with cold PBS. Tumors were cut into 1-2 mm3 cubes and transplanted to the back of NSG mice subcutaneously. The tumor growth was monitored by measuring the diameters with calipers and the tumor volume was calculated as the flowing formula: volume (mm3) = ½ (width * width * length). Dissected tumors will be frozen at -80ºC or fixed in 10% formalin for biochemical and pathology evaluation. For SNIPER-11 treatment, the tumor bearing mice were intraperitoneally (IP) administrated with SNIPER-11 (5 µM or 5.31 mg/kg, every two days, for 3 weeks) or DMSO when the tumors grew to about 100 mm3. Mice treated with 5 µM of MV1 or RA were included as controls in some of the experiments.
Orthotopic PDX models were established as reported [11]. In brief, limited passaged PDX tumors were resected from mice and rinsed in serum free RPMI 1640. Tumors were minced into the smallest possible fragments and digested into single-cell suspension with collagenase IV solution (200 U/mL, fresh prepared in RPMI 1640). After digestion, tumor cells were washed with RPMI 1640 and stained through a 70 µm strainer. Red blood cells (RBC) were removed using RBC lysis buffer and the resulting cell solution was washed and stained through a 40 µm strainer. Single cells were collected by centrifuge, counted and resuspended in serum free RPMI 1640 at 2x106 cell/100 µL. 50 µL of cell suspension was mixed with matrigel at 1:1 ratio and injected to pancreas in NSG mice. Tumor growth was assessed by MRI imaging at the Case Center for Imaging Research (CCIR). Starting from 4-5 weeks after implantation, tumor bearing mice were IP treated with SNIPER-11 (5 µM, every two days), gemcitabine (20 mg/kg weekly) or a combination of SNIPER-11 and gemcitabine for 3 weeks, DMSO was used as negative control.
Statistics
The Kaplan-Meier method was used to determine overall survival with respect to CRABP-II expression based on the data from Kaplan-Meier Plotter database. The CRABP-II or ADRP staining comparison between primary tumors and relapsing tumors were conducted with a paired sample t-test. Two-way ANOVA was used for tumor growth comparison with respect to different treatments. Student’s t-test was used for other two means comparison. All the statistics were performed using Graphpad Prism software or Excel and p value < 0.05 was regarded as significant difference.