All commercially available starting materials and solvents were purchased from commercial vendors and used without further purification. Reactions were monitored using analytical thin-layer chromatography (TLC) on precoated silica gel GF254 plates (Qingdao Haiyang Chemical Plant, Qingdao, China) and visualized under ultraviolet light (254 nm and 365 nm). Column chromatography was performed on silica gel (200–300 mesh). 1H and 13C NMR spectra were recorded on the Broker AVANCE NEO and Agilent DD2 500 with 400 or 500 MHz for proton (1H NMR) and 100 or 125 MHz for carbon (13C NMR) with tetramethylsilane (Me4Si) as the internal standard, respectively. The chemical shifts (δ) were expressed in parts per million (ppm). Abbreviations used: s = singlet; d = doublet; t = triplet; q = quartet; m = multiplet. The coupling constant (J) values were described as hertz. High-resolution (ESI) MS spectra were recorded using a QTOF-2 Micromass spectrometer. The purity of the final compounds for biological evaluation was higher than 95% by analytical HPLC analysis with the Primaide 1210 system. General procedure to synthesis compounds (Supplementary information)
The human full-length SIRT6 gene was inserted into the prokaryotic expression vector (pET-28a (+)). The verified recombinant plasmid was transformed into Escherichia coli BL21 (DE3) expression strain and cultured overnight at 37℃ on LB plates supplemented with 50 µg/mL kanamycin. Single colonies were inoculated in fluid kanamycin-containing LB mediums. It was shaken at 160 rpm for 16 hours at 37°C and transferred to kanamycin-containing LB medium at a ratio of 1:100. It was shaken at 160 rpm at 37°C until the OD600 reached 0.6. The Isopropyl-β-d-thiogalactopyranoside (IPTG) was added to make the final concentration of solution 0.5 mmol/L, cultured for 15°C and 160 rpm for 24 hours.
Bacterial liquid was harvested at 12,000 rpm at 4°C for 20 min, and then re-suspended in Lysis buffer (20 mmol/L Hepes pH 7.5, 20 mmol/L imidazole, 5% Glycerol, 500 mmol/L NaCl). Cell pellets were disrupted by high-pressure crusher (JNBIO, Guangzhou China) and centrifuged (4°C, 12000 rpm, for 20 min). Supernatant was collected and purified by FPLC with a 1 mL Ni–NTA Sepharose column (GE Healthcare, Stamford, USA). The SIRT6 protein was eluted at containing 50 mM, 100 mM, 150 mM, 200 mM, and 500 mM imidazole in elution buffer. The purified SIRT6 protein was dialyzed in dialysis buffer (50 mM Tris-HCl, 137 mM NaCl, 2.7 mM KCl, 1 mM MgCl2). The purified SIRT6 protein was analyzed by SDS-PAGE.
Fluor-De-Lys (FDL) assays were used for the examination of the deacetylation activities of sirtuins as described previously [24, 25]. For the detection of SIRT6 activity, the 50-µL reaction mixture contained 5 µM SIRT6, 2.5 mM NAD+ (β-Nicotinamide adenine dinucleotide hydrate, Sigma, N1636), 75 µM RHKK-Ac-AMC (GL Biochem, Shanghai), compounds/DMSO, and assay buffer. The reactions were incubated at 37°C for 2 h, added 50 µL stop solution (40 mM nicotinamide, 6 mg/mL trypsin) at 25°C for 30 min. Fluorescence intensity was measured at an excitation wavelength of 360 nm and an emission wavelength of 460 nm. IC50 values were calculated in GraphPad Prism version 8.00.
Biolayer interferometry (BLI)
Binding assays were performed using Ni-NTA biosensors on the the Octet® Red96 system (ForteBio, Menlo Park, CA) in 96-well microplates and measuring changes wavelength shift (in nm) over time (sec). All steps were carried out at 25℃ with the plate shaking speed of 1000 rpm. The total working volume was 200 µL in 96-well microplate (Greiner Bio-one, Germany). The SIRT6 protein and compounds were diluted with phosphate buffered saline (PBS, pH 7.4). Ni-NTA biosensors were prewetted for at least 10 min in PBS prior to the assay. Afterwards, the Ni-NTA biosensors were immobilized for 180 sec with 200 µL culture containing 20 µg/mL SIRT6. After loading, sensors were washed in PBS buffer for another 120 sec and transferred to wells containing different concentrations of compound 8a (0, 12.5, 25, 50 ,100 µM) for binding. Finally, the dissociation step was carried out in kinetic buffer for 180 sec. The assay data were processed and calculated using the ForteBio Octet RED analysis software.
The cellular thermal shift assay (CETSA).
The CETSA [26] approach builds on the thermal shift assay concept where ligand binding to its target protein affects protein stability at high temperatures. Hela cells were plated in 100 mm plates. After 12 h attachment, Hela cells were incubated with 8a (25 µM) for 6 h, and then the cells were digested with trypsin and collected. The collected cells were subsequently aliquoted into six parts, and incubated each part at different temperatures (40, 41, 42, 43, 44, 45, 46, 47, 48, 49 and 50°C) for 3 minutes. The heated cells were then snap frozen in liquid nitrogen three times to lyse. After that, the lysed cells were then centrifuged at 12,000 rpm for 20 minutes. Then the level of SIRT6 was detected by western blot.
Cell culture.
The BXPC-3 (RRID:CVCL_0186), PANC-1 (RRID:CVCL_0480), and Hela (RRID:CVCL_0030) cell lines were obtained from the Cell Resource Center of the Shanghai Institute for Biological Sciences, Chinese Academy of Sciences. BXPC-3 were cultured in RPMI1640 medium (Gibco) supplemented with 10% fetal bovine serum, sodium pyruvate (1 mmol/L), penicillin (50 units/mL), and streptomycin (50 µg/mL). PANC-1 and Hela were maintained in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% (v/v) FBS, streptomycin (50 µg/mL) and penicillin (50 units/mL). The cells were maintained in 37°C incubator with 5% CO2.
Proliferation Assay.
BXPC-3 and PANC-1 cells were plated in 96-well plates at 5,000 cells per well. After 12 h attachment, varying concentrations of 8a were added into the culture medium for 72 h. Then 10 µL MTT (5 mg/ml) solution was added into per well for 4 h at 37℃. After removing medium, formazan crystals were dissolved in 150 µL DMSO. The cells viability was quantified by measuring the absorbance at 570 nm.
Colony Formation Assay.
The BXPC-3 cells were plated in six-well plates at 1000 cells per well. After 12 h, different concentrations of 8a, Gemcitabine or their combination were added for 7–10 days. The cells were fixed for 15 min with paraformaldehyde (4%) and then stained with crystal violet (0.1%) for 15min at room temperature. Surviving colonies consisting of 50 or more cells were counted under microscope.
Cell cycle analysis.
The BXPC-3 cells were seeded onto six-well plates at 1x105 cells per well and different concentrations of 8a, Gemcitabine or their combination were added for 48 h. The cells were washed with PBS, collected after trypsinization and fixed in cool 70% ethanol at -20℃. After fixation, the cells were incubated in the dark with PBS containing 50 µg/mL propidium iodide (PI) and 100 µg/mL RNase A for 30 min at room temperature. Finally, the samples were analyzed on a flow cytometer (Beckman Coulter).
Cell apoptosis analysis.
To quantify cell apoptosis, BXPC-3 cells were treated with different concentrations of compound 8a, Gemcitabine or their combination for a further 72 h. Harvested cells were incubated with Muse™ Annexin V &Dead Cell assay kit (Muse TM Cell Analyzer, Millipore (catalog no. MCH100105)). Cells were then analyzed by flow cytometry (Muse TM Cell Analyzer, Millipore).
Hoechst 33342 stain.
BXPC-3 cells were seeded on the chamber-slides at 12-well plates and treated with the indicated concentration of compound 8a for 48 h. After treatment, cells were fixed with 4% paraformaldehyde for 15 min. Then, cells were stained with 10 µg/mL Hoechst 33342 for 30 min. After washing with PBS, slides were observed with a fluorescent microscopy.
Western blot analysis.
Cells were harvested and lysed in a lysis buffer (solarbio, R0010) supplementing 1 mM PMSF and protease inhibitor (TargetMol, USA). Protein was separated on 7.5%-15% SDS-PAGE gels and transferred to the 0.45 µm or 0.22 µm PVDF membrane (Merck Millipore), and subsequently blocked with 5% bovine serum albumin (BSA) in TBST. The blocked membrane was incubated with primary antibodies:SIRT6 (1:1000, Cell Signaling Technology, Cat#12486S, RRID:AB_2636969), Histone H3 Antibody (1:1000, Cell Signaling Technology, Cat#9715S, RRID:AB_331563), β-Actin (1:2000, Cell Signaling Technology, Cat#4970S, RRID:AB_2223172), Cleaved caspases-3 (1:1000, Cell Signaling Technology, Cat#9661S, RRID:AB_2341188), Cleaved Caspase-9 (Asp315) (1:1000, Cell Signaling Technology, Cat#20750S, RRID:AB_2798848), Cleaved PARP (1:1000, Cell Signaling Technology, Cat#5625S, RRID:AB_10699459), AKT (1:1000, Cell Signaling Technology, Cat#9272S, RRID:AB_329827), Phospho-AKT (Ser473) (D9E) (1:1000, Cell Signaling Technology, Cat#4060S, RRID:AB_2315049), ERK1/2 (1:1000, Cell Signaling Technology, Cat#4695S, RRID:AB_390779), Phospho-p44/42 MAPK (ERK1/2) (Thr202/Tyr204) (1:1000, Cell Signaling Technology, Cat#4370S, RRID:AB_2315112), Cyclin D1 (E3P5S) (1:1000, Cell Signaling Technology, Cat#55506S, RRID:AB_2827374), Phospho-mTOR(Ser2448)(1:1000,Cell Signaling Technology, Cat#5536S, RRID:AB_10691552), Phospho-p70S6Kinase (Thr421/Ser424) (1:1000, Cell Signaling Technology, Cat#9204S, RRID:AB_2265913), Phospho-Histone H2A.X (Ser139) (1:1000, Cell Signaling Technology, Cat#9718S, RRID:AB_2118009) at 4℃ overnight. After being washed with TBST, the membranes were incubated with horseradish peroxidase-conjugated secondary antibodies at room temperature for one hour. Then, the membranes were detected by Tanon 5200 (Tanon, Beijing, China).
Immunofluorescence assay.
Cells were plated in the chamber-slides in the same conditions, and treated with compound 8a, gemcitabine or their combination for a further 48 h. Cells were washed twice with PBS and fixed with paraformaldehyde (4%) at room temperature for 15 min. After washing twice with PBS, cells were blocked in PBS containing 0.2% Triton X-100, 10% goat serum and 5% BSA for 2 h, and then incubated with primary antibodies (1:500, γ-H2AX, Ser139, Cell Signaling Technology, Cat#9718, RRID:AB_2118009) overnight at 4°C. Cells were stained with secondary antibodies (1:1000, Alexa Fluor 488-conjugated anti-rabbit IgG, Abcam, Cat#ab150077, RRID:AB_2630356) for 2 h at 4℃ in dark. After washing with PBS, the cells were stained with hoechst and observed by confocal microscopy (Leica TCS SP8 STED 3X).
Tumor nude mice model.
All animal experiments were performed in accordance with the principles and procedures approved by the Animal Ethics Committee of the Ocean University of China. Female BALB/c nude mice (14 ± 2 g, 4 weeks old) were purchase from Beijing Vital River Laboratories (Beijing, China). All animals were allowed to adapt to the environment for one week upon arrival. These animals were kept in a standard environment with a 12-hour light/dark cycle and allowing free drinking and eating. About 2 × 106 of BXPC-3 cells were inoculated into nude mouse flanks. When the tumor volume reached approximately 100 mm3, mice were randomly divided into four groups with 6 mice per group before treatment and then treated with vehicle, 8a (20 mg/kg), gemcitabine (10 mg/kg) and their combination every 2 days. Tumor volumes and body weight were measured every 3 days and use the formula to calculate the tumor volume: V = (L × W× W) /2 (V, tumor volume; L, length; and W, width). Mice were sacrificed and tumor tissues were removed. Tumor tissues were fixed with paraformaldehyde (4%) and embedded with paraffin.