Plasmids
The oligonucleotides and plasmids used in this study are listed in Supplementary Table 1 and Table 2. For gene overexpression, human KHK-C was cloned into pLVX-puro. For gene knockout, guide RNA sequences of KHK or SLC2A5 were cloned into the pLVX-CRISPR-v2-puro vector. Human KHK was cloned into the pET28a vector for recombinant KHK expression in E. coli. All plasmids were sequenced and verified.
Cell lines and cell culture conditions
The cell line WRL68 was purchased from Mingzhou Biotechnology. Other cell lines were obtained from the Cell Bank of the Type Culture Collection of Chinese Academy of Sciences. Huh7, HepG2, A549, HeLa, K562, MCF7 and HEK293T cells were cultured in DMEM high glucose medium (Biological Industries, 0023119), and T24 and WRL68 cells were cultured in RPMI 1640 medium (Biological Industries, 2035127) with 10% fetal bovine serum (FBS) (Biological Industries, 1841924) in 5% CO2 at 37 ℃. All cells were identified without mycoplasma infection.
To overexpress genes in cultured cells, expression plasmids were stably transfected with a retrovirus-mediated transfection method. For this, HEK293T cells were transfected with pLVX-puro (empty plasmid) or pLVX-KHK-C-puro and the two packaging plasmids, psPAX.2 and pMD2.G. Two days later, the virus particles were collected, filtered, and used to infect the target cells. After 48 hours of infection, transfected cells were selected with 2 mg/mL puromycin. Cells stably transfected with pLVX-puro were used as a control cell line.
CRISPR–Cas9-mediated gene knockout
HEK293T cells were transfected with pLVX-CRISPR-v2-puro (empty plasmid), pLVX-CRISPR-v2-SLC2A5-puro or pLVX-CRISPR-v2-KHK-puro and the two packaging plasmids, psPAX.2 and pMD2.G. Two days later, the virus particles were collected, filtered, and used to infect the target cells. After 48 hours of infection, 2 mg/mL puromycin (InvivoGen, QLL-41-03) was used to select positive cells. The selected cells were subjected to limiting dilution to obtain knockout cells. Western blotting was used to verify the expression of the target genes. Selected knockout clones were analyzed and verified by DNA sequencing.
Metabolomic analysis
For central carbon metabolomic analyses, cells were seeded on 10 cm culture plates and cultured to a density sufficient for 70-80% confluence. After l-sorbose treatment for 12 h, the cells were collected and added to 400 mL water. Cells were vortexed for 1 min after the addition of 500 mL precooled chloroform/methanol (1/3, v/v) and then homogenized for 4 min at 35 Hz and sonicated for 10 min. The homogenization and sonication circles were repeated three times, vibrating at 4 ℃ for 15 min, and incubating at -80 ℃ for 1 h. The samples were centrifuged at 9,000 × g for 10 min. The supernatants were collected and dried under a gentle stream of nitrogen. Then, they were dissolved in 200 mL ultrapure water. Reconstituted samples were vortexed before filtration through a centrifuge tube filter and subjected to HPIC-MS/MS analysis.
The HPIC separation was performed using a Thermo Scientific Dionex ICS-6000 HPIC System (Thermo Scientific) equipped with Dionex IonPac AS11-HC (2 × 250 mm) and AG11-HC (2 mm × 50 mm) columns. Mobile phase A was 100 mM NaOH in water, and mobile phases C and D were methanol and water, respectively. Another pumping system was used to supply the solvent (2 mM acetic acid in methanol), and the solvent was mixed with the effluent before entering electrospray ionization (ESI) (flow rate of 0.15 mL/min). The column temperature was 30 ℃. The temperature of the autosampler was 4 ℃ and the injection volume was 5 mL. An AB SCIEX 6500 QTRAP+ triple quadrupole mass spectrometer (AB Sciex) equipped with an ESI interface was used for analysis and development. Typical ion source parameters were as follows: ion spray voltage = -4500 V, curtain gas = 30 psi, ion source gas 1 = 45 psi, ion source gas 2 = 45 psi, and temperature = 450 ℃. By injecting a standard solution of a single analyte into the API source of the mass spectrometer, flow injection analysis was used to optimize the MRM parameters for each target analyte. AB SCIEX Analyst Workstation Software (1.6.3 AB SCIEX), MultiQuant 3.0.3. software and Chromeleon 7 were employed for MRM data collection and processing.
Quantitative analysis of intracellular l-sorbose-1-phosphate
Cells were seeded on 35 mm culture plates and cultured to a density of approximately 70%. After l-sorbose treatment for 24 h, the medium was aspirated, and 1 mL of 80% methanol:20% water mixture was added to extract metabolites. The plate was placed at -20 degrees for 20 minutes. Then, the cell material was scraped into a 1.5 mL test tube prechilled on ice. The cell debris was pelleted by centrifugation at 15,000 × g at 4 ℃ for 10 min, and the supernatant was transferred to a new tube and stored at -20 ℃ until analysis.
HPLC separation was carried out using a WATERS ACQUITY UPLC System (WATERS) equipped with HILIC-Z 2.7 μm (2.1×100 mm) columns. The mobile phases were acetonitrile and 0.1% formic acid. The column temperature was 45 ℃. The injection volume was 5 mL. The mass spectrometric data were collected on a MALDI SYNAPT Q-TOF mass spectrometer (WATERS) connected with an electrospray ionization interface in positive ion mode (ESI+). Typical ion source parameters were as follows: capillary voltage of 3.5 kV, cone voltage of 30 V, source block temperature of 100 ℃, desolvation temperature of 400 ℃, desolvation gas flow of 700 l/h, cone gas flow of 50 l/h, and collision energy of 6/20 eV. The mass range of m/z 20–2000 was scanned. MassLynx V4.2 software (WATERS) was employed for data acquisition and processing.
Xenograft model
All procedures were conducted in compliance with all the relevant ethical regulations and were approved by the Jiangnan University Animal Welfare and Ethical Review Body. Male BALB/c nude mice aged 5 weeks were purchased from Charles River Laboratories and placed in the Animal Experiment Center of Jiangnan University. Mice were placed five per cage with free access to water and food (chow diet). After habituation for a week, mice were inoculated subcutaneously with Huh7 cells (1.0 ×107 cells in 100 μL PBS per mouse). When the tumors reached 50-100 mm3, the mice were randomly assigned to different groups. Sorafenib was dissolved in dimethyl sulfoxide (DMSO) and diluted in 5% sodium carboxymethyl cellulose. Mice received 20% l-sorbose in water according to their body weights (200 mL/20 mg) by oral gavage once every day. Sorafenib was intragastrically administered to mice at a dose of 50 mg/kg every day. Mice were sacrificed when the xenograft tumor size reached 1,000 mm3. None of the mice showed severe weight loss or signs of infection or wounds. The tumor volume was measured every other day until the endpoint and calculated according to the equation: Volume = Length × Width2 × 1/2.
Western blot
Cells were lysed in RIPA buffer (Solarbio, R0020) supplemented with protease inhibitor cocktail (MCE, HY-K0010) on ice for 30 min. After centrifugation at 15,000 × g for 10 min at 4 ℃, the protein lysates were collected. Then the protein lysates were separated by SDS–PAGE and transferred onto PVDF membranes. The membranes were blocked with 5% nonfat powdered milk for 1 h and incubated with primary antibodies against b-actin (Proteintech, 23660-1-AP, 1:1000), Bax (CST, 2772T, 1:1000), Bcl-2 (CST, 3498T, 1:1000), b-tubulin (Proteintech, 10068-1-AP, 1:1000), PCNA (BBI, D220014, 1:1000), Nrf2 (Abcam, ab137550, 1:1000), HK2 (Proteintech, 66974-1-lg, 1:5000), GLUT5 (Santa Cruz, sc-271005, 1:1000), KHK (Santa Cruz, sc-377411, 1:1000), KHK-A (SAB, 21709, 1:500), and KHK-C (SAB, 21708, 1:500) at 4 ℃ overnight and then probed with the appropriate secondary antibodies for 1 h at room temperature. The bands were visualized with western ELC substrate (BIO-RAD, 1705060), and the images were captured on a Tanon-5200Multi visualization instrument.
Quantitative real-time PCR
Total RNA was isolated from cells using CellAmpTM Direct RNA Prep Kit for RT–PCR (TaKaRa, 3732), and complementary DNA was synthesized from 5 mg total RNA using PrimeScriptTM RT Master Mix (TaKaRa, RR036A). qPCR was performed using TB Green® Premix Ex TaqTM II (Tli RNaseH Plus) (TaKaRa, RR420A) on a Prism 7000 Sequence Detection System (Applied Biosystems) according to the manufacturer’s instructions. Primer sequences for qPCR are listed in Supplementary Table 3.
Cell viability assay
Cells were seeded in 96-well culture plates at a density of 5.0 × 103 cells/well and grown overnight. After treatment with l-sorbose or other reagents, the cells were incubated with CCK-8 (Cell Counting Kit-8, DOJINDO Laboratories, CK04) solution and cultured at 37 ℃ for another 1 h. The absorbance was detected at 450 nm wavelength using a microplate reader (BIO-RAD).
Colony formation assay
Cells were seeded in 6-well plates at a density of 1000 cells/well. Cells were incubated for 24 h to allow attachment to the plates, after which l-sorbose was added to the cells and incubated for 24 h. The cells were cultured for 14 days in the absence of l-sorbose and fixed with a 4% paraformaldehyde fix solution (Beyotime Biotechnology, P0099) for 20 min. Then, the cells were stained with crystal violet (Beyotime Biotechnology, C0121) solution diluted in water. After 10 minutes, the plate was washed with water left to dry and scanned.
Cell apoptosis assay
The cellular apoptosis rate was determined using the Annexin V/PI double-staining Kit (Dojindo Laboratories, AD10). Cells were seeded in 6-well culture plates at a density of 2.0 × 105 cells/well. After incubation with different concentrations of l-sorbose for 24 h, the cells were washed twice with annexin V binding buffer and double-stained with annexin V and PI. Cell apoptosis was examined on a FACSCalibur flow cytometer (BD Accuri C6) and analyzed by FlowJo software.
Measurement of ROS and mitochondrial volume
The Intracellular ROS levels were detected by DCFH-DA (Beyotime Biotechnology, S0033S). Cells were cultured in 6-well plates at a density of 2.0 × 105 cells/well. After being treated with l-sorbose, the cells were gently washed with D-PBS (BBI, E607009) followed by incubation with DCFH-DA at 37 ℃ for 30 min.
To obtain microscopy images, cells were seeded on a confocal dish overnight and treated with l-sorbose for 6 h. After being treated with l-sorbose, the cells were gently washed with D-PBS followed by incubation with MitoBright LT (Dojindo Laboratories, MT11) and DCFH-DA (Dojindo Laboratories, CK04) at 37 ℃ for 30 min. Then the cells were washed with D-PBS twice. Images were obtained with a Nikon C2 Eclipse Ti-E inverted confocal microscope equipped with NIS-Element AR software.
To measure the volume of mitochondria, cells were cultured in 6-well plates at a density of 2.0 × 105 cells/well. After being treated with l-sorbose, the cells were gently washed with D-PBS followed by incubation with MitoBright at 37 ℃ for 30 min. Then the cells were washed twice with D-PBS. The fluorescence of the cells was measured immediately on a FACSCalibur flow cytometer and analyzed by FlowJo software.
Measurement of mitochondrial membrane potential
Cells were cultured in 6-well plates at a density of 2.0 × 105 cells/well. After being treated with l-sorbose, the cells were gently washed with D-PBS. The changes in mitochondrial membrane potential were detected by staining with JC-1 dye (Beyotime Biotechnology, C2006). After incubating with 10 mM JC-1 staining solution in a cell incubator at 37 ℃ for 15 minutes, the cells were washed twice with JC-1 staining buffer and then analyzed by flow cytometry.
Intracellular HK activity measurement
Cells were seeded into 6-well dishes at a density of 2.0 × 105 cells/well. Cells were treated with l-sorbose for 24 h and then collected for the measurement of HK activity using a Hexokinase Activity Detection Kit (Solarbio, BC0740) according to the manufacturer’s protocol. HK activity was calculated and normalized to the cell number.
Production of recombinant ketohexokinase
The plasmid pET28a-KHK was transformed into Escherichia coli BL21 cells. BL21 cells were grown to an absorbance of 0.6-0.8 at 600 nm and then induced with 0.1 mM IPTG for 20 h at 16 ℃. Cells were lysed in lysis buffer (50 mM Tris-HCl pH 7.5), and the cell lysate was precipitated by centrifugation. The enzyme was purified by batch binding to Ni-NTA resin (Qiagen, 30721). The resin was then washed with lysis buffer contain ing 250 mM imidazole, and 2His-tagged KHK was eluted with 500 mM imidazole. The purified enzyme was concentrated and desalted with an Amicon Ultra centrifugal filter (10 kDa) using 50 mM Tris-HCl (pH 7.5). The amounts of purified protein were determined by BCA protein assay kit (Beyotime Biotechnology, P0011).
Statistics and reproducibility
GraphPad Prism8 was used for statistical analysis. At least three independent or parallel experiments were performed for statistical analysis. Data were analyzed by the unpaired Student's t test or one-way ANOVA with Dunnett’s test. Three levels of significance were determined: *P < 0.05, **P < 0.01, ***P < 0.001.