Cell culture and chemicals
Human HCT 116 cells were purchased from Cell Bank/Stem Cell Bank, Chinese Academy of Sciences. Human HEK293T and A549 cells were obtained from American Type Culture Collection. All cells were grown at 37 ℃ with 5% CO2. HCT116 cells were cultured in McCoy’s 5A Medium, HEK293T cells were cultured in Dulbecco’s Modified Eagle Medium (DMEM), and A549 cells were cultured in RPMI 1640 medium purchased from Gibco (Grand Island, NY, USA). All media were supplemented with 10% fetal bovine serum (FBS) (Biological Industries, Kibbutz, Israel), 100 unit/mL penicillin and 100 μg/mL streptomycin (Thermo Fisher Scientific, San Jose, CA, USA). LC-MS grade water and acetonitrile (ACN) were obtained from Merck (Darmstadt, Germany). All chemicals were purchased from Sigma Aldrich (St Louis, MO, USA) unless otherwise specified.
TRAP workflow for purified/recombinant proteins
Model proteins including purified bovine RNase A and human recombinant PKM2 were incubated with their corresponding ligands or solvent for 1 hr at room temperature. The proteins were labeled with 2 μL of 0.5% CD2O and 10 μL of 10 mM Borane Pyridine Complex (BPC) (J&K Scientific Ltd., Beijing, China, cat. no. 121499) for 30 min, and the reaction was quenched by the addition of NH4HCO3 to a final concentration of 50 mM and incubation for 20 min. The resultant mixtures were filtered with 10 kDa molecular weight cut-off (Merck, Darmstadt, Germany) by centrifugation at 12,000 g for 15 min. Then, the enriched proteins were denatured by 8 M urea for 30 min followed by incubation with 5 mM DTT at 56 ℃ for 30 min and subsequent incubation with IAM at room temperature in dark for 30 min. DTT was added again to neutralize the excess IAM for another 10 min. The sample was then diluted with 25 mM NH4HCO3 to make the concentration of urea less than 1 M, and overnight digested by trypsin in an enzyme/protein ratio of 1:40 (wt/wt) at 37 ℃. The digestion was quenched by formic acid (FA) followed by desalting with C18 Ziptip (Waters, Milford, MA, USA). The desalted digest was dried and stored until further LC-MS/MS analysis.
TRAP workflow for human cancer cell line proteome
Cell lysates preparation
Cells were washed with cold phosphate buffered saline (PBS) for three times, scraped and centrifuged at 1,000 rpm for 5 min. The cell pellets were resuspended in mammalian protein extraction reagent (M-PER, Pierce/Thermo Fisher Scientific, cat. no. 78503) containing protease inhibitor (ApexBio Technology, Houston, TX, USA, cat. no. K1007) and phosphatase inhibitor (ApexBio Technology, cat. no. K1013), and lysed on ice for 30 min. The supernatants were collected by centrifugation at 18,000 g for 10 min. Protein concentration was determined by bicinchoninic acid (BCA) Protein Assay (Beyotime Biotechnology, Beijing, China, cat. no. P0011). The cell lysates were then diluted with M-PER lysis to 3 μg/μL and incubated with given metabolites or solvents for 1 hr at 25 ℃. Specifically, the glycolytic metabolites were administered at dosages detailed as follows: 200 μM D-Fructose 1,6-bisphosphate (FBP), 100 μM D-fructose 6-phosphate (F6P), 100 μM D-glucose 6-phosphate (G6P), 30 μM D-ribose 5-phosphate (R5P), 30 μM D/L-glyceraldehyde-3-phosphate (G3P), 10 μM D-2-phosphoglycerate (2PG), 10 μM D-3-phosphoglycerate (3PG), 5 μM phosphoenolpyruvate (PEP), 300 μM sodium pyruvate (Pyr), 2 mM sodium L-lactate (Lac). 2PG was purchased from Shanghai yuanye Bio-Technology Co., Ltd. The dosages of these metabolites were set based on their intracellular concentrations as previously reported52,53.
Proteome labeling and preparation for the label-free quantification-TRAP workflow
Cell lysates (150 μL, 3 μg/μL) were labeled by the addition of 6 μL of 1% CD2O and 90 μL of 10 mM BPC for 30 min. The reaction was quenched by incubating with 50 mM NH4HCO3 for 20 min. Then, methanol, chloroform and water were added to the labeled lysate in order according to a ratio of 4:1:3:1 by volume followed by centrifugation at 12,000 rpm for 10 min to precipitate the proteome. The flaky precipitate was washed twice with methanol and re-solubilized by 8 M urea in 25 mM ammonium bicarbonate solution. Proteins were then reduced by 10 mM DTT by incubation at 56 ºC for 30 min, and alkylated by 40 mM IAM at 25 ºC for 20 min in dark. Additional DTT was added and allowed to react with excess IAM at room temperature for 10 min. The mixture was added with 25 mM NH4HCO3 to dilute urea to 1 M followed by overnight digestion with sequencing-grade trypsin (Promega, Madison, WI, USA) at an enzyme/protein ratio of 1:40 (wt/wt) at 37 ºC. The digestion was quenched by FA addition to pH=3. The mixture was desalted with C18 SepPak cartridges (Waters) and evaporated to dryness with vacuum centrifuge (Thermo Fisher Scientific). Samples were stored at -80 ºC prior to analysis.
Sample preparation for TMT-based multiplexed TRAP
Cell lysates were labeled and precipitated using the same protocol as the label-free quantification-TRAP workflow. The precipitate was resuspended in 8 M urea solution (in 50 mM Tris-HCl containing 10 mM EDTA, pH 8.0) followed by DTT reduction and IAM alkylation. To this was added LysC (Signalchem, cat. no. L585-31N-05) in an enzyme/substrate ratio of 1:400 (wt/wt), followed by incubation at 25 ºC for 4 h. The mixture was diluted with 25 mM NH4HCO3 so that the final concentration of urea is less than 2 M, and then sequencing grade trypsin was added in an enzyme/substrate ratio of 1:50 for overnight digestion at 37 ºC. After quenching by FA, the mixture was desalted with reversed phase SPE cartridges (Waters) and dried with vacuum centrifuge. The sample was resuspended in 300 μL of 50 mM HEPES (pH 8.5) with the concentration of peptides determined by Pierce quantitative colorimetric peptide assay (Thermo Scientific, cat. no. 23275). A 60 μg peptide aliquot of each sample was labeled with TMT reagent for 1.5 hr according to the manufacturer’s instructions. The reaction was quenched by incubating with 10 μL 5% hydroxylamine for 15 min. Aliquots labeled with the 6-plex TMT reagents were combined and acidified with FA followed by evaporation to dryness. The labeled proteome is then desalted again by SPE cartridges and evaporated to dryness. The proteome was dissolved in HPLC phase A buffer (10 mM ammonium formate containing 5% ACN) and then injected into the sample loop of the UPLC system (Acuity, Waters) for fractionation. Phase B consists of ACN with 20% 10 mM ammonium formate aqueous buffer. The flow rate was set as 0.2 mL/min and the gradient was as follows: 0-5 min, 1% B; 5-79 min, 1-50% B; 79-81 min, 50-100% B; 81-98 min, 100% B; 98-100 min, 100-1% B; 100-120 min, 1% B. The effluent was collected every 1.5 min. Every 12 fractions were set as a cycle, and each fraction was combined with the fractions collected in the following cycles. The lyophilized fractions were dissolved in 60 μL of 0.1% FA followed by desalting with C18 Ziptips and storage at -80 ºC prior to analysis.
Mass spectrometry
Intact mass measurement
RNase A (100 μM) and CDP/CTP (1 mM) was both dissolved in 25 mM ammonium acetate buffer and incubated for 30 min, and native MS measurement of the formed holo-complex was conducted on a TripleTOF 5600 system (SCIEX, Framingham, MA, USA) by direct infusion. The instrument was set to acquire over the m/z range of 100−2000 Da for TOF-MS scan.
For intact mass measurement of the dimethylated RNase A with and without ligand incubation, labeled RNase A was desalted by 3 kDa MWCO and analyzed on a C4 column (4.6×150 mm, 3 µm, 300 Å, Sepax Technologies, Newark, DE, USA) on an LC-30 HPLC system (Shimadzu, Kyoto, Japan). The mobile phase consisting of 0.1% FA in water (phase A) and 0.1% formic acid in ACN (phase B) was delivered at a flow rate of 0.4 mL/min using a 15 min gradient program. The eluent was then introduced via ESI ion source into the TripleTOF 5600 system (AB SCIEX, Framingham, MA, USA) for mass measurement. Q-TOF analyzer was set to scan over the m/z range of 100-2000. The spectra were combined by summing across the chromatographic peak of labeled RNase A and deconvoluted using the SCIEX BioPharma View Software.
Label-free proteomic quantification
Data used for label-free quantification was acquired on a nanoACQUITY UPLC system coupled to SYNAPT G2-Si mass spectrometer (Waters). A C18 trapping column (Waters Acquity UPLC M-Class, 0.18×20 mm, 5 μm, 100 Å) and a HSS T3 analytical column (Waters Acquity UPLC M-Class, 75 μm×150 mm, 1.8 μm, 100 Å) were employed. Mobile phases A and B consist of 0.1% FA in water and 0.1% FA in ACN, respectively. A 60 min and 120 min length gradient of 1-40 % ACN at a flow rate of 300 nL/min was used for separation of recombinant protein digests and cell lysates samples, respectively. MS scan range was set to m/z 350-1500 with a scan time of 0.2 s, and MS/MS scan range was set to m/z 50-2000 using data-dependent acquisition (DDA). The top 10 abundant precursors were subjected to MS/MS fragmentation with a ramp CE set between low energy (14-19 eV) and elevated energy (60–90 eV) using a scan time of 0.15 s per function.
TMT-based MS3-level multiplexed quantification
Data used for the multiplexed TRAP workflow was collected on an Orbitrap Fusion Lumos mass spectrometer equipped with an EASY-nano LC 1200 liquid chromatography system (Thermo Fisher Scientific). Mobile phase A consisting of 0.1% FA in water and B consisting of 0.1% FA in ACN-H2O (8:2 by volume) were delivered at a flow rate of 300 nL/min. The 75 μm capillary column was packed with 35 cm of Accucore 150 resin (2.6 μm, 150 Å, Thermo Fisher Scientific). Peptides were analyzed using a 150 min chromatography gradient from 0%-50% phase B during 5-79 min. For MS data acquisition, MS1 spectra were collected at the m/z range of 375-1500 at a resolution of 120,000 in the Orbitrap with a maximum injection time of 50 ms or a maximum automated gain control (AGC) value of 4e5. For MS2 acquisition, fragmentation was conducted by collision-induced dissociation with a normalized collision energy (NCE) at 35. MS2 spectra were collected at the mass range of 400-1200 in ion trap with a maximum AGC of 1e4 or a maximum injection time of 50 ms. For accurate quantification, MS3 were conducted for TMT reporter ion quantification by high energy collision-induced dissociation (HCD) with NCE at 65. The MS3 spectra were collected over the mass range of 100-500 at a resolution of 50,000 with the maximum injection time set at 105 ms and AGC target value at 1e5.
Proteomic data analysis and bioinformatics
Protein identification and quantification
The acquired label-free DDA data and TMT-MS3 data were searched against the Homo sapiens UniProt database (version 2018) using PEAKS Studio 8.5. Due to the necessary lysine labeling step employed by TRAP, we allowed up to two missed cleavages and semi-specific tryptic digestion. Carboxyamidomethylation on cysteines (+57.02 Da) was selected as fixed modification, and methionine oxidation (+15.99 Da), CD2O-mediated dimethylation (+32.06 Da) and mono-methylation (+16.03 Da) on lysines were set as variable modifications. For label-free quantification data, precursor mass tolerance was set to 20 ppm, and fragment mass tolerance was set to 0.1 Da. For TMT-based MS3 quantification data, precursor mass tolerance was set to 10 ppm, MS2 fragment mass tolerance was set to 0.6 Da, and MS3 fragment mass tolerance was set at 0.02 Da. The identified proteins were filtered with 1% FDR and the quantified proteins must include at least one unique peptide. Regarding label-free quantification, we set 50 ppm mass tolerance and 3 min retention time shift tolerance for peptide alignment.
Classification of quantified peptides and proteins
We assessed potential glycolytic targetome by removing proteins associated with keratin firstly from the pool of quantified proteins, and only considered the quantified peptides which belong to the following three types as TRP candidates that are described as follows: a. peptides contain K residues that carry dimethylation or methylation and are not located at the C-terminus; b. peptides contain K, and the K does not carry dimethylation or methylation modification and are located at the C-terminus; c. peptides may not contain K, but the amino acid next to the N-terminal residue is K. Accessibility of peptides that can fit into the above types can be probed by TRAP, and are thus considered for TRP screening.
Determination of TRPs for drugs
As for target screening based on label-free quantification data, the screening standard of TRPs was set as peptides displaying p value < 0.01 and TRAP Ratio Rdrug/control > 2 or < 0.5 in the presence of the assayed drugs relative to the control. For drug targets discovery made based on the TMT-aided multiplexed TRAP data, a more stringent cutoff was utilized for TRP screening: peptides displaying p value < 0.0001 and TRAP Ratio Rdrug/control) > 2 or < 0.5 in the presence of ligands relative to the control.
Determination of TRPs for metabolites
For quantitative screening of the TRPs for the metabolites of interest, we first classified all quantified peptides into two categories, loose and compact. The loose category refers to the TRPs that become more chemically accessible to TRAP labeling reagents CD2O after incubation with given metabolites. The increased chemical accessibility is assigned based on the increased abundance of peptides that carry dimethylated K (type a) or decreased abundance of peptides belonging to type b and c. Conversely, the compact category refers to the peptides that become less accessible to TRAP labeling after the administration of given metabolites. The reduced chemical accessibility is judged by the decreased abundance of type a peptides or increased abundance of type b and c peptides.
We set the standard of TRP screening for metabolites as peptides that display significant abundance changes with q value < 0.03 (q values are used to adjust for multiple testing using the Benjamini-Hochberg method to control the FDR at the cut-off level of 0.03) and the TRAP Ratio Rtreated/control > 1.5 or < 0.67 for compact peptides while Rtreated/control > 2 or < 0.5 for loose peptides. We posit that loose peptides reflect indirect binding events or conformational changes induced by direct binding, so more stringent criteria was given for this category.
Quality assessment of TRAP results
In order to assess the quality of our results, we estimated true positive rate generated by the TRAP approach by modifying previous assessment method9. We collected the known interactions from the BRENDA repository and set the species as homo sapiens. Among all potential enzyme-metabolite interactions, 140 known enzyme-metabolite interactions for homo sapiens in BRENDA were retrieved. Our TRAP results detected 30 known enzyme-metabolite interactions that are classified as true positive hits, which confer a true positive rate as 21.43% (calculated by 30/140).
Volcano plot of the TRAP-identified targetome
In the volcano plot, each point accords to a protein that is represented by a peptide selected based on a scoring system. The score of each quantified peptide for given proteins is obtained by consideration of both the TRAP ratio Rtreated/control and p/q value of the peptide abundances between samples with and without given ligands shown as follows.
TRAP Score = -Log10 (p value/q value) * Abs[log2 (Ratiotreated/control)]
After scoring, the peptide with the maximum TRAP score was selected to represent the given protein.
Secondary structure analysis
UniProt identifiers of all quantified proteins were matched with PDB accession number from Protein Data Bank (PDB) (http://www.rcsb.org/pdb/search/searchModels.do) and only entries with >90% sequence identity were retrieved for analysis. Secondary structure information of the compiled protein pool was downloaded from the DSSP (https://swift.cmbi.umcn.nl/gv/dssp) database and Python script was used to extract the secondary structure for each quantified lysine residue54. The extracted secondary structure classes were classified into four categories, namely helix (DSSP classes H, G, I), sheet (DSSP classes B, E), loop (T, S) and no structure (“ ”).
Conservation analysis
To estimate the sequence conservation of the obtained Lysine sites from TRPs, we calculated the Lysine sequence identity across 11 representative vertebrate species (Human, Rhesus monkey, Mouse, Rat, Cow, Dog, Opossum, Chicken, Frog, Zebrafish, and Fugu) using an in-house perl script. Specifically, we downloaded multiple amino acid sequence alignment of coding sequence (CDS) region across 100 species (multiz100way) from UCSC Genome Browser55. For each gene, its CDS region alignment across the selected 11 representative vertebrate species was further extracted. For each lysine from TRPs, its sequence conservation was estimated using the percentage of sequence conservation across 11 representative vertebrate species. To further examine whether the obtained TRP lysine sites were more conserved than random expectation, we calculated the sequence conservation of all quantified Lysine sites to estimate the background Lysine conservation, and then use Kolmogorov-Smirnov (KS) test to evaluate the statistical significance of excessive sequence conservation of the obtained TRP Lysine residues. The KS test p <0.05 was considered as significant.
Measurements of Euclidean distances
For functional sites analysis, PyMOL-Python scripts was used to measure the Euclidean distances between the atoms of lysine in TRPs and any atoms of annotated ligands (such as substrates, cofactors, products) in Å for enzymes assigned as glycolytic metabolites targets by TRAP for those with available structures retrieved from PDB files. The minimum distance was recorded to represent each ligand-target pair. Further, if the minimum distance is less than 10 Å, the lysine is categorized as functional and otherwise as unfunctional.
The active site boundary detectable by TRAP is defined based on the median of the minimum distances measured between the TRPs of known enzymatic targets that use the examined metabolites as substrates and their corresponding active sites.
For evaluation of the metabolites’ influence on given enzymes’ activities, the minimum distance between all TRPs from the identified targets involved in the carbohydrate metabolism pathway and their corresponding active sites were measured and shown in Fig. 5c.
GO and KEGG pathway analysis
All the TRAP-identified target proteins were annotated to non-overlapping GO molecule function (MF) terms. The MF classification was performed using the functional annotation tool of PANTHER (http://pantherdb.org/). GO MF terms include Transcription regulator activity (GO: 0140110), Catalytic activity (GO:0003824), Transporter activity (GO:0005215), Molecular transducer activity (GO:0060089), Translation regulator activity (GO:0045182), Structural molecule activity (GO:0005198), Molecular function regulator (GO:0098772), Binding (GO:0005488). If a protein belongs to more than one class, they are categorized based on the above order to prioritize. Proteins that do not match any of the above GO terms were sorted into the category “Uncategorized”.
Specifically, for proteins sorted in the “Catalytic activity” category, ClueGO app included in Cytoscape (version 3.7.1) was employed to perform the KEGG pathway annotation network analysis. A setting of group p value <0.001 and inclusion of at least three genes in each group was used for filtering.
GO biological process (BP) analysis of the druggable glycolytic targetome was performed by BiNGO app included in Cytoscape with a setting of group p value <1e-15.
DrugBank analysis for ligandability classification
The DrugBank database (v. 5.1.3 released on 2019-04-02; group "all_ target_polypeptide_ids") were downloaded and used to classify the glycolytic targetome into DrugBank (ligandable) and non-DrugBank (unligandable) proteins.
Analysis of lysine PTM
The PTM information of lysines from all quantified proteins was collected from the iPTMnet database (https://research.bioinformatics.udel.edu/iptmnet). Glycolytic metabolite-target interaction is considered to potentially affect the lysine-bearing PTM status if a given lysine in TRPs is reported to carry PTM.
Co-immunoprecipitation experiment
Sample preparation
Cultured HEK293T cells were first transfected with plasmids encoding FLAG-TRIM28 or hygro-Negative Control vector using Polyjet transfection reagents for 16 hr at 37 °C following the manufacturer’s instructions. After transfection, the cells were treated with 10 mM sodium lactate or the matching solvent for 24 hr. Then, the cells were harvested and lysed in 1% NP-40 lysis buffer (P0013F, Beyotime Biotechnology, Shanghai, China) with protease inhibitor cocktail and phosphatase inhibitor cocktail. The supernatants were collected by centrifugation at 18,000 g for 10 min. Protein concentrations were determined by BCA assay. For co-immunoprecipitation (co-IP) experiments, approximately 1 mg of protein lysates were incubated overnight at 4°C with anti-FLAG antibody. The lysates were then incubated with Protein A/G agarose beads (Invitrogen, cat. no. 15918014) for 4 hr at 4°C. The beads-bound proteins were then eluted with SDS loading buffer, heated at 95°C for 10 min and then collected for immunoblotting. For analysis of the enriched interactome, on beads-digestion and reductive dimethylated-based labeling were performed. Briefly, the enriched proteins were washed with PBS for three times followed by incubation with urea, DTT, IAA and trypsin for overnight digestion. The resultant mixtures were centrifuged at 10,000 rpm, and the supernatants were collected and desalted with C18 SepPak cartridges. The digested peptides were re-solubilized in 100 mM TEAB buffer (ThermoFisher, cat. no. 90114) and labeled by the addition of 4 μL 4% CH2O (light-label group) (Sigma, cat. no. 47608-250ML-F) or CD2O (heavy-label group) and 4 μL 0.6 M NaBH4CN (Sigma, cat. no. 452882) for 1 hr at room temperature. The dimethylation reaction was quenched by with the addition of 16 μL 1% ammonia for 15 min followed by further termination with 4 μL FA. Then, the light and heavy-labeled samples were combined, evaporated, and reconstituted in 0.1% FA aqueous solution followed by desalting with C18 ZipTips. The labeled interactome samples were then evaporated to dryness and subjected to proteomic analysis as previously described.
Data acquisition and analysis:
The acquired light and heavy-dimethylated labeled proteomics data were searched against the Homo sapiens UniProt database (version 2021.09.15, entry 20413) using PEAKS Studio 8.5. Up to three missed cleavages and semi-specific tryptic digestion were allowed. Carboxyamidomethylation on cysteines (+57.02 Da) was selected as fixed modification, and methionine oxidation (+15.99 Da), CD2O-mediated dimethylation (+32.06 Da) and CH2O-mediated methylation (+28.03 Da) on lysines were set as variable modifications. For dimethylation labeling-based quantitative proteomics data, precursor mass tolerance was set to 10 ppm, and fragment mass tolerance was set to 0.02 Da. The interacting proteins were screened following the criteria: the protein must have at least 2 unique peptides and can only be detected in the TRIM28-overexpression group (detactable in all replicates) rather than the vector control group. Then, the abundances of the TRIM28-interacting proteins were normalized to the abundance of TRIM28 in each group to obtain the Relative Affinity. Affinity Ratio is further obtained by comparing the Relative Affinity of each TRIM28-interacting proteins with and without lactate. The protein-protein network was built by Cytoscape and the annotated known PPIs were retrieved from databases including STRING, BioGRID, PrePPI, and IID (Integrated Interactions Database). The colors of the nodes represent the changed binding affinity to TRIM28 following lactate treatment, and the colors of the frames represent the frequency of retrieving the identified PPIs in this study from the four databases.
Immunoblotting
Cells were lysed in RIPA lysis buffer supplemented with protease inhibitor cocktail. The protein concentrations were then determined by the BCA assay. The lysates were diluted by 4× XT Sample Buffer, heated to 95 ℃ for 5 min, cooled and separated by 8%-12% sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE). After being transferred onto polyvinylidene difluoride (PVDF) membranes, proteins were blocked using 5% non-fat dry milk in Tris-buffered saline with 0.1% Tween 20 detergent (TBST) and incubated with primary antibodies at 4 °C overnight. After being washed five times with TBST, the membranes were incubated with HRP-conjugated secondary antibodies for 1 hr at 37 ℃. The immunoblotted bands were detected by the addition of horseradish peroxidase (HRP) substrate and captured on a ChemDoc XRS+ System (Bio-Rad). The primary antibodies used in this study include antibodies against β-tubulin (AP0064, Bioworld Technology), TRIM28 (, 4123, Cell Signaling Technology), ENO1 (3810, Cell Signaling Technology), α-tubulin (11224-1-AP, Proteintech), FLAG tag for WB (8146, Cell Signaling Technology), FLAG tag for immunoprecipitation (14793, Cell Signaling Technology), Histone H3 (4499, Cell Signaling Technology, Acetylated H3K27 (PTM-116RM, PTM Biolabs), cleaved PARP (5625, Cell Signaling Technology), cleaved caspase-3 (9661, Cell Signaling Technology).
Subcloning and mutagenesis
The synthetic genes encoding wild type (WT) human ENO1 (UniProt: P06733) and PKM2 (UniProt: P14618) with an N-terminal hexahistidine purification (His6) tag were subcloned into the GV296 vector (Genechem, Shanghai, China). The synthetic genes encoding WT human TRIM28 with an N-terminal maltose binding protein (MBP) affinity tag (UniProt: Q13263) were subcloned to the pMAL-p2X vector (New England Biolabs, Beverly, MA, USA). The ENO1 K330E and TRIM28 K337E/K340E double mutants were obtained by site-directed mutagenesis. PCR primer sequences used for ENO1 and TRIM28 mutagenesis are listed as below:
Human ENO1 K330E
|
F: GATCGCCGAGGCCGTGAACGAGAAGTCCTG
|
|
R: ACGGCCTCGGCGATCCTCTTTGGGTTGGTC
|
Human TRIM28 K335E
|
F: GAACGAGGAGTCCTGCAACTGCCTCCTGCT
R: CAGGACTCCTCGTTCACGGCCTTGGCGATC
|
Human TRIM28 K337E/K340E
|
F: GAGATCCAGGAGCACCAGGAGCACATTCTG
R: CTGGTGCTCCTGGATCTCGGTCATGGTCCAGTGCTGCC
|
Recombinant protein expression and purification
All His-tagged recombinant protein expression and purification steps are generally as follows: plasmids were transformed into BL21(DE3) E. coli cells and the transformed bacteria were selected on LB plate containing 50 µg/mL kanamycin. The isolated colonies were grown in LB medium at 37 ℃ and 220 rpm until the OD600 of culture arrive 0.8. The target protein expression was induced with 0.5 mM isopropyl β-D-thiogalactoside (IPTG) by shaking overnight at 16 ℃. Cells were harvested by centrifugation followed by resuspension in ultrapure water containing protease inhibitor cocktail, and then lysed by sonication. Lysates were centrifuged for 20 min at 18,000 g at 4 ℃ and the resultant supernatants were discarded. The His-tagged target proteins were then purified with a His-tag Protein Purification Kit (Beyotime Biotechnology, cat. no. P2226). After purification, proteins were concentrated by filter with a 10 kDa cut-off and the purity was evaluated by SDS-PAGE.
For the expression of TRIM28 constructs, cultures were supplemented with LB medium containing 50 µg/mL ampicillin and 0.5% glucose. Protein expression was induced at OD600 = 0.8 with 0.2 mM IPTG by shaking at 100 rpm overnight at 16 ℃. To purify the MBP-tagged TRIM28, cells were resuspended in lysis buffer (50 mM Tris pH 7.4, 50mM NaCl, 5 mM DTT, 1:10,000 (v/v) benzonase solution, 1×protease inhibitor cocktail and lysed by sonication. The lysate was clarified by centrifugation. The supernatant was transferred to amylose resin (New England Biolabs) and incubated overnight at 4 ℃. The protein was purified using a New England Biolabs kit according to the manufacturers’ protocol. The bound fusion protein was eluted with 10 mM maltose (New England Biolabs). The yield of the fusion proteins was evaluated by separation on SDS-PAGE and visualization via Coomassie blue staining.
PKM2 activity assay
In vitro activity of PKM2 was performed using luminescent Kinase-Glo Plus reagent (Promega, cat. no. V3773) and used according to the manufacturer’s instructions. In brief, human recombinant PKM2 was diluted in the assay buffer (50 mM Tris HCl, 100 mM KCl, 10 mM MgCl2, PH 7.4) and incubated with each metabolite/drug/vehicle at 25 ℃ for 40 min. Substrate solution was prepared by mixing ADP at 200 μM and PEP at 200 μM with the assay buffer. Then, a 50 μL aliquot of substrate solution was added to 50 μL recombinant PKM2 solution, and allowed to react for 10 min at room temperature in 96-well plates. The luminescence response of each sample was measured due to the formation of ATP at 37 ℃ in a Gen5 platform (BioTek). The measurements were fitted by a nonlinear fitting algorithm (log [agonist] vs. response-variable slope with 4 parameters) in Prism 8.0.1 (GraphPad, San Diego, CA, USA).
GAPDH activity assay
In vitro GAPDH enzymatic activity assay was performed in 96-well plates at room temperature by measuring the reduced NAD+ level. The recombinant GAPDH (Abnova, Taiwan, China, cat. no. P4547) was first diluted in 10 mM sodium pyrophosphate buffer (pH 8.5) to 30 U/mg. A 100 μL of reaction mixture containing 20 mM sodium arsenate, 1 mM NAD+ and 2.88 mM G3P was readily added to GAPDH. The NAD+ level was examined by measuring the absorbance at 340 nm every 20 s for 20 min. Reaction rate was calculated by curve fitting of the time course measurements by linear regression.
NAMPT activity assay
In vitro activity of NAMPT was measured using a NAMPT colorimetric assay kit (CycLex, MBL International, cat. no. CY 1251V2) in 96-well plates by the one-step method. In brief, recombinant NAMPT was diluted in NAMPT assay buffer and subsequently incubated with solvent/metabolites/FK866 for 30 min at 30 ℃. The reaction was initiated by the addition of 60 μL One-Step Assay Mixture to each well. NAMPT activity was measured photometrically in absorbance at 450 nm for 60 min with a 1 min-interval.
Thermal shift assay and isothermal dose-response assay
For thermal shift assay conducted using cell lysate samples, cultured HCT116 cells were harvested and washed with PBS. The washed cells were diluted in 0.1% NP-40 lysis buffer supplemented with 1× protease inhibitor cocktail and 1× phosphatase inhibitor cocktail. The cell suspensions were freeze-thawed five times using liquid nitrogen and passed through a 27” gauge needle five times. The samples were snap-frozen in liquid nitrogen for > 1 min and placed onto a heating block set at 25 °C until 60% of the confluent was thawed. Then, the samples were subjected to centrifugation at 20,000 g for 20 min at 4 °C. The supernatant was collected and divided into two aliquots, with one aliquot being treated with given glycolytic metabolites and another aliquot with the solvent as the control. After 60 min-incubation at room temperature, each sample were further divided into 8 aliquots and heated individually at their designated temperature for 3 min in a 96-well thermal cycler followed by cooling at room temperature for 3 min. The heated lysates were centrifuged at 20,000 g for 20 min at 4 °C in order to separate the soluble fractions from precipitates. Each resultant soluble fraction of the proteome was transferred to a new low-adsorption 1.5 mL microtube and analyzed by SDS-PAGE followed by western blotting for target proteins.
For the cell lysate-level isothermal dose-response (ITDR) experiments, G3P were serially diluted to generate a 9 points dose–response curve. HCT116 cells lysate were incubated with the assayed metabolite of serial concentrations (at least eight concentrations) and one vehicle as control in 1.5 mL low absorption microtubes for 1 hr at room temperature. Then, the samples were aliquoted to 120 μL and transferred into 200 μL PCR microtubes followed by heating at designated temperatures for 3 min in a 96-well thermal cycler and subsequent cooling for 3 min at room temperature. Then, isolation of the soluble fractions and immunoblotting of given target protein are repeated as described for thermal shift assays.
For cell lysate-level thermal shift assay, the band intensities detected at increasing temperatures were normalized to that of the lowest temperature, and the Boltzmann sigmoid equation was fitted using GraphPad Prism. For the ITDR experiments, the band intensities at increasing doses were normalized to the intensity at the highest concentration of the assayed metabolites and analyzed by the saturation binding curve function in Prism.
Surface plasmon resonance analysis
SPR analysis was conducted on a Biacore T200 system (GE Healthcare, Sweden). Target protein was diluted in 10 mM sodium acetate and immobilized via the amine coupling method on a CM5 sensor chip. Metabolites were dissolved in H2O and diluted to a serial concentration with running buffer (PBS with 0.05% tween 20). Then, the metabolites were injected through the reference and active channels at a flow rate of 30 μL/min. The association and dissociation times were both set at 60 s. The affinity fitting was carried out on a Biacore T200 evaluation software by global fitting via a steady-state affinity model to obtain the equilibrium dissociation constant KD.
Protein thermal stability analysis by nanoDSF
Both the WT-ENO1 (0.5 μg/μL) and the mutant-ENO1 (0.5 μg/μL) were incubated with 2 mM G3P for 30 minutes firstly. Samples were filled within the nanoDSF capillaries (n=3), and subsequently loaded into the Prometheus NT.48 device (NanoTemper Technologies, Germany). Samples were heated from 20 °C to 95 °C with a slope of 1 °C/min and the unfolding transition temperatures were automatically identified by the PR. ThermControl software (NanoTemper Technologies). Raw data was exported for plotting the thermal stability curve in GraphPad Prism.
In vitro lysine acetylation (Kac) assay
The in vitro Kac assay was performed by following literature56. Briefly, the human recombinant ENO1 and Histone 3.3 (10 μg) were diluted with the reaction buffer (50 mM Tris-HCl, pH 8.0, 100 nM trichostatin A, 0.1 mM EDTA, 1 mM DTT and protease inhibitor cocktail) and incubated with G3P and pyruvate, respectively, for 30 min. Recombinant Histone 3.3 was purchased from New England Biolab (cat. no. M2507S).Then, the mixtures were incubated with 2 mM Acetyl-CoA (Shanghai yuanye Bio-Technology Co., Ltd, cat. no. Y34045) and 0.55 μg of the recombinant human p300 protein (Active Motif, cat. no. 81158) at 30 °C for 45 min for in vitro Kac. The Kac process was terminated by adding 8 M urea, and subsequently digestion was performed as previously described. The digested proteins were desalted with C18 SepPak cartridges, evaporated to dryness and subjected to bottom-up analysis.
Flow cytometric analysis of cell apoptosis
Cell apoptotic rate was examined using an Annexin-V/FITC Kit (BD Biosciences, San Jose, CA, USA). Cells were pretreated with pyruvate for 4 hr and then treated with TSA for 24 hr. The cells were harvested, washed with PBS, resuspended in 1× Annexin V binding buffer, and stained with annexin V and PI for 15 min at room temperature in darkness. The rates of apoptotic cells were determined using an Accuri C6 flow cytometer (BD Biosciences).
Statistical analysis
Statistical analysis was performed in GraphPad Prism. All data represent mean ± SEM (n = 3 or 5 per group as indicated in legend). The statistical significance of differences between two groups was determined using Student’s t-test (unpaired, two-tailed) unless otherwise specified, *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.
References
52. Chen, W. W., Freinkman, E., Wang, T., Birsoy, K., & Sabatini, D. M. (2016). Absolute quantification of matrix metabolites reveals the dynamics of mitochondrial metabolism. Cell 166, 1324–1337.e11.
53. Miyo, M., Konno, M., Nishida, N., Sueda, T., Noguchi, K., Matsui, H., Colvin, H., Kawamoto, K., Koseki, J., Haraguchi, N., Nishimura, J., Hata, T., Gotoh, N., Matsuda, F., Satoh, T., Mizushima, T., Shimizu, H., Doki, Y., Mori, M., & Ishii, H. (2016). Metabolic adaptation to nutritional stress in human colorectal cancer. Sci. Rep. 6, 38415.
54. Feng, Y., De Franceschi, G., Kahraman, A., Soste, M., Melnik, A., Boersema, P. J., de Laureto, P. P., Nikolaev, Y., Oliveira, A. P., & Picotti, P. (2014). Global analysis of protein structural changes in complex proteomes. Nat. Biotechnol. 32, 1036–1044.
55. Haeussler, M., Zweig, A. S., Tyner, C., Speir, M. L., Rosenbloom, K. R., Raney, B. J., Lee, C. M., Lee, B. T., Hinrichs, A. S., Gonzalez, J. N., Gibson, D., Diekhans, M., Clawson, H., Casper, J., Barber, G. P., Haussler, D., Kuhn, R. M., & Kent, W. J. (2019). The UCSC Genome Browser database: 2019 update. Nucleic Acids Res. 47, D853–D858.
56. Huang, H., Tang, S., Ji, M., Tang, Z., Shimada, M., Liu, X., Qi, S., Locasale, J. W., Roeder, R. G., Zhao, Y., & Li, X. (2018). p300-mediated lysine 2-hydroxyisobutyrylation regulates glycolysis. Mol. Cell 70, 663–678.e6.