Patients and tissue samples
The study was granted approval from the Ethics Committee of the Third Xiangya Hospital of Central South University. In accordance with the Declaration of Helsinki, informed consent was obtained from the patients. Both tissue samples and clinical data were obtained from the patients diagnosed with DLBCL before treatment between December 2018 and August 2020. The diagnosis of DLBCL was confirmed by at least two pathologists in line with the World Health Organization classification27. After collection, the tissues were immediately frozen and stored in liquid nitrogen. Cryopreserved tissues contained both cancerous and paired distant normal tissues. This study was carried out in a retrospective series of 56 DLBCL cases with cryopreserved tissues, with follow-up lasting until December 2020.
Cell culture and reagents
The human DLBCL cell lines U2932, OCI-ly7 and OCI-ly8, and human normal B-lymphocyte cell line GM12878 were provided by the Cancer Research Institute of Central South University. The human DLBCL cell line SU-DHL-2 and the R-CHOP-resistant DLBCL cell line SU-DHL-2/R were sourced from Xiangya Hospital of Central South University and then characterized as reported previously28. All of the cell lines were cultured in RPMI 1640 (Gibco) with 12% foetal bovine serum, 100 U/mL penicillin and 100 mg/mL streptomycin.
Rituximab-resistant DLBCL cell line OCI-ly8/R was established as previously reported29. OCI-ly8 cells were exposed to rituximab for weeks to obtain rituximab-resistant lines. In brief, sensitive parental cell lines were cultured in RPMI-1640 and the cells were exposed to an increasing dose of rituximab for 24 hours immediately after the log phase of growth was reached (from 0.1 to 128 μg/mL). After 24 hours of incubation with rituximab, the cells were centrifuged and recultured in fresh RPMI 1640. Then, the cells were allowed to redevelop for a minimum of 3 days, and the procedure was repeated for 10 times in total once the log phase of growth was reached. The reagents used include rituximab (Roche Pharma), sodium dichloroacetate (#S8615, Selleckchem), puromycin (#S7417, Selleckchem) and G418 Sulfate (#S3028, Selleckchem).
Cell growth ability assay
For cell growth ability assay, 2000 cells in a 200 µL volume were added per well of a 96-well plate and treated with rituximab (50 μg/ml) for a specified period of time. Countess Automated Cell Counter (Thermo Fisher Scientifc) was applied to evaluate count cells. 100% cell viability was measured at hour 0, while relative cell growth was indicated by the ratio of hour X to hour 0.
Cell viability assay
Cell viability assay was performed using CCK-8 solution (Dojindo, Japan) in line with the manufacturer’s instructions. In brief, the cells were seeded in 96-well plates (5 × 103 cells per well) and exposed to rituximab (50 μg/ml) for 48 hours either at increased doses, alone or in combination. CCK-8 assay was carried out according to the standard procedure and the plate was measured at 450 nm using a microplate reader (PerkinElmer, USA).
Cell apoptosis assay
The cells were harvested after 48 hours of treatment, washed with ice-cold PBS, and re-suspended in 400 μL of binding buffer. Then, 20 μg/mL FITC-Annexin V and 5 μg/mL propidium iodide (BD Biosciences) were added to each sample and incubated for 15 minutes. Flow cytometry (Becton Dickinson, USA) was employed to analyse the stained cells, with apoptotic cells treated as Annexin V-positive cells.
Western blotting
Total proteins were extracted with the assistance of RIPA lysis buffer (NCM Biotech) containing freshly added proteinase inhibitor. Then, the proteins obtained were separated by 10–12 % sodium dodecyl sulphate-polyacrylamide gelelectrophoresis and transferred to 0.22 μm PVDF membranes (Millipore). The membranes were blocked with 5 % skim milk and then incubated with primary antibodies overnight at 4 ºC. The respective horseradish peroxidase-conjugated secondary antibodies were added, and protein signals were detected using enhanced chemiluminescence reagents (Affinity Biosciences). ChemiDox XRS Chemiluminescence imaging system (Bio-Rad Laboratories, USA) was adopted to capture and analyse the developed images. The primary antibodies are detailed as follows: anti-PDK4 (DF7169, Affinity Biosciences, 1:1000), anti-CD20 (DF13319, Affinity Biosciences, 1:1000), anti-β-actin (AF7018, Affinity Biosciences, 1:3000).
real-time quantitative polymerase chain reaction (qRT-PCR)
Total RNA was extracted from tissues and cells using the FastPure Cell/Tissue Total RNA Isolation Kit (Vazyme, Nanjing, China). Then, 2 μg of RNA was converted to cDNA with the HiScript III RT SuperMix for qPCR (+gDNA wiper) Kits (Vazyme) according to the instructions from the manufacturer. Gene expression (mRNA) was analysed using the ChamQ Universal SYBR qPCR Master Mix (Vazyme) and LightCycler 480 real-time PCR instrument (Roche) in a two-step qRT-PCR (95 ºC for 30 s, which is followed by 40 cycles of 95 ºC for 10 s and 60 ºC for 30 s). The specific primers (Supplementary Table S2) were synthesized in the Beijing Genomics Institute. Human B-lymphocyte GM12878 cells were taken as a calibrator. The mRNA relative levels of the target genes were calculated using the 2-ΔΔCt method.
RNA-seq and bioinformatics
Total RNA was derived from the tissues as mentioned above. Total mRNA preparation and sequencing were performed in the Beijing Genomics Institute (BGI, Shenzhen, Guangdong, China)25,30. In brief, the quality of the RNA samples was assessed using an Agilent Bioanalyzer (Agilent). cDNA libraries were generated using TruSeq RNA Sample Preparation (Illumina), with each library sequenced using single-reads on a HiSeq2000/1000 (Illumina). The levels of gene expression were measured in RPKM using Cufflinks31. Differentially expressed genes (DEGs) were identified using the DESeq2 R package. The criteria for DEGs were set up as fold change (FC, log2) >2 or <−2, Q-value <0.05, and FDR<0.05. RNA sequencing data was analysed by Partek Inc. (St. Louis, MO).
Determination of glucose consumption, lactate production, and ATP levels
The glucose consumption, lactate production, and ATP level assays were conducted according to the previous study32. The cells were cultured for a period of 20 hours. The culture media were then harvested, while the lactate and glucose concentrations were measured using a Lactate Assay kit (BioVision, CA, USA) and glucose assay kit (Sigma-Aldrich), respectively. ATP levels were quantified using a colorimetric ATP Assay Kit (Beyotime, Jiangsu, China) according to the instructions from the manufacturer. In brief, cells were harvested and lysed, which is followed by the mixing of supernatant with luciferase reagent, thus catalyzing the light production from ATP and luciferin. Linearly related to the level of ATP concentration, the emitted light was measured using a microplate luminometer. Protein concentration, as measured using a bicinchoninic acid (BCA) protein assay (Beyotime), was referenced to normalize all lactate, glucose, and ATP measurements.
Analysis of mitochondrial ATP synthesis and energy budget calculations.
The rate of mitochondrial ATP synthesis was determined according to previous description33. In brief, 2.5 × 105 cells were resuspended in 1 ml of buffer containing 150 mM KCl, 25 mM Tris-HCl, 2 mM EDTA, 0.1% BSA, 10 mM K3PO4, 0.1 mM MgCl2, 40 g/ml digitonin, 0.15 mM P1, P5-Di (adenosine) pentaphosphate (an inhibitor of adenylate kinase), 10 mM malate, 10 mM pyruvate, and 1 mM ADP, either with or without 1 M oligomycin. The cells were incubated at 37°C for 15 minutes. At 0, 5, 10, and 15 min, 50 μl of aliquots of the reaction mixture were quenched in 450 μl of boiling buffer containing 100 mM Tris-HCl and 4 mM EDTA (pH 7.75) for 2 minutes. Then, the aliquots were diluted 1/10 in the quenching buffer and the quantity of ATP was determined using the colorimetric ATP Assay Kit. The rate of mitochondrial ATP synthesis was calculated on the basis of difference in ATP content in the presence and absence of oligomycin.
As for the calculation of energy budget, the contribution of mitochondria and glycolysis to total cellular ATP was measured as previously reported9,34. In brief, oxidative ATP turnover was calculated at a presumed P/O ratio of 2.36, where P represents nmol of ATP produced per nmol of oxygen consumed (O), which was in turn derived from the oligomycin-sensitive proportion of OCR. The contribution of glycolysis to energy budget was calculated at a presumed stoichiometric ratio of 1:1 between glycolytically-derived lactate (as described above) and glycolytically-derived ATP. OCR and lactate measurements were performed over a 2-hour period. For each cell line, nmol of ATP derived from glycolysis or oxidative phosphorylation was then pooled and the contributions to the total ATP production were calculated in percentage.
Measurement of ECAR and OCR
The cells were seeded in a 96-well plate with a density of 5 × 104 cells/well and treatment as indicated. Then, they were treated with ECAR reagents according to the recommendations made by the manufacturer (Abcam, UK). The ECAR measurements were performed at an interval of 5 minutes for a total assay time of 120 minutes in a micro-plate reader system (PerkinElmer), with the excitation and emission wavelengths of 380 and 615 nm used, respectively. The level of OCR was measured in real time using the XF24 extracellular flux analyzer instrument (Seahorse Bioscience, USA) as described previously9,35. In brief, the cells were equilibrated with bicarbonate-free RPMI 1640 medium supplemented with 25 mM glucose, 1 mM pyruvate, and 2 mM glutamine and incubated overnight. Three metabolic inhibitors were sequentially loaded at a number of specific time points: 1 μM oligomycin, 0.5 μM FCCP, and a combination of 100 nM rotenone with 100 nM myxothiazol. The level of OCR was measured both at the baseline and after the addition of each reagents.
Lentiviral infection
Two pairs of short hairpin RNA sequences (shRNA1 and shRNA2) targeting human PDK4 were annealed and ligated into the Plk0.1-puro lentiviral vector in line with the manufacturer’s instruction. The targeting sequences of PDK4 shRNA-1 and shRNA-2 were determined as 5′-ACTGCAACGTCTCTGAGGTG-3′ and 5′-AAGCAGATCGAGCGCTACTC-3′, respectively. A scrambled shRNA was applied as a control. HEK293T cells were transfected with 16 μg of plasmid containing the expected construct, 12 μg of packing vector pSPAX2 and 8 μg of envelope vector pMD2G in a 10 cm dish for obtaining lentivirus. After the elapse of 48 hours, the culture supernatant was filtered through the 0.45 μm filter and virus concentrated using the PEG 8000 method. Lymphoma cells were incubated with the virus prepared overnight, and the culture medium was replaced with 5 mL of fresh medium. Then, puromycin (2 μg/mL) was used to select infected cells.
Human PDK4 coding sequence was cloned into the pcDNA3.1 plasmid for the generation of a pcDNA/PDK4 expression plasmid. The pcDNA3.1 was treated as empty vector control for subsequent analysis. In order to generate PDK4 stable over expression cells, the lymphoma cells were transfected with pcDNA/PDK4 or vector control through liposome-mediated transfection. G418 (700 μg/ml) was applied to select the transfected cells. After two weeks of selection, the survived cells were seeded individually into 96-well plate for further expansion. The transfection efficacy was determined by means of RT-qPCR and western blotting analysis.
Immunofluorescence analysis
The cells were collected and placed on glass substrates for 20 minutes of fixing with 4 % paraformaldehyde. Then, the fixed cells were rinsed thrice, permeabilized with 0.1 % Triton X-100 (Sigma, USA) for 15 minutes, and blocked with 5 % BSA in phosphate buffered saline (PBS) for 1 hour at room temperature (15-25 ºC). Afterwards, the cells were incubated overnight at 4 ºC with primary antibody against CD20 (DF13319, Affinity Biosciences, 1: 200) and stained with secondary antibody conjugated with Alexa Fluor 488 goat anti-rabbit (S0018, Affinity Biosciences, 1: 200) for 1 hour at room temperature in the dark, which is followed by counterstaining with 4′,6-diamidino-2-phenylindole (Sigma). After three PBS rinses, the samples were covered with coverslip using Antifade Solution (Solarbio). The images were analysed and captured by confocal fluorescence microscope (Olympus, Japan).
Tumour xenografts in mice
All experiments involving animals were approved by the Institutional Animal Care and Use Committee of Central South University, China. The female B-NDG mice (18–20g, 5–7 weeks old) used in this study were obtained from Jiansu Biocytogen Co., Ltd (Nantong, China) 36-38. DLBCL xenograft mouse model was constructed through the subcutaneous injection of 1 × 107 PDK4 overexpressing OCI-ly8 cells (OCI-ly8 PDK4 OE) or OCI-ly8 cells transfected with empty vector (OCI-ly8 EV) into the right flank of B-NDG mice. At day 10 after the injection of the DLBCL cells, when the tumours became palpable, half of the mice from both the PDK4 overexpressing group and empty vector group were treated with rituximab (12.5 mg/kg, equivalent to 225-275 μg/mice) injected intraperitoneal on a daily basis for 2 weeks, with PBS injected as the control for the rest half of mice. Calliper measurement was performed to determine the diameter of tumour, and the volume of tumour was calculated using the following formula: 0.5 × length × width2. Tumour volume and body weight were measured every 2 days for the mice. They were sacrificed when the volume of tumour reached 2000 mm3. Then, the tumors were excised and weighed.
Statistical analysis
Expressed as mean ± standard deviation (SD), all data are representative of at least three separate experiments. SPSS 19.0 Student’s t-test was conducted to perform comparison between two independent groups, and the corresponding bar chart or line chart was drawn using GraphPad Prism 7 software. To determine the differences between continuous variables, unpaired t test or the Mann-Whitney U test was carried out. Probability values < 0.05 were treated as statistically significant. The mice were randomly assigned to their groups using the random number table.