Cell lines
Raji and MCF7 cell lines were purchased from ATCC or the European Collection of Cell Cultures (Wiltshire, UK). The cells were cultured in RPMI-1640 medium (Sigma-Aldrich, St Louis, MO, USA) supplemented with 10% fetal bovine serum (FBS) (Sigma-Aldrich), 2 mM L-glutamine (Sigma-Aldrich) and 1% antibiotics – penicillin/streptomycin (Sigma-Aldrich) (referred as full RPMI medium) in a humidified atmosphere containing 5% CO2. All the cell lines were maintained through continuous passaging and were confirmed to be free of contamination with Mycoplasma spp. For luciferase-based assays cell lines (Raji and MCF7) were modified with plasmid pLenti7.3/V5 TOPO-RedLuc encoding the red luciferase gene and green fluorescent protein, as described previously [20].
Reagents
All tested compounds GPX4 inhibitors: RSL3 (Selleckchem, S8155) and ML162 (Merck, SML2561), iFSP1 (Selleckchem, S9663), liproxstatin-1 (Merck, SML1414), liproxstatin-1 for in vivo experiments (Chem-Norm, TBW04068), Z-VAD-FMK (Selleckchem, S7023), necrostatin-1 (Sigma, N9037) were dissolved in DMSO to obtain 10 mM stock solutions and kept at -20 or -80°C according to manufacturer’s recommendations. Further dilutions were performed in culture medium, directly before each experiment. Deferoxamine (Desferal, Novartis leftovers donated by patients) was dissolved in dH2O and was kept at -20°C.
T cell isolation and stimulation
Human primary T cells were isolated from buffy coats of healthy donors obtained from the Regional Blood Center in Warsaw, Poland, with the knowledge of the Bioethics Committee at the Central Clinical Hospital of the Ministry of Interior and Administration in Warsaw (approved on 12/10/2022). Initially, mononuclear cells were isolated by density gradient centrifugation using Lymphoprep (STEMCELL Technologies Canada, Inc.). Subsequently, T cells were magnetically separated from mononuclear cells with negative selection using EasySep™ Human T Cell Isolation Kit (STEMCELL Technologies Canada, Inc.). T cells were stimulated with the magnetic beads coated with monoclonal antibodies against CD3 and CD28 molecules (Dynabeads Human T-Activator CD3/CD28, Thermo Fisher Scientific) and were cultured in full RPMI medium supplemented with 100 U/mL of IL-2 (Peprotech). After 5 days the beads were removed and T cells were further cultured in the presence of IL-2 only.
T cell viability assays
Unstimulated T cells, stimulated T cells or CAR-T cells were treated with tested compounds for an indicated time, 24 or 48 h. Subsequently, the viability of the cells was assessed by propidium iodide (PI, 1 µg/mL, Sigma-Aldrich) staining, followed by flow cytometry analysis BD FACSCantoII and HTS sampler.
T cell proliferation assay
Human primary T cells were resuspended in PBS (1 × 106/ml) and stained with Cell Trace Violet (CTV) dye (Thermo Fisher Scientific) for 20 min at 37°C at a final CTV concentration of 2.5 µM. Subsequently, T cells were washed and seeded onto a 96-round-bottom plate (2 × 104 cells per well) in full RPMI medium in the presence of IL-2 (100 U/mL), Dynabeads Human T-Activator CD3/CD28 (beads to cell ratio 2:1) and increasing concentrations of iFSP1 or RSL3 with or without Lip-1. After 3 and 6 days of incubation, the T cells were stained with DRAQ7 viability stain (BioLegend) and analyzed on BD FACSCantoII and HTS sampler.
Flow cytometry staining
Extracellular staining for surface antigen was performed in a staining buffer (PBS supplemented with 1mM EDTA and 2% FBS) for 20 min at room temperature onto a 96-round-bottom plate. For intracellular staining, the cells were fixed with BD Cytofix buffer, permeabilized with BD Perm/Wash buffer and stained at 4°C for 30 min. Next, cells were washed in Perm/Wash buffer, resuspended in staining buffer and analyzed with BD FACSCantoII and HTS sampler. Antibodies used for staining are listed in Table 1.
Table 1
Flow cytometry antibodies
Antibody | Fluorochrome | Catalog number | Company |
CD71 (transferrin receptor) antibody | APC | 17-0719-42 | Invitrogen |
Ferroportin/SLC40A1 antibody | PE | NBP1-21502PE | Novus Biologicals |
CD340 (erbB2/HER-2) antibody | APC | 324408 | BioLegend |
goat anti-human IgG, Fcγ fragment specific antibody | AF647 | 109-606-098 | Jackson ImmunoResearch Labs |
Lipid peroxidation assay
Lipid peroxidation was evaluated in T cells either at a steady state or following RSL3 treatment using the fluorescent lipid peroxidation sensor named BODIPY 581/591 C11 (ThermoFisher). Briefly, T cells were seeded onto a 96-round-bottom plate at cell density 1 × 106/ml and incubated with increasing concentrations of RSL3 in the presence or absence of Lip-1 for 20 h. The next day, T cells were centrifuged and resuspended in full RPMI medium containing 0.5 µM BODIPY 581/591 C11 reagent (100 µl/well) and incubated at 37°C for 30 min. Subsequently, the cells were washed three times and analyzed on BD FACSCantoII and HTS sampler. The level of lipid ROS was assessed as an increase in oxidized C11-BODIPY (green fluorescence).
Labile iron pool detection
Intracellular Fe2+ level was evaluated with the fluorescent probe FerroOrange (Dojindo). The cells were seeded onto a 96-round-bottom plate at cell number 2 × 105/well, washed three times with PBS, and then resuspended in HBSS buffer. In experiments with deferoxamine pretreatment, the cells were preincubated with 1 mM or 2 mM deferoxamine and incubated for 30 min at 37°C. Subsequently, 2 times concentrated FerroOrange probe was added to the wells at a final concentration of 1 µM and incubated for 30 min at 37°C. After incubation, cells were analyzed without washing on a BD LSRFortessa X20 instrument (BD Biosciences) and PE channel.
Intracellular ROS detection
Intracellular ROS were determined with fluorescent probes CellROX Deep Red and CellROX Green (Thermo Fisher Scientific). Briefly, T cells were seeded onto a 96-round-bottom plate at the density of 5 × 105 cells/mL and incubated with the CellROX Deep Red or CellROX Green reagent at 37°C, 5% CO2 for 30 minutes. After washing, the cells were analyzed on a BD FACSCantoII flow cytometer (BD Biosciences).
GSH detection
To determine GSH level in T cells the Intracellular glutathione (GSH) Detection Assay Kit (ab112132) was applied. Briefly, T cells were seeded onto a 96-round-bottom plate at cell density 1 × 106/ml and incubated with Thiol Green fluorescent probe (diluted 1:10000) for 30 min at 37°C. After incubation, the cells were washed and analyzed using flow cytometry and green fluorescence.
CAR constructs and lentiviral T cell modification
In this study, we utilized two CD19 CAR (FMC63 clone) constructs, generously provided by M. Pule from UCL, UK. The first construct includes the CD8 hinge and transmembrane domain, the 41BB costimulatory domain, the CD3ζ signaling domain, and the rituximab recognized-RQR8 epitope for CAR detection. The second CD19 CAR construct consists of the IgG1 half-hinge, the CD28 transmembrane and co-stimulatory domain, and CD3ζ. PD-L1-targeting CAR consists of an atezolizumab-based scFv sequence following an IgG1 half-hinge, CD28 transmembrane region, CD28 costimulatory domain, and CD3ζ signaling domain. HER2 CAR construct consists of a trastuzumab-based scFv sequence following a CD8 hinge and transmembrane domain and a 4-1BB-CD3ζ signaling tail. All constructs were subcloned into the lentiviral pSEW plasmid. T cells were modified with the CAR constructs using a lentiviral transduction system as described previously [21]. The CAR expression on the surface of the T cells was evaluated by flow cytometry 48–72 h after transduction as described in [21].
RTCA-based killing assay
The HER2 CAR-mediated killing of T cells was monitored with a real-time cell analysis (RTCA) assay. Adherent target MCF7 cells (3 × 104 cells/well) were seeded onto 16-well E-Plate (ACEA Biosciences) in 150 µl of a full RPMI medium. The proliferation of MCF7 cells was monitored in the incubator at 37°C (5% CO2, 95% humidity) for 24 h with the xCELLigence impedance-based RTCA system (ACEA Biosciences). The next day, 100 µl of the medium was aspirated and replaced with the full RPMI medium containing effector cells (control unmodified T cells or HER2 CAR-T cells) at effector to target ratio E:T 2:1. T cells and CAR-T cells were pretreated with increasing concentrations of RSL3 for 5 h and transferred onto target cells without washing out the RSL3-containing medium. The CAR-mediated killing of target cells was monitored for the next 12 h. Analysis was performed using RTCA Software Pro (ACEA Biosciences). The impedance changes (cell index) were normalized to the end value of the target cells' proliferation and plotted over time as normalized cell index.
Luciferase-based cytotoxicity assay
Cell lines previously modified to express the luciferase reporter gene (Red-Luc), were seeded onto the 96-well black plates with a clear bottom (Perkin Elmer) at a cell density of 3 × 104 per well in 100 µl of full RPMI in three or four technical replicates. MCF-7 cells were allowed to adhere for 24 h while suspension Raji cells were directly used in the experimental procedures. For cytotoxicity assays, increasing concentrations of RSL3 or iFSP1 were added to the wells and the cells were incubated for 48 h. For luciferase-based killing assays, effector CAR-T cells and control unmodified T cells were added to the wells at different E:T ratios and were cocultured for the next 18 h. For bioluminescence readout Bright-Glo™ Luciferase Assay System (E2610, Promega) was used. The plate was incubated for 5 min in darkness at room temperature and luminescence was measured using Tecan INFINITE M1000 (TECAN).
Flow cytometry-based killing assay
PD-L1 CAR-T cells were pretreated with or without 20 µM iFSP1 for 48 h. For cytotoxicity assay, a target (Raji PD-L1) cells were stained with Cell Trace Violet (CTV) and seeded onto the 96-well plate at a cell density of 1 × 105 per well in 100 µl of full RPMI in two technical replicates. Next, PD-L1 CAR-T cells were added for 24 h at E:T ratios 0.25:1, 0.5:1 and 1:1. Propidium iodide (PI) was used to discriminate live/dead cells. Cytotoxicity of effector cells was evaluated as an increase in a percentage of violet-CTV positive, PI positive target cell population.
Degranulation and cytokine production assay
Before degranulation and cytokine production assay CAR-T cells were preincubated with either RSL3 for 16 h or iFSP1 for 24 h. The next day, tumor cells expressing recognized antigen on the surface were added to appropriate wells at E:T (effector to target) ratio 0.25:1 and 1:1. CD19 CAR-T cells were incubated with Burkitt’s lymphoma cell line Raji (CD19+), HER2 CAR-T cells with breast cancer cell line MCF7 (low HER2+) and PD-L1 CAR-T cells with Raji cells, genetically modified to overexpress PD-L1 molecule [21]. Subsequently, Golgi Stop (BD Biosciences, dilution 1:250), Golgi Plug (BD Biosciences, dilution 1:200) and anti-CD107a-PE antibody (BD Biosciences, dilution 1:40) were added and the assay plate was incubated for 4 h at 37°C and 5% CO2. After incubation, the cells were stained with anti-CD3-BV421 antibody and Fixable viability stain 510 (BD Biosciences, dilution 1:200) followed by fixation and permeabilization procedures. Eventually, the cells were stained for cytokines with anti-IFNy-APC (BD Biosciences, dilution 1:100) and anti-TNFα-PECy7 (BD Biosciences, dilution 1:100) antibodies. Degranulation and cytokine production by effector cells was assessed using flow cytometry.
Seahorse analysis
Cell metabolism was measured using Seahorse XF HS Mini Analyzer (Agilent). Seahorse analysis was performed on T cells and CAR-T cells pretreated with iFSP1. Stimulated T cells or CAR T cells were seeded onto a 24-well plate with 20 µM of iFSP1 and incubated for 48 h. iFSP pretreated T/CAR T cells were subsequently resuspended in Agilent Seahorse XF RPMI medium, pH 7.4 supplemented with Agilent Seahorse XF glucose (10 mM) and glutamine (2 mM) solutions and seeded at 2 × 105 cells/well onto Agilent Seahorse XFp PDL Cell Culture Miniplates. Metabolic parameters were measured under basal conditions upon treatment with oligomycin A (1.5 µM), BAM15 (2.5 µM) and rotenone/antimycin A (0.5 µM each) (Seahorse XF T Cell Metabolic Profiling Kit, Agilent). All steps were performed following the manufacturer’s recommendations.
Western Blotting
For Western blotting, cells were lysed with RIPA lysis buffer (Tris-HCL pH 7.4, NaCl 150 mM, NP-40 1% (v/v), sodium deoxycholate 1% (v/v), SDS 0.1% (v/v)) supplemented with Complete Protease Inhibitor Cocktail and Phosphatase Inhibitor Cocktail (Roche Diagnostics). Protein concentration was measured using the Pierce™ Rapid Gold BCA Protein Assay Kit (Thermo Fisher Scientific) according to the manufacturer's instructions with minor modifications on the TECAN Infinite M1000 Pro microplate reader. 20 µg of cell lysates were separated in 10, 12 or 15% (v/v) (depending on the molecular weight of detected protein) reducing SDS-polyacrylamide gel, then transferred onto nitrocellulose membranes and blocked with either 5% (w/v) nonfat milk or 5% Bovine Serum Albumin (w/v) (Kenilworth) in TBST (Tris-buffered saline, pH 7.4 and 0.05% (v/v) Tween-20) and then incubated with the following primary antibodies: anti-GPX4 (Cell Signaling cat. 52455S, dilution 1:1000), anti-FSP1 (Abcam cat. ab302673, dilution 1:1000), anti-ACSL4 (Santa Cruz cat. sc-271800, dilution 1:1000), anti-LOX15 (Abclonal cat. A6864, dilution 1:1000), anti-LC3 (Cell Signaling cat. 4108, dilution 1:1000), anti-perforin (MABTECH cat. Pf-344, dilution 1:1000), and anti-β-actin-HRP (A222, Sigma-Aldrich; dilution 1:50,000). For detection of primary protein bands HRP-conjugated secondary antibodies were used. The blots were exposed to the Super Signal chemiluminescent substrates (Thermo Fisher Scientific). The signal was detected using the ChemiDoc Imaging System (Bio-Rad ChemiDoc MP Imaging System).
Animal studies
All in vivo experiments were performed with 8–12-week old male NSG (NOD.Cg-PrkdcscidIl2rgtm1Wjl/SzJ) mice obtained from Charles River Laboratories, which were bred at the Animal Facility of the Mossakowski Medical Research Institute, Polish Academy of Sciences. All experiments were performed in accordance with the guidelines and approved by The Second Local Ethics Committee for the Animal Experimentation, Warsaw University of Life Sciences (number: WAW2/100/2023, WAW2/027/2024). The experiments were carried out in an SPF animal facility with IVC systems. To avoid confounders, all mice were labeled and kept in tagged cages. The cages had an assigned, unchanging place in the rack. Results obtained from individual mice according to the treatment method are presented. The blinding was not applied. The distribution of mice to the experimental groups was random and no animals were excluded during the experiment. The sample size was determined based on the assumed increase in tumor diameter. The experimental group size was calculated by power analysis (for assumed test power 80%) or resource equation approach as described in [22].
In vivo experiment
Mice were inoculated subcutaneously with 2 × 106 Raji cells in 50% Matrigel Growth Factor Reduced (Corning) on day 0 of the experiment. Subsequently, on days 4, 7, 10, and 13, 5 × 106 CD19 CAR-T cells were administrated intravenously. Liproxstatin-1 (Chem-Norm) was injected intraperitoneally with 10 mg/kg every day or every other day for 2 consecutive weeks. Control mice received medium or solvent respectively. Tumor growth was monitored three times per week with caliper starting from day 7 of the experiment. Tumor volume was calculated according to the formula volume (mm3)= (width2 [mm] × length [mm])/2. Mice were sacrificed when the tumor diameter reached 15 mm in at least one dimension. Total number of mice used within this study was 37.
RNAseq and bioinformatics analysis
RNAseq analysis was done using the dataset - GSE 59846, which comprises expression data for two cell types - CD4+ naïve and CD4+ memory T-cells, each derived from 3 individuals. Each cell type was additionally stimulated for 48 h using beads coated with monoclonal antibodies against the CD3 and CD28. Thus, 12 RNA-seq experiments were carried out; however, due to possible mislabeling of two runs - SRR1531315 and SRR1531316, we excluded them from the downstream analysis. Initially, we assessed library quality with FastQC [23] (v. 0.11.9). Next, trimmomatic [24] (v. 0.39) was used to trim fragments of reads from the 3’ and 5’ ends if their average quality fall below 30. Additionally, reads shorter than 40 bp were excluded at this stage. Processed reads were mapped to the human genome (GRCh38) using the align function available from the Rsubread package [25] (v. 2.12.3). Samtools [26] (v. 1.10) was used to remove unmapped reads. The number of reads associated with human genes, as defined in the Gencode GTF annotation file V40, was calculated using the featureCounts function from the Rsubread package and converted to counts per million (in log2 scale). The differential gene expression analysis was carried out using the limma program [27] (v. 3.54.2) for samples divided into two groups ‘stimulated’ or ‘unstimulated’ (Table 2). P-values were adjusted for multiple tests with the Benjamini–Hochberg procedure. All analyses were carried out in R (v. 4.2.2). Results of the statical test are shown as * for p-value ≤ 0.1, ** p-value ≤ 0.05; ***, p-value < 0.01.
Table 2
Sample labelling in RNAseq reanalysis
Sample ID | Source name | Activation | Donor | Final label |
SRR1531325 | Memory CD4 T cell | 48 h | 5134 | stimulated |
SRR1531324 | Memory CD4 T cell | 48 h | 5009 | stimulated |
SRR1531323 | Memory CD4 T cell | 0 h | 5134 | unstimulated |
SRR1531322 | Memory CD4 T cell | 0 h | 5009 | unstimulated |
SRR1531321 | Naive CD4 T cell | 48 h | 5134 | stimulated |
SRR1531320 | Naive CD4 T cell | 48 h | 5009 | stimulated |
SRR1531319 | Naive CD4 T cell | 0 h | 5134 | unstimulated |
SRR1531318 | Naive CD4 T cell | 0 h | 5009 | unstimulated |
SRR1531317 | Memory CD4 T cell | 48 h | 5053 | stimulated |
SRR1531314 | Naive CD4 T cell | 0 h | 5053 | unstimulated |
Statistical analysis
Statistical analysis was performed with GraphPad Prism 9 (GraphPad Software). To determine data distribution, the Shapiro-Wilk normality test was performed. If data passed the normality test, parametric statistics were used. In all analyses, the tests were two-tailed. For comparison between 2 groups either unpaired or paired t-test was performed, depending on data sets. For the differences between three or more independent groups one-way ANOVA was applied, followed by multiple comparisons tests. For comparison differences between groups with two independent variables, twoway ANOVA with post hoc analysis was performed. All statistically significant differences (p-value < 0.05) and their p-values were marked on the graphs. Non-significant differences with p-value ≥ 0.05 were marked as ns. Data are represented as means with standard deviation. Each dot on the graphs represents the average of 2 technical replicates for a particular donor.