Abstract Although oxidative stress is closely associated with tumor invasion and metastasis, its’ exact role and mechanism in the initial stage of oral cancer remain ambiguous. Glutamine uptake mediated by alanine-serine-cysteine transporter 2 (ASCT2) participates in glutathione synthesis to resolve oxidative stress. Currently, we firstly found that ASCT2 deletion caused oxidative stress in oral mucosa and promoted oral carcinogenesis induced by 4-Nitroquinoline-1-oxide (4-NQO) using transgenic mice of ASCT2 knockout in oral epithelium. Subsequently, we identified an upregulated gene Thbs1 linked to macrophage infiltration by mRNA sequencing and immunohistochemistry. Importantly, multiplex immunohistochemistry showed M1-like tumor-associated macrophages (TAMs) were enriched in cancerous area. Mechanically, targeted ASCT2 effectively curbed glutamine uptake and caused intracellular reactive oxygen species (ROS) accumulation, which upregulated Thbs1 in oral keratinocytes and then activated p38, Akt and SAPK/JNK signaling to polarize M1-like TAMs via exosome-transferred pathway. Moreover, we demonstrated M1-like TAMs promoted malignant progression of oral squamous cell carcinoma (OSCC) both in vitro and in vivo by a DOK transformed cell line induced by 4-NQO. All these results establish that oxidative stress triggered by ASCT2 deletion promotes oral carcinogenesis through Thbs1-mediated M1 polarization, and indicate that restore redox homeostasis is a new approach to prevent malignant progression of oral potentially malignant disorders. Keywords: ROS, ASCT2, Thbs1, Macrophages, Oral carcinogenesis Graphical abstract [37]Image 1 [38]Open in a new tab Highlights * • ASCT2 knockout induced oxidative stress in oral epithelium and promoted oral carcinogenesis. * • ASCT2 knockout simultaneously promoted high expression of Thbs1 and enrichment of M1-like TAMs in the cancerous sites. * • Oxidative stress caused by ASCT2 knockdown promoted the polarization of M1-like TAMs through exosome-transferred Thbs1. * • M1-like TAMs accelerated oral carcinogenesis by enhancing cell proliferation, stemness, migration and invasion. 1. Introduction Oxidative stress is defined as deregulation of reactive oxygen species (ROS) production and ROS limitation, characterized by unspecific oxidation of biomolecules and alteration of normal physiological function. Aberrant redox homeostasis plays important roles in regulating diverse aspects of cell behavior (e.g., proliferation, differentiation, migration and angiogenesis) [[39]1]. It has been proven that oxidative stress is closely associated with tumor progression. On the one hand, oxidative stress triggers tumor development by causing DNA damage and genomic instability [[40]2,[41]3]; on the other hand, the rapid growth of tumor enhances ROS production [[42]4,[43]5]. However, it remains obscure on the role and mechanism of oxidative stress in the initiation of tumor carcinogenesis. Glutamine (Gln) is the most abundant and widely used non-essential amino acid in the human body, which involves in tricarboxylic acid (TCA) cycle and inter-organ nitrogen exchange [[44]6]. In addition, Gln can balance redox homeostasis by participating in de novo biosynthesis of glutathione (GSH) to cope with oxidative stress [[45][7], [46][8], [47][9]]. Alanine-serine-cysteine transporter 2 (ASCT2, encoded by gene Slc1a5) has been acknowledged as the major Gln transporter [[48]10,[49]11], although there have been numerous membrane transporters generally recognized as four Solute Carrier (SLC) families (SLC1, SLC6, SLC7 and SLC38) [[50]12,[51]13]. ASCT2 is an obligatory Na^+-dependent amino acid transporter which mediates the cotransport of Na^+ with Gln or other neutral amino acids (alanine, serine and cysteine) to support the biosynthetic pathways [[52]12,[53]14]. There have been numerous studies showed that targeted interference with ASCT2-mediated Gln uptake significantly suppressed Gln metabolism, especially GSH production, which contributed to redox imbalance and induced cellular oxidative stress [[54][15], [55][16], [56][17], [57][18]]. Oral leukoplakia (OLK) is one of the most common oral potentially malignant disorders (OPMDs) in oral mucosa and has the potential to develop into oral squamous cell carcinoma (OSCC) which affects nearly 377713 people worldwide in 2020 [[58][19], [59][20], [60][21]]. Although the etiology and anatomical locations are varied, a hallmark of OSCC is redox imbalance characterized by decreased antioxidant and increased oxidant [[61]22,[62]23]. 4-Nitroquinoline-1-oxide (4-NQO) is a synthetic quinolone-derived molecule which is used to mimic the carcinogenic effect of tobacco [[63]21,[64]24]. It can induce intracellular oxidative stress, leading to the production of ROS metabolic substances that bind to DNA and generating DNA adducts [[65]24,[66]25]. So far, the 4-NQO-induced mouse model remains the most ideal model that accurately reflects the process of oral carcinogenesis. In current study, we firstly induced oxidative stress in oral mucosa by utilizing ASCT2 conditional knockout mice, and subsequently found that oxidative stress promoted oral mucosa carcinogenesis. Mechanically, it was associated with the polarization of M1-like tumor-associated macrophages (TAMs) activated by exosome-transferred Thbs1. Altogether, the results established restoring redox homeostasis as a new approach targeting ROS-Thbs1-M1 pathway to prevent oral carcinogenesis. 2. Materials and method 2.1. Transformation of human dysplastic oral keratinocyte cells by 4-NQO and RNA transfection The human dysplastic oral keratinocytes (DOK) were obtained from Dr. Juan Xia (Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University). DOK cells were cultured in high-glucose DMEM (Gibco, NY, USA) supplemented with 10 % fetal bovine serum (FBS, Gibco, NY, USA) and 5 μg/mL hydrocortisone (Sigma-Aldrich, St. Louis, MO, USA). DOK cells were transformed by 4-NQO (Sigma-Aldrich, St. Louis, MO, USA) in our laboratory. Cultures were exposed to 2.6 μM 4-NQO for 3 h as described previously [[67]26]. After passed 30 generations of intermittently 4-NQO-treatment over 2 months, the DOK-4-NQO cells were expanded for tumorgenicity in vitro and in vivo. To generate stable silenced DOK cells, shRNA lentiviral vectors targeting ASCT2 were purchased from Genechem Co., Ltd (Shanghai, China) and transfected into DOK cells with corresponding control vectors according to the manufacturer's protocols. After transfection, DOK cells were incubated in conditioned medium (CM) with 2 μg/mL puromycin (Aladdin Co., Ltd, Shanghai, China) to select the stably transfected cells. The sequences of the shRNAs were presented in [68]Table S1 (Supporting information). To determine the function of Thbs1 in ASCT2 knockdown DOK cells, small interfering RNA (siRNA) oligonucleotide (Hanbio Co., Ltd, Shanghai, China) was used. RNAFit Transfection Reagent (Hanbio Co., Ltd, Shanghai, China) was used to facilitate the transfection of siRNA oligonucleotides into the targeted cells in accordance with manufacturer's instruction. The Thbs1 siRNA sequences were listed in [69]Table S1 (Supporting information). 2.2. Human macrophage differentiation in vitro The human monocyte cells THP-1 were provided as a gift from Dr. Zhengmei Lin (Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University). THP-1 cells were maintained in RPMI 1640 medium (Gibco, NY, USA) with 10 % FBS and 5 mM β-mercaptoethanol (Macklin Co., Ltd, Shanghai, China). M0 macrophages were differentiated from THP-1 cells using 100 ng/mL phorbol 12-myristate 13-acetate (PMA, Sigma-Aldrich, St. Louis, MO, USA) for 48 h. Then, the THP-1-derived macrophages were further differentiated into M1-like cells cultured in medium with 20 ng/mL IFN-γ (PeproTech Inc., NJ, USA) and 100 ng/mL LPS (Absin Bioscience Inc., Shanghai, China), or differentiated into M2-like cells cultured in medium with 20 ng/mL IL-4 (PeproTech Inc., NJ, USA) and 20 ng/mL IL-13 (PeproTech Inc., NJ, USA) for 48 h. To obtain conditioned medium (CM) of M0 or M1 macrophages, cells, about 70%–80 % confluence were washed and cultured for additional 24 h with fresh RPMI 1640 medium with 10 % FBS and 5 mM β-mercaptoethanol. Then the supernatants were centrifuged at 3000 g (4 °C) for 15 min and finally were filtrated through 0.22 μm Filter Units (Merk Millipore, MA, USA). 2.3. Animal models Slc1a5^tm1.1cyagen mice (CKOCMP’-02923-Slc1a5) were purchased from Cyagen Biosciences Inc (SF, USA). K14-CreER^tam mice (JAX-005107) were kindly given as a gift by Dr. Juan Xia (Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University), which expressed Cre recombinase in squamous epithelium under the control of the K14 promoter. Slc1a5^f/f Cre ^± mice were generated by crossing the floxed Slc1a5 allele Slc1a5^tm1.1cyagen (Slc1a5^f/f) mice with K14-CreER^tam (K14-Cre^+/−) mice. Genotype of transgenic mice were identified by the One Step Geno Typing Kit (Vazyme Biotech Co, Nanjing, China) following the manufacturer's protocol. For acquiring Slc1a5 conditional knockout mice, Slc1a5^f/f Cre ^± mice were intraperitoneally injected with tamoxifen (TAM, Sigma Aldrich, St. Louis, MO, USA) five times every other day at a dose of 120 mg/kg. The PCR primers of genotyping were provided in [70]Table S2 (Supporting information). For mouse model of 4-NQO-induced oral carcinogenesis, 4-NQO was dissolved in propylene glycol (Macklin, Shanghai, China) to prepare stock solution (1 mg/mL) and further diluted to working solution (100 μg/mL) in drinking water every week. For inducing oral carcinogenesis, mice with the same gene background (Slc1a5^f/f Cre^+/−) were fed with working solution for 16 weeks and maintained with regular drinking water for a further 6 weeks. All animal experiments were approved by the Laboratory Animals Ethics Committee of Sun Yat-sen University (SYSU-IACUC-2019-000228). For xenograft model, male BALB/c nude mice (nu/nu, aged 3–5weeks) were purchased from the Laboratory Animal Center of Sun Yat-sen University (Guangzhou, China). In brief, 1*10^6 DOK or DOK-4-NQO cells/per site were subcutaneously injected under the skin of the axillary to establish the nude mouse xenograft tumor models (n ≥ 6 for each group). For assessing the effects of CM of macrophages on the tumorigenesis of DOK-4-NQO cells, mice with xenografts were intraperitoneally injected with 500 μL CM from M1-like TAMs, M0 or RPMI 1640 every other day. Tumor volumes (length * width^2/2) were monitored and compared. The above animal experiments were approved by the Laboratory Animals Ethics Committee of Sun Yat-sen University (SYSU-IACUC-2023-001462, SYSU-IACUC-2024-000773). 2.4. Histological hematoxylin-eosin (HE) saining and immunohistochemistry (IHC) For tissue HE staining, tissues were dissected and fixed overnight with 4 % paraformaldehyde (Biosharp, Chongqing, China). Samples were then embedded in paraffin. For the HE staining of cells, cells were just fixed with 4 % paraformaldehyde for 15 min. Paraffin sections with 4 mm thick or cells were prepared and stained with hematoxylin and eosin (Servicebio Co, Ltd, Wuhan, China) for histology. IHC was carried out in accordance with the manufacturer's guidelines of the IHC Kit (Gene Tech, Shanghai, China) and quantified as our previous published methods [[71]17,[72]27]. The antibodies used in IHCassays were listed in [73]Table S3 (Supporting information). 2.5. Western blot The protocol was conducted according to the previous studies [[74]17,[75]28]. The primary antibodies used were listed in [76]Table S3 (Supporting information). The secondary antibodies were anti-rabbit or anti-mouse HRP-conjugated antibodies (1:5000, EMAR, Beijing, China). The bands were visualized by chemiluminescent HRP substrate (Millipore, MA, USA). 2.6. Tissue immunofluorescence (IF) staining and multiplex IHC (mIHC) Tissue IF was conducted as previously described [[77]27]. In short, sections were first incubated with primary antibodies against ASCT2, 8-Hydroxyl-2-deoxyguanosine (8-OHdG) or γ-H2AX which was from DNA Damage Assay Kit (C2035S, Beyotime, Shanghai, China). Then these sections were incubated with corresponding fluorochrome-conjugated secondary antibodies for 1 h. Sections were then counterstained with DAPI (Solarbio, Beijing, China) and evaluated in an invert laser confocal microscope (Olympus FV3000, TKY, Japan). mIHC was performed using a Neon DendronFluor Kit (Histova Biotechnology Co, Ltd, Jilin, China) as previously described [[78]27]. The antibodies used in these sections were listed in [79]Table S3 (Supporting information). 2.7. Assessment of oxidative stress in tissue and cells Visualization of ROS in tongue epithelium samples was conducted by staining for oxidation of cytosolic dihydroethidium (DHE) (Applygen, Beijing, China) as described previously [[80]29]. In brief, 10 μm tongue cryosections were air-dried for 30 min and then incubated with 0.5 μM DHE for 30 min at 37 °C. Then the sections were incubated with primary antibody against ASCT2 overnight at 4 °C, which were followed by incubation with fluorochrome-conjugated secondary antibody Alexa Flour488 for 1 h at room temperature. Sections were next counterstained with DAPI and evaluated in Olympus FV3000. The production of H[2]O[2] in tongue mucosa epithelium was assessed by H[2]O[2] Content Assay Kit (Solarbio, Beijing, China) according to the manufacture's guideline. GSH/GSSG ratio in tongue mucosa epithelium was determined by using GSH and GSSG Assay Kit (Beyotime, Shanghai, China) as the instruction described. For assessing ROS level in cells, intracellular ROS were detected using the DHE Kit (Applygen, Beijing, China) by flow cytometry as we previously described [[81]17,[82]27]. 2.8. RNA sequencing analysis For mouse model of 4-NQO-induced oral carcinogenesis, mouse tongues were treated with dispase Ⅱ enzyme (Roche, Basel, Switzerland) to obtain tongue mucosa epithelium for RNA-Seq analysis, which was conducted at OE biotech Co., Ltd. (Shanghai, China). The libraries were sequenced on an Illumina HiSeq X Ten system and 125/150bp paired-end reads were generated. Raw data (raw reads) of the fastq format were first processed using Trimmomatic. Then about 40–55 million clean reads for each sample were acquired by abolishing reads containing adapter or ploy-N and low-quality reads. The clean reads were next mapped to the human genome using HISAT2. The fragments per kb of transcript per million reads (FPKMs) value of each gene were calculated using Cufflinks, and the read counts of each gene were obtained by HTSeq-count. Differentially expressed genes (DEGs) analysis was performed using the DESeq (2012) R package. A P value of <0.05 and a fold change of >2 or <0.5 were set as the threshold for significantly differential expression. Hierarchical cluster analysis of differentially expressed genes was conducted to analyze transcript expression patterns. Gene ontology enrichment (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis of DEGs were performed using R based on the hypergeometric distribution. 2.9. Bioinformatics analysis Bioinformatics analysis was conducted on the basis of the TCGA cohort of head and neck squamous cell carcinoma (HNSCC). Gene expression data of corresponding number pairs of adjacent and cancer samples was extracted to analyze the Thbs1 expression in HNSCC samples. The survival data was analyzed using the “survival” (v3.3.1) R package. K-M survival curve analysis was used to identify the survival outcomes of HNSCC patients based on the Thbs1 expression levels. The ssGSEA algorithm in the “GSVA” (v1.46.0) R package [[83]30] was used to assess the tumor infiltration status of 24 immune cell types [[84]31]. Besides, we used the spearman's correlation analysis to evaluate the relationship between Thbs1 expression level and the immune cell infiltration status. 2.10. Flow cytometry For cell analysis, Zombie NIR™ Fixable Viability Kit (Biolegend, CA, USA, diluted at 1:500) was used to label dead cells at room temperature for 30 min firstly. Then, cells were incubated with specific antibodies for 30 min on ice. Data was acquired on LSRFortessa (BD Bioscience, NJ, USA) and was analyzed using Flowjo software (BD Bioscience, NJ, USA). The antibodies were listed in [85]Table S3 (Supporting information). 2.11. RNA extraction and quantitative real-time PCR (qPCR) Total RNA was isolated from cells with RNA-Quick Purification kit (ES Science, Shanghai, China). RNA concentration was measured by NanoDrop One (Thermo Fisher Scientific Inc., CA, USA). HiScript Ⅲ RT SuperMix for qPCR kit (Vazyme Biotech Co., Nanjing, China) was used to reversed-transcribe 1 μg of RNA to acquire cDNA. Quantitative RT-PCR was conducted to quantify gene expression level of β-actin, ASCT2 and Thbs1 by applying ChamQ Universal SYBR qPCR Master Mix (Vazyme Biotech Co., Nanjing, China) and quantified by 2^−ΔΔCT method. The primers used in this study, purchased from GeneRay biotechnology (Guangzhou, China) were listed in [86]Table S1 (Supporting information). 2.12. Exosome isolation, characterization and tracing Exosomes were isolated from the cell culture supernatants by ultracentrifugation. In brief, equal amounts of DOK cells treated differently were seeded on 15-cm dishes and cultured in high glucose DMEM medium with 10 % exosome-free FBS for 48 h. These supernatants were collected and centrifuged at 3000 g for 30 min at 4 °C to abolish dead cells and cell fragments, and then filtered through a 0.22 μm filter (Merk Millipore, MA, USA). Then the solution was ultracentrifuged at 110000 g for 2 h at 4 °C. The precipitate was collected and washed using sterilized PBS (Solarbio, Beijing, China), followed by next ultracentrifugation at 110000 g for 2 h at 4 °C. The pellets of exosome were resuspended with 200 μL PBS. The morphology and size of exosomes were determined by transmission electron microscopy (TEM, Hitachi, Tokyo, Japan). The size distribution and concentration were examined by nano flow cytometry (nanoFCM, Agilent, CA, USA). The expression of exosome markers (CD63, CD81, TSG101 and Histone 3) was analyzed by WB. Exosome-tracing experiments were conducted by EvLINK 505 Exosome-labeling Kit (Tingo Regenerative Medicine, Tianjin, China) according to the manufacturer's instruction. Exosomes labeled with fluorescent were incubated with THP-1 cells for 24 h. Then, cells were fixed in 4 % paraformaldehyde and permeabilized in 0.25 % Triton X-100 (Solarbio, Beijing, China) for 30 min respectively. Cells were next incubated with primary antibody against β-Tubulin overnight at 4 °C, followed by incubated with Alex Fluor 594 fluorescent-conjugated secondary antibody for 1 h and counterstained with DAPI. The cells were finally observed by Olympus FV3000. The antibodies were listed in [87]Table S3 (Supporting information). Exosomes were incubated with recipient cells at a concentration of 50 μg exosome protein/10^5 cells in following experiments. 2.13. Glutamine uptake assay ^3H-glutamine (Perikin Elmer, MA, USA, NET55120UC) was used to examine glutamine uptake as we previously described [[88]16]. 2.14. Metabolite determination Whole cell lysates of DOK cells were prepared for determined a-ketoglutarate (α-KG), citrate, malate, fumarate and succinate levels using the following ELISA kits: Human a-KG ELISA Kit (Jianglaibio, Co., Ltd, Shanghai, China), Human Citrate ELISA Kit (Jianglaibio, Co., Ltd, Shanghai, China), Human Malate ELISA Kit (Jianglaibio, Co., Ltd, Shanghai, China), Human Fumarate ELISA Kit (Jianglaibio, Co., Ltd, Shanghai, China), and Human Succinate ELISA Kit (Jianglaibio, Co., Ltd, Shanghai, China). The optical density (OD) at 450 nm was examined by a microplate reader and the concentrations were calculated according to respective standard curves. Cellular glutamate, GSH and ATP levels were identified by using Glutamate Assay Kit (Jiancheng Engineering Research Institute, Nanjing, China), Reduced Glutathione Assay Kit (Solarbio, Beijing, China) and ATP Assay Kit (Beyotime, Shanghai, China) respectively according to the manufacturer's instructions. 2.15. Papanicolaou staining assay and cytoskeleton staining assay The morphology of cells was observed by using Papanicolaou Staining Kit (LBP Medicine Science and Technology Co., Ltd) in accordance with the manufacturer's guideline. For cytoskeleton staining assay, Cells with different treatment were fixed in 4 % paraformaldehyde for 15 min and then permeabilized in 0.25 % Triton X-100 for 30 min at room temperature. The cells were subsequently stained with Actin-Tracker Green-555 (Beyotime, Shanghai, China) for 30 min and counterstained with DAPI. Olympus FV3000 was used for observing. 2.16. Cell cycle assay and cell proliferation assay For cell cycle assay, DOK cells were seeded on 6-well plates at a density of 2*10^5 per well. Cell Cycle Staining Kit (Multi Sciences, Hangzhou, China) was applied to examine the DNA content of cells in different stages of cell cycle in accordance with the manufacture's instruction. For cell proliferation assay, DOK cells were plated on a 96-well plate at a density of 1*10^3 cell per well and cultured in the corresponding conditions. The cell counting kit-8 solution (CCK-8, Dojindo, Kumanmoto, Japan) was added into wells and incubated for 1 h in a 37 °C incubator in accord with the instruction. the optical density (OD) value was measured at 450 nm and 600 nm. 2.17. Colony formation assay and tumor sphere formation assay For colony formation assay, 1000 cells/well were seeded in 6-well plates and cultured under the indicated conditions. After incubated for 7 days, cells were fixed with 4 % paraformaldehyde for 15 min, and stained with the crystal violet staining solution (Beyotime, Shanghai, China) for 15 min. The number of colonies (>50 cells) were counted using Image J analysis software. For tumor sphere formation assay, the cells were cultured in ultra-low attachment 96-well plates (Corning Inc., NY, USA) at a density 1000 cells/well in serum-free tumor stem cell medium (TSCM, QIDABIO, Shanghai, China) in a humidified 5 % CO2 incubator at 37 °C. Fresh medium was replenished every 3 days. The number of the valid spheres (>75 μm) was assessed under microscopy 1 week later. 2.18. Cell migration and invasion assay The in vitro cell migration and invasion were measured with the Corning Biocoat Matrigel Invasion Chamber (Corning Inc., NY, USA) according to the manufacturer's instruction. Briefly, 1*10^5 cells resuspended in 200 μL of serum-free medium were seeded in the upper chambers, and 500 μL culture medium with 20 % FBS was added into the lower chambers. After incubated with 24 h, cells of the reverse side of inserts were stained with the crystal violet staining solution and quantified with Image J analysis software. 2.19. Statistical analysis The results were presented as the mean ± SD from replicated experiments. Two-tailed Student's t-test or one-way ANOVA was performed to compare the means of different groups based on the raw data when the data conformed to normal distribution or homogeneity of variance. P-values <0.05 were considered statistically significant with Prism 9 (GraphPad Software, San Diego, CA, USA), and the levels of significance were further defined at level of *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001. 3. Results 3.1. ASCT2 conditional knockout triggered oxidative stress in oral mucosa It has been reported in vitro that intracellular Gln is mainly converted to glutamate which participates in the synthesis of GSH to protect cells from oxidative stress [[89][15], [90][16], [91][17],[92]32,[93]33]. However, it remains unknown whether in vivo targeted interference with ASCT2-mediated Gln uptake causes redox homeostasis imbalance. We therefore selectively deleted ASCT2 specially in oral mucosa epithelium, as briefly described in Methods ([94]Fig. S1a and b, Supporting information). As shown in [95]Fig. S1c (Supporting information),the tissue-specific gene deletion band (470 bq) was confirmed by the genotyping. The expression of ASCT2 gradually decreased and stabilized 2 weeks after intraperitoneal injection of tamoxifen according to Western blot and IHC experiments ([96]Fig. 1a and b). Fig. 1. [97]Fig. 1 [98]Open in a new tab The identification of oxidative stress in oral mucosa epithelium of ASCT2 knockout mice Western blot (a) and IHC (b) were used to detect the efficiency of ASCT2 knockout in day 1,3,7,14,21 after tamoxifen injection (120 mg/kg). ([99]Fig. 1–b, magnification × 100 for the left, magnification × 100 for the right, scale bars, 400 μm for 100* magnification and 100 μm for 400* magnification. the red box showed the area of interest). (c) The GSH/GSSG ratio was measured in the 3 groups. (d) H[2]O[2] levels were evaluated in oral epithelium using the H[2]O[2]Content Assay Kit. (e) ROS levels in oral epithelium of the same gene background (Slc1a5^f/f Cre^+/−) were assessed by DHE staining in cryosections. The markers were ASCT2 (green), ROS (red) and nucleus (blue) respectively. Left: Representative captures from 3 mice for each group. (Scale bar 100 μm). Right: Quantification of ASCT2 and DHE staining using image J. (f) Staining of γ-H2AX of the 3 groups in paraffin sections. The markers were ASCT2 (green), γ-H2AX (red) and nucleus (blue) respectively. Left: Representative captures for each group (Scale bar 100 μm). Right: Quantification of ASCT2 and γ-H2AX staining using image J. (g) Staining of 8-OHdG of the 3 groups in paraffin sections. The markers were ASCT2 (green), 8-OHdG (red) and nucleus (blue) respectively. The above mice used to detect oxidative stress were all male. (For interpretation of the references to colour