Abstract Arsenic (As^3+) is a well-established environmental carcinogen known to induce malignant transformation and cancer stem-like cell (CSC) properties in somatic cells, with Nrf2 functioning as a central regulator. However, the impact of chronic As^3+ exposure on pluripotent stem cells, particularly through Nrf2-mediated epigenetic and metabolic reprogramming, remains largely unexplored. In this study, we chronically exposed human induced pluripotent stem cells (iPSCs, Nips-B2) to an environmentally relevant concentration of trivalent arsenic (0.25 μM, As^3+) for three months. The tumorigenic potential of exposed iPSCs was evaluated using anchorage-independent growth assays and xenograft models, while mechanistic insights were gained via chromatin immunoprecipitation sequencing (ChIP-seq) for Nrf2 and key histone modifications (H3K4me3, H3K9me3, H3K27me3, H3K36me3, and H4K20me3), alongside transcriptomic profiling by RNA sequencing (RNA-seq). Prolonged exposure markedly enhanced tumor sphere formation in vitro and accelerated tumor growth in vivo, indicating the acquisition of CSC-like traits. Integrated ChIP-seq and RNA-seq analyses revealed widespread Nrf2 chromatin binding and global epigenetic remodeling, characterized by increased levels of H3K27me3, H3K36me3, and H4K20me3, a modest rise in H3K9me3, and reduced H3K4me3. Notably, As^3+ exposure enhanced Nrf2 binding at loci regulating glycolysis, cholesterol biosynthesis, self-renewal, and oncogenesis. Functional analyses confirmed that transcriptional and metabolic changes were Nrf2-driven and closely linked to H3K36me3 and H3K27me3 dynamics. Collectively, our findings demonstrate that chronic As^3+ exposure reprograms iPSCs through Nrf2 activation and coordinated epigenetic remodeling, revealing a novel mechanism by which environmental carcinogens exploit stem cell plasticity to initiate CSC-like transformation. Keywords: Arsenic (As^3+), Nrf2, iPSCs, CSCs, Histone methylation Graphical abstract [43]Image 1 [44]Open in a new tab Highlights * • Chronic arsenic exposure induces CSC-like transformation in human iPSCs. * • Nrf2 activation drives metabolic and transcriptional reprogramming in iPSCs. * • Arsenic alters histone marks, enhancing H3K27me3 and H3K36me3 deposition. * • Upregulated genes are enriched in glycolysis, cholesterol biosynthesis, and stemness. * • Nrf2 and histone remodeling coordinately regulate CSC-associated gene expression. 1. Introduction Environmental exposure to arsenic, particularly in regions with contaminated drinking water, poses a major public health risk worldwide. Inorganic arsenic exists primarily as trivalent (As^3+) and pentavalent (As^5+) species, with human exposure typically involving a mixture of both. Among these, As^3+ is recognized as the more toxic and biologically active form and is the predominant intracellular species following cellular uptake and reduction [[45]1]. Extensive epidemiological and case-control studies have established a strong link between arsenic exposure and elevated risks of cancers in the lung, liver, prostate, skin, and other organs [[46]2]. Although As^3+ has limited mutagenic potential, experimental evidence from in vitro and in vivo models indicates that it is highly effective in promoting malignant transformation and carcinogenesis. Mechanistically, As^3+ activates a board range of stress-responsive and oncogenic kinases through both oxidative stress-dependent and -independent mechanisms. Using prolonged low-dose exposure to mimic environmental relevant conditions, our previous studies and that of others have demonstrated that As^3+-transformed non-cancerous cells acquire features of cancer stem-like cells (CSCs). These include enhanced tumor sphere formation, resistance to apoptosis, elevated expression of stemness transcription factors, and metabolic reprogramming—specifically, reduced mitochondrial activity and a shift toward glycolysis [[47][3], [48][4], [49][5]]. We previously showed that this metabolic switch from mitochondrial tricarboxylic acid (TCA) cycle activity to cytosolic glycolysis is largely driven by Nrf2-dependent transcriptional activation of glycolytic enzymes and antioxidant genes, thereby conferring a survival advantage under As^3+-induced stress [[50]5]. In parallel, As^3+-induced CSCs display a distinct epigenetic landscape, with increased levels of the transcriptionally active histone mark H3K4me3 and reduced levels of the repressive mark H3K27me3 [[51]6]. Nrf2 (nuclear factor erythroid 2–related factor 2) is traditionally known for orchestrating the cellular antioxidant response [[52]7,[53]8]. However, emerging evidence positions Nrf2 as a key modulator of redox-sensitive transcriptional networks, metabolic plasticity, carcinogenesis, and CSC maintenance [[54][9], [55][10], [56][11]]. In As^3+-exposed cells, Nrf2 is frequently activated and has been implicated in driving oncogenic transformation by coordinating both metabolic and epigenetic reprogramming [[57]12,[58]13]. Given this dual role in stress adaptation and oncogenesis, we hypothesized that Nrf2 play a pivotal role in initiating As^3+-induced transformation. To test this hypothesis, we investigated genome-wide Nrf2 binding patterns alongside histone methylation dynamics, aiming to delineate the transcriptional and epigenetic landscape associated with chronic As^3+ exposure. Most prior studies investigating As^3+ carcinogenicity have relied on terminally differentiated or immortalized cancer cell lines, which often harbor genetic abnormalities and chromosomal instability that may confound mechanistic interpretation. In contrast, human induced pluripotent stem cells (iPSCs) offer a genetically stable and renewable model capable of differentiating into all three germ layers [[59]14]. Although As^3+ is predominantly metabolized in the liver into monomethylarsonic acid (MMA) and dimethylarsinic acid (DMA), iPSCs lack substantial hepatic metabolic capacity and are unlikely to efficiently methylate As^3+. Therefore, iPSCs provides a unique opportunity to investigate the direct effects of unmetabolized As^3+ on early stem cell states and epigenetic regulation. Recent studies have highlighted the value of iPSCs in toxicology and carcinogenicity testing. For instance, Jahan et al. utilized human episomal iPSCs to assess the neuroprotective effects of Nobiletin against sodium arsenate-induced toxicity [[60]15]. Similarly, Graham et al. reported that As^3+ alters morphology, reduces viability, and induces DNA damage in foreskin fibroblast-derived iPSCs [[61]16]. Bao et al. further showed that As^3+ disrupts cardiomyocyte differentiation by downregulating key mesodermal and cardiac progenitor markers, including TBX1, EOMES, GATA4, MESP1, TBX5, and ACTN4 [[62]17]. Additionally, Wang et al. demonstrated that chronic As^3+ exposure drives transformation in a mouse intestinal organoid model [[63]18]. Importantly, emerging attention has turned to non-classical oncogenic drivers. Many cancer hallmarks, such as metastasis, therapy resistance, and CSC persistence, are sustained by pathways not traditionally classified as oncogenic. This concept of “non-oncogenic addiction” highlights molecular dependencies, like sustained Nrf2 activation, that may represent actionable vulnerabilities in tumors lacking canonical mutations. Understanding these alternative mechanisms is essential for identifying novel therapeutic targets and elucidating early carcinogenic processes. In the present study, we employed the human iPSCs (Nips-B2), derived from nasal epithelial cells of an atopic asthmatic patient using Sendai virus-mediated delivery of OCT3/4, SOX2, KLF4, and MYC [[64]19]. We exposed the cells to a chronic low dose of As^3+ (0.25 μM) for three months to model environmental exposure and investigated the resulting alterations in genome-wide Nrf2 occupancy and histone methylation patterns. Consistent with our previous findings in human bronchial epithelial cells (BEAS-2B cells), we observed that chronic As^3+ exposure induces profound metabolic and epigenetic reprogramming in iPSCs. These data provide new insights into the early molecular events underpinning As^3+-induced transformation and support a central role for Nrf2 in mediating the stem cell malignancy associated with chronic As^3+ exposure. 2. Materials and methods 2.1. Cell culture The human iPSC line Nips-B2 (HPS0223) [[65]19] was obtained from the RIKEN Bioresource Center (Tsukuba, Japan). Cells were maintained on iMatrix-511-coated 60-mm dishes (TaKaRa, cat#T304) in StemFit® Basic04 Complete Stem Cell Culture Medium (amsbio, cat#SF041-001), supplemented with 80 ng/mL basic fibroblast growth factor (bFGF) and 1 % penicillin/streptomycin (Gibco, cat#15140122). The culture medium was refreshed daily. For routine passaging, iPSCs were dissociated either as cell clumps using a pipette or cell scraper, or as single cells using TrypLE™ Select (Gibco, cat#12563011). For cryopreservation, cells were enzymatically dissociated with TrypLE™ Select and frozen at −80 °C or in liquid nitrogen using STEM-CELLBANKER® freezing medium (amsbio, cat#11924). 2.2. Establishment of As^3+-transformed iPSCs As^3+-transformed iPSCs were generated by continuously exposing Nips-B2 cells to 0.25 μM arsenite (As^3+) for 105 days. Culture medium was replenished daily, and cells were passaged twice weekly. Untreated Nips-B2 cells were cultured in parallel as parental controls. To assess the malignant transformation potential, anchorage-independent growth assays were performed. iPSCs were dissociated into single cells, and 500 cells were seeded in 3 mL StemFit® Basic04 medium onto PolyHEMA-coated (5 mg/mL) dishes. After two weeks of incubation, bright-field microscopy was used to quantify sphere formation. The sphere size was measured by ImageJ ([66]https://imagej.net/ij/index.html). 2.3. In vivo tumorigenicity assay All animal procedures were conducted in compliance with the guidelines of the Division of Laboratory Animal Resources (DLAR) at Stony Brook University (IACUC protocol #IACUC202100104). Mice were housed in a maximum isolation facility with unrestricted access to food and water and maintained on a 12-h light/dark cycle. Eight-week-old female NSG mice (NOD.Cg-Prkdc^scid Il2rg^tm1Wjl/SzJ; Jackson Laboratories) were randomly assigned to two groups (n = 5 per group): control and As^3+-transformed iPSCs. Cells were harvested using TrypLE™ Select and resuspended on ice in phosphate-buffered saline (PBS) mixed with phenol red-free Cultrex basement membrane extract (R&D Systems, cat#3432-005-01) at a final BME concentration of 10 mg/mL. A total of 1 × 10^6 cells in 200 μL of the cell/BME mixture was injected subcutaneously into each mouse. Tumor growth was monitored every other day, and tumor volumes were measured starting two weeks post-injection. Mice were euthanized if the subcutaneous tumor reached 2 cm in diameter or if no tumor developed within 30 days. Harvested tumor tissues were fixed in 10 % formalin and processed for histological analysis using Hematoxylin and Eosin (H&E) staining. 2.4. Western blotting Proteins were extracted using 1 × RIPA buffer (CST, cat#9806) supplemented with protease and phosphatase inhibitor cocktail (Thermo Fisher Scientific, cat#A32957) and PMSF (Thermo Fisher Scientific, cat#36978). Protein concentrations were determined using the BCA Protein Assay Kit (Thermo Fisher Scientific, cat#23225). Samples were mixed with 4 × LDS sample buffer (Thermo Fisher Scientific, cat#NP0008) and dithiothreitol (Thermo Fisher Scientific, cat#R0861), then denatured at 95 °C for 10 min. Equal amounts of protein were separated on 10 % SDS-PAGE gels and transferred to PVDF membranes (Thermo Fisher Scientific, cat#88518). Membranes were blocked with 5 % non-fat milk for 1 h at room temperature, then incubated overnight at 4 °C with primary antibodies. The following day, membranes were washed three times (10 min each) and incubated with species-appropriate HRP-conjugated secondary antibodies for 1 h at room temperature. Protein bands were visualized using the ChemiDoc MP Imaging System (Bio-Rad). The following antibodies were used: anti-NANOG (CST, cat#3580, 1:1000), anti-KCTD10 (Sigma, Cat#HPA014273, 1:1000), anti-CD44 (CST, Cat#37259, 1:1000), anti-GAPDH (CST, cat#97166, 1:1000), anti-rabbit (Thermofisher, cat#31460, 1:3000), and anti-mouse (Thermofisher, cat#31430, 1:3000). 2.5. Chromatin immunoprecipitation sequencing (ChIP-seq) A total of 5×10^6 iPSCs, treated with or without As^3+ for 3 months, were collected for ChIP-seq analysis. The experiment was performed following protocols previously described [[67]20,[68]21]. All reagents and ChIP-grade antibodies were obtained from Active Motif (Carlsbad, CA). Antibodies used in this study were pre-validated by the manufacturer to ensure high specificity and a satisfactory signal-to-noise ratio. Genome-wide chromatin profiling focused on histone 3 or 4 trimethylation marks, H3K9me3, H3K27me3, H3K36me3, and H4K20me3, which were used to segment and annotate distinct chromatin states across the genome. ChIP-seq data quality was assessed through multiple metrics, including mapping ratio, read depth, normalized strand coefficient (NSC), background uniformity, GC content bias, and signal enrichment at summits. Data visualization and inspection of signal distribution were carried out using the UCSC Genome Browser. 2.6. Bulk RNA sequencing (RNA-seq) A total of 2×10^6 cells were collected for RNA sequencing services following the Active Motif (Carlsbad, CA) protocol. Total RNA was extracted and assessed for quality and integrity using an Agilent Bioanalyzer. Library preparation and sequencing were performed by Active Motif using the Illumina platform. Raw reads were aligned to the human reference genome (hg19) using the STAR aligner (v2.5.2b). Transcript assembly and quantification were conducted with Cufflinks, and gene expression levels were normalized using geometric FPKM (Fragments Per Kilobase of transcript per Million mapped reads) values. For differential expression analysis, read counts were derived from exon-mapped fragments, quantified at the annotated 3′ ends of genes. Differentially expressed genes (DEGs) were identified using DESeq2 package (v1.14.1) in R 4.3.1 following standard preprocessing steps, including data normalization and filtering. 2.7. Bioinformatics analysis To explore the biological relevance of DEGs identified from both ChIP-seq and RNA-seq datasets, functional enrichment analysis was conducted using Enrichr ([69]https://maayanlab.cloud/Enrichr). DEGs were uploaded and analyzed across multiple curated gene set libraries, including Gene Ontology (GO) categories—Biological Process, Molecular Function, and Cellular Component—as well as KEGG pathways, Reactome pathways, Gene Set Enrichment Analysis (GSEA) gene sets, and the Molecular Signatures Database (MSigDB). Additionally, transcription factor regulatory networks were explored using ChEA and ENCODE databases. Enrichment results were ranked by the combined score, which the p-value from Fisher's exact test with a z-score reflecting deviation from the expected rank. Terms with an adjusted p-value (padj) < 0.05 and strong biological relevance were considered significantly enriched. The top-ranking pathways and gene sets were selected for further interpretation and visualization. 2.8. Kaplan-meier survival analysis To assess the prognostic significance of specific genes in lung cancer, Kaplan–Meier survival analysis was conducted using the Kaplan–Meier Plotter online tool ([70]https://kmplot.com/analysis/index.php?p=home). This platform integrates gene expression and clinical outcome data from publicly available datasets, including The Gene Expression Omnibus (GEO), European Genome-phenome Archive (EGA), and The Cancer Genome Atlas (TCGA). For the current analysis, the lung cancer gene chip dataset (Affymetrix microarray) was selected. Gene symbols of interest were queried individually, and patients were stratified into high- and low-expression groups based on the median expression level. Overall survival (OS) was used as the primary endpoint, and the “Auto select best cutoff” option was applied unless otherwise specified. The hazard ratio (HR) with 95 % confidence interval (CI) and the log-rank p-value were calculated and displayed on the corresponding Kaplan–Meier plots, which were generated for all selected genes. 2.9. Statistical analysis Unless otherwise specified, all experiments were independently repeated at least three times, each with a minimum of three biological replicates. Statistical comparisons between groups were conducted using either Student's t-test or one-way analysis of variance (ANOVA), depending on the experimental design. A two-tailed p-value less than 0.05 was considered statistically significant. All data were analyzed using GraphPad Prism 8.0, and results are expressed as mean ± standard deviation (SD). 3. Results 3.1. As^3+ enhances sphere formation and tumorigenesis of iPSCs To assess the cytotoxicity of As^3+ in iPSCs (Nips-B2), we initially treated the cells with a concentration range of 0–2.5 μM As^3+ in StemFit Basic04 medium for 10 days. No viable iPSCs were observed after 4 days of exposure to 2.5 μM As^3+ or after 10 days of exposure to 1.0 μM As^3+. Based on these findings, we selected lower, sub-lethal concentrations (0.1–0.25 μM As^3+) for subsequent experiments. As depicted in [71]Fig. 1A, iPSCs tolerated 0.25 μM As^3+ well over a period of 1, 2, and 3 months of continuous treatment. To examine whether As^3+ affects the expression of stemness-related proteins, we performed Western blot analysis of CD44 and NANOG in iPSCs at passage 24 (day 91) and passage 26 (day 105) following 0.25 μM As^3+ exposure. While As^3+ only slightly reduced the level of full-length CD44, it markedly increased the cleaved form of CD44 in both passages ([72]Figure 1B), suggesting enhanced CD44 processing. The effect of As^3+ on NANOG expression was inconclusive. Despite the absence of significant morphological differences between control and treated cells, As^3+ treatment led to a notable increase in sphere size in an in vitro sphere formation assay ([73]Figure 1C), indicating enhanced self-renewal potential. To determine whether As^3+ exposure promotes tumorigenicity, we subcutaneously injected 1 × 10^6 cells from both untreated and 3-month As^3+-treated iPSCs into NSG mice and monitored tumor development. Tumors were detectable 20–25 days post-injection in both groups. However, by days 32–39, tumors derived from As^3+-treated iPSCs were significantly larger than those from control cells ([74]Figure 1D). Histopathological analysis of excised tumors revealed that those from control iPSCs displayed typical teratoma-like structures containing tissues representative of all three germ layers (ectoderm, mesoderm, and endoderm), along with inflammatory infiltration, ductal hyperplasia, and some differentiated papillary structures. In contrast, tumors from 3-month As^3+-treated iPSCs exhibited additional malignant features, including keratinized epithelium, mucin-filled cysts, pigment deposition, and nests of neoplastic squamous cells ([75]Figure 1E). Together, these findings demonstrate that chronic low-dose As^3+ exposure enhances the tumorigenic potential of iPSCs. Fig. 1. [76]Fig. 1 [77]Open in a new tab Chronic low-dose As^3+ exposure enhances sphere formation and tumorigenicity of iPSCs. A Bright-field images of iPSCs (Nips-B2) after continuous exposure to 0.25 μM As^3+ for 1, 2, and 3 months. Cells maintained normal morphology and confluency, indicating tolerance to prolonged low-dose As^3+ treatment. B Western blot analysis of stemness markers CD44 and NANOG in iPSCs at passage 24 (day 91) and passage 26 (day 105) following 0.25 μM As^3+ exposure. As^3+ treatment reduced full-length CD44 while markedly increased its cleaved form; effects on NANOG expression were variable. CIn vitro sphere formation assay showing increased sphere size in iPSCs treated with As^3+ for 3 months, indicating enhanced self-renewal capacity under anchorage-independent conditions. Data are presented as mean ± SD (N = 10 per group). D Tumor growth curves in NSG mice subcutaneously injected with control or 3-month As^3+-treated iPSCs (1 × 10^6 cells/mouse, N = 5 per group). Tumors derived from As^3+-transformed iPSCs grew significantly larger between days 32 and 39 post-injection. Data are presented as mean ± SD. E Representative H&E-stained tumor sections from control and As^3+-transformed iPSC-derived xenografts. Control tumors exhibited typical teratoma architecture with ectodermal, mesodermal, and endodermal elements. In contrast, tumors from As^3+-transformed iPSCs showed malignant features, including keratinized epithelium, mucin-filled cysts, pigment deposition, and nests of neoplastic squamous cells. 3.2. As^3+ treatment alters histone methylation landscapes in iPSCs To investigate the molecular basis of the enhanced tumorigenicity observed in 3-month As^3+-treated iPSCs, we performed ChIP-seq to profile histone trimethylation patterns. Specifically, we analyzed five key histone modifications: H3K4me3, H3K9me3, H3K27me3, H3K36me3, and H4K20me3, using antibodies specific to each mark. In our previous studies involving As^3+-induced cancer stem-like cells (CSCs) derived from human bronchial epithelial cells (BEAS-2B), we observed a global increase in H3K4me3 enrichment [[78]2,[79]6]. Interestingly, the pattern in iPSCs diverged significantly: prolonged exposure to As^3+ led to a marked decrease in H3K4me3 levels across gene-enriched regions. Conversely, the levels of H3K27me3, H3K36me3, and H4K20me3 were substantially elevated in As^3+-transformed iPSCs ([80]Figure 2A). Given the established role of Nrf2 as a central regulator of redox homeostasis, metabolic reprogramming, and stemness—particularly in the context of As^3+-induced transformation [[81]5,[82]13,[83]22]—we additionally profiled genome-wide Nrf2 binding to determine its contribution to the observed chromatin alterations. Consistent with its proposed function in driving As^3+-mediated CSC phenotypes, we detected a marked increase in Nrf2 occupancy across the genome following As^3+ exposure ([84]Figure 2A). To further characterize the effects of As^3+ on the chromatin landscape, we visualized the distribution of methylation peaks across the genome using the UCSC Genome Browser. As expected, over 90 % of H3K4me3 peaks localized sharply to promoter regions or the transcription start site (TSS), while H3K36me3 exhibited broad enrichment across gene bodies—as exampled by the TBCK locus on chromosome 4 ([85]Figure 2B, left panels). TBCK encodes a putative kinase implicated in mTOR signaling and cell proliferation [[86]23]. It has not been previously reported as a canonical Nrf2 target. In the region of As^3+-enhanced Nrf2 enrichment peak, there is a conserved Nrf2 binding element TGAGTGA, suggesting TBCK is an undocumented Nrf2 target gene. We selected the TBCK locus as a representative site to illustrate the spatial distribution of active and repressive histone marks due to its well-annotated structure, detectable expression in iPSCs, and dynamic epigenetic response to As^3+ exposure observed in our dataset. On the entire chromosome level, H3K27me3 peaks were more variably distributed, appearing at promoters, gene bodies, and flanking regulatory regions. In contrast, both H3K9me3 and H4K20me3—histone marks classically associated with transcriptional repression and heterochromatin—formed large clusters in gene-poor regions lacking active marks like H3K4me3, H3K27me3, and H3K36me3. These clusters were predominantly localized to pericentromeric regions and the terminal ends of the q arms on several chromosomes, including chromosome 22, a representative small autosome often used to visualize compact heterochromatin domains [[87]24]—as highlighted by red filled triangles in the right panels of [88]Figure 2B. While As^3+ had minimal impact on H3K9me3 distribution, it significantly enhanced H4K20me3 enrichment, with new or enlarged clusters aligning with existing H3K9me3 domains. These results not only highlight the mutually exclusive nature of active (H3K4me3, H3K27me3, H3K36me3) and repressive (H3K9me3, H4K20me3) chromatin marks but also underscore the profound remodeling induced by chronic As^3+ exposure in iPSCs. Notably, due to the female origin of the Nips-B2 iPSC line, ChIP-seq analysis revealed no significant methylation peaks on the Y chromosome, except for a few non-specific background signals (data not shown). Fig. 2. [89]Fig. 2 [90]Open in a new tab Chronic As^3+ exposure alters histone methylation landscapes and Nrf2 chromatin binding in iPSCs. A ChIP-seq analysis showing average enrichments of H3K4me3, H3K9me3, H3K27me3, H3K36me3, H4K20me3, and Nrf2 in control and 0.25 μM As^3+-treated iPSCs after 3 months. Chronic As^3+ treatment led to a global reduction in H3K4me3, accompanied by increases in H3K27me3, H3K36me3, H4K20me3, and Nrf2 occupancy. B Representative genome browser views of the TBCK locus (left panel) showing gene-specific enrichment of H3K4me3 and Nrf2 at the promoter and H3K36me3 across the gene body in control and As^3+-transformed iPSCs. A conserved Nrf2 binding element in the Nrf2 peak region, TGAGTGA, inserted in the panel. The right panel displays genome-wide histone methylation profiles—H3K4me3, Nrf2, H3K27me3, H3K36me3, H3K9me3, and H4K20me3—across chromosome 22. Red filled triangles indicate regions of broad H3K9me3 and H4K20me3 enrichment predominantly localized to gene-poor heterochromatic domains, including pericentromeric regions and the distal ends of chromosome arms. These repressive clusters become further intensified following As^3+ exposure. 3.3. H3K4me3 is enriched on genes involved in differentiation and stemness regulation H3K4me3 is widely recognized as an active histone modification associated with gene transcription. However, its functional impact is often cell context-dependent and influenced by the surrounding chromatin environment, including DNA methylation, additional histone modifications (e.g., H3K27me3, H3K79me3), and transcription factor binding dynamics [[91]25]. In line with the global reduction observed in H3K4me3 enrichment across gene bodies ([92]Figure 2A), we also observed a marked decrease in H3K4me3 signals at gene promoters and transcription start sites (TSSs) following chronic As^3+ exposure in iPSCs ([93]Figure 3A, left panel). To illustrate these changes, we selected representative loci on chromosomes 10 and 3 for genome browser visualization. These chromosomes harbor several biologically relevant genes—including MYOD1 and DPPA4—that showed prominent and readily interpretable alterations in H3K4me3 enrichment in our dataset. Moreover, many of the genes with the most significant changes in H3K4me3 following As^3+ exposure were located on these two chromosomes. Chromatin peak analysis further confirmed that approximately 90 % of H3K4me3 peaks showed reduced intensity after As^3+ treatment ([94]Figure 3A, right panel). However, a small fraction of peaks displayed increased intensity post-treatment, highlighted by red filled triangles. To further explore the functional relevance of H3K4me3 loss, we performed pathway enrichment analysis (WikiPathways) on 9325 genes that showed a log[2] fold change < -0.1 in H3K4me3 levels. The top enriched pathways included dopaminergic neurogenesis, epinephrine (EPI)/non-EPI signaling, and various cell differentiation programs ([95]Figure 3B, left panel). Notably, many of these genes are well-established tumor suppressors—such as Myogenic Differentiation 1 (MYOD1), Interferon Regulatory Factor 4 (IRF4), Inhibitor Of DNA Binding 2 (ID2), Cyclin Dependent Kinase Inhibitor 2A (CDKN2A), CDKN2B, and BRCA1 Associated Deubiquitinase 1 (BAP1)—which exhibited substantial H3K4me3 loss ([96]Figure 3B, right panels). These findings suggest that epigenetic repression of differentiation-related tumor suppressor genes may contribute to the acquisition of malignant potential in As^3+-exposed iPSCs. Fig. 3. [97]Fig. 3 [98]Open in a new tab Chronic As^3+ exposure reshapes H3K4me3 enrichment at genes regulating cell differentiation and stemness. A ChIP-seq analysis showing the average profile of merged H3K4me3 peaks across regions spanning -5 kb to +5 kb relative to gene loci in control and As^3+-transformed iPSCs. The right panel illustrates a global reduction in H3K4me3 peak intensities across chromosomes 3 and 10 in iPSCs following 3 months of chronic 0.25 μM As^3+ exposure. A subset of peaks displayed increased intensity post-treatment, highlighted by red triangles. B WikiPathways enrichment analysis of 9325 genes with decreased H3K4me3 levels (log[2] fold change < -0.1) revealed top pathways related to dopaminergic neurogenesis, norepinephrine signaling, and cell differentiation. The right panel shows genome browser tracks with reduced H3K4me3 enrichment at several tumor suppressor genes, including MYOD1, IRF4, ID2, CDKN2A, CDKN2B, and BAP1, in As^3+-transformed iPSCs. C WikiPathways analysis of 1050 genes with increased H3K4me3 levels (log[2] fold change >0.1) highlighted enrichment in RNA processing and metabolic pathways (left). The right panel (highlighted by red triangles) displays representative genome browser views showing elevated H3K4me3 enrichment at core stemness genes such as NANOG, SLC2A3, DPPA4, NR6A1, and TDGF1 in As^3+-transformed iPSCs. Interestingly, although As^3+ broadly diminished H3K4me3, a subset of 1050 genes exhibited notable elevation of H3K4me3 enrichment. This small group represents a focal upregulation module that stands out against the backdrop of global loss. Pathway analysis revealed that these genes were enriched in RNA metabolism, pre-mRNA splicing, and RNA/rRNA processing pathways ([99]Figure 3C, left panel). Importantly, several core pluripotency regulators, including NANOG, Developmental Pluripotency Associated 4 (DPPA4), Nuclear Receptor Subfamily 6 Group A Member 1 (NR6A1), Teratocarcinoma-Derived Growth Factor 1 (TDGF1), and Solute Carrier Family 2 Member 3 (SLC2A3), were among the top genes showing increased H3K4me3 levels in response to As^3+ ([100]Figure 3C, right panels). While enhanced H3K4me3 does not necessarily lead to increased transcription, this modification likely poises these genes for rapid activation, thereby supporting the maintenance of a stem-like or CSC-like state in As^3+-transformed iPSCs. This dual effect—global repression of differentiation programs and selective activation of stemness-associated genes—highlights a reprogramming mechanism that may underlie As^3+-induced malignant transformation. 3.4. Overlapping of As^3+-induced Nrf2 activation with H3K36me3 and H3K27me3 At the chromosome-wide level, we observed co-localization of H3K4me3, H3K27me3 and H3K36me3 clusters in both control and As^3+-transformed iPSCs ([101]Figure 2B). This overlap, particularly between H3K36me3 and H3K27me3, was further validated through the pathway enrichment analysis of genes targeted by H3K36me3 (5943 genes) and H3K27me3 (6511 genes) in the As^3+-transformed iPSCs. The top ten pathways enriched in genes with As^3+-induced H3K36me3 included thromboxane A2 receptor (TXA2R), lysophosphatidic acid receptor (LPAR), C-X-C Motif Chemokine Receptor 3 (CXCR3), Protein Tyrosine Phosphatase Non-Receptor Type 1 (PTP1B), Hedgehog, endothelin, RhoA, ephrin B (EPHB), EPHA, and α−Synuclein (SNCA) ([102]Figure 4A). Notably, eight of these pathways overlapped with those enriched in H3K27me3-targeted genes ([103]Figure 4B, highlighted by pink dots in left panel). Approximately 54 % (3208/5943) of the genes enriched with H3K36me3 and 49 % (3208/6511) of those enriched with H3K27me3 also showed co-enrichment for both marks upon As^3+ treatment ([104]Figure 4B, right panel). Interestingly, transcription factor perturbation assays revealed that six of the top ten pathways in the H3K27me3-targeted genes were enriched in Nrf2 signaling and Nrf2-regulated genes ([105]Figure 4C). We had previously shown that As^3+ activates Nrf2, contributing to metabolic reprogramming in As^3+-induced CSCs from human bronchial epithelial cells BEAS-2B [[106]5]. In iPSCs, As^3+ also strongly activated Nrf2, as indicated by enhanced Nrf2 peaks in the ChIP-seq data, which were detected in both gene bodies ([107]Figure 2A, bottom right) and promoters ([108]Figure 4D, left panel). While As^3+ treatment amplified the existing Nrf2 peaks in control iPSCs, approximately 10–15 % of Nrf2 peaks in As^3+-transformed iPSCs were newly formed and absent in control cells ([109]Figure 4D, peaks marked with red triangle in the right panels). These de novo Nrf2 peaks may represent a shift in Nrf2 distribution on the genome in As^3+-transformed cells. Specifically, As^3+ reduced Nrf2 binding in the proximal promoter and 5′ UTR regions, while increasing its distribution in the exon and intron regions ([110]Figure 4E). Consistent with these findings, WikiPathways analysis of 1905 genes with enhanced Nrf2 enrichment following As^3+ treatment revealed a strong association with Nrf2 signaling, glycolysis, aryl hydrocarbon receptor (AHR) pathways, oxidative stress, and amino acid metabolism ([111]Figure 4F). Fig. 4. [112]Fig. 4 [113]Open in a new tab Overlap of As^3+-induced Nrf2 activation with H3K36me3 and H3K27me3 enrichment in iPSCs. A Pathway enrichment analysis of 5943 genes marked by H3K36me3 following chronic 0.25 μM As^3+ exposure revealed activation of signaling pathways including TXA2R, LPAR, CXCR3, PTP1B, Hedgehog, endothelin, RhoA, EPHB, EPHA, and SNCA. B Comparative pathway analysis showing that 8 of the top 10 pathways enriched in H3K36me3-marked genes also overlapped with those enriched in H3K27me3-marked genes (6511 genes), suggesting epigenetic crosstalk (left). Right: Venn diagrams showing that ∼54 % of H3K36me3-marked genes and ∼49 % of H3K27me3-marked genes were co-enriched for both modifications in As^3+-transformed iPSCs. C Transcription factor perturbation analysis revealed that 6 of the top 10 pathways enriched in H3K27me3-targeted genes are regulated by Nrf2 signaling, supporting a potential link between Nrf2 activity and repressive chromatin remodeling. D ChIP-seq analysis showing average Nrf2 peak distribution across ±5 kb of gene loci in control and As^3+-treated iPSCs. Right: Genome browser views highlighting amplified Nrf2 peaks and de novo As^3+-specific Nrf2 peaks (red triangles), not present in control. E Genomic distribution of Nrf2 binding sites before and after 3-month As^3+ exposure. Chronic As^3+ treatment reduced Nrf2 occupancy at promoter and 5′ UTR regions while increasing its binding within exons and introns, indicating a redistribution of Nrf2 across the genome. F WikiPathways enrichment analysis of 1905 genes with increased Nrf2 binding following As^3+ treatment revealed significant associations with Nrf2 signaling, glycolysis, aryl hydrocarbon receptor (AHR) signaling, oxidative stress response, and amino acid metabolism—highlighting widespread transcriptional reprogramming induced by As^3+. 3.5. As^3+ induces limited cross-talk between Nrf2 and H3K4me3 in iPSCs Nrf2 is widely recognized as a master regulator of oxidative stress response genes. More recently, its oncogenic potential has emerged in various cellular and animal models [[114]8,[115]26]. At the human γ-globin gene locus, Nrf2 binding has been shown to be a prerequisite for the enrichment of the active transcription marker H3K4me3 and for facilitating long-range chromatin interactions between the locus control region and the γ-globin gene promoter [[116]27]. Genome-wide analysis revealed that Nrf2 binding sites largely overlapped with clusters of H3K4me3, H3K27me3, and H3K36me3 ([117]Fig. 2, [118]Fig. 5C). To explore the interplay between Nrf2 and H3K4me3 at the gene level, we compared the gene sets with elevated Nrf2, H3K4me3, and H3K36me3 following As^3+ exposure in iPSCs. Notably, only 6 % of Nrf2 target genes showed concurrent increases in H3K4me3, suggesting a limited but potentially functionally significant correlation. Despite this modest overlap, ChIP-seq Enrichment Analysis (ChEA) of the genes with concurrent Nrf2 and H3K4me3 enrichment identified pathways associated with key transcriptional regulators, including FOXP3, SOX2, EST1, and MYC ([119]Figure 5A). FOXP3 had been implicated in promoting metastasis in human lung cancer and maintaining stemness in hematopoietic stem cells [[120]28,[121]29]. Similarly, SOX2, MYC, and EST1 are well-established factors involved in stem cell self-renewal and differentiation. These findings suggest that the limited cooperation between Nrf2 and H3K4me3 under As^3+ exposure may preferentially enhance tumorigenic stemness in iPSCs. H3K27me3, a well-characterized repressive histone modification, also displayed large-scale co-localization with H3K36me3 and H3K4me3 across the genome ([122]Fig. 2, [123]Fig. 5C). However, at the gene-specific level, less than 1 % of genes enriched for H3K27me3 also exhibited increased H3K4me3 ([124]Figure 5B). Among the 47 genes that were marked by both modifications—indicative of a bivalent chromatin state—many were involved in pluripotency-associated pathways such as those governed by SOX2 and NANOG ([125]Figure 5B, right panel). This supports the notion that key developmental and pluripotent genes remain transcriptionally poised via bivalent histone configurations. Furthermore, Nrf2 binding clusters generally paralleled those of H3K9me3, H3K27me3, and H3K36me3 ([126]Figure 5C, green-filled circles), while being largely excluded from regions enriched with H3K9me3 and H4K20me3, which are typically associated with heterochromatin and transcriptional silencing ([127]Figure 5C, yellow box). Across all autosomes and the X chromosome, most Nrf2 clusters were not co-located with H3K9me3 and H4K20me3 domains ([128]Figure 5C, yellow box in left panel), except those in gene-poor regions or loci expressing long non-coding RNAs ([129]Figure 5C, right panels). Fig. 5. [130]Fig. 5 [131]Open in a new tab Enrichment analysis of genes co-targeted by Nrf2 and H3K4me3 following As^3+ exposure. A ChEA pathway enrichment analysis of genes with concurrent Nrf2 binding and H3K4me3 enrichment in iPSCs after chronic As^3+ treatment. Identified transcriptional regulators include FOXP3, SOX2, EST1, and MYC, implicating their potential roles in promoting stemness and tumorigenic features. B ChEA analysis of bivalent chromatin states in As^3+-transformed iPSCs. Left: Venn diagram showing minimal overlap (<1 %) between genes marked by both H3K4me3 and H3K27me3. Right: Enrichment analysis of 47 genes exhibiting a bivalent chromatin state (co-enrichment of H3K4me3 and H3K27me3), revealing strong association with pluripotency-related factors such as SOX2 and NANOG, suggesting these genes are poised for developmental regulation. C Genome browser snapshot showing the alignment of Nrf2 binding peaks with clusters of H3K27me3 and H3K36me3, as well as selected overlap with H3K9me3 (indicated by green-filled circles). These Nrf2-enriched regions are largely excluded from domains marked by H3K9me3 and H4K20me3, as highlighted in the yellow box. Right panels illustrate rare instances of Nrf2 occupancy within H3K9me3-enriched regions, all of which are located in gene-poor areas or near long noncoding RNA (lncRNA) loci. 3.6. Upregulated oncogenic gene expression in As^3+-transformed iPSCs Above data demonstrated that As^3+ exposure induces both Nrf2 activation and broad epigenetic reprogramming in iPSCs. However, these chromatin-level changes do not necessarily reflect direct transcriptional outcomes related to oncogenesis or CSC formation. To investigate this, we performed RNA-seq to profile gene expression changes in control and As^3+-transformed iPSCs. Using a shrunken log[2] fold change cutoff of >0.1 for upregulation and < -0.1 for downregulation, we identified 2133 upregulated and 2369 downregulated genes following As^3+ exposure. The top 20 most significantly altered genes are presented in [132]Figure 6A. Among the most upregulated, ALPPL2 (ALPG), a folate metabolism-associated alkaline phosphatase, is a known surface marker for the naïve pluripotent state and plays a critical role in both establishing and maintaining pluripotency [[133]30]. XYLT1 and XXYLT1, which encode xylosyltransferases involved in glycosaminoglycan biosynthesis, are essential for extracellular matrix function; mutations in XYLT1 cause Desbuquois dysplasia [[134]31]. Other upregulated genes include TRNP1, a neural stem cell DNA-binding protein, and PLA2G2A, a niche factor involved in intestinal stem cell regulation [[135]32]. Gene Ontology analysis using EnrichNet showed that the top five enriched biological processes among upregulated genes were associated with cholesterol metabolism and vesicle trafficking—including COPI and COPII vesicle coating, intra-Golgi transport, isoprenoid biosynthesis, and cholesterol biosynthesis ([136]Figure 6B). Notably, 20 out of 34 genes in the EnrichNet cholesterol biosynthesis pathway were significantly upregulated, suggesting a potential role of cholesterol homeostasis in the tumorigenicity of As^3+-transformed iPSCs (data not shown). Consistent with our earlier studies [[137]26,[138]33], Molecular Signatures Database (MSigDB) analysis further revealed enrichment of upregulated genes in pathways associated with Myc signaling, glycolysis, hypoxia, and cholesterol metabolism ([139]Figure 6C), highlighting an oncogenic transcriptional program linked to stemness and metabolic reprogramming. In contrast, over 2000 genes were downregulated by As^3+. Transcription factor perturbation analysis revealed enrichment of repressed genes downstream of regulators such as GATA6, SPDEF, ATF6, and FOXQ1 ([140]Figure 6D). GATA6 is a key transcription factor for early cardiac differentiation [[141]34] and endodermal lineage commitment in human iPSCs [[142]35]. Similarly, SPDEF, ATF6, ETV2, and FOXQ1 have been implicated in stem cell or cancer cell differentiation [[143][36], [144][37], [145][38], [146][39]]. Thus, repression of these factors may diminish differentiation potential while reinforcing the pluripotent and self-renewing phenotype of As^3+-transformed iPSCs. MSigDB analysis of downregulated genes further supported this interpretation, showing significant enrichment in pathways related to p53 signaling, apoptosis, and myogenesis ([147]Figure 6E), all of which are commonly associated with cell differentiation and tumor suppression. Fig. 6. [148]Fig. 6 [149]Open in a new tab Oncogenic transcriptional reprogramming in As^3+-transformed iPSCs. A Heatmap showing the top 20 differentially expressed genes in iPSCs chronically exposed to 0.25 μM As^3+ for 3 months, as identified by RNA-seq. Significantly upregulated genes include ALPPL2, XYLT1, TRNP1, XXYLT1, and PLA2G2A. B Gene Ontology (GO) analysis using EnrichNet revealed significant enrichment of upregulated genes in pathways related to cholesterol biosynthesis, isoprenoid metabolism, and vesicle trafficking (COPI/COPII coating and intra-Golgi transport). C MSigDB enrichment analysis showing upregulated genes are associated with oncogenic and stemness-related pathways, including Myc signaling, glycolysis, hypoxia response, and cholesterol metabolism. D Transcription factor perturbation analysis of downregulated genes revealed enrichment for targets of GATA6, SPDEF, ATF6, ETV2, and FOXQ1, indicating suppression of key regulators involved in lineage specification and differentiation. E MSigDB analysis of downregulated genes showing significant association with tumor suppressive and differentiation-related pathways, including p53 signaling, apoptosis, and myogenesis, suggesting impaired differentiation potential and enhanced self-renewal in As^3+-transformed iPSCs. 3.7. As^3+-induced Nrf2 amplifies glycolytic metabolism via the pentose phosphate pathway (PPP) and hexosamine biosynthetic pathway (HBP) in iPSCs We previously demonstrated that As^3+-induced Nrf2 drives a metabolic reprogramming from mitochondrial TCA cycle activity toward cytosolic glycolysis in CSCs derived from bronchial epithelial cells. This metabolic shift diverts glycolytic intermediates into ancillary biosynthetic routes, including the HBP and the serine/glycine pathway [[150]5]. Consistent with this, we observed a marked enhancement of Nrf2 signaling in As^3+-transformed iPSCs. ChIP-seq data confirmed widespread enrichment of Nrf2 binding across the genome ([151]Fig. 2, [152]Fig. 4D), while RNA-seq revealed increased expression of key Nrf2 regulators and target genes ([153]Figure 7A). These include the upstream activators VCP, SQSTM1 (p62), and KLF2, as well as canonical Nrf2 downstream genes involved in antioxidant defense such as PRDX1, FTL, HMOX1, and NQO1. Interestingly, ChIP-seq also showed strong Nrf2 occupancy at the promoter regions of SQSTM1, VCP and PRDX1, highlighted by red arrows in [154]Figure 7B, suggesting the presence of a positive feedback loop that amplifies Nrf2 signaling in response to As^3+. This autoregulatory circuit likely reinforces a broader oncogenic and metabolic program encompassing redox homeostasis, glycolysis, stemness, and transcriptional plasticity. Interestingly, TXNRD2, which was downregulated following As^3+ exposure, lacked Nrf2 binding, further supporting a selective transcriptional program. RNA-seq also revealed marked upregulation of genes encoding rate-limiting enzymes in glycolysis, especially those involved in early steps, such as HK2 and TKT, and in the glycolytic shunt pathways—PPP and HBP ([155]Figure 7C). In contrast to our previous findings in CSCs derived from As^3+-transformed BEAS-2B cells [[156]5], several genes critical for the serine/glycine pathway were downregulated in As^3+-transformed iPSCs, suggesting cell-type-specific metabolic rewiring. The oxidative branch of the PPP, crucial for the generation of nucleotides and NADPH in rapidly proliferating cells, was strongly activated. All three rate-limiting enzymes—G6PD, PGLS, and PGD—were significantly upregulated, accompanied by robust Nrf2 and H3K36me3 enrichment at the gene bodies and/or first exon regions ([157]Figure 7D, top panels), indicating transcriptional activation through coordinated Nrf2 signaling and chromatin remodeling. Similarly, most key enzymes in the HBP—GFPT, PGM3, UAP1, and UAP1L1—were also upregulated in As^3+-transformed iPSCs, with corresponding Nrf2 and H3K36me3 co-enrichment at their loci ([158]Fig. 7C and bottom panel of [159]Figure 7D). GNPNAT1 was the only HBP gene that was not induced. As the HBP is crucial for protein glycosylation, including modification of stemness regulators and SCAP (SREBP cleavage-activating protein), its activation supports cholesterol and fatty acid biosynthesis [[160]40]. Collectively, these findings suggest that Nrf2-mediated activation of the PPP and HBP contributes to the acquisition of stem-like and tumorigenic properties in As^3+-transformed iPSCs through metabolic reprogramming and epigenetic regulation. Fig. 7. [161]Fig. 7 [162]Open in a new tab Nrf2-mediated metabolic reprogramming in As^3+-transformed iPSCs. A RNA-seq analysis reveals significant upregulation of key Nrf2 target genes in As^3+-treated iPSCs, including Nrf2 regulators (VCP, SQSTM1, KLF2) and antioxidant defense genes (PRDX1, FTL, HMOX1, NQO1), confirming robust activation of Nrf2 signaling. B ChIP-seq analysis demonstrates strong Nrf2 binding at the promoter regions of SQSTM1, VCP and PRDX1 in As^3+-transformed iPSCs, highlighted by red arrows, suggesting a positive feedback loop that amplifies Nrf2 signaling. No Nrf2 binding was detected at the promoter region of TXNRD2, indicating selective regulation. C Bar graph showing significant upregulation of key enzymes involved in glycolysis and glycolytic shunt pathways, including the pentose phosphate pathway (PPP) and hexosamine biosynthetic pathway (HBP). Key enzymes such as HK2 and TKT (glycolysis) and G6PD, PGLS, PGD, GFPT, PGM3, and UAP1 (PPP/HBP) are markedly upregulated in As^3+-transformed iPSCs. D ChIP-seq analysis shows significant Nrf2 binding and H3K36me3 histone modification at the gene bodies and first exons of key metabolic genes in the PPP (G6PD, PGLS, PGD) and HBP (GFPT, PGM3, UAP1), supporting transcriptional activation through both Nrf2 signaling and histone modification. GNPNAT1 in the HBP pathway was not induced. 3.8. Nrf2 promotes cholesterol metabolism and oncogenic stemness in As^3+-transformed iPSCs The pathological role of elevated blood cholesterol in metabolic diseases such as atherosclerosis, myocardial infarction, and stroke is well-established. More recently, aberrant cholesterol homeostasis has also been implicated in cancer progression, with studies linking dysregulated cholesterol metabolism to breast cancer metastasis and poor prognosis across various malignancies [[163]41]. Intriguingly, increased cholesterol efflux associated with hypercholesterolemia has been shown to expand hematopoietic stem and progenitor cells, as well as leukemia-initiating cells [[164]42]. Our previous study revealed that chronic As^3+ exposure reprograms non-cancerous epithelial cells into CSCs, characterized by persistently upregulated cholesterol metabolism [[165]33]. Consistent with these findings, cholesterol metabolism emerged as one of the top 10 enriched pathways in As^3+-transformed iPSCs ([166]Figure 6C). RNA-seq data indicated that genes involved in every step of cholesterol biosynthesis were upregulated following As^3+ treatment, including key regulators of sterol regulatory element-binding protein (SREBP) signaling ([167]Fig. 8A and B, highlighted in red). Moreover, ChIP-seq revealed pronounced Nrf2 enrichment at the promoter or gene body regions of several cholesterol biosynthetic genes such as ACAT2, HMGCS1, MMAB-MVK, and FDPS, supporting a direct transcriptional role of Nrf2 in driving cholesterol metabolism in iPSCs ([168]Figure 8C). We previously reported that BEAS-2B cells transformed by chronic As^3+ exposure exhibit suppressed ROS production, reduced oxidative phosphorylation, enhanced glycolysis, and stem-like properties [[169]2,[170]4,[171]43]. Given that a major function of Nrf2 is to promote antioxidant defense and reduce intracellular ROS—a hallmark of CSCs—and that accumulated cholesterol can further shift cellular metabolism from oxidative phosphorylation to glycolysis [[172]44], it is plausible that Nrf2-driven cholesterol accumulation enhances the CSC-like potential of iPSCs in response to As^3+. Gene expression profiles of As^3+-transformed iPSCs support this hypothesis. Several transcription factors essential for oncogenic stemness were markedly upregulated, including PRDM14, KLF4, NR6A1, STAT5A, NANOG, and SOX2 ([173]Figure 8D). In contrast, the expression of MYC, VEGFA, and PDGFB was significantly suppressed by As^3+, though the underlying mechanism remains unclear. For the upregulated genes, increased Nrf2 occupancy may contribute to their transcriptional activation. For example, within the STAT5 gene cluster on chromosome 17, As^3+ induced prominent Nrf2 peaks specifically at STAT5A (highlighted by red arrows), which showed elevated expression, whereas neighboring genes STAT5B and STAT3 showed no Nrf2 binding and no change in expression ([174]Figure 8E). Another notable feature of As^3+-transformed iPSCs is the reprogramming of histone trimethylation marks ([175]Figure 2A), likely resulting from altered expression of genes encoding chromatin-modifying enzymes. As^3+ treatment increased the expression of DNA methyltransferases DNMT3B and DNMT3L, as well as demethylation-associated enzymes TET and GADD45G, while reducing expression of GADD45A and GADD45B ([176]Figure 8F). These changes may shift the balance of DNA methylation. It is known that DNA hypermethylation is positively associated with histone marks H3K9me3, H3K27me3, and H4K20me3, and negatively associated with H3K4me3, due to methylation-dependent repulsion of H3K4 methyltransferases [[177]45]. Thus, the observed reduction of H3K4me3 and increase in H3K27me3 and H4K20me3 may result from elevated DNMT3B and DNMT3L activity in response to As^3+. As^3+ also modulated expression of several histone methyltransferases and demethylases, including EZH2, SETD4, and the KDM4 family ([178]Figure 8F). However, the extent to which these transcriptional changes directly contribute to altered histone methylation remains to be elucidated. Notably, changes in expression of these epigenetic regulators did not consistently correlate with Nrf2 binding, suggesting involvement of additional regulatory mechanisms beyond Nrf2. Fig. 8. [179]Fig. 8 [180]Open in a new tab Nrf2 directly regulates cholesterol biosynthesis and oncogenic stemness transcriptional programs in As^3+-transformed iPSCs. A Schematic representation of the cholesterol biosynthetic pathway, with genes upregulated in As^3+-transformed iPSCs highlighted in red. B Bar graph showing RNA-seq-based differential expression of key cholesterol biosynthetic genes in control and As^3+-transformed iPSCs. Gene expression is presented as average log[2] fold change. C ChIP-seq tracks illustrating Nrf2 occupancy at promoter or gene body regions of representative cholesterol biosynthetic genes—including ACAT2, HMGCS1, MMAB-MVK, and FDPS—in control and As^3+-transformed iPSCs. D RNA-seq analysis showing upregulation of oncogenic stemness-associated transcription factors, including PRDM14, KLF4, NR6A1, STAT5A, NANOG, and SOX2, in As^3+-transformed iPSCs. Gene expression is shown as average log[2] fold change. E ChIP-seq tracks of the STAT5 gene cluster showing specific Nrf2 enrichment at the STAT5A locus (indicated by red arrows), which correlates with increased expression. No Nrf2 binding or expression changes were observed at neighboring genes STAT5B or STAT3. F Pie chart depicting the altered expression profiles of genes involved in DNA methylation and histone modification in As^3+-transformed iPSCs compared to controls, including upregulated genes such as DNMT3B, DNMT3L, TET, GADD45G, EZH2, SETD4, and members of the KDM4 family. 3.9. High expression of cholesterol biosynthesis genes correlates with poorer survival in lung cancer patients While scattered studies have linked genetic polymorphisms in cholesterol biosynthesis genes to cancer risk [[181]46,[182]47], the prognostic value of these genes remains largely underexplored. To evaluate the clinical relevance of our findings, we assessed the association between cholesterol biosynthesis gene expression and first progression (FP) survival in a cohort of 982 lung cancer patients using the Kaplan-Meier Plotter database. Among 15 cholesterol biosynthesis genes examined, high expression of nine genes—including SREBF2, ACAT2, HMGCS1, MVK, MVD, FDFT1, SQLE, LSS, and DHCR7—was significantly associated with shorter FP survival ([183]Figure 9A). Conversely, elevated expressions of HMGCR, IDI1, FDPS, and DHCR24 correlated with improved FP survival. No survival correlation was observed for SC5DL. These findings suggest a general trend wherein upregulation of specific cholesterol biosynthesis genes may predispose to more aggressive tumor behavior, supporting the pro-oncogenic role of hypercholesterolemia. To further underscore this relationship, we evaluated the expression of three cytochrome P450 family members involved in cholesterol metabolism or catabolism: CYP51A1 that catalyzes conversion of lanosterol to cholesterol; CYP11A1, which catalyzes cholesterol side-chain cleavage potentially linked to the generation of methylmalonic acid (MMA) for stemness factor activation [[184]48]; and CYP27A1, which promotes cholesterol excretion via bile acid synthesis ([185]Figure 9B). High expressions of CYP51A1 and CYP11A1 were linked to worse FP survival, while elevated CYP27A1 expression predicted better survival outcomes ([186]Figure 9C). These results reinforce the notion that disrupted cholesterol homeostasis—especially increased synthesis and impaired clearance—may contribute to tumor progression and poor clinical prognosis. Fig. 9. [187]Fig. 9 [188]Open in a new tab Increased expression of cholesterol biosynthesis genes is associated with poorer prognosis of lung cancer. A First Progression (Progression Free, FP) survival of lung cancer patients (n = 2167; data from the Kaplan-Meier Plotter) stratified by expression levels of key cholesterol biosynthesis genes. The optimal cutoff was used to define high and low expression groups for survival comparison. B Schematic diagram showing contribution of the indicated CYP family members to cholesterol metabolism. C FP survival curves of lung cancer patients stratified by expression levels of the indicated CYP genes, based on Kaplan–Meier analysis. 4. Discussion The association between environmental As^3+ exposure and human cancers is well-established. A key mechanism underlying As^3+ carcinogenicity is its ability to induce cancer stem-like cell (CSC) formation, either by reprogramming normal stem cells or by generating CSCs de novo [[189]2,[190]49]. In this study, we used human iPSCs to investigate the impact of chronic As^3+ exposure on metabolic and epigenetic programs. Our findings suggest that the transcription factor Nrf2 may contribute to these reprogramming events, particularly through the modulation of glycolytic and cholesterol biosynthesis pathways. Building on previous work demonstrating that As^3+ activates Nrf2 and induces a metabolic shift from mitochondrial oxidative phosphorylation to glycolysis [[191]5], we observed robust upregulation of glycolytic genes in iPSCs following long-term As^3+ exposure. Genome-wide Nrf2 binding was enriched at the loci of these genes. Interestingly, whereas As^3+-transformed bronchial epithelial cells redirected glycolytic intermediates toward the hexosamine biosynthetic and serine/glycine synthesis pathways [[192]5], transformed iPSCs preferentially funneled these metabolites into the pentose phosphate pathway (PPP) and the hexosamine biosynthetic pathway (HBP). We also observed upregulation of genes involved in cholesterol biosynthesis, which aligns with our recent metabolomic data and further implicates cholesterol metabolism as a conserved response to chronic As^3+ stress [[193]33]. Epigenetic remodeling—particularly alterations in histone methylation—represents a hallmark of both stem cells and CSCs. While the exact influence of As^3+ on epigenetic regulation remains incompletely understood, our data reveal a surprising global reduction in H3K4me3 levels and concurrent increases in H3K27me3, H3K36me3, and H4K20me3 in iPSCs following As^3+ exposure. H3K4me3, typically associated with transcriptional activation, was still enriched at transcription start sites (TSS), albeit at lower levels. This reduction may be attributed to increased expression of DNA methyltransferases such as DNMT3B and DNMT3L, which were upregulated by As^3+. Interestingly, colocalization patterns revealed enrichment of H3K4me3, H3K27me3, H3K36me3, and Nrf2 within active chromatin regions for a number of genes, suggesting coordinated regulation of gene expression. These epigenetic markers showed minimal overlap with repressive marks such as H3K9me3 and H4K20me3, which were largely confined to heterochromatin or gene-poor regions. One of the most striking findings of this study is the robust activation of cholesterol biosynthesis in iPSCs following As^3+ exposure. This mirrors our previous observations in As^3+-transformed bronchial epithelial cells and aligns with emerging evidence linking environmental As^3+ exposure to elevated serum cholesterol levels in humans and cardiovascular pathology in animal models [[194]33,[195]50,[196]51]. While the direct contribution of cholesterol to As^3+-induced carcinogenesis remains uncertain, several studies suggest a role for cholesterol as a pro-oncogenic metabolite. For example, hypercholesterolemia has been correlated with increased cancer risk and progression [[197]52]. Statins, cholesterol-lowering drugs, have been shown to reduce recurrence and mortality in breast cancer patients [[198]53,[199]54]. In triple-negative breast cancer (TNBC), metastatic tumors exploit SREBP2-driven cholesterol synthesis to support angiogenesis [[200]55]. Estrogen receptor-positive (ER^+) breast cancers overexpress CYP27A1, converting cholesterol into 27-hydroxycholesterol (27HC), which mimics estrogen to promote tumor growth and metastasis [[201]56,[202]57]. 27HC also modulates immune evasion by recruiting immunosuppressive myeloid populations and depleting CD8^+ T cells [[203]57]. Notably, CYP27A1 was the most highly expressed cholesterol metabolism gene in As^3+-transformed iPSCs ([204]Figure 8B). Cholesterol also promotes survival in glioblastoma, and its depletion leads to apoptosis via liver X receptor (LXR) signaling [[205]58]. In diffuse large B-cell lymphoma (DLBCL), cholesterol enhances B-cell receptor (BCR) clustering and PI3K/AKT signaling [[206]59]. Moreover, cholesterol derived from tumor cells can induce CD8^+ T-cell exhaustion, facilitating immune evasion [[207]60,[208]61]. Whether iPSCs and CSCs actively produce cholesterol and how this impacts their self-renewal and differentiation remain open questions. Previous studies have highlighted a role for cholesterol in stem cell biology. For instance, Gu et al. demonstrated that hypercholesterolemia drives hematopoietic stem and progenitor cell (HSPC) expansion via SREBP2 [[209]62]. In zebrafish, cholesterol efflux activates endothelial SREBP2 and Notch signaling to promote HSPC emergence during development. Similarly in mammalian systems, cholesterol boosts intestinal stem cell (ISC) proliferation, as shown in mouse models where high-cholesterol diets increase ISC numbers and promote tumorigenesis [[210]63]. Meta-analyses have further linked hypercholesterolemia with clonal hematopoiesis and an increased risk of hematologic malignancy [[211]64]. In our study, both As^3+-induced CSCs and iPSCs exhibited enhanced Nrf2 binding to key cholesterol metabolic genes [[212]5], along with a marked upregulation of cholesterol metabolism, suggesting that cholesterol serves as a critical metabolic node supporting stemness. While the full extent of Nrf2's role in regulating cholesterol metabolism remains to be fully recognized currently, previous studies in LDLR^-/- mice implicate Nrf2 in cholesterol-driven pathologies. In the context of atherosclerosis, a disease characterized by excessive cholesterol accumulation in arterial walls, Nrf2 deficiency led to a reduction in atherosclerotic lesions [[213]65]. Overall, our findings identify a novel mechanism by which As^3+ enhances oncogenic stemness through Nrf2-mediated metabolic reprogramming. This rewiring features heightened PPP and HBP activity, supplying key metabolites for nucleotide biosynthesis, glycosylation, and methylation. For example, S-adenosylmethionine (SAM), derived from the methionine cycle linked to PPP, serves as the universal methyl donor for DNA and histone methylation. UDP-GlcNAc, produced via HBP, supports glycosylation of stemness-related transcription factors such as OCT4, SOX2, and MYC, as well as epigenetic regulators like TET enzymes and H3K4 methyltransferase complexes [[214][66], [215][67], [216][68]]. SCAP glycosylation is also essential for SREBP activation, enabling Nrf2-SREBP synergy in promoting cholesterol biosynthesis [[217]40]. In the nucleus, SREBP acts in concert with Nrf2 and other transcription factors to drive the transcription of genes involved in cholesterol biosynthesis. Once synthesized, cholesterol integrates into both the plasma membrane and endoplasmic reticulum (ER) membrane, where it facilitates the assembly and activation of membrane-associated growth signaling pathways—including B-cell receptor (BCR), Notch, and Hedgehog (Hh)—which are critical for promoting metabolic and epigenetic reprogramming, as well as the proliferation of stem cells and CSCs [[218]69]. Notably, cholesterol metabolites can directly modulate epigenetic machinery. For instance, the nuclear metabolite 25-hydroxycholesterol (25HC) has been shown to act as a direct activator of DNMT1, enhancing DNA methylation [[219]70]. In contrast, sulfated cholesterol derivatives such as 25HC3-sulfate (25HC3S) and 27HC3S function as potent inhibitors of DNMT1, DNMT3A, and DNMT3B [[220]71]. Moreover, an earlier study demonstrated that cholesterol could upregulate DNMT1 expression and promote the DNA-binding activity of methyl-CpG binding protein 2 (MeCP2) and the histone methyltransferase EZH2 [[221]72]. While our data support a potential role for Nrf2 in orchestrating these interconnected pathways, we cannot rule out the contribution of other regulatory mechanisms beyond Nrf2 for the As^3+-induced metabolic reprogramming. Moreover, neither Nrf2 nor As^3+ can be equalized to an independent reprogramming factor capable of inducing pluripotency in somatic cells. Instead, our findings reveal a previously unrecognized mechanism involved in As^3+-induced carcinogenesis, consistent with prior reports that Nrf2-dependent metabolic shift improves the efficiency of iPSC reprogramming from human dermal fibroblast cells [[222]73]. Indeed, the supportive role of Nrf2 to the self-renewal, pluripotency, and survival has been widely reported across various stem cells and progenitor cell types, including human embryonic stem cells, mouse hematopoietic stem cells, human mesenchymal stem cells, human endothelial progenitor cells, and mouse neural stem cells, among others [[223]74]. Although complete disruption of the Nrf2 gene NFE2L2 through CRISPR-Cas9 gene editing in the iPSCs used in this study may pose challenges due to severe cell death, which precludes further analysis of As^3+- or other carcinogen-induced metabolic reprogramming and malignant transformation, it is imperative to pursue alternative loss-of-function approaches to clarify the specific contributions of Nrf2 and its downstream effectors in this process. Nonetheless, our findings suggest that As^3+ exposure, at least in part through Nrf2-associated metabolic rewiring, engages a complex network of interrelated pathways, including glycolysis, nucleotide biosynthesis, protein glycosylation, cholesterol metabolism, and epigenetic regulation, that collectively promote cancer development and the emergence of CSCs ([224]Figure 10). Fig. 10. [225]Fig. 10 [226]Open in a new tab Schematic representation of potential mechanisms underlying Nrf2-mediated metabolic rewiring in response to As^3+exposurein iPSCs. As^3+ induces Nrf2-dependent metabolic reprogramming that coordinates a network of interconnected pathways—including enhanced glycolysis, nucleotide biosynthesis, protein glycosylation, cholesterol metabolism, epigenetic modifications, and weakened mitochondrial citric acid cycle (TCA or Krebs cycle). Together, these alterations contribute to cancer development and promote the acquisition of CSC properties. CRediT authorship contribution statement Akimasa Seno: Methodology, Investigation, Data curation. Zhuoyue Bi: Methodology, Investigation, Formal analysis. Lisa Polin: Methodology, Investigation. Ziqi Liu: Validation, Software, Resources. Yiran Qiu: Methodology, Investigation, Data curation. Wenxuan Zhang: Validation, Methodology, Data curation. Aashna Pawar: Software, Investigation. Chitra Thakur: Resources, Investigation, Data curation. Masaharu Seno: Resources. Ziwei Wang: Writing – original draft, Visualization, Validation, Methodology, Investigation. Fei Chen: Writing – review & editing, Supervision, Funding acquisition, Conceptualization. Declaration of competing interest The authors declare no competing financial interests or personal relationships that could have appeared to influence the work reported in this paper. Acknowledgements