Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2025 Oct;44(40):3774-3786.
doi: 10.1038/s41388-025-03530-w. Epub 2025 Aug 8.

ALDH1A3 promotes aggressive basal-like pancreatic cancer through an AP-1/RUNX2 enhancer network

Affiliations

ALDH1A3 promotes aggressive basal-like pancreatic cancer through an AP-1/RUNX2 enhancer network

Xiaoping Zou et al. Oncogene. 2025 Oct.

Abstract

The basal-like transcriptional subtype of pancreatic ductal adenocarcinoma (PDAC) is linked to therapy resistance and poor prognosis. The cancer stem cell marker aldehyde dehydrogenase 1A3 (ALDH1A3) is a critical enzyme in acetaldehyde metabolism, but the interconnection to the basal-like subtype is poorly understood. Here, we identified ALDH1A3 as a key gene, which correlates with reduced survival and increased tumor growth. Functional studies revealed interaction of ALDH1A3 with genes like FAM3C, MCC, PMEPA1, and IRS2, forming a network driving PDAC progression. Chromatin profiling showed that ALDH1A3 affects acetylation of histone 3, mediating AP-1 activity, particularly via FOS family members, activating oncogenic pathways such as MAPK and TNF signaling. RUNX2 emerged as a therapeutic target within this network, as its knockdown disrupted MAPK signaling and reduced tumor growth. These findings emphasize the role of ALDH1A3 in linking nuclear metabolic-epigenetic programming in basal-like PDAC, highlighting it as a promising therapeutic target for novel treatment strategies.

PubMed Disclaimer

Conflict of interest statement

Competing interests: The authors declare no competing interests. Ethics: Clinical samples used in this study were approved by the Ethics Committee of the Technical University Munich (approval number: 80/17S) and the Ethics Committee of the Affiliated Drum Tower Hospital of Nanjing University (approval number: 2019-015-01). All mouse experiments and procedures were approved by the Institutional Animal Care and Use Committee of the Technical University of Munich (approval numbers: 55.2-1-54-2532-197-2016, ROB-55.2-2532.Vet_02-17-83), which address the ARRIVE guidelines. All procedures involving transgenic mice were performed in accordance with the Office of Laboratory Animal Welfare and German Federal Animal Protection Laws. BALB/c nu/nu athymic mice were approved by the Animal Care and Use Committee of the Nanjing Drum Tower Hospital (approval number: 20180102). All methods were performed in accordance with the relevant guidelines and regulations.

Figures

Fig. 1
Fig. 1. Enriched ALDH1A3-network signature in aggressive PDAC subtypes.
A Venn diagram showing differentially expressed genes between ALDH1A3-positive and -negative samples in patient-derived xenografts (PDXs), cell lines, and tissues. B Spearman´s correlation matrix of 8 genes. ***p < 0.001, **p < 0.01, *p < 0.05. C Interaction network analysis depicting relationships among the genes: ALDH1A3 (A), EMP1 (E), FAM3C (F), IRS2 (I), MAML2 (MA), MCC (MC), PMEPA1 (P), and SP100 (S). D Subcutaneous xenotransplantation of parental AsPC-1 cells (NC) to immunodeficient mice or genetically manipulated AsPC-1 cells with knockdown (KD) of ALDH13 (A), FAM3C (F), MCC (MC), or PMEPA1 (P), demonstrating effects on tumor 1size and weight. Left: images of tumor xenografts; Right: mean weights and standard deviations. Control (NC, n = 18), ALDH1A3KD (n = 5), FAM3CKD (n = 7), MCCKD (n = 11), PMEPA1KD (n = 7); p values via unpaired Student’s t test. E Metastatic lung colonization following tail vein injection in mice; NC (n = 20), ALDH1A3KD (n = 22), FAM3CKD (n = 13), MCCKD (n = 12), PMEPA1KD (n = 15); p values by unpaired t test. F Pie chart detailing weight parameters. A (ALDH1A3: 23.08%), F (FAM3C: 19.23%), E (EMP1:15.38%), P (PMEPA1: 11.54%), MC (MCC: 11.54%), I (IRS2: 7.69%), MA (MAML2: 7.69%), S (SP100: 3.85%). G Survival analysis for ALDH1A3High versus ALDH1A3Low patients in TCGA and Compass (stage I-III) datasets. H Heatmap demonstrating the ALDH1A3 network score and molecular subtype in TCGA and Compass datasets; High-risk PDAC subtypes are highlighted in purple. I Proportion of aggressive subtypes versus others in the ALDH1A3High and ALDH1A3Low groups from TCGA and Compass datasets. J ALDH1A3 network score in TCGA and Compass datasets, divided by aggressive subtype. p values via unpaired t-tests.
Fig. 2
Fig. 2. ALDH1A3 regulates AP-1 activity through the FOS family.
A Screening results for AP-1 binding sites in the promoters of candidate genes. B Heatmap illustrating Pearson correlation coefficients between candidate genes and AP-1 subunits from the FOS and JUN families. ***p < 0.001, **p < 0.01, *p < 0.05. C Western blot analysis showing protein levels before (NC) and after ALDH1A3 knockdown (AKD#1; AKD#2) in HPAC and AsPC-1 cells. Key proteins detected include JNK activity markers (p-JNKT183/Y185; p-c-JUNS73) as well as expression of FOS subunits (FOSB, FOSL1, FOSL2). This panel shows one representative experiment out of three conducted. D TF enrichment analysis in PANC-1 and AsPC-1 cells affected by altered ALDH1A3 expression. Analysis based on RNA-seq data from three biological replicates. E Contingency table analysis for co-expression of ALDH1A3 and FOSL2 in PDAC sections; statistical significance assessed by Chi-square (χ2) test). IHC images displaying FOSL2/ALDH1A3 staining in PDAC sections, scale bars represent 50 μm. F Results from AP-1 luciferase reporter assays in cells subjected to dual or triple knockdown of FOS subunits. Data are presented as mean values from three independent experiments: p values calculated via unpaired t test. G RT-qPCR (left) and western blot analysis (right) performed on AsPC-1 cells before and after knockdown of multiple FOS subunits, examining the expression of the previously described 8 candidate genes. One representative result from three independent experiments is displayed.
Fig. 3
Fig. 3. Aldh1a3 is crucial for Jnk/AP-1 activation and pancreatic carcinogenesis in vivo.
A Scheme of treatment of KC mice with cerulein. Western blot analysis of pancreata from KC mice 0, 3, 96 h (h) and 14 days (d) after cerulein treatment and detection of Jnk activity (p-JnkT183/Y185, p-c-JunS73), expression of Fos subunits (Fosb, Fosl1, Fosl2), and Aldh1a3 ; n = 3/group. B IHC displaying Aldh1a3 expression in KC pancreata 14 days after cerulein treatment. Immunofluorescence revealing Aldh1a3/α-amylase and Aldh1a3/Krt19 co-staining; scale bar: 50 μm; n = 3. C KC mice and KC; Aldh1a3-/- underwent cerulein treatment and subsequent analysis of protein expression in their pancreas by western blot, as described above. n = 3. D Pancreas proteins analyzed by western blot post-cerulein treatment, n = 5. E H&E- and α-SMA-stained sections from KCERT; Aldh1a3OE or KCERT control mice 14 days post-treatment; scale bars: 50 μm, n = 5/group. F Differential gene expression analyzed in RNA-seq between KCERT; Aldh1a3OE (n = 3) and KCERT pancreata, (n = 4). Among 987 upregulated genes the top ten enriched TFs were shown. Among 1, 610 downregulated genes five enriched TFs were shown. (G) H&E-stained pancreas sections depicting KC and KC; Aldh1a3–/– pancreata at 30 weeks. IHC showing Krt19 and α-amylase-positive cells; scale bars: 50 μm, n = 3 (KC), n = 4 (KC; Aldh1a3–/–). p values by unpaired t test. H IHC results of H&E, Krt19, or α-amylase-stained pancreas sections from one-year-old KC and KC; Aldh1a3–/– mice. Scale bars: 50 μm, n = 5/genotype.
Fig. 4
Fig. 4. ALDH1A3 promotes an oncogenic, basal-like specific transcriptional program by regulating AP-1-mediated enhancer activity.
A Heatmap showing differential ATAC-seq results that identify ALDH1A3-associated accessible or inaccessible chromatin regions in PANC-1 and AsPC-1 cells. B Identification of the top five TF motifs found at ALDH1A3-accessible or -inaccessible chromatin sites in AsPC-1 and PANC-1 cells, analyzed with n = 2 biological replicates. C Pie charts illustrating the genomic distribution of ATAC-seq peaks associated with ALDH1A3, highlighting enrichment of intergenic and intronic sites within open peaks, p = 4.0×10-6, chi-squared test. D KEGG pathway analysis of genes corresponding to ALDH1A3 open peaks derived from ATAC-seq data. E Quality control analysis of H3K4me1 and H3K27ac CUT&Tag profiles in PANC-1 and AsPC-1 cells, detailing the distribution of all peaks relative to transcription start sites (TSS) in base pairs (bp), with n = 2 biological replicates. F Identification of the top five enriched TF motifs in H3K27ac CUT&Tag data comparing ALDH1A3OE overexpressing and control PANC-1 cells, as well as ALDH1A3KD knockdown versus control AsPC-1 cells, with n = 2 biological replicates. G Visualization of overlapping data showing upregulated genes alongside H3K27ac-associated peaks in specific cellular comparisons; accompanied by KEGG pathway analysis, with n = 2 biological replicates in each group. H GSVA displaying expression profiles of basal-like and classical markers in 22 patient-derived xenografs (PDXs) with high (ALDH1A3High) and low (ALDH1A3Low) ALDH1A3 expression. I H3K27ac ChIP-seq intensity profiles for basal-like and classical markers across ALDH1A3High and ALDH1A3Low PDXs groups. J Top five enriched TF motifs identified in H3K27ac CUT&Tag profiles comparing ALDH1A3High versus ALDH1A3Low PDXs groups. K Pie chart showing RNA-seq-based upregulated gene overlap with ALDH1A3-associated H3K27ac peaks in ChIP-seq data between ALDH1A3High versus ALDH1A3Low groups, including KEGG pathway analysis.
Fig. 5
Fig. 5. Convergence of the AP-1-dependent enhancer network on the oncogenic MAPK pathway in ALDH1A3High PDAC.
A Quality control (QC) analysis of FOSL2 CUT&Tag in PANC-1 and AsPC-1 cells, illustrating peak distances from the transcription start side (TSS) in base pairs (bp). Data shown for n = 2 biological replicates. B Overlapping pie charts depicting ALDH1A3-associated histone modifications (H3K4me1, H3K27ac) and FOSL2 peaks in PANC-1 cells with ALDH1A3 overexpression (ALDH1A3OE) versus control, and AsPC-1cells with ALDH1A3 knockdown (ALDH1A3KD) versus control. Analysis conducted with n = 2 biological replicates per group. C Top five enriched TF motifs identified in FOSL2 CUT&Tag analysis with open or closed chromatin states in PANC-1/ALDH1A3OE versus PANC-1/control cells and AsPC-1/ control versus AsPC-1/ALDH1A3KD cells. Performed with n = 2 biological replicates. D Overlapping charts illustrating upregulated genes co-localized with ALDH1A3-associated FOSL2 peaks across different cell line comparisons including KEGG pathway analysis of these genes. Performed with n = 2 biological replicates. E Heat map showing significantly upregulated genes and their association with FOSL2-binding sites linked to the MAPK pathway in PANC-1/ALDH1A3OE versus PANC-1/control cells, and AsPC-1/ control cells versus AsPC-1/ALDH1A3KD cells. IGV tracks showed open chromatin peaks of MAP2K3 and EREG in PANC-1/ALDH1A3OE versus PANC-1/control cells, and NFκB1 and EGFR in AsPC-1/control versus AsPC-1/ALDH1A3KD cells.
Fig. 6
Fig. 6. RUNX2 as druggable target of ALDH1A3High PDAC.
A Integrated analysis of RNA-seq up, ATAC-seq open, and FOSL2 peaks open under ALDH1A3 regulation in PANC-1 and AsPC-1 cells, identifying intersections with RUNX2 and CD55 as shared candidate targets. B ATAC-seq data revealing open chromatin sites at the RUNX2 locus in PANC-1 cells overexpressing ALDH1A3 (ALDH1A3OE) compared to controls. C RUNX2 mRNA expression levels in ALDH1A3High versus ALDH1A3Low groups from TCGA and Compass datasets. D Western blot analysis showing RUNX2 protein levels in KC; Aldh1a3–/– pancreata against KC controls, conducted with n = 3 per genotype. E Western blot indicating RUNX2 levels post-knockdown of dual or triple FOS subunits in AsPC-1 cells; representative of three similar experiments. F Quality control (QC) analysis of RUNX2 CUT&Tag in HPAC cells, showing all peak distances from the transcription start side (TSS) in base pairs (bp). Data from n = 2 biological replicates. G Top five enriched TF motifs in RUNX2-binding open chromatin sites in HPAC cells, identified from the RUNX2 CUT&Tag experiment with = 2 biological replicates. H Western-blot analysis demonstrating RUNX2 expression in HPAC cells transduced with negative controls (NC) or RUNX2-specific shRNAs; one of three independent experiments is shown. I Overlapping charts displaying upregulated genes in HPAC/NC versus HPAC/RUNX2KD and RUNX2-binding genes in HPAC cells, accompanied by KEGG pathway analysis of overlapping genes. Performed with n = 2 biological replicates. J Western-blot analysis illustrating activation levels of oncogenic MAPK pathways (p-ERKT202/Y204, p-c-JUNS73, p-p38T180/Y182 and p-JNKT183/Y185) and expression of RUNX2 in HPAC/RUNX2KD and control cells treated with FBS for 1 h or irradiated with UV for 30 min, representative of three independent experiments with similar outcome. K Xenograft model of HPAC cells demonstrating the effect of RUNX2 knockdown on tumor growth, with n = 4, p values by unpaired Student’s t test. L Tumor growth curves, treated with CADD522 (a RUNX2 inhibitor, n = 4) or control (n = 4), in a subcutaneous tumor model generated by HPAC cells; p values by unpaired Student’s t test.

References

    1. Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA Cancer J Clin. 2023;73:17–48. 10.3322/caac.21763. - PubMed
    1. Collisson EA, Sadanandam A, Olson P, Gibb WJ, Truitt M, Gu S, et al. Subtypes of pancreatic ductal adenocarcinoma and their differing responses to therapy. Nat Med. 2011;17:500–3. 10.1038/nm.2344. - PMC - PubMed
    1. Moffitt RA, Marayati R, Flate EL, Volmar KE, Loeza SG, Hoadley KA, et al. Virtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinoma. Nat Genet. 2015;47:1168–78. 10.1038/ng.3398. - PMC - PubMed
    1. Bailey P, Chang DK, Nones K, Johns AL, Patch AM, Gingras MC, et al. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature. 2016;531:47–52. 10.1038/nature16965. - PubMed
    1. Chan-Seng-Yue M, Kim JC, Wilson GW, Ng K, Figueroa EF, O’Kane GM, et al. Transcription phenotypes of pancreatic cancer are driven by genomic events during tumor evolution. Nat Genet. 2020;52:231–40. 10.1038/s41588-019-0566-9. - PubMed

MeSH terms

Substances

LinkOut - more resources