Published in last 50 years
Related Topics
Articles published on Expression In Stromal Cells
- New
- Research Article
- 10.1016/j.canlet.2025.218101
- Oct 30, 2025
- Cancer letters
- Saugato Rahman Dhruba + 11 more
Enhanced prediction of breast cancer patient response to chemotherapy by integrating deconvolved expression patterns of immune, stromal and tumor cells.
- New
- Research Article
- 10.1177/10935266251376089
- Oct 28, 2025
- Pediatric and developmental pathology : the official journal of the Society for Pediatric Pathology and the Paediatric Pathology Society
- Weiming Yu + 2 more
Nevus lipomatosus superficialis (NLS) is an uncommon benign skin lesion and characterized by aggregates of mature adipose tissue ectopically among the collagen bundles of the dermis. The precise etiology of NLS remains poorly understood, and it is classified as a cutaneous hamartoma in the WHO Classification of Skin Tumors, 5th edition. This is a case report of recurrent classical Hoffmann-Zurhelle form of NLS on a 3-year-old male child's hand. Immunohistochemical stains revealed diffusely PLAG1 positive expression in the lesional stellate stromal cells and adipocytes. Ancillary molecular testing by RNA fusion panel showed COL3A1::PLAG1 rearrangement. These findings suggest that some cases of NLS may share genetic alterations with neoplastic processes involving PLAG1 rearrangements.
- New
- Research Article
- 10.1007/s00428-025-04302-3
- Oct 20, 2025
- Virchows Archiv : an international journal of pathology
- Sohei Kitazawa + 11 more
Foveolar-type gastric adenoma with raspberry-like appearance (FGA-RA) is a rare subtype of Helicobacter pylori-naïve gastric neoplasm characterized by its small, reddish, polypoid morphology. This study aimed to clarify the molecular basis of its epithelial phenotype and the superficial capillary proliferation characteristic of FGA-RA's reddish appearance by examining alterations in mucin gene expression and angiogenesis-related pathways using spatial transcriptomic profiling and immunohistochemistry. Formalin-fixed, paraffin-embedded tissue from a representative case of FGA-RA was analyzed using the Visium spatial transcriptomics platform (10 × Genomics). Spatial gene mapping revealed diffuse MUC5AC expression throughout the tumor epithelium, consistent with a foveolar phenotype. Notably, MUC17-typically restricted to intestinal enterocytes-was aberrantly coexpressed with MUC5AC in the superficial tumor layer, while MUC2 was absent, suggesting skewed differentiation toward absorptive enterocytes without goblet cell lineage. To validate these findings, immunohistochemical staining for MUC5AC and MUC17 was performed on six biopsy specimens from genetically validated FGA-RA cases harboring the characteristic KLF4 mutation, five of which showed overlapping superficial expression of both MUC5AC and MUC17. Spatial transcriptomic analysis also demonstrated elevated VEGFA expression in tumor and inflammatory cells, and FLT1 expression in superficial stromal endothelial cells. This expression pattern correlated with vascular markers and may contribute to the prominent capillary proliferation seen in FGA-RA. FGA-RA exhibits a distinctive mucin profile reflecting incomplete intestinal differentiation due to KLF4 dysfunction. The characteristic raspberry-like morphology may be a consequence of VEGF-driven angiogenesis induced by chronic stimulation of a superficially compromised epithelium, suggesting that this unique appearance arises from sequential epithelial and stromal alterations orchestrated by KLF4 mutation.
- Research Article
- 10.1097/md.0000000000044921
- Oct 10, 2025
- Medicine
- Jianlei Wu + 5 more
T-cell immunoglobulin and mucin domain-containing molecule 3 (TIM-3) has been reported to be overexpressed and associated with poor prognosis in solid tumors. However, its features and prognostic value in epithelial ovarian cancer (EOC) remain undetermined. In this study, we aimed to characterize TIM-3 expression and its prognostic significance in patients with EOC. A total of 134 EOC patients and 20 healthy controls from North China were included. TIM-3 mRNA and protein expression in EOC tumor tissues and benign ovarian tissues were detected by real-time quantitative PCR and immunohistochemistry. The distribution of TIM-3 protein in different regions of EOC tissue (tumor cells and the tumor microenvironment) were evaluated by multicolor immunofluorescence. Associations between their expression and clinicopathological parameters as well as survival analyses were performed. The results showed that high expression levels of TIM-3 mRNA were significantly associated with shorter progression-free survival (PFS; P < .001, hazard ratio [HR] = 1.57, 95% confidence interval [CI] = 1.29–1.91) and overall survival (OS; P = .013, HR = 1.31, 95% CI = 1.06–1.63) durations in EOC patients. High TIM-3 expression levels in tumor cells had shorter PFS (HR = 1.62, 95% CI = 1.09–2.46, P = .018) and OS (HR = 1.81, 95% CI = 1.19–2.75, P = .006) compared with those low TIM-3 expression levels. Similarly, TIM-3 in the tumor microenvironment was also an independent factor that affected the clinical outcome of EOC patients (PFS: HR = 1.99, 95% CI = 1.29–3.08, P = .002; OS: HR = 2.13, 95% CI = 1.37–.30, P = .001). These findings indicated that IM-3 may be a potential biomarker for predicting prognosis and immunotherapy efficacy in patients with EOC, exerting different roles on tumor cells and tumor microenvironment.
- Research Article
- 10.1097/01.tp.0001170876.59958.b4
- Oct 1, 2025
- Transplantation
- Kohei Tatsumi + 4 more
P.54: Effect of cell culture medium on the tissue factor expression and prothrombotic potential of mesenchymal stromal cell.
- Research Article
- 10.1093/humrep/deaf174
- Sep 26, 2025
- Human reproduction (Oxford, England)
- Mingjuan Zhou + 6 more
How does allograft inflammatory factor-1 (AIF-1) affect endometrial receptivity in women with recurrent implantation failure (RIF)? Significant upregulation of AIF-1 in the endometrial stromal cells of women with RIF inhibits cell proliferation and decidualization via the p38 mitogen-activated protein kinase (MAPK) phosphorylation, thereby reducing endometrial receptivity. RIF is a challenging clinical issue, with AIF-1, a cytokine-inducible protein linked to allograft rejection, potentially contributing to its pathogenesis. However, the precise mechanisms remain elusive. This study analyzed endometrial tissue samples from women diagnosed with RIF and a control group of fertile women from December 2018 to December 2023. Single-cell RNA sequencing (scRNA-seq) data from public datasets (GSE111976, GSE250130, GSE183837) were integrated to characterize AIF-1 expression patterns in endometrium. Isolated human endometrial stromal cells (HESCs) from the human endometrium and an endometrial stromal cell line were used for in vitro analysis, and an in vivo mouse model with AIF-1 overexpression in the uterus was employed to evaluate implantation outcomes. Mid-secretory endometrial samples were collected from the 18 patients with RIF and 18 control patients; endometrial samples from another five different phases during the menstrual cycle were collected from 30 additional control patients. Quantitative PCR, western blot, immunohistochemical and immunofluorescence analyses, and RNA sequencing were conducted to determine the expression levels of AIF-1 and various markers. Cell proliferation, decidualization, and trophoblast outgrowth were measured. AIF-1 overexpression and gene silencing were achieved by plasmid injection and short hairpin RNA, respectively. For in vivo experiments, CD-1 mice with intrauterine injection of an AIF-1 plasmid were used. Phosphorylation of p38 was inhibited by PD169316. Based on scRNA-seq analysis and our own endometrial tissue detection, AIF-1 was significantly increased in HESCs in patients with RIF compared with their control group during the mid-secretory phase. AIF-1 overexpression resulted in reduced cell proliferation, inadequate cell decidualization, and diminished embryo outgrowth in in vitro experiments, and it reduced the number of embryo implantation sites in CD-1 mice; these effects were mitigated by PD169316, an inhibitor of p38 MAPK. Although the study establishes a link between increased AIF-1 expression in endometrial stromal cells and reduced endometrial receptivity, the role of AIF-1 in endometrial macrophages during embryo implantation remains unclear. The findings suggest that targeting the AIF-1 and p38 MAPK pathway could serve as a promising therapeutic strategy to improve endometrial receptivity in RIF patients. This study was supported by the National Key R&D Program of China (No. 2022YFC2703800) and the National Natural Science Foundation of China [Nos. 82271703; 82371704; 82071596; 82071712; 82101800; 81701513]. The authors declare that they have no conflict of interests with the contents of this manuscript. N/A.
- Research Article
- 10.3390/cancers17172905
- Sep 4, 2025
- Cancers
- Parisa Ghahremanifard + 11 more
The SUMO E3 ligase PIAS1 (Protein Inhibitor of Activated STAT1) regulates pathways such as TGFβ signaling and has been implicated in multiple cancers. However, its role in the tumor microenvironment (TME), particularly in non-malignant stromal and immune cells, remains poorly understood. This study aimed to characterize the expression and functional relevance of PIAS1 within the TME of oral squamous cell carcinoma (OSCC). PIAS1 protein expression was assessed via immunohistochemistry (IHC) on OSCC tissue microarrays. Single-cell RNA-sequencing (scRNA-seq) datasets from OSCC tumors and normal tissues were analyzed to map cell-type-specific PIAS1 expression. Downstream effects were evaluated using differential gene expression, Ingenuity Pathway Analysis (IPA), gene set enrichment analysis (GSEA), and cell-cell communication inference. IHC analysis revealed that higher stromal PIAS1 levels correlated with improved survival. scRNA-seq analysis showed an increase in the proportion of PIAS1-expressing cells across most stromal and immune cell populations within OSCC-derived tumors compared to their counterparts in adjacent normal tissue. However, when comparing PIAS1-positive cells, expression levels were significantly reduced in cancer cells, CAFs, TAMs, T cells, and endothelial cells within the TME. PIAS1-positive CAFs, TAMs, and T cells exhibited activation of apoptotic and tumor-suppressive pathways, while PIAS1-negative counterparts showed enrichment of immunosuppressive signaling and immune checkpoint expression. Cell-cell communication analyses indicated that PIAS1 fosters an immune-activated TME by promoting pro-inflammatory signaling, M1-like TAM polarization, and T cell activation. PIAS1 expression in stromal and immune cells is associated with tumor-suppressive reprogramming of the OSCC microenvironment. These findings position PIAS1 as a potential modulator of anti-tumor immunity and candidate target for therapeutic intervention.
- Research Article
- 10.1016/j.labinv.2025.104193
- Sep 1, 2025
- Laboratory investigation; a journal of technical methods and pathology
- Shu-Ichiro Sasaki + 7 more
Lacking TRPA1 Cation Channel Impairs Primary Closure of a Stromal Incision Injury in a Mouse Cornea.
- Research Article
- 10.1016/j.placenta.2025.07.085
- Sep 1, 2025
- Placenta
- Yusmaris Cariaco + 6 more
Ferroptosis as a mechanism of placenta dysfunction in inflammation-driven preeclampsia.
- Research Article
- 10.1101/2025.07.29.667434
- Aug 1, 2025
- bioRxiv : the preprint server for biology
- Claire E Tocheny + 14 more
Oligonucleotide therapeutics are a new class of drugs that enable robust and sustained modulation of gene expression. However, achieving efficient delivery of siRNAs to tumors is a challenge for therapy. Here, we demonstrate that fully chemically modified siRNAs conjugated with an albumin-binding dendrimer are efficiently delivered to both neoplastic and stromal/immune cells within primary TNBC mammary tumors. siRNAs were designed to selectively target IRS2, a signaling adaptor of insulin and insulin-like growth factor signaling that has been implicated in aggressive breast cancers. These siRNAs reduced Irs2 expression in tumor and stromal cells without causing hyperglycemia, resulting in reduced tumor growth that was associated with decreased vascularization and alterations in macrophage polarization and the expression of EMT proteins. This work demonstrates that siRNAs can be delivered to neoplastic and specific stromal populations in mammary tumors and that they can effectively and specifically silence a driver of aggressive breast cancer.
- Research Article
- 10.1016/j.jcyt.2025.04.066
- Aug 1, 2025
- Cytotherapy
- Tiffany C Heard + 7 more
Cold storage effects on mitochondrial bioenergetics and protein expression in human mesenchymal stromal cells.
- Research Article
- 10.7150/jca.110597
- Jul 24, 2025
- Journal of Cancer
- Yuzhe Hu + 6 more
Head and neck squamous cell carcinoma (HNSCC) ranks among the most prevalent malignancies with a poor prognosis. The underlying mechanisms driving HNSCC carcinogenesis are not fully elucidated. In this study, we identified dual specificity phosphatase 9 (DUSP9) as a carcinogenic factor in HNSCC development. According to the public data, DUSP9 was significantly up-regulated in HNSCC tumor tissues compared to normal tissues, confirmed by clinical data and single-cell RNA sequencing (scRNA-seq) data. Survival analysis revealed that high levels of DUSP9 expression contribute to poor prognosis in HNSCC patients. Knockdown of DUSP9 decreased, but overexpression of DUSP9 increased the proliferation and migration of HNSCC cells. ScRNA-seq data analysis suggested that DUSP9 was selectively expressed in tumor cells, with negligible expression in immune cells and stromal cells, and showed an elevated trend from primary tissues to metastatic tissues. Enrichment analyses of DUSP9-correlated genes suggested the involvement of DUSP9 in cell adhesion, wound healing, cell migration, transcription regulation and metabolic process. Furthermore, DUSP9 expression in tumor tissues exhibited an inverse relationship with immune cell infiltration within the tumor microenvironment (TME). In conclusion, this study provided evidence that DUSP9 was up-regulated in HNSCC tissues and may play a pivotal role in HNSCC progression, suggesting its potential as a novel biomarker.
- Research Article
- 10.1007/s10815-025-03596-1
- Jul 24, 2025
- Journal of assisted reproduction and genetics
- Jie Sheng + 7 more
Endometriosis (EM) and recurrent spontaneous abortion (RSA) exhibit clinical associations, yet their shared molecular mechanisms remain unclear. This study aimed to identify shared molecular mechanisms and potential hub genes underlying EM and RSA. Differentially expressed genes (DEGs) were identified from EM (GSE7305) and RSA (GSE165004) datasets. Functional enrichment and weighted gene co-expression network analysis (WGCNA) revealed shared pathways and key modules. Venny software was used to identify hub genes between DEGs and key module genes. The diagnostic value of FXYD1 was assessed via ROC analysis. Regulatory networks and immune cell infiltration were explored. Pan-cancer analysis was conducted to assess FXYD1's expression profile across tumor types. Single-cell RNA sequencing validated FXYD1 expression in EM tissues, maternal-fetal interface and RSA samples. DEGs in EM and RSA were enriched in pathways associated with abnormal proliferation, immune dysfunction, and developmental regulation. FXYD1 was identified as a shared hub gene, upregulated in both conditions, with potential diagnostic value. It was correlated with immune cell populations, particularly natural killer (NK) cells. Pan-cancer analysis revealed widespread FXYD1 downregulation across multiple cancer types. Single-cell RNA sequencing confirmed elevated FXYD1 expression in stromal and decidual cells of RSA tissues, implicating its role in impaired decidualization. FXYD1 emerges as a critical molecular link between EM and RSA, potentially contributing to decidualization dysfunction. Its dysregulation may underlie the shared pathophysiology of these conditions, offering new insights into their molecular mechanisms.
- Research Article
- 10.1038/s41392-025-02308-6
- Jul 21, 2025
- Signal Transduction and Targeted Therapy
- Jie Qin + 19 more
The treatment of hepatocellular carcinoma (HCC) faces challenges of low response rates to targeted drugs and immune checkpoint inhibitors, which are influenced by complicated microenvironment of HCC. In this study, the complex tumor microenvironment was identified by using tissue microarray (TMA), spatial transcriptomes and single-cell sequencing. High expression of CC chemokine receptor 7 (CCR7) in tumor cells predicted lower Overall Survival (OS). Conversely, CRISPR-Cas9-mediated knockout of CCR7 enhanced the sensitivity of HCC to sorafenib in preclinical experiments, resulting from the inhibition of epithelial-mesenchymal transition through the AKT and ERK signaling pathways. Simultaneously, we revealed CCR7 expression in stromal cells, with increased infiltration of CCR7+ immune cells into the tumor mesenchyme associated with high CCL21 expression at tumor sites. Subsequently, VEGF-C was identified as an independent predictor of higher patient OS and showed a significant positive correlation with CCR7 signaling. Interestingly, exogenous VEGF-C was found to promote the formation of tertiary lymphoid structures (TLSs) by activating lymphatic angiogenesis and the CCL21/CCR7 axis. As a result, VEGF-C treatment enhanced the efficacy of anti-PD-1 immunotherapy. This study highlights the opposing effects of tumor cell-derived versus stromal cell-derived CCR7 expression and guides the precision treatment for HCC.
- Research Article
1
- 10.1101/2024.06.14.598770
- Jul 10, 2025
- bioRxiv
- Saugato Rahman Dhruba + 11 more
ABSTRACTThe tumor microenvironment (TME) is a complex ecosystem of diverse cell types whose interactions govern tumor growth and clinical outcome. While multiple studies have extensively charted the TME’s impact on immunotherapy, its role in chemotherapy response remains less explored. To address this, we developed DECODEM (DEcouplingCell-type-specificOutcomes usingDEconvolution andMachine learning), a generic computational framework leveraging cellular deconvolution of bulk transcriptomics to associate gene expression of individual cell types in the TME with clinical response. Employing DECODEM to analyze gene expression of breast cancer patients treated with neoadjuvant chemotherapy across three bulk cohorts, we find that the expression of specific immune cells (myeloid, plasmablasts, B-cells) and stromal cells (endothelial, normal epithelial, CAFs) are highly predictive of chemotherapy response, achieving the same prediction levels as the expression of malignant cells. Notably, ensemble models integrating the estimated expression of different cell types perform best and outperform models built on the original tumor bulk expression. These findings and the models’ generalizability are further tested and validated in two single-cell (SC) cohorts of triple negative breast cancer. To investigate the possible role of immune cell-cell interactions (CCIs) in mediating chemotherapy response, we extended DECODEM to DECODEMi to identify such key functionally important CCIs, validated in SC data. Our findings highlight the importance of active pre-treatment immune infiltration for chemotherapy success. DECODEM and DECODEMi are made publicly available to facilitate studying the role of the TME in mediating response in a wide range of cancer indications and treatments.
- Research Article
- 10.1007/s12672-025-03068-z
- Jul 3, 2025
- Discover Oncology
- Boyang Li + 4 more
BackgroundTMEM71 is a poorly characterized transmembrane protein with unclear roles in cancer. This study aimed to explore its prognostic value, immune relevance, and therapeutic implications in breast cancer.MethodsWe integrated transcriptomic, single-cell RNA-seq, immune infiltration and drug response data from TCGA, GTEx, GEO and public scRNA-seq datasets. Prognostic analysis, immune correlation (ssGSEA, CIBERSORT), functional enrichment (GO, KEGG, GSEA) and chemotherapy/immunotherapy sensitivity assessments were conducted.ResultsTMEM71 was significantly downregulated in breast cancer and correlated with poor clinical outcomes. Single-cell analysis revealed its predominant expression in malignant and stromal cells. TMEM71 expression was associated with enhanced infiltration of immune cells, upregulation of immune checkpoints and enrichment of immune-related pathways. High TMEM71 expression predicted better response to immune checkpoint inhibitors (anti-PD-1/PD-L1, anti-CTLA-4) and greater sensitivity to chemotherapeutics, including Gemcitabine, 5-fluorouracil and paclitaxel.ConclusionTMEM71 is a novel immune-related gene with potential as a prognostic biomarker and predictor of therapy response in breast cancer. These findings offer new insights into the immunological role of TMEM71 and support its potential utility in guiding personalized treatment strategies.
- Research Article
- 10.1016/j.tjog.2025.04.009
- Jul 1, 2025
- Taiwanese journal of obstetrics & gynecology
- Esra Çobankent Aytekin + 3 more
Placental mesenchymal dysplasia: A cause of intrauterine growth restriction and intrauterine death.
- Research Article
- 10.5493/wjem.v15.i2.102761
- Jun 20, 2025
- World Journal of Experimental Medicine
- Evgeniya Yu Zubareva + 2 more
BACKGROUNDBreast cancer (BC) continues to occupy a leading position in terms of morbidity and mortality from malignant neoplasms among the female population. One of the promising markers associated with BC progression is programmed death ligand 1 (PD-L1). Previously, we investigated PD-L1 expression in BC via a new antibody against programmed cell death protein 1 ligand 1 (PDCD1 LG1) and reported that high PDCD1 LG1 expression in tumor cells is an independent factor for a high risk of regional metastasis in patients with BC. However, the prognostic significance of PDCD1 LG1 expression in BC stromal cells has not been adequately studied.AIMTo study the features of PDCD1 LG1 expression in BC stromal cells and its relationship with BC clinicopathological characteristics.METHODSIn a prospective single-center observational study, tumor samples from 148 patients with newly diagnosed BC were examined. The tumor sections were immunohistochemically stained with antibodies against PDCD1 LG1. In the tumor samples, the PDCD1 LG1-positive lymphocyte (PDCD1 LG1+ LF) score, presence of nuclear PDCD1 LG1 expression in the LFs, PDCD1 LG1 expression in polymorphic cell infiltrates (PDCD1 LG1+ polymorphic cell infiltrates [PCIs]), and cells of the fibroblastic stroma and endothelial cells of the tumor microvessels were assessed. Statistical analyses were performed using Statistica 10.0 software.RESULTSA PDCD1 LG1+ LF score ≥ 3 was detected more often at stages N0 and N3 than at N1 and N2 (P = 0.03). Moderate and pronounced PDCD1 LG1+ PCIs and the presence of PDCD1 LG1+ fibroblastic stroma were associated with negative estrogen receptor status (P = 0.0008 and P = 0.03, respectively), human epidermal growth factor receptor 2-positive (HER2+) BC (P < 0.00001 and P = 0.0005), and luminal B HER2+, non-luminal HER2+ and triple-negative BC (P < 0.00001 and P = 0.004). The risk of metastasis to regional lymph nodes (RLNs) depend on lymphovascular invasion (LVI) and the PDCD1 LG1+ LF score. In the absence of LVI and a PDCD1 LG1+ LF score < 3 or ≥ 3, metastases in RLNs were absent in 66.6% and 93.9% of patients with BC, respectively. In the presence of LVI and a PDCD1 LG1+ LF score < 3 or ≥ 3, metastases in RLNs were detected in 82.6% and 92.7% of patients with BC, respectively.CONCLUSIONThe results indicated that the combined assessment of the PDCD1 LG1+ LF score and LVI can improve the accuracy of predicting the risk of metastasis to RLNs in patients with BC.
- Research Article
- 10.2337/db25-239-or
- Jun 13, 2025
- Diabetes
- Chenxin Gu + 1 more
Introduction and Objective: Bone disease is a major type of complication of diabetes. However, the molecular and metabolic basis of how insulin deficiency or resistance affects bone is still incompletely known. Here, we tested if O-linked N-Acetylglucosamine (O-GlcNAc) signaling prevents diabetic osteoporosis in preclinical models. Methods: Post-translational modification of intracellular proteins by a single O-GlcNAc moiety at serine or threonine residues is mediated by O-GlcNAc transferase (OGT). We first used single-cell RNA sequencing and Flow Cytometry to examine the levels of Ogt gene expression and protein O-GlcNAc modification in osteogenic bone marrow stromal cells (BMSCs). We then generated adult perivascular BMSC-specific OGT knockout (OgtΔLepr) mice and characterized their bone microstructure at the steady state and during streptozotocin (STZ)-induced diabetes. Finally, we treated cultured BMSCs with chemicals to induce O-GlcNAcylation or overexpressed OGT in BMSCs in mice to test whether O-GlcNAc augmentation alleviates diabetes-induced osteopathy. Results: OGT and protein O-GlcNAcylation are enriched in adipo-osteogenic BMSCs that are marked by the leptin receptor gene (Lepr). LeprCre-mediated deletion of OGT in adult BMSCs (OgtΔLepr) did not evidently change bone microarchitecture or mineral density in young or old mice. Instead, STZ-induced diabetes diminished stromal O-GlcNAcylation and OgtΔLepr mice showed exacerbated bone loss during diabetes. In culture, OGT was required for insulin and intermittent parathyroid hormone (iPTH) to stimulate BMSC proliferation and osteogenic differentiation. In mice, OGT overexpression in BMSCs alleviated induced bone loss. Mechanistically, OGT modified and activated RUNX2 to stimulate osteogenesis. Conclusion: Protein O-GlcNAcylation modulates the differentiation fate and metabolic fitness of BMSCs, thus impeding diabetic osteopathy. Disclosure C. Gu: None. H. Ruan: None.
- Research Article
- 10.1186/s12935-025-03810-1
- Jun 10, 2025
- Cancer Cell International
- Fereshteh Rezagholizadeh + 12 more
BackgroundPancreatic ductal adenocarcinoma (PDAC) has a poor prognosis and most patients are diagnosed at a stage where the disease is unresectable, locally advanced, or has already metastasized. Invasive pancreatic cancer is believed to arise through a progression of noninvasive ductal lesions referred to as pancreatic intraepithelial neoplasia (PanIN). The mechanisms driving the transition from PanIN, to invasive PDAC are not fully understood. Moreover, extensive stromal involvement of pancreatic cancer tissue complicates bulk analysis, hindering precise tumor-specific molecular data.MethodsThis issue was addressed through a comprehensive in-silico analysis of laser-capture microdissected (LCM) pure tumor epithelial cells from transcriptomic PDAC cohort datasets, with survival outcomes further validated using TCGA. We employed LCM to evaluate mRNA expression in PanIN lesions, tumor epithelial, and stromal cells, using RT-qPCR in 20 PDAC patients. Immunohistochemistry (IHC) on 353 PDAC patients, including 73 PanIN lesions and 280 tumor tissues on tissue microarray (TMA) slides, provided further confirmation.ResultsBased on in-silico analysis, RAD51AP1 was identified as a tumor-specific marker associated with aggressive tumor behavior in captured PDAC samples. RT-qPCR validation demonstrated significantly elevated RAD51AP1 expression in tumor epithelial cells compared to tumor stromal cells, PanIN lesions, and adjacent normal tissues. Consistently, IHC findings for nucleus, cytoplasm, and membrane localization revealed higher RAD51AP1 protein expression in tumor tissues compared to PanIN lesions and normal tissues. This increased expression was correlated with tumor progression and aggressiveness in PDAC patients, as well as reduced survival and poor prognosis in patients with PanIN lesions.ConclusionsRAD51AP1 is a tumor-specific molecule that associated with more aggressive behavior, advanced disease, and a worse survival rate.Graphical