Abstract
Background
Pancreatic cancer (PC) presents significant challenges in oncology, with metastasis critically affecting patient outcomes. Autophagy-related genes (ARGs)’s involvement in influencing immune activity and metastasis in PC remains inadequately understood.
Aim
This study seeks to identify and validate five ARGs that could serve as immune targets, enhancing enhancing Pancreatic cancer metastasis (PCM)’s prognostic models and informing immunotherapy strategies.
Methods
ARGs that were diffentially expressed were screened, followed by Cox regression and LASSO analyses to pinpoint five genes linked to overall survival (OS). A prognostic model was developed and validated using ROC curves. Functional analyses, including GO and KEGG, were performed to elucidate ARG mechanisms. Immune infiltration and TFs/microRNA/mRNA networks were assessed to understand ARG-immune cell interactions. Experimental validation employed real-time PCR, IHC, and Western blotting, supported by TCGA data. Functional assays explored RHEB’s role in PC, particularly its interaction with LC3.
Results
Five ARGs (CASP1, RHEB, CHMP2B, MYC, and HDAC6) were identified, contributing to a robust prognostic model where low-risk individuals showed significantly longer OS. The model demonstrated high AUC scores, indicating strong prognostic capability. CD8 T cells and Treg cells’ elevated levels were observed in metastatic subjects. RHEB knockdown suppressed cancer cell proliferation and invasion, with a negative correlation between RHEB and LC3, suggesting a role in autophagy-mediated modulation of PC metastasis.
Conclusion
This study introduces a novel prognostic model incorporating five ARGs, highlighting their potential as immune targets for cancer immunotherapy. The negative correlation between RHEB and LC3 suggests a therapeutic pathway for PCM intervention, laying the groundwork for more effective anti-cancer strategies. These findings advance the identification of novel immune targets and signaling pathways, aligning with precision medicine goals in cancer treatment.
Supplementary Information
The online version contains supplementary material available at 10.1007/s12672-025-02190-2.
Keywords: Autophagy-related genes, Pancreatic cancer, Metastasis, Prognostic model, Immune infiltration, RHEB
Introduction
Pancreatic cancer (PC) represents a considerable global health dilemma, ranking as the 12th most popular and 7th most deadly malignant tumor according to GLOBOCAN 2020 statistics [1]. The disease is characterized by a notably short median survival time and a low 5-year survival rate, primarily due to factors such as late diagnosis, high invasiveness, and resistance to chemotherapy [2]. These factors underscore the imperative need for creative strategies to advance PC prognosis and patient endings.
The possibility of improving the immune system’s ability to detect and battle cancer cells has been highlighted by recent developments in cancer immunotherapy. To design effective immune targets, it is essential to identify the distinct molecular properties of cancer cells, such as their genetic, epigenetic, and protein expression patterns. Within this framework, autophagy—a defense mechanism in eukaryotes that includes phases such as autophagic lysis and maturation—is essential for preserving the homeostasis of cells. A number of malignancies, including PC, have been related to dysregulated autophagy. This is because dysregulated autophagy is connected to enhanced invasion and metastasis, which worsens prognosis and treatment results [3–6].
Autophagy-related genes (ARGs) have emerged as key regulators of PC progression [5, 7], yet their precise mechanisms remain unclear. While ARGs have been explored in the prognosis of other cancers such as colorectal cancer, grade IV astrocytoma, non-small cell lung cancer, and prostate carcinoma, investigation specific to PC is limited [8–11]. Notably, autophagy may influence the tumor immune microenvironment, suggesting that ARGs could serve as novel immune targets for cancer immunotherapy [12]. This potential makes them attractive candidates for developing aimed therapies that coud strengthen the immune system’s ability to combat cancer.
In this study, we investigate the impact of ARGs on pancreatic cancer metastasis (PCM) and elucidate their mechanisms of action through functional enrichment assays, immune cell function analysis, and transcription factor (TF)/mRNA/microRNA (miRNA) network evaluations. We propose small-molecule compounds targeting ARGs as potential therapeutic agents for PC. Moreover, we build a prognostic model grounded in five ARGs to predict PC outcomes. Our findings reveal that the expression of these ARGs in PC cells or tissues aligns with both experimental and bioinformatic analyses, with RHEB identified as a key player in promoting PC proliferation and invasion. This research offers new insights into early clinical characteristics of PCM and suggests innovative approaches for more effective treatment strategies. By focusing on the identification of immune targets and signaling pathways, this introduction aligns with the special issue’s emphasis on cancer immunotherapy and immunoprecision medicine.
Materials and methods
Data Collection and Differentially Expressed ARGs (DEARGs) Screening ARGs associated with PCM were selected from GEO’s gene expression profile GSE19279 dataset [13, 14]. mRNA profiles from 15 samples (3 normal pancreatic samples, 4 primary PDAC samples (primary group), 3 normal liver samples, and 5 liver metastasis samples (metastatic group)) were collected. To determine how ARGs affect metastatic PC patients’ outcomes, RNA-seq-batch effects data and clinicopathological information were collected [15] (https://xenabrowser.net/datapages/). ARGs were obtained from HADb [16]. GEO2R was used for data analysis. Differentially expressed mRNAs (DEmRNAs) were characterized with the standard: (adj. p-values) < 0.05 and |log2FC (fold-change)|> 1. DEARGs were obtained by intersecting 243 human ARGs in HADbs with the DEmRNAs.
The 5-ARGs prognostic model’s analysis
For the purpose of determining how OS affects DEARGs, Kaplan–Meier (KM) plots were used [17]. Univariate Cox was applied to retain OS-related DEARGs and LASSO Cox was applied to delve OS-related DEARGs. Expression of hub genes was confirmed in TCGA-PAAD. “ggplot2” package in R (version 4.2.1) was used. A risk score was provided to every subject, and its median was selected to separate subjects into low- or high-risk groups. Survival difference was used to assess the model’s effectiveness.
Subsequently, a prognostic model grounded in five autophagy-related genes (ARGs) was developed. The risk scores for this model were computed by the formula: , where Coefi represents the coefficient of each of the five ARGs, and Xi denotes the expression level of every gene. The prognostic value of the TCGA cohort was evaluated using R. Survival of PC patients was evaluated by conducting KM analysis. Time-ROC plots were created. AUC was determined to evaluate this model’s prognostic ability. Additionally, PC patients were divided into subgroups based on: age, gender, stage, and T staging. Survival in subgroups was performed using the KM curve. To comparatively evaluate the prognostic ability between clinical characteristics and risk score, independent risk factors were identified using Xiantao study forest plot tool. ROC curves were created by “pROC”, and AUC was used to evaluate the prognostic ability. DCA was performed and calibration curves were used to experiment the prognostic ability of the model using the “stdca.R” and “rms” packages in R.
Predicting DEARGs’ mechanism of action
GO and KEGG enrichment analyses were used to predict hub genes’ function using Metascape [18]. To predict PC drugs, HDAC6 was placed in the downregulation group, the other four were placed in the upregulation group, and they were uploaded to L1000FWD [19]. A list that includes the top 10 potential compounds and the top 3 drugs was uploaded to PubChem [20] for visualization. Next, to elucidate the roles of DEARGs on PCM, a TF/miRNA/mRNA network was created. miRNA-mRNA data were extracted from the online platform Starbase [21], and TF-mRNA data were from chEA3 [22]. Cytoscape was used to create the whole TF/miRNA/mRNA network. Additionally, Pearson analyses were performed to determine the relationship between autophagy’s key component and ARGs. The correlation between ARGs and MAP1LC3B was evaluated by “ggplot2”.
Immuno-microenvironment assay
Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data (ESTIMATE) was performed by “estimate” of R. Ratio of the stromal-immune element in TME was assessed to get stromal score, immune score, ESTIMATE score (the total of stromal and immune score), and tumor purity (# of tumor cells). A lower score indicated a lower ratio of TME. To evaluate immunoinfiltration, CIBERSORT was used to calculate 22 subsets via CIBERSORTx [23]. The relationship between ARGs and levels of infiltrating subsets was evaluated via Pearson analyses using “ggplot2”.
Cell culture and tissue specimens
Capan-1, COLO357, PANC-1, CFPAC-1, MIA PaCa-2, and HPDE6-C7 from the Chinese Academy of Sciences Cell Bank were maintained in DMEM with 10% FBS at 37 °C. The tissue samples used for RT-qPCR, including primary tumor samples and paired normal samples (n = 5), were collected from the 1st Affiliated Hospital of Chongqing Medical University. This study has the approval from the hospital’s ethics committee (2023–0104).
RNA extraction, qPCR, and RNA interference
RNAs were isolated by Trizol (Biyotime). RT-PCR was carried out with 1 µg of total RNA. qPCR assays were carried out by a SYBR kit and a 7500 cycler (ABI). Each sample had three replicates. The result was calculated by the 2−ΔΔCt formula. Primers were designed for the 5 hub genes (MYC, CHMP2B, RHEB, and CASP1, Supplementary Table 1). The siRNA that targeted human RHEB was purchased from Tsingke Biotech. PANC-1 and MIA PaCa-2 cells were transfected with siRHEB and siNC Lipo 2000 (Invitrogen) for 48 h following the user manual. Cells were gathered 48 h later. WB assays were carried out to estimate the knockdown efficiency, using GAPDH as a loading control. The RHEB-siRNA sequences used were as follows:
RHEB-SiRNA1: GTGTATTCTGTTACATCAA.
RHEB-SiRNA2: CACAGTAAATGGACAAGAA.
RHEB-SiRNA3: GAAAGACCTGCATATGGAA.
Immunohistochemistry (IHC)
The paraffin-embedded tissues were used to perform immunohistochemical analysis. Sections were incubated with anti-RHEB antibody (ab25873) overnight at 4 °C. After secondary antibody detection, the antigen was visualized by standard DAB staining (Dako, Sweden) followed by hematoxylin counterstaining.
Immunofluorescence (IF)
The tissue slices were used to perform immunofluorescence staining by incubating them with anti-RHEB antibodies (Abcam, ab25873) at a dilution of 1:1000, LC3 antibodies (Wanleibio, WL01506) at a dilution of 1:400, and the second antibodies at a dilution of 1:5000. The stained samples were counterstained with DAPI (2 µg/mL, Beyotime). Finally, slices were observed with a fluorescence microscope (Leica Microsystems GmbH).
Western blotting (WB) analysis
Whole-cell lysates were put on ice with RIPA buffer and quantified using a BCA Kit (Beyotime, China). The extracts were mixed with a 5 × loading buffer (Mengbio, China) and denatured by boiling. Lysates (40 µg) underwent 10% SDS-PAGE (Epizyme, China) and were transferred to PVDF membranes (Invitrogen, USA). After blocking with a buffer (Beyotime Technology, China), primary antibodies against RHEB (Abcam, ab25873) and GAPDH (Huabio, 12D6) were incubated for 12 h at 4 °C. Samples were then treated with HRP-conjugated secondary antibodies (Servicebio, G1213, China) for 2 h. Blots were visualized using the HRP Substrate kit (Merck Millipore Corporation, Germany).
Cell proliferation and colony formation assays
CCK-8 (Biosharp, China) was added to 96-well plates containing 2000 cells per well, and the cells were incubated for 2 h at 0/24/48/72 h. At 450 nm, absorbance was measured. For 10–14 days, the cells that were implanted in six-well plates were grown at densities of 1000 cells/well. The surviving colonies were scanned and counted using a CanoScan 8800F MOEL-85 scanner (CanoScan, Japan) following fixation with 4% paraformaldehyde (Servicebio, China). The colonies were then visualized using Gentian violet (Servicebio, China). There were three independent duplicates of each sample.
Transwell cell migration and aggression experiments
Transwell chambers with an 8-µm pore size (Corning Inc.) were utilized to assess cell migration and aggression. For the aggression assay, a Matrigel layer (Becton, Dickinson, and Company, USA) was applied to the Transwell membrane. After 48 h of cell incubation, the migrated or invaded cells were photographed and documented using a Leica camera (Germany). Each experiment was conducted three times.
Data analysis
Data were analyzed by SPSS 26.0 R (4.2.1), and GraphPad Prism 9.0. Differences in categorical data were determined by Fisher’s exact test or the Chi-Square test. Distinctions in continuous data were determined by the Wilcoxon test or Student’s t-test. The distinctions in all groups of continuous variables were determined by ANOVA. Survival was analyzed by regression assay. Correlation was performed by Pearson analysis. p < 0.05 was designated as significance.
Results
Study flowchart
The study’s flowchart is illustrated in Fig. 1a. Initially, we selected a dataset focused on liver metastasis in PC to analyze differentially expressed genes, intersecting them with the autophagy database HADb to identify 22 ARGs. Through regression analysis, five genes closely linked to overall survival (OS) were identified. These genes were then used for survival analysis and to construct a risk model for diagnostic efficacy. Additionally, we conducted functional enrichment analysis, constructed a TF/mRNA/miRNA regulatory network, and assessed immune infiltration to understand its mechanism of action. We also identified promising small-molecule compounds, such as Apicidin, Simvastatin, and PD-0325901, targeting ARGs.
Fig. 1.
Flow chart and hierarchical clustering analysis of autophagy-related genes (ARGs). a Study design. b Venn diagrams highlighting ARGs common to both GSE19279 and HADb datasets. c Heatmaps of differentially expressed ARGs (DEARGs), with orange indicating up-regulation and gray indicating down-regulation
DE ARGs and ca 5-ARGs prognostic model
From the GSE19279 dataset, 1113 differentially expressed mRNAs (DEmRNAs) were identified. After intersecting with 243 human ARGs from HADb, 22 differentially expressed ARGs (DEARGs) were obtained (Fig. 1b, c). The Kaplan–Meier (KM) method and Cox regression analysis distinguished seven DEARGs (Fig. 2a–h). LASSO Cox regression further refined this to five prognostic ARGs: CASP1, RHEB, CHMP2B, MYC, and HDAC6, which were included as hub genes in this model (Fig. 2i–j).
Fig. 2.
Clinical correlation of ARGs. a–g Kaplan–Meier (KM) plots. h Forest plot. i Univariate Cox regression analysis of hub genes. j LASSO coefficients profiling
The 5-ARGs prognostic model’s application
Risk score distribution, survival, and heat maps for the five ARGs were created (Fig. 3a–c). The data indicated that higher risk scores correlated with increased mortality. Four ARGs, except HDAC6, had lower levels in low-risk subjects. KM curves demonstrated significantly higher OS in low-risk subjects (Fig. 3d). Risk scores for 1-, 3-, and 5-year OS had AUCs of 0.68, 0.69, and 0.70, respectively (Fig. 3e). Stratification based on risk scores effectively differentiated between favorable and unfavorable prognoses for PC patients, with low-risk groups showing better outcomes across different genders, stages, and T staging (Supplementary Fig. 1). Thus, the 5-ARGs prognostic model accurately predicted PC patient survival.
Fig. 3.
Evaluation of the prognostic model. a Distribution of risk scores. b Correlation between ARGs. c Correlation between survival rates. d KM survival assay. e Risk scores
The 5-ARGs model and clinicopathological features’ prognostic value
For the purpose of estimating predictive values, univariate Cox regression identified prognostic factors. Forest plots indicated that only risk score and stage were typical independent risk factors (Fig. 4a). ROC curves are presented in Fig. 4b, excluding T/M staging due to insufficient data. AUCs for pathological indices were lower than risk scores. Decision curve analysis (DCA) suggested that the risk score was a high quality prognostic marker at 1, 3, and 5 years (Supplementary Fig. 2). Calibration curves, plotted to assess predictive accuracy, revealed that at 1, 2, and 3 years, the model’s predictions closely matched the criteria curve, demonstrating excellent prognostic capability (Fig. 4c).
Fig. 4.
Comparison of diagnostic efficiency. a Regression analyses of prognostic factors, specifically T stage and N stage. b AUC values. c Calibration curves
Functional enrichment analysis and screening small-molecule drugs
GO and KEGG analysis identified macroautophagy, autophagy, and autophagic processes as the most enriched biological processes (BPs). The most enriched cellular components (CCs), molecular functions (MFs), and KEGG pathways are shown in Fig. 5a, suggesting that hub genes regulate tumor progression by modulating autophagy. To identify promising drugs, the five hub genes were submitted to L1000FWD. The most promising small-molecule drugs, including Apicidin, Simvastatin, and PD-0325901, are listed in Table 1. These drugs, with their 3D/2D structures modeled using PubChem (Supplementary Fig. 3), can downregulate ARG expression by inducing autophagy, thereby advancing targeted PC treatment.
Fig. 5.
Mechanism of action of hub genes. a GO and KEGG analyses. b TF/mRNA/miRNA networks, with light purple representing TFs, green for mRNAs, and deep purple for miRNAs. c Relationships between hub genes and LC3
Table 1.
The screened drugs for PAAD treatment
| Drug | Similarity score | p-value | q-value | Z-score | Combined score | MOA | Predicted MOA |
|---|---|---|---|---|---|---|---|
| Apicidin | − 0.6 | 6.90E-05 | 2.74E-01 | 1.59 | − 6.6 | Unknown | HDAC inhibitor |
| Simvastatin | − 0.6 | 5.36E-05 | 2.74E-01 | 1.63 | − 6.98 | HMGCR inhibitor | Glucocorticoid receptor |
| PD-0325901 | − 0.6 | 4.55E-05 | 2.74E-01 | 1.76 | − 7.63 | MEK inhibitor | MEK inhibitor |
| Auranofin | − 0.6 | 5.11E-05 | 2.74E-01 | 1.81 | − 7.78 | NFkB panthway inhibitor | Calcium channel |
| Panobinostat | − 0.4 | 3.69E-03 | 2.76E-01 | 1.55 | − 3.77 | HDAC inhibitor | HDAC inhibitor |
| PP-2 | − 0.4 | 2.83E-03 | 2.74E-01 | 1.62 | − 4.13 | Scr inhibitor | Cyclooxyenase inhibitor |
| BMS-387032 | − 0.4 | 3.12E-03 | 2.74E-01 | 1.6 | − 4.01 | CDK inhibitor, MCL1 inhibitor | CDK inhibitor |
| LY-294002 | − 0.4 | 2.94E-03 | 2.74E-01 | 1.65 | − 4.19 | mTOR inhibitor, PI3K inhibitor | PI3K inhibitor |
| Olvanil | − 0.4 | 3.11E-03 | 2.74E-01 | 1.68 | − 4.2 | TRPV agonist | Adrenergic receptor |
| THM-I-94 | − 0.4 | 3.49E-03 | 2.74E-01 | 1.64 | − 4.04 | HDAC inhibitor | HDAC inhibitor |
TF/miRNA/mRNA regulatory network and correlation between ARGs and MAP1LC3B
To explore how ARGs influence PCM, interactions between miRNA-DEARG and TF-DEARG were analyzed using Starbase and chEA3, resulting in a TF/miRNA/mRNA network (Fig. 5b). This network indicated that most hub genes are regulated by FOS and MYC. MAP1LC3B is indentified as a crucial component of autophagy [24]. Pearson correlation analysis between MAP1LC3B and five ARGs (Fig. 5c) showed significant associations with RHEB, CHMP2B, MYC, and CASP1, implying their potential role in regulating PCM through autophagy.
Assay of immunoinfiltration and co-expression of ARGs and metastasis-related immune cells
We examined the principle that five autophagy-related genes (ARGs) influence pancreatic cancer (PC) prognosis. Autophagy is linked to immune infiltration [5, 25, 26] and tumor progression [27, 28]. Using Sangerbox, we analyzed the relationship between ARG expression and immune components. The data revealed that metastatic subjects had significantly lower stromal, immune, and ESTIMATE scores (Fig. 6a–c) but higher tumor purity (Fig. 6d), suggesting a connection between hub gene levels and the tumor microenvironment (TME). CIBERSORT analysis identified differences in 22 immune cell subsets between elementary and metastatic cases (Fig. 6e). T cell follicular helpers and CD8 T cells were prevalent in primary subjects, while CD8 T cells and Treg cells dominated in metastatic subjects (Fig. 6i). Principal component analysis (PCA) indicated big differences in immune cell percentages, with plasma cells or Tregs being higher in primary or metastatic subjects, respectively (Fig. 6f). We applied Pearson analysis to exploring the relevance between immune cells and ARGs. We examined 22 immune cell subsets (Fig. 6g), revealing a link with infiltration. Specifically, CASP1, HDAC6, and RHEB showed a strong correlation with infiltrating plasma cells, while RHEB, MYC, and CHMP2B were closely associated with infiltrating Treg cells (Fig. 6h). This represents that these hub genes function in modulating immune cell infiltration, particularly Treg cells, in pancreatic cancer.
Fig. 6.
Immune phenotype of PAAD, hub genes’ co-expression patterns, and immune cell subsets. a Stromal score. b Immune score. c ESTIMATE score. d Tumor purity. e PCA of GEO samples. f Immune cell abundance in PAAD. g Correlations among different infiltrating immune cells. h Relationships between ARGs and infiltrating immune cells. i Relative proportions of 22 immune cells
Expression characteristics of the 5 hub genes in PC cells or tissues
We assessed the levels of five differentially expressed ARGs in PC and normal tissues (n = 179) using TCGA + GTEx-PAAD data. CASP1, CHMP2B, MYC, and RHEB were significantly elevated in PC tissues, while HDAC6 was decreased (Fig. 7a). Five ARGs were measured for their mRNA levels using qPCR in five pancreatic cancer (PC) cell lines, five normal pancreatic cell lines, and PC and normal tissues. The outcomes of the research indicated that PC cells and tissues had considerably higher levels of the mRNAs of four ARGs, with the exception of HDAC6 (Fig. 7b–k). These results are consistent with the five differentially expressed ARGs (DEARGs) that we identified during our first screening.
Fig. 7.
Validation of hub genes expression. a Gene expressions in TCGA + GTEx-PAAD. b–f mRNA levels of five ARGs in five PC cell lines. g–k mRNA levels in five paired PC tissues
RHEB silencing suppresses proliferation, migration, and aggression of pancreatic cells
Given the significant increase in RHEB mRNA, we selected it to validate its role in pancreatic cancer (PC) cells. Immunohistochemistry (IHC) and Western blot (WB) assays confirmed that RHEB protein expression was markedly more in PC tissues and cells in comparison to normal counterparts (Fig. 8a, b). Upon silencing RHEB (siRHEB) (Fig. 8c), there was a notable reduction in PC cell growth (Fig. 8d–g). Furthermore, RHEB silencing led to significantly fewer cells traversing the membrane and reduced invasion of the Matrigel barrier compared to controls (Fig. 8h–i). These findings represent that RHEB may facilitate the proliferation, migration, and aggression of PC cells.
Fig. 8.
RHEB enhances proliferation, invasion, and migration in PC cells. a RHEB immunohistochemical staining. b Protein levels of RHEB. c siRNA knockdown of RHEB. d–e Cell proliferation. f–g Representative images and quantitative analyses of colony formation and invasion (*p < 0.05, **p < 0.01, ***p < 0.001)
RHEB silencing upregulates LC3
In our final analysis, we explored the relationship between RHEB and the autophagy marker LC3. Immunofluorescence experiments revealed that RHEB and LC3 co-localize on the cell membrane, with LC3 expression significantly increasing when RHEB is silenced (Fig. 9a). Western blot assays further confirmed elevated LC3 levels in RHEB-silenced cells compared to controls (Fig. 9b). This negative correlation between RHEB and LC3 implies that RHEB may influence pancreatic cancer cell proliferation and metastasis through autophagy, positioning RHEB as a potential target for pancreatic cancer therapy.
Fig. 9.
RHEB silencing upregulates the expression of LC3. a Immunofluorescence experiments of RHEB and LC3 in the PANC-1–1 and MIA PaCa-2 cells transfected with the siRNA or the siRHEB. b Western blot assays of RHEB and LC3 in the PANC-1–1 and MIA PaCa-2 cells transfected with the siRNA or the siRHEB
Discussion
PC is still a daunting challenge due to its aggressive character, with patients facing a grim 5–11-month survival period and a mere 9% 5-year survival rate [1]. Metastasis is a major contributor to the high mortality rate associated with this disease. In our study, we identified differentially expressed autophagy-related genes linked to pancreatic cancer metastasis and prognosis. Through regression analysis, we pinpointed five ARGs that either promote or inhibit PCM. Our survival curve analysis demonstrated that this model provides a more precise prognosis than traditional clinicopathological data by accurately classifying patients based on risk. We employed enrichment assays, network assays, and immune infiltration assays, which indicated that these ARGs influence PCM by modulating autophagy and immune infiltration. Additionally, we proposed several small-molecule drugs targeting these ARGs.
We created a prognostic model grounded in these five ARGs and utilized it for functional enrichment analysis. The oncogene MYC, known for its amplification in various tumors, lacks a fully understood mechanism [29]. CASP1, part of the cysteinyl aspartate protease family, is implicated in tumor metastasis [30, 31]. Previous studies have shown CASP1’s role in promoting liver cancer metastasis, potentially through autophagy [32]. CHMP2B, associated with receptor degradation or recycling, has limited information regarding its role in cancer [33], although it has been suggested as a prognostic marker for PC [34]. Our findings indicate that CHMP2B exacerbates PCM, likely through autophagy, leading to poor prognosis. HDAC6 is involved in cell adhesion, invasion, and migration through deacetylation processes [35]. While its relationship with PC is less explored compared to other tumors, some studies have established a connection between HDACs and PC [36, 37]. Our results suggest that HDAC6 is downregulated in PCM tissues, indicating its potential inhibitory role in PCM. Notably, three of the 10 proposed small-molecule drugs are HDAC inhibitors. Further research is required to elucidate its precise mechanism of action.
RHEB, a GTPase known for activating mTORC1, has been identified as an oncogenic protein [38]. Previous studies have demonstrated that silencing RHEB can restrain the growth of colon tumor cells and accelerate apoptosis by suppressing the mTOR pathway [39]. Moreover, elevated levels of RHEB have been linked to poor prognosis in PC [40]. Our study corroborates these findings, showing that RHEB silencing reduces the proliferation and metastatic potential of PC cells. Although our study confirmed the role of RHEB in driving PC advance and metastasis, the precise mechanisms remain to be elucidated. Future research will focus on unraveling these pathways to enhance our understanding of immune signaling in cancer. It is still important to mention that our study’s sample size was limited, and the clinical data was predominantly derived from TCGA, which mainly includes Caucasian populations. Additionally, immune cell infiltration was assessed using a single algorithm, necessitating further verification. By addressing these limitations, we aim to help to indentify new immune targets and signaling pathways, aligning with the goals of improving cancer immunotherapy.
In our research, enrichment assays of five ARGs indicated significant enrichment in necroptosis, senescence, and the thyroid hormone (TH) axis, which affects mitophagy and other biological processes [41]. The THR axis, known to play a role in hepatic autophagy, is also implicated in liver cancer [42]. This suggests that ARGs might influence PCM through these pathways. Furthermore, transcription factors and microRNAs have been found to be tightly associated with PC progression [43–52]. Our network assays revealed that most of the five ARGs are linked to FOS/MYC and regulated by five miRNAs, supporting the idea that FOS or miR-369-3p could significantly impact PCM by modulating RHEB, a hypothesis we plan to explore in future research.
Immune infiltration is crucial for tumor progression, as it aids in immune surveillance evasion and recruitment of immunosuppressive cells, which suppress tumor immunogenicity [53]. Although immune infiltration and autophagy are strongly associated with PC evolution [5], little research has focused on immune infiltration in metastatic PC patients. Our comprehensive study of immune infiltration mechanisms and ARGs in PCM revealed increased Tregs and decreased plasma cells in metastatic subjects, with the five ARGs linked to various cell subsets. Dendritic cells, B cells, and cytotoxic lymphocytes, components of tertiary lymphoid structures (TLSs), are generally associated with better cancer prognoses [54]. Consistent with this, decreased B cell numbers have been linked to poor PC prognosis, a trend observed in other cancers as well [55–57]. While the role of B cells in autophagy remains unclear, Tregs are known to be influenced by autophagic depletion and rely heavily on autophagy to function [58]. Despite limited studies on Tregs in PCM, data suggest that in pancreatic ductal adenocarcinoma, Tregs are upregulated early but restricted to paracancerous lymph nodes in advanced stages [59]. Tregs modulate CD4 + T cell infiltration in the tumor microenvironment through a CTLA-4/CD8-dependent axis [59], though further verification is needed. Our findings suggest a regulatory relationship between immune infiltration and 5 ARG levels, indicating that PC cells might modulate immune infiltration via autophagy during invasion or metastasis, potentially attracting immunosuppressive cells to facilitate immune escape and aggression. This notion could explain the bad prognosis observed in metastatic PC patients, though further experiments are necessary to validate this hypothesis.
In our pursuit of advancing cancer immunotherapy, we explored the potential of small-molecule drugs as modulators of ARGs to identify novel therapeutic biomarkers. Gene alterations present a promising avenue for altering tumor therapy outcomes and targeting ARGs could unveil new immune targets for precision medicine. For instance, Apicidin, a histone deacetylase inhibitor, interacts with class I HDACs, disrupting deacetylation processes and modifying protein acetylation levels, thereby exerting therapeutic effects on PC [60]. Similarly, simvastatin, known for its lipid-lowering properties, has shown potential in counteracting gemcitabine resistance in PC by disrupting the TGF-β1/Gfi-1 signaling pathway [61], although its direct effects on PC remain to be amply explored. These findings underscore the need for further validation of small-molecule drugs in the context of cancer immunotherapy.
Conclusion
In this study, we have identified and validated five ARGs that are significantly associated with pancreatic cancer metastasis and prognosis. Through rigorous RT-qPCR and Western blot experiments, we confirmed that these genes’ expression levels align with our initial findings. Notably, our research highlights the negative correlation between RHEB and LC3, suggesting that RHEB may enhance pancreatic cancer cells’ proliferation, aggression and migration. This insight into the molecular interplay between RHEB and LC3 could unveil novel immune targets for cancer immunotherapy. Moreover, we created an innovative prognostic model grounded in these five ARGs, which holds promise for advancing precision medicine and enhancing the efficacy of anti-cancer treatments. This model could serve as a pivotal tool in identifying new immune targets and signaling pathways, aligning with the goals of improving cancer immunotherapy and personalized immune medicine.
Supplementary Information
Acknowledgements
The experiments part of this study was supported by the First Affiliated Hospital of Chongqing Medical University Key Laboratory of Molecular Oncology and Epigenetics.
Abbreviations
- ARGs
Autophagy-related genes
- DEARGs
Differentially expressed autophagy-related genes
- GEO
Gene Expression Omnibus
- TCGA
The cancer genome database
- PDAC
Pancreatic ductal adenocarcinoma
- UCSC
The University of California Santa Cruz
- HADb
Human Autophagy Database
- DEmRNAs
The mRNAs with different expression levels
- OS
Overall survival
- LASSO
Least absolute shrinkage and selection operator
- PAAD
Pancreatic ductal adenocarcinoma
- time-ROC
Time-dependent Receiver Operating characteristic curves
- AUC
The area under the ROC curve
- DCA
Decision curve analysis
- GO
Gene Ontology
- KEGG
Kyoto Encyclopedia of Genes and Genomes
- BP
Biological process(s)
- CC
Cellular
- MF
Molecular function
- L1000FWD
L1000 fireworks display
- PubChem
Published Chemical information component
- ESTIMATE
The Estimation of STromal and Immune cells in MAlignant Tumors tissues using Expression data
- TME
The tumor microenvironment
- RT-PCR
Reverse transcriptase-polymerase chain reaction
Author contributions
QD, ZJ: conception and design. QD, JH and FW: performed experiments. QD, LW: collect data and analyzed data. QD, LW: drafted the manuscript. ZJ, JH: reviewed data and finalized the manuscript. All authors reviewed and approved the final version.
Funding
This study was jointly supported by Sichuan Medical and Health Care Promotion Institute Youth Research Project (KY2023QN0129), Research Project of Early Gastrointestinal Cancer Physician Co-Growth Program (GTCZ-2023-SC-06) and The Third Hospital of Mianyang Research Project (202206). The funding agencies had no role in study design, data collection, and analysis, decision to publish, or preparation of the manuscript.
Data availability
The datasets used and/or analysed during the current study are available from NCBI Gene Expression Omnibus (GEO: GSE19279) https://www.ncbi.nlm.nih.gov/geo/ and the cancer genome database (TCGA -PAAD) https://portal.gdc.cancer.gov.
Declarations
Ethics approval and consent to participate
Informed consent was obtained from each participant included in the study. This study was ratified by the Ethics Committee of the Third Hospital of Mianyang on September 27, 2022 (Approved notice: 2022-14-2), and the experimental procedures followed the Declaration of Helsinki.
Consent for publication
Not applicable.
Competing interests
The authors declare no competing interests.
Footnotes
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
References
- 1.Park W, Chawla A, O’Reilly EM. Pancreatic cancer: a review. JAMA. 2021;326(9):851–62. 10.1001/jama.2021.13027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Globocan, Cancer today. https://gco.iarc.fr/today/home. Accessed 10 Jan 2024.
- 3.Pohl C, Dikic I. Cellular quality control by the ubiquitin-proteasome system and autophagy. Science. 2019;366:818–22. 10.1126/science.aax3769. [DOI] [PubMed] [Google Scholar]
- 4.Fujii S, Mitsunaga S, Yamazaki M, et al. Autophagy is activated in pancreatic cancer cells and correlates with poor patient outcome. Cancer Sci. 2008;99(9):1813–9. 10.1111/j.1349-7006.2008.00893.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Gorgulu K, Diakopoulos KN, Ai J, et al. Levels of the autophagy-related 5 protein affect progression and metastasis of pancreatic tumors in mice. Gastroenterology. 2019;156:203-17 e20. 10.1053/j.gastro.2018.09.053. [DOI] [PubMed] [Google Scholar]
- 6.Liang C, Xu J, Meng Q, et al. TGFβ1-induced autophagy affects the pattern of pancreatic cancer progression in distinct ways depending on SMAD4 status. Autophagy. 2020;16:486–500. 10.1080/15548627.2019.1628540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Han H. Dosage of Atg5 gene affects pancreatic tumorigenesis and progression. Gastroenterology. 2019;156(1):17–9. 10.1053/j.gastro.2018.11.038. [DOI] [PubMed] [Google Scholar]
- 8.Xu J, Dai S, Yuan Y, et al. A prognostic model for colon cancer patients based on eight signature autophagy genes. Front Cell Dev Biol. 2020;8: 602174. 10.3389/fcell.2020.602174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Wang Z, Gao L, Guo X, et al. Development and validation of a nomogram with an autophagy-related gene signature for predicting survival in patients with glioblastoma. Aging (Albany NY). 2019;11(24):12246–69. 10.18632/aging.102566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Liu Y, Wu L, Ao H, et al. Prognostic implications of autophagy-associated gene signatures in non-small cell lung cancer. Aging (Albany NY). 2019;11(23):11440–62. 10.18632/aging.102544. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Hu D, Jiang L, Luo S, et al. Development of an autophagy-related gene expression signature for prognosis prediction in prostate cancer patients. J Transl Med. 2020;18(1):160. 10.1186/s12967-020-02323-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Galluzzi L, Buque A, Kepp O, et al. Immunological effects of conventional chemotherapy and targeted anticancer agents. Cancer Cell. 2015;28:690–714. 10.1016/j.ccell.2015.10.012. [DOI] [PubMed] [Google Scholar]
- 13.Barry S, Chelala C, Lines K, et al. S100P is a metastasis-associated gene that facilitates transendothelial migration of pancreatic cancer cells. Clin Exp Metastasis. 2013;30(3):251–64. 10.1007/s10585-012-9532-y. [DOI] [PubMed] [Google Scholar]
- 14.The Gene Expression Omnibus database. https://www.ncbi.nlm.nih.gov/gds. Accessed 23 Mar 2024.
- 15.The University of California Santa Cruz. https://xenabrowser.net/datapages/. Accessed 27 Mar 2024.
- 16.Human Autophagy Database. https://autophagy.lu/clustering/index.html. Accessed 2 Apr 2024.
- 17.Kaplan-Meier plotter. https://autophagy.lu/clustering/index.html. Accessed 10 Apr 2024.
- 18.Metascape. https://metascape.org/. Accessed 1 May 2024.
- 19.The L1000 fireworks display. https://maayanlab.cloud/L1000FWD. Accessed 3 May 2024.
- 20.The Published Chemical Information website. https://pubchem.ncbi.nlm.nih.gov. Accessed 3 May 2024.
- 21.Starbase. https://starbase.sysu.edu.cn/. Accessed 28 Apr 2024.
- 22.ChEA3. https://maayanlab.cloud/chea3/. Accessed 28 Apr 2024.
- 23.CIBERSORTx. https://cibersortx.stanford.edu/runcibersortx.php. Accessed 8 May 2024.
- 24.Levine B, Kroemer G. Biological functions of autophagy genes: a disease perspective. Cell. 2019;176(1–2):11–42. 10.1016/j.cell.2018.09.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Xia H, Green DR, Zou W. Autophagy in tumour immunity and therapy. Nat Rev Cancer. 2021;21(5):281–97. 10.1038/s41568-021-00344-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Fan Y, Wang Y, Zhang J, et al. Breaking bad: autophagy tweaks the interplay between glioma and the tumor immune microenvironment. Front Immunol. 2021;12: 746621. 10.3389/fimmu.2021.746621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Kitamura T, Qian BZ, Pollard JW. Immune cell promotion of metastasis. Nat Rev Immunol. 2015;15(2):73–86. 10.1038/nri3789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Tauriello DVF, Palomo-Ponce S, Stork D, et al. TGFβ drives immune evasion in genetically reconstituted colon cancer metastasis. Nature. 2018;554(7693):538–43. 10.1038/nature25492. [DOI] [PubMed] [Google Scholar]
- 29.Beroukhim R, Mermel CH, Porter D, et al. The landscape of somatic copy-number alteration across human cancers. Nature. 2010;463:899–905. 10.1038/nature08822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Hong W, Gu Y, Guan R, et al. Pan-cancer analysis of the CASP gene family in relation to survival, tumor-infiltrating immune cells and therapeutic targets. Genomics. 2020;112(6):4304–15. 10.1016/j.ygeno.2020.07.026. [DOI] [PubMed] [Google Scholar]
- 31.Li J, Zhang Y, Ruan R, et al. The novel interplay between CD44 standard isoform and the caspase-1/IL1B pathway to induce hepatocellular carcinoma progression. Cell Death Dis. 2020;11(11):961. 10.1038/s41419-020-03158-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Deng L, Chen L, Zhao L, et al. Ubiquitination of Rheb governs growth factor-induced mTORC1 activation. Cell Res. 2019;29(2):136–50. 10.1038/s41422-018-0120-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Zhang Q, Lv L, Ma P, et al. Identification of an autophagy-related pair signature for predicting prognoses and immune activity in pancreatic adenocarcinoma. Front Immunol. 2021;12: 743938. 10.3389/fimmu.2021.743938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Deng X, Sun X, Yue W, et al. CHMP2B regulates TDP-43 phosphorylation and cytotoxicity independent of autophagy via CK1. J Cell Biol. 2021;221(1): e202103033. 10.1083/jcb.202103033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Valenzuela-Fernández A, Cabrero JR, Serrador JM, et al. HDAC6: a key regulator of cytoskeleton, cell migration and cell-cell interactions. Trends Cell Biol. 2008;18(6):291–7. 10.1016/j.tcb.2008.04.003. [DOI] [PubMed] [Google Scholar]
- 36.Edderkaoui M, Chheda C, Soufi B, et al. An inhibitor of GSK3B and HDACs kills pancreatic cancer cells and slows pancreatic tumor growth and metastasis in mice. Gastroenterology. 2018;155(6):1985–1998.e5. 10.1053/j.gastro.2018.08.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Ge LP, Jin X, Yang YS, et al. Tektin4 loss promotes triple-negative breast cancer metastasis through HDAC6-mediated tubulin deacetylation and increases sensitivity to HDAC6 inhibitor. Oncogene. 2021;40(12):2323–34. 10.1038/s41388-021-01655-2. [DOI] [PubMed] [Google Scholar]
- 38.Tian Y, Shen L, Li F, et al. Silencing of RHEB inhibits cell proliferation and promotes apoptosis in colorectal cancer cells via inhibition of the mTOR signaling pathway. J Cell Physiol. 2020;235(1):442–53. 10.1002/jcp.28984. [DOI] [PubMed] [Google Scholar]
- 39.Tan J, Liu W, Li J, et al. Over-expressed RHEB promotes the progression of pancreatic adenocarcinoma. Life Sci. 2021;277: 119462. 10.1016/j.lfs.2021.119462. [DOI] [PubMed] [Google Scholar]
- 40.Olszewska DA, Lonergan R, Fallon EM, et al. Genetics of frontotemporal dementia. Curr Neurol Neurosci Rep. 2016;16(12):107. 10.1007/s11910-016-0707-9. [DOI] [PubMed] [Google Scholar]
- 41.Yau WW, Singh BK, Lesmana R, et al. Thyroid hormone (T3) stimulates brown adipose tissue activation via mitochondrial biogenesis and MTOR-mediated mitophagy. Autophagy. 2019;15(1):131–50. 10.1080/15548627.2018.1511263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Chi HC, Chen SL, Tsai CY, et al. Thyroid hormone suppresses hepatocarcinogenesis via DAPK2 and SQSTM1-dependent selective autophagy. Autophagy. 2016;12(12):2271–85. 10.1080/15548627.2016.1230583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Sereno M, Haskó J, Molnár K, et al. Downregulation of circulating miR 802–5p and miR 194–5p and upregulation of brain MEF2C along breast cancer brain metastasization. Mol Oncol. 2020;14(3):520–38. 10.1002/1878-0261.12632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Li P, Lin Z, Liu Q, et al. Enhancer RNA SLIT2 inhibits bone metastasis of breast cancer through regulating P38 MAPK/c-Fos signaling pathway. Front Oncol. 2021;11: 743840. 10.3389/fonc.2021.743840. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Yan Z, Zhou Y, Yang Y, et al. Zinc finger protein 384 enhances colorectal cancer metastasis by upregulating MMP2. Oncol Rep. 2022;47(3):49. 10.3892/or.2022.8260. (Epub 2022 Jan 14). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Yang Y, Jiang H, Li W, et al. FOXM1/DVL2/Snail axis drives metastasis and chemoresistance of colorectal cancer. Aging (Albany NY). 2020;12(23):24424–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Fang L, Zhang B, Zhu N. MiR-877 suppresses tumor metastasis via regulating FOXM1 in ovarian cancer. J BUON. 2021;26(1):229–34. 10.18632/aging.202300. [PubMed] [Google Scholar]
- 48.Jeong HS, Lee DH, Kim SH, et al. Hyperglycemia-induced oxidative stress promotes tumor metastasis by upregulating vWF expression in endothelial cells through the transcription factor GATA1. Oncogene. 2022;41(11):1634–46. 10.1038/s41388-022-02207-y. [DOI] [PubMed] [Google Scholar]
- 49.Bai J, Hu Y, Chen X, et al. miR-144-3p inhibits the invasion and metastasis of lung adenocarcinoma cells by targeting IRS1. Zhongguo Fei Ai Za Zhi. 2021;24(5):323–30. 10.3779/j.issn.1009-3419.2021.104.05. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Li T, Tang C, Huang Z, et al. miR-144-3p inhibited the growth, metastasis and epithelial-mesenchymal transition of colorectal adenocarcinoma by targeting ZEB1/2. Aging (Albany NY). 2021;13(13):17349–69. 10.3779/j.issn.1009-3419.2021.104.05. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Escuin D, López-Vilaró L, Mora J, et al. Circulating microRNAs in early breast cancer patients and its association with lymph node metastases. Front Oncol. 2021;11: 627811. 10.3389/fonc.2021.627811. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Niu Y, Liu F, Wang X, Chang Y, Song Y, Chu H, Bao S, Chen C. miR-183-5p promotes HCC migration/invasion via increasing aerobic glycolysis. Onco Targets Ther. 2021;14:3649–58. 10.2147/OTT.S304117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Chen DS, Mellman I. Elements of cancer immunity and the cancer-immune set point. Nature. 2017;541(7637):321–30. 10.1038/nature21349. [DOI] [PubMed] [Google Scholar]
- 54.Schumacher TN, Thommen DS. Tertiary lymphoid structures in cancer. Science. 2022;375(6576):eabf9419. 10.1126/science.abf9419. [DOI] [PubMed] [Google Scholar]
- 55.van der Sijde F, Mustafa DAM, Vietsch EE, et al. Circulating immunological biomarkers: prognosis of pancreatic cancer patients reflected by the immune system. Pancreas. 2021;50(7):933–41. 10.1097/MPA.0000000000001862. [DOI] [PubMed] [Google Scholar]
- 56.Tan G, Wu A, Li Z, et al. Bioinformatics analysis based on immune-autophagy-related lncRNAs combined with immune infiltration in bladder cancer. Transl Androl Urol. 2021;10(8):3440–55. 10.21037/tau-21-560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Cao J, Wu L, Lei X, et al. A signature of 13 autophagy−related gene pairs predicts prognosis in hepatocellular carcinoma. Bioengineered. 2021;12(1):697–707. 10.1080/21655979.2021.1880084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Raza IGA, Clarke AJ. B cell metabolism and autophagy in autoimmunity. Front Immunol. 2021;12: 681105. 10.3389/fimmu.2021.681105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Bengsch F, Knoblock DM, Liu A, et al. CTLA-4/CD80 pathway regulates T cell infiltration into pancreatic cancer. Cancer Immunol Immunother. 2017;66(12):1609–17. 10.1007/s00262-017-2053-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Bauden M, Tassidis H, Ansari D. In vitro cytotoxicity evaluation of HDAC inhibitor Apicidin in pancreatic carcinoma cells subsequent time and dose dependent treatment. Toxicol Lett. 2015;236(1):8–15. 10.1016/j.toxlet.2015.03.017. [DOI] [PubMed] [Google Scholar]
- 61.Xian G, Zhao J, Qin C, et al. Simvastatin attenuates macrophage-mediated gemcitabine resistance of pancreatic ductal adenocarcinoma by regulating the TGF-β1/Gfi-1 axis. Cancer Lett. 2017;385:65–74. 10.1016/j.canlet.2016.11.006. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
The datasets used and/or analysed during the current study are available from NCBI Gene Expression Omnibus (GEO: GSE19279) https://www.ncbi.nlm.nih.gov/geo/ and the cancer genome database (TCGA -PAAD) https://portal.gdc.cancer.gov.









