Abstract
Background
Prostate adenocarcinoma (PRAD) is a leading cause of male mortality,
with NK/T cell communication being key areas of the research.
Methods
The Seurat package was utilized to normalize and reduce the
dimensionality of the single-cell data, and CellMarker 2.0 was employed
for cell annotation. CellChat was utilized to construct the
ligand-receptor interaction network of cell subsets. Differentially
expressed genes (DEGs) were filtered by the limma package. Univariate
Cox and the LASSO regression in the glmnet package were used to obtain
biomarkers and develop a risk model. The survminer package was used to
calculate the optimal threshold for dividing patients into high-risk
and low-risk groups, and then Kaplan-Meier (KM) survival analysis was
performed. Single-sample GSEA (ssGSEA), TIMER, and ESTIMATE packages
were employed for immune infiltration analysis. Pathway analysis was
conducted for the low- and high-risk groups using GSEA. Immunotherapy
responses were evaluated by adopting TIDE method. Additional cellular
validation (quantitative real-time PCR, CCK-8, Transwell, and scratch
assay) was implemented to confirm the effects of feature genes on PRAD.
Results
Compared with the benign group, NK/T cells were the cell type with the
greatest changes in the tumor group, and their communication intensity
was relatively high among all cell types. A RiskScore model was
constructed as follows:
[MATH:
0.579*FOXS<
mi
mathvariant="italic">1 + 0.345
*GPC6 + 0.385
*ISYNA1 + 0.418
*ITGAX + 0.792*MG<
/mi>AT4B +
0.368*PRR7 + 0.458
*REXO2 :MATH]
. Analysis of the differences between the two risk groups showed that
the level of immune infiltration was higher in the high-risk group, and
it was significantly enriched in immune-correlated pathways, while the
low-risk group was mainly enriched in metabolism-related pathways. TIDE
analysis indicated that the high-risk group had higher immune escape
potential. The cellular validation assays have revealed the higher
expression of seven biomarkers in PRAD groups. Further, ISYNA1
knockdown inhibited the proliferation, migration, and invasion ability
of PRAD cells.
Conclusion
The current research reveals key communication genes in PRAD, offering
new possibilities for the exploration of new therapeutic targets.
Keywords: NK/T cell communication, tumor microenvironment, prostate
cancer, immune escape, machine learning
1. Introduction
Globally, prostate cancer (PCa) is a common cause of death among men.
Statistics show that in 2022, there were 1,466,680 new PCa cases
globally, accounting for 7.3% of new cancers and ranking fourth
([29]1). More than 95% of PCa cases are adenocarcinomas, with the
majority originating from acini and a minority originating from ducts
([30]2). Its incidence is closely related to location and age ([31]3,
[32]4). In the early stage, prostate adenocarcinoma (PRAD) has
relatively mild symptoms, with the most common being dysuria and
increased frequency of urination ([33]5). In the advanced stage, PRAD
may present with urinary retention and back pain ([34]5). Currently,
the main treatment method for PCa is androgen deprivation therapy, but
it is difficult to completely cure metastatic castration-resistant
prostate cancer (mCRPC) caused by cancer metastasis ([35]6). The
research on immunotherapy for PRAD mainly focuses on Sipuleucel-T and
monoclonal antibodies ([36]7), but experiments have found that their
efficacy in PRAD patients is not yet significant ([37]8, [38]9). Given
the long natural course of PRAD and the relatively high mortality rate
of moderate or high-risk local, locally advanced or metastatic cancers
([39]10), it is necessary to establish a prognostic model related to
PRAD, which helps in the stratified treatment of patients and the
selection of medical regimens.
The single-cell RNA sequencing (scRNA-seq) technique has offered
unprecedented opportunities for studying intercellular interactions
within the tumor microenvironment (TME) in recent years ([40]11),
particularly in the research of immune cells such as T cells and
natural killer (NK) cells. NK/T cells play pivotal roles in tumor
immune evasion and immune surveillance ([41]12). Studies have found
that combinations of NK-cell-based immunotherapy and T-cell-based
immunotherapy may be beneficial for tumor immunotherapy ([42]13).
Nguyen et al. suggest that further detection of tumour-infiltrating
lymphocytes in prostate cancer may help to gain insight into the
regulatory mechanisms of T cells in the TME ([43]14). In addition, NK/T
cells interact with tumor cells and other immune cells through cellular
communication ([44]15). Currently, the systematic inference of
ligand-receptor-mediated intercellular communication has become a
research hotspot, which is closely linked to tumorigenesis ([45]16,
[46]17). Understanding these intercellular communication mechanisms can
uncover new immune escape pathways and provide novel strategies for
immunotherapy in PRAD.
In this study, by analyzing single-cell data from PRAD, we identified
the cell communication receptor-ligand genes between NK/T cells and
other cell types, and constructed their interaction networks. Through
bioinformatics methods, we screened for differential genes related to
the communication receptor-ligands between NK/T cells and other cells.
This study developed a risk model with these key genes and conducted
prognostic analysis. Then, we performed in vitro experimental
verification on the obtained biomarkers and evaluated the immune
infiltration levels and immune therapy responses of different risk
groups. These findings not only provide potential biomarkers for early
detection and prognostic assessment of PRAD but also open up new
possibilities for future stratified and personalized treatment
developments in PRAD.
2. Material and methods
2.1. Collection and processing of data
The gene expression data and related clinical data of PRAD patients
were obtained from the The Cancer Genome Atlas (TCGA,
[47]https://portal.gdc.cancer.gov) database ([48]18). The FPKM values
of TCGA-PRAD RNA sequencing (RNA-Seq) data was transformed into TPM and
then log2-converted. After retaining the samples with complete
progression-free interval (PFI), 495 PRAD samples and 52 adjacent
normal control samples were recruited. The expression data and clinical
data (MSKCC, Cancer Cell 2010) of PRAD were extracted from the
cBioPortal website ([49]https://www.cbioportal.org/). After screening,
a sum of 131 tumor samples were included. From Gene Expression Omnibus
(GEO, [50]https://www.ncbi.nlm.nih.gov/geo/), we sourced four benign
and four PRAD single-cell samples in [51]GSE193337. The library
construction platform was 10x Genomics, and the sequencing platform was
Illumina HiSeq 4000.
2.2. ScRNA-seq analysis
The Seurat object was created by using the CreateSeuratObject function
in the Seurat package ([52]19). Cells with the number of retained genes
ranging from 200 to 8000 and the quantity of mitochondrial genes less
than 20% were reserved. Next, the NormalizeData function ([53]19) was
used for normalization. After the principal component analysis (PCA)
dimensionality reduction, batch effect was eliminated by the harmony
package ([54]20). Then, the RunUMAP function ([55]19) was utilized for
dimensionality reduction by uniform manifold approximation and
projection (UMAP). Finally, cells were clustered by the FindNeighbors
and FindClusters functions ([56]19) with the parameters of dims = 1:30
and resolution = 0.1. According to the marker genes provided by the
CellMarker2.0 database, the cell types were annotated ([57]21).
2.3. Cell communication
CellChat ([58]22) was used to develop the ligand-receptor interaction
network of cell subpopulations. The netVisual_bubble and
netVisual_circle packages ([59]22) were utilized to display the bubble
plots of receptors and ligands between NK/T cells and other cells and
the number of communications between them.
2.4. Screening of differently expressed genes differentially expressed genes
(DEGs)
Differential analysis between the tumor and control sample groups in
the TCGA cohort was performed in the limma package ([60]23). The
screening criteria were that |log 2fold change (FC)| > log2(1.5) and p
< 0.05, thus obtaining the DEGs. Then, the intersection was taken
between the mRNAs related to the receptor and ligand genes of the
communication between NK/T cells and other cells (cor > 0.5, p < 0.01)
and the TCGA DEGs to obtain the mRNAs related to the communication of
NK/T cells.
2.5. Development of the risk model and validation
The TCGA-PRAD samples were assigned randomly at the ratio of 5:5 into
training set and test set. The survival package ([61]24) was utilized
to conduct univariate Cox regression analysis to determine the
prognostic relevance of the intersection genes and p < 0.05 was
selected for filtering. Subsequently, the glmnet package ([62]25) was
employed to carry out LASSO COX regression analysis and stepwise
regression to further narrow down the gene scope. Through multivariate
analysis, the key genes and their corresponding coefficients were
calculated, and the RiskScore for patients was calculated based on the
formula:
[MATH:
RiskScore = Σβi ×
Expi :MATH]
where βi refers to the regression coefficient of each key gene, and
Expi represents the expression of the corresponding gene. Then, the
survminer package ([63]26) was employed to calculate the optimal
threshold to classify the patients into low- and high-risk groups. KM
survival analysis was performed, and receiver operating characteristic
(ROC) curve model was constructed to predict the prognostic performance
of the TCGA training set and test set. The area under the curve (AUC)
value is a robust metric for assessing the classification accuracy of
the RiskScore model. By comparing the AUC values to the assessment
criteria, we can determine the effectiveness of the model in
distinguishing between high- and low-risk patients. To better validate
whether the model was robust, the same method was employed to the MSKCC
dataset for verification.
2.6. Analysis of the tumor immune microenvironment (TIME)
To analyze the association between RiskScore and the TIME, the
infiltration status of immune cells was calculated using different
methods. The ssGSEA function of GSVA ([64]27, [65]28) was utilized to
compute the scores of 28 types of tumor infiltrating immune cells
(TIICs) (1). The TIMER online tool ([66]http://cistrome.org/TIMER) was
utilized to calculate six immune scores, and the ESTIMATE package
([67]29) was used to score the immune cells.
2.7. Pathway enrichment analysis
The gene set enrichment analysis (GSEA) ([68]30) was employed for
pathway analysis to explore the pathways of diverse biological
processes in the two risk groups. The candidate gene sets from the
Kyoto Encyclopedia of Genes and Genomes (KEGG) database were used to
conduct GSEA ([69]31).
2.8. Evaluation of immunotherapy
The TIDE method was adopted to evaluate immunotherapy response. The
standardized transcriptome data were uploaded into the TIDE website
([70]http://tide.dfci.harvard.edu/) to calculate the TIDE score, with a
higher TIDE prediction score indicating greater immune escape
possibility and lower immunotherapy efficacy.
2.9. Cell culture
In this study, the cell lines, namely human normal prostate epithelial
cells PNT1A (cat.no RE59812) and human PCa cells DU145 (cat.no YS101C),
were sourced from Shanghai Yaji Biotechnology Co., Ltd (Shanghai,
China) ([71]http://www.yajimall.com/). All cells were cultured in
Roswell Park Memorial Institute (RPMI) 1640 (11875093, Gibco, Grand
Island, NY, USA) with the supplementation of 10% fetal bovine serum
(FBS) (S9020, Solarbio Lifesciences, Beijing, China). All cells were
incubated in the incubator at 37°C with 5% CO[2]. The cells underwent
short tandem repeat (STR) identification, and the results of mycoplasma
detection for these cells were found to be negative.
2.10. Quantitative real-time PCR
Following the instructions, total RNA was isolated from PNT1A and DU145
cells utilizing the TriZol total RNA extraction kit (15596026CN,
Invitrogen, Carlsbad, CA, USA). Subsequently, the concentration of the
isolated RNA was measured. Then, complementary DNA was synthesized by
reverse transcription with a relevant assay kit (D7178S, Beyotime,
Shanghai, China). After that, SYBR Green qPCR Mix (D7260, Beyotime,
Shanghai, China) was used for the PCR assay according to the protocols.
The conditions for qPCR included an initial step at 94°C for 30
seconds, succeeded by 40 cycles consisting of 5 seconds at 94°C and 30
seconds at 60°C. Finally, the relative level was calculated by the
2^-ΔΔCT method ([72]32), with GAPDH as a reference gene. Based on
National Center for Biotechnology Information (NCBI) sequences, the
qRT-PCR primers used in this study were designed using Primer Premier 6
software. The primer sequences were presented in [73]Supplementary
Table 1 .
2.11. Cell transfection
For the liposome transfection, the small interfering RNA against ISYNA1
(si-ISYNA1) and the negative control small interfering RNA (si-NC) were
all purchased from Merck KGaA (Shanghai, China) and transfected into
DU145 cells utilizing lipofectamine 2000 transfection reagent
(11668027, Invitrogen, Carlsbad, CA, USA) as per the manuals. The
sequences applied for the transfection were
5’-GCACCCATCATGCTGGACCTA-3’(si-ISYNA1#1) and
5’-CAGAAGAATGGTACAAATCCAAG-3’(si-ISYNA1#2).
2.12. Cell viability
DU145 cells in the logarithmic growth phase were plated into a 96-well
dish at a density of 1 × 10^4 cells per well and incubated at 37°C with
5% CO[2] for durations of 0, 24, 48, or 72 hours. Following this, 10 μL
of CCK-8 reagent was added to each well, and the samples were incubated
at 37°C for 2 hours. For the construction of the CCK-8 curve,
absorbance measurements were taken at 450 nm, which served as the
y-axis, while time was plotted on the x-axis.
2.13. Cell invasion assay
For the cell invasion assay, 1 × 10^5 DU145 cells were suspended in 200
μL of serum-free medium and cultured in the upper chamber of the
Transwell (3422, Corning, Inc., Corning, NY, USA) coated with Matrigel
(C0372, Beyotime, China), while 700 μL of medium containing 10% bovine
serum were supplemented to the lower Transwell chamber. After
incubation, the chamber was taken out and gently rinsed with phosphate
buffered saline (PBS) buffer (P1010, Solarbio Lifesciences, Beijing,
China) to remove the non-invasive cells. Subsequently, 4%
paraformaldehyde (P1110, Solarbio Lifesciences, Beijing, China) was
employed for fixing the cells, which were dyed by 0.1% crystal violet
(G1063, Solarbio Lifesciences, Beijing, China) at room temperature for
20 minutes. The invasive cells were observed with an optical microscope
(DP27, Olympus, Tokyo, Japan) in three randomly selected fields of view
([74]33). Finally, to ensure the accuracy and reproducibility of cell
counting, we used ImageJ software for automated cell counting.
2.14. Cell migration assay
The DU145 cells (5 × 10^5 cells/well), after being transfected, were
grown in a 6-well plate containing media devoid of serum. Upon reaching
full confluence, an artificial wound was introduced into the monolayer
using a 200-μL sterile pipette tip. Following a 48-hour incubation
period, the cells were photographed under an inverted optical
microscope (DP27, Olympus, Japan). Subsequently, the percentage of
wound closure (%) was calculated to reflect the migration ability of
the PRAD cells, ensuring uniqueness in reporting ([75]34).
2.15. Statistical analysis
All the statistical data were analyzed in R language (version 3.6.0).
Experimental data were analyzed using GraphPad Prism 8.0 software. The
Wilcoxon rank-sum test was used to compare the differences between
two-group continuous variables, the Spearman method was employed to
calculate the correlations, the log-rank test was utilized to compare
the survival differences among patients in each grouping. Unpaired
t-test and one-way analysis of variance were applied for the comparison
on the experimental data, and p < 0.05 signified a statistical
significance.
3. Results
3.1. Single-cell atlas of PRAD
Single-cell clustering and annotation analysis were conducted on
samples from benign and tumor tissues of PRAD, identifying a total of
seven cell subpopulations: NK/T cells, Macrophage cells, Epithelial
cells, Fibroblast cells, Mast cells, Endothelial cells, and B cells
([76] Figure 1A ). [77]Figures 1B, C depict the marker genes for each
of these cell subpopulations. Among them, high expression of CD3D and
NKG7 was observed in NK/T cells; EPCAM in epithelial cells; VWF in
endothelial cells; CD163 in macrophages; COL1A1 and ACTA2 in
fibroblasts; CD79A in B cells; and TPSAB1 and CPA3 genes in mast cells.
Analysis of the distribution of various cell types between the benign
and tumor groups revealed that, compared to the benign group, NK/T
cells exhibited the highest proportional increase in the tumor group
([78] Figure 1D ).
Figure 1.
[79]Figure 1
[80]Open in a new tab
Single-cell atlas of PRAD. (A) UMAP dimensionality reduction plot for
single-cell clustering and annotation of benign and tumor samples from
PRAD. (B) Bubble plot for annotating the expression of marker genes in
subpopulations. (C) Violin plot illustrating the expression of marker
genes. (D) Proportion of each cell subpopulation within each sample
between the two groups.
3.2. Signal communication among various cell types
Analysis of the number of communications among various cell types
revealed that B cells had the least number of communications among all
cell types ([81] Figure 2A ). Examination of the intensity of
communications among cells showed that NK/T cells exhibited high
communication intensity, indicating their active role in the TME ([82]
Figure 2B ). To visually present this point more clearly, the
communication intensity of each cell type was further detailed, and the
results demonstrated that the communication intensity between NK/T
cells and Macrophage cells, Epithelial cells, as well as Endothelial
cells was particularly prominent ([83] Figure 2C ). These findings
showed that NK/T cells played a pivotal role in immune regulation
within the TME and have close interactions with other key cell types.
[84]Figure 2D further illustrates the receptors and ligands involved in
cellular communications.
Figure 2.
[85]Figure 2
[86]Open in a new tab
Analysis of signaling communication between NK/T Cells and other cell
types. (A) Number of communications between cells. (B, C) Intensity of
communications between cells. (D) Ligand-receptor mediated cellular
communications between NK/T cells and other cell types.
3.3. Identification of DEGs related to communication receptors and ligands
Differential analysis was conducted between tumor and control sample
groups in the TCGA cohort, resulting in 1951 downregulated genes and
971 upregulated genes ([87] Figures 3A, B ). Next, these DEGs were
intersected with mRNAs linked to communication receptors and ligands
between NK/T cells and other cells (correlation coefficient > 0.5, p <
0.01). This intersection identified a total of 2026 overlapping genes
that are associated with communication between NK/T cells ([88]
Figure 3C ).
Figure 3.
[89]Figure 3
[90]Open in a new tab
Identification of DEGs. (A) Volcano plot on the distribution of DEGs
between PRAD and control samples in TCGA. (B) Heatmap illustrating the
expression of the top 50 DEGs in TCGA. (C) Venn diagram showing the
intersection between mRNAs related to communication receptors and
ligands between NK/T cells and other cells, as well as the DEGs in
TCGA.
3.4. Model construction and validation in PRAD
The TCGA-PRAD samples were assigned at the ratio of 5:5 into a training
set and a testing set to construct the model. Using the data from the
training set, the intersected genes were subjected to univariate Cox
proportional hazards regression. Subsequently, LASSO COX and stepwise
regression were applied to further shrink the range of genes, and seven
genes independently associated with prognosis were screened out for
constructing the RiskScore model ([91] Figures 4A, B ). The formula for
this RiskScore model is ([92] Figure 4C ):
Figure 4.
[93]Figure 4
[94]Open in a new tab
Construction and validation of the risk model. (A, B) Changes in the
regression coefficients of gene features in the LASSO regression model
and the optimal penalty parameter (λ) determined by cross-validation.
(C) Genes and their coefficients in the risk model. (D-G) 1 - 5-year
ROC curves of the risk model and differences in PFI between the
high-risk and low-risk groups in the training set (D), test set (E),
TCGA cohort (F), and MSKCC cohort (G). (H) Expression of seven
prognostic genes in the TCGA cohort. (I) Expression of seven prognostic
genes in the MSKCC cohort. ns denotes p > 0.05, **p < 0.01, ****p <
0.0001. * means vs. High-risk.
[MATH:
RiskScore = 0.579
* FOXS1 + 0.345
* GPC6 + 0.385
* ISYNA1 + 0.418
* ITGAX + 0.79
2 * MGAT4B +
0.368 * P
mi>RR7 + 0.458
* REXO2 :MATH]
Patients were classified into high- and low-risk group by the optimal
cut-off value of the RiskScore. The ROC curve demonstrated that the
TCGA cohort, training set, and testing set all exhibited relatively
high AUC values at various time points, which verified its good
classification accuracy. Moreover, compared with those with a low-risk
score, the PFI of patients with a high-risk score was significantly
lower ([95] Figures 4D-F , p < 0.001). The same method was used to
validate the MSKCC dataset, and the model also had a relatively high
AUC value. There was a significant difference in prognosis between two
risk groups ([96] Figure 4G ). By analyzing the expressions of
prognostic genes in the TCGA cohort and the MSKCC cohort, it was found
that, except for the REXO2 gene which showed no significant difference
between the two risk groups in the MSKCC dataset, the expressions of
the other genes in the low-risk group were all remarkably lower ([97]
Figures 4H-I , p < 0.05).
3.5. Correlation between the RiskScore and TIME
To analyze the relationship between RiskScore and TIME, different
methods were used to calculate the infiltration of immune cells.
Analyzing the results of immune cell infiltration assessment by
ESTIMATE, it was found that the low-risk group had lower StromalScore,
ImmuneScore and ESTIMATEScore than the high-risk group, indicating that
the immune infiltration level was lower in the low-risk group ([98]
Figure 5A , p < 0.05). Based on TIMER, the immune cell scores of the
TCGA dataset were calculated, and the results showed that the scores of
Neutrophil, B_cell, Dendritic, Macrophage, CD4_Tcell were all
remarkably lower in the low-risk group ([99] Figure 5B , p < 0.01).
Using ssGSEA functional analysis to score 28 types of immune cells
([100]35), it could be seen that the infiltration scores of multiple
immune cells such as myeloid derived suppressor cells (MDSC), activated
CD8 T cell, NKT cell, regulatory T cell (Treg), NK cell in the low-risk
group were all significantly lower ([101] Figure 5C , p < 0.05).
Figure 5.
[102]Figure 5
[103]Open in a new tab
Differences in immune infiltration between the high-risk and low-risk
groups. Differences in ESTIMATE (A), Timer (B), and ssGSEA (C) scores
between the high-risk and low-risk groups in the TCGA cohort. *p <
0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. ns (abbreviation of
"non-significant") means p > 0.05.
3.6. Differences in enriched pathways between high-risk and low-risk groups
The results of KEGG pathway enrichment analysis demonstrated that the
high-risk group was significantly enriched in primary immunodeficiency,
malaria, complement and coagulation cascades, ECM−receptor interaction,
staphylococcus aureus infection pathways ([104] Figure 6A ). The
low-risk group was significantly enriched in arginine and proline
metabolism, beta−Alanine metabolism, Butanoate metabolism, mineral
absorption and valine, leucine and isoleucine degradation pathways
([105] Figure 6B ).
Figure 6.
[106]Figure 6
[107]Open in a new tab
KEGG enrichment pathway analysis. (A, B) KEGG enrichment pathway
analysis of high-risk and low-risk groups.
3.7. Characterization of immune escape potential and immunosuppression in
PRAD patients from different risk groups
TIDE was employed to estimate the potential clinical immunotherapy
benefit to the two risk groups. The results demonstrated that in the
TCGA cohort, the high-risk group had the highest TIDE score, showing a
higher likelihood of immune escape in the high-risk group and less
immunotherapy benefit ([108] Figure 7 , p < 0.05). Moreover, in the
high-risk group, the scores of dysfunction, exclusion, MDSC, and cancer
associated fibroblast (CAF) were significantly higher than those in the
low-risk group, which revealed the significant immunosuppressive
features and complex mechanisms in the TME ([109] Figure 7 , p < 0.05).
Figure 7.
[110]Figure 7
[111]Open in a new tab
Differences in TIDE scores between high-risk and low-risk groups in
TCGA.
3.8. In vitro experiments to validate the function of key genes in PRAD cells
and the potential role of ISYNA1
Subsequently, the roles of seven characteristic genes in PRAD were
validated by in vitro experiments. The results showed that all six
genes (GPC6, ISYNA1, ITGAX, MGAT4B, PRR7, and REXO2), except FOXS1,
were significantly highly expressed in PRAD cells ([112] Figure 8A , p
< 0.01). Since ISYNA1 is highly expressed in DU145 cells and to date,
there are few studies on ISYNA1 in tumor research, especially PRAD. For
this reason, we chose to silence ISYNA1 in order to validate its
potential effect on PRAD cell development ([113] Figure 8B , p < 0.01).
Subsequently, we observed a significant decrease in the proliferation,
migration and invasion ability of DU145 cells after silencing ISYNA1
relative to controls ([114] Figures 8C-E , p < 0.001). These results
suggest the potential impact of our NK/T cell-related key gene-based
screen on PRAD cell genesis and development.
Figure 8.
[115]Figure 8
[116]Open in a new tab
Cell validation results. (A) Quantified mRNA level of seven feature
genes in PRAD cell line DU145 and the control cell line PNT1A. (B) The
qRT-PCR to verify the transfection efficiency of ISYNA1 between the
si-negative control (si-NC) and si-ISYNA1 groups. (C) Assessment of the
proliferation level of DU145 cells after ISYNA1 silencing based on the
CCK-8 assay. (D) The results of Transwell assay on the invasion of
DU145 cells after the silencing of ISYNA1. (E) The results of scratch
assay on the migration of DU145 cells after the silencing of ISYNA1.
**p < 0.01, ***p < 0.001, ****p < 0.0001, and ns stands for no
significant difference.
4. Discussion
According to global data in 2022, although the mortality rate of PRAD
ranks only eighth, its incidence rate ranks fourth ([117]36), and it
has a high recurrence rate, with about one-third of men experiencing
recurrence after local treatment, ultimately leading to the generation
of malignant cells ([118]37). NK cells are cytotoxic immune cells
capable of killing cancer cells in the innate immune system ([119]38).
Research has found that cellular communication functions importantly in
the TME ([120]39), particularly involving NK/T cells, which are
associated with the immune evasion mechanisms of cancers ([121]40). In
this study, by analyzing PRAD-related single-cell data, the interaction
between NK/T cells and other cells was identified, and a risk model
related to communication receptors and ligands was established. These
results provided new insights into the immunotherapy of PRAD and
opening up new potential pathways for personalized cancer treatment.
The risk model in this study includes a total of seven related
molecular markers, namely FOXS1, GPC6, ISYNA1, ITGAX, MGAT4B, PRR7, and
REXO2. As a type of DNA-binding proteins, Forkhead-box (FOX) family
proteins participate in cell growth, embryogenesis, differentiation,
and lifespan ([122]41). FOXS1 has a high expression in pan-cancers, and
the gene is linked to a worse surviva. Its overexpression is related to
high infiltration levels of tumor-associated macrophages (TAM) and
Tregs ([123]42), which is consistent with the result of a higher
infiltration level of Tregs in the high-risk group in this study. GPC6
belongs to the glypican gene family, and its function depends on
interactions with cytoplasmic and/or extracellular ligands ([124]43).
Chen et al.’s research has confirmed that GPC2 in the GPC family can
promote the progression of PRAD ([125]44). ISYNA1 encodes an enzyme
essential for inositol biosynthesis. Silencing ISYNA1 can inhibit the
growth of tumor cells ([126]45). Jia et al.’s research has demonstrated
that it can serve as a biomarker related to pan-cancer immunomodulation
prognosis ([127]46). The in vitro experiments in this study also
confirmed that the knockout of ISYNA1 would influence both the
migration and invasion abilities of PRAD cells. ITGAX usually functions
as a receptor for the extracellular matrix and is associated with tumor
angiogenesis ([128]47, [129]48). Williams et al.’s research has
confirmed that it can act as a susceptibility gene for aggressive PRAD
([130]49). The MGAT4B protein belongs to the glycosyltransferase family
and can recognize and bind to both donor and receptor substrates
([131]50). In liver cancer cells, studies have found that the
expression of MGAT4B is upregulated and it is related to immune evasion
([132]51). PRR7 is a transmembrane adaptor protein (TRAP) and an
important organizer and regulator of immune receptor-mediated signal
transduction ([133]52). It mediates T cell receptor signaling by
assembling the membrane-proximal signalosome ([134]53). REXO2 is a
homologue of the prokaryotic oligoribonuclease existing in human
mitochondria and cytoplasm ([135]54). Previous experiments have
confirmed that REXO2 can serve as a biomarker for PRAD ([136]55). The
above evidence demonstrates the possible roles that these cell
communication ligand-related genes may play in cancer and also implies
their potential as biomarkers for PRAD.
Analysis on the correlation between the RiskScore and the immune
microenvironment revealed that most of the immune scores in the
high-risk group were higher. However, despite the higher scores of NK
cells, the scores of Tregs in the high-risk group were also higher.
Tregs are a type of T lymphocyte with immunosuppressive functions and
functions crucially in the process of the immune system eliminating
tumor cells as well as in immune self-tolerance ([137]56). Tregs play
an immunosuppressive role in the TIME. Studies have confirmed that
Tregs can promote tumor immune evasion through the activation of
transforming growth factor-β (TGF-β) mediated by integrin αvβ8
([138]57, [139]58). Therefore, although there is a strong immune
response in the high-risk group, the high scores of Tregs indicate that
tumors may “escape” immune attacks by inducing an immunosuppressive
environment. In addition, the biomarkers FOXS1 and PRR7 obtained in
this study are both closely related to Treg signal transduction or
infiltration ([140]42, [141]53), which further implies that high-risk
patients affect the TME through the immune evasion mechanism.
The results of pathway enrichment showed that the pathways in the
low-risk group were mainly enriched in metabolic pathways, whereas the
high-risk group were mainly enriched in immune-related pathways. This
reflects that the high-risk group is linked to immune dysfunction,
while the low-risk group is in a relatively healthy state. The TIDE
analysis revealed that the exclusion score of the high-risk group was
high, suggesting that effector immune cells have difficulty effectively
infiltrating into the interior of the tumor, which may be related to
physical barriers or chemical repellent factors in the TME. Meanwhile,
the score of MDSC in the high-risk group was also relatively high. MDSC
is a heterogeneous group of immature bone marrow cells with the
potential to effectively target T cells and suppress autoimmune
responses ([142]59, [143]60). Studies have already demonstrated that
MDSC can influence the suppression of specific T cell responses in
myeloma by inducing the development of Tregs in the TME ([144]61).
Combined with the results of immune infiltration, the high score of
MDSC indicates that these cells may suppress effector immune responses
by influencing Tregs, thereby helping the tumor achieve immune evasion.
The increase in the score of CAF further reflects the activity of CAFs.
CAF activated by TGF-β is one of the most abundant stromal cell types
in almost all solid tumors, and its special role is widely recognized
([145]62). Moreover, TGF-β is closely related to Tregs, NK cells, and
immune evasion ([146]63, [147]64). In conclusion, these characteristics
of the high-risk group jointly reveal the tumor’s strategy of enhancing
its survival ability through multi-level immune suppression mechanisms,
providing important implications for in-depth research on tumor
immunotherapy.
The limitations of this study require further consideration and
refinement. Firstly, the sample size and representativeness of this
study may not fully encapsulate the heterogeneity and diversity of PRAD
within the general population, potentially limiting the
generalizability of the study results. To address this issue, future
research should include larger and more diversified cohorts of PRAD
samples, encompassing different stages and subtypes of the disease as
well as various ethnic groups. Additionally, the data verification in
this study mainly relies on bioinformatics techniques and in vitro
experiments, lacking the support of in vivo experiments. To verify our
research results more comprehensively, the construction of relevant
animal models is an essential step. Furthermore, the clinical
application of the identified biomarkers and risk models needs to be
further explored and validated in clinical trials.
5. Conclusion
The present study successfully developed a risk model closely related
to communication receptors and ligands, marking significant progress in
exploring the immune mechanisms of PRAD. The establishment of this
model not only deepened our understanding of the complex immune
microenvironment of PRAD, but also provided strong theoretical support
and practical tools for the formulation of stratified and personalized
strategies for PRAD. Furthermore, we confirmed a crucial role of NK/T
cells in the development of PRAD, offering new targets and ideas for
the progression of novel immunotherapies. In the future, we anticipate
enhancing the antitumor activity of NK/T cells by modulating the
expression or function of communication receptors and ligands, or
designing more precise targeted drugs to inhibit the immune escape
mechanisms of tumor cells, thereby further improving the treatment
outcomes of PRAD and bringing more effective therapy and better quality
of life to patients.
5.1. Scope statement
Compared with the benign group, NK/T cells were the cell type with the
greatest changes in the tumor group, and their communication intensity
was relatively high among all cell types. A RiskScore model was
constructed as follows:
[MATH:
0.579*FOXS<
mi
mathvariant="italic">1 + 0.345
*GPC6 + 0.385
*ISYNA1 + 0.418
*ITGAX + 0.792*MG<
/mi>AT4B +
0.368*PRR7 + 0.458
*REXO2 :MATH]
. Analysis of the differences between the two risk groups showed that
the level of immune infiltration was higher in the high-risk group, and
it was significantly enriched in immune-correlated pathways, while the
low-risk group was mainly enriched in metabolism-related pathways. TIDE
analysis indicated that the high-risk group had higher immune escape
potential. The cellular validation assays have revealed the higher
expression of seven biomarkers in PRAD groups. Further, ISYNA1
knockdown inhibited the migratory and invasive capabilities of PRAD
cells. The current research reveals key communication genes in PRAD,
offering new possibilities for the exploration of new therapeutic
targets.
Glossary
PCa
prostate cancer
PRAD
prostate adenocarcinoma
mCRPC
metastatic castration-resistant prostate cancer
scRNA-seq
single-cell RNA sequencing
TME
tumor microenvironment
NK
natural killer
TCGA
The Cancer Genome Atlas
FPKM
ragments per kilobase of exon model per million mapped fragments
RNA-Seq
RNA sequencing
TPM
transcripts per million
PFI
progression-free interval
GEO
Gene Expression Omnibus
PCA
principal component analysis
UMAP
uniform manifold approximation and projection
DEGs
differentially expressed gene
LASSO
least absolute shrinkage and selection operator
KM
Kaplan-Meier
ROC
receiver operating characteristic
TIME
tumor immune microenvironment
RPMI
Roswell Park Memorial Institute
FBS
fetal bovine serum
qRT-PCR
quantitative real-time PCR
NCBI
National Center for Biotechnology Information
PBS
phosphate buffered saline
ssGSEA
single sample gene set enrichment analysis
TIIC
tumor infiltrating immune cell
KEGG
kyoto encyclopedia of genes and genomes
TIDE
tumor immune dysfunction and exclusion
AUC
Area Under the Curve
CAF
cancer associated fibroblast
FOX
forkhead-box
TAM
tumor-associated macrophages
Treg
regulatory T cell
TRAP
transmembrane adaptor protein
TGF-β
transforming growth factor-β.
Funding Statement
The author(s) declare that financial support was received for the
research and/or publication of this article. This study was supported
by National Natural Science Foundation of China (81802580).
Data availability statement
The datasets presented in this study can be found in online
repositories. The names of the repository/repositories and accession
number(s) can be found in the article/[148] Supplementary Material .
Ethics statement
Ethical approval was not required for the studies on humans in
accordance with the local legislation and institutional requirements
because only commercially available established cell lines were used.
Author contributions
KZ: Data curation, Formal Analysis, Methodology, Project
administration, Validation, Visualization, Writing – original draft,
Writing – review & editing. HX: Conceptualization, Data curation,
Formal Analysis, Methodology, Project administration, Resources,
Software, Visualization, Writing – original draft, Writing – review &
editing. FZ: Formal Analysis, Investigation, Methodology, Resources,
Supervision, Visualization, Writing – review & editing. YH:
Conceptualization, Data curation, Formal Analysis, Funding acquisition,
Methodology, Project administration, Supervision, Visualization,
Writing – original draft, Writing – review & editing.
Conflict of interest
The authors declare that the research was conducted in the absence of
any commercial or financial relationships that could be construed as a
potential conflict of interest.
Generative AI statement
The author(s) declare that no Generative AI was used in the creation of
this manuscript.
Publisher’s note
All claims expressed in this article are solely those of the authors
and do not necessarily represent those of their affiliated
organizations, or those of the publisher, the editors and the
reviewers. Any product that may be evaluated in this article, or claim
that may be made by its manufacturer, is not guaranteed or endorsed by
the publisher.
Supplementary material
The Supplementary Material for this article can be found online at:
[149]https://www.frontiersin.org/articles/10.3389/fimmu.2025.1564784/fu
ll#supplementary-material
[150]Table1.docx^ (17.8KB, docx)
References