2,394
Views
2
CrossRef citations to date
0
Altmetric
Research Article

Integrative exploration of the mutual gene signatures and immune microenvironment between benign prostate hyperplasia and castration-resistant prostate cancer

ORCID Icon, , , &
Article: 2183947 | Received 03 Nov 2022, Accepted 17 Feb 2023, Published online: 28 Mar 2023

Figures & data

Figure 1. Weighted gene co-expression network analysis (WGCNA).

(A) This study conducted a systematic review of the literature and integrative analysis of 10 bulk-RNA sequencing or microarray studies and three single-cell RNA sequencing; (B) with weighted gene co-expression network analysis, seventeen modules were identified in 206 cases; (C) the expression of the above-clustered genes were highly correlated; (D) the cells from the prostate tissues were clustered into nine populations according to classical markers for indicated cell types; (E) the gene signature significance between significant modules and indicated prostate lesion were highly correlated; (F–H) the gene ontology biological process analyses of significant module genes showed that the chromatin remodeling and DNA damage repair pathways were exclusively overexpressed in the prostate cancer.
Figure 1. Weighted gene co-expression network analysis (WGCNA).

Figure 2. Analysis of copy number variations in benign prostate hyperplasia and prostate cancer.

(A) The canonical molecular markers for nine subpopulations from prostate tissues was presented; (B) UMAP embedding of jointly analyzed single-cell transcriptomes from normal and malignant prostate; (C) The single-cell RNA sequencing data was classified by disease state or cell types; (D) The comparison of CNV scores in luminal and basal cells between BPH and CaP showed that CNV was higher in the luminal cells. (E) The estimation of CNV by inferCNV in BPH and CaP was compared with normal epithelial of the prostate. (F) Recurrent CNV in the prostate cancer cohort was identified with GISTIC 2.0 (q-value < 0.2). (G) The top 5 recurrently altered regions are annotated, when the 10q23.31 was frequently deleted in prostate cancer, where PTEN was located.
Figure 2. Analysis of copy number variations in benign prostate hyperplasia and prostate cancer.

Figure 3. The difference in transcription factors between BPH and prostate cancer.

(A) Heatmap of regulon activity was analyzed by SCENIC with default thresholds for binarization in BPH and CaP. The "regulon" refers to the regulatory network of transcription factors (TFs) and their target genes; (B) most TFs were generally active in both BPH and prostate cancer, except for the indicated exclusive TFs; (C,D) ridge plot visualizes the comparison of indicated regulon activity in basal cells and luminal cells among normal prostate, BPH tissues, and prostate cancer tissues. FOSL1 and MYC were active in BPH; (E) SPDEF was exclusively activated in luminal cells of prostate cancer; (F) XBP1 was active in both BPH and prostate cancer.
Figure 3. The difference in transcription factors between BPH and prostate cancer.

Figure 4. The unique gene signatures and signaling pathways in BPH.

PPI, protein–protein interaction; DEGs, differential expression of genes; BPH, benign prostate hyperplasia; CaP, prostate cancer; CRPC, castration-resistant prostate cancer; G.O., gene ontology.

(A) The genes of differential expression of genes (DEGs) and significant modules from WGCNA were selected to find the overlap signatures; (B) five overexpressed genes (H2BC15, PSMA2, H4C3, LIMD2, ATP1B2) were found in the BPH DEGs and significant modules; (C) the five gene signatures were not significantly associated with the prognosis of prostate cancer; (D) the shared genes were found to be functionally active in molecular functions toward translation, post-translational modification, or metabolic pathways.
Figure 4. The unique gene signatures and signaling pathways in BPH.PPI, protein–protein interaction; DEGs, differential expression of genes; BPH, benign prostate hyperplasia; CaP, prostate cancer; CRPC, castration-resistant prostate cancer; G.O., gene ontology.

Figure 5. The role of mutual gene signatures between BPH and CRPC in prognosis and anti-PD1 response.

Forty-two genes were shared between the BPH and CRPC based on differential gene analysis and WGCNA; (B) The shared genes were mainly focused on MAPK phosphorylation and MYC signaling; (C) The protein-protein interaction network and clusters analysis of the shared gene signatures were conducted; (D) The cross-validation error rates were good. (E) Lasso coefficient profiles of the shared gene signatures associated with the overall survival of prostate cancer (F) Boxplots indicating the expression of DDA1 varied in various prostatic lesions; (G) The association with the overall survival and disease-free survival in prostate cancer was not significantly different; (H) The expression of DDA1/C14orf1 were significantly and negatively correlated with the survival probability of cancer patients receiving anti-PD1 treatment.
Figure 5. The role of mutual gene signatures between BPH and CRPC in prognosis and anti-PD1 response.

Figure 6. The comparison of immune microenvironment between BPH and CRPC.

(A) To further explore the role of immunotherapy, the immune infiltration was statistically compared among prostate lesions; (B) correlation of the immune cells in prostate tissues was high in macrophage and T cells; (C–F) the violin plot showed the difference in the Immune Score (C), ESTIMATE Score (D), Stromal Score (E), and Tumor Purity (F) of every case was presented and colored by the type of prostatic lesions; (G,H) the box plot showed the difference in expression of CD274 (G) and TIGIT (H) among different prostate lesions were increased in prostate cancer, but not BPH and CRPC; (I) compared with CaP, the infiltration of CD8 + T cells, M1 macrophage, and NK cells were significantly increased in BPH and CRPC.
Figure 6. The comparison of immune microenvironment between BPH and CRPC.

Figure 7. The correlation between selected gene signatures with the tumor immune microenvironment.

(A–D) The expression of OXAL1 (A), ERG28 (B), DDA1 (C), and OGFOD1 (D) were all negatively and significantly correlated with the infiltration of CD8+ T cells according to the TCGA dataset; (E) the mRNA expression of OXAL1, ERG28, DDA1, and OGFOD1 and CD8+ T cells were significantly correlated in prostate cancer tissues with adjacent normal tissues; (F) the copy number variations (CNV) of OXAL1, ERG28, and OGFOD1 were significantly correlated with CD8+ T cells in prostate cancer tissues; (G) the GSVA score of the four gene signatures was significantly correlated with infiltration of CD8 + T, DC, or NK cells in prostate cancer tissues.
Figure 7. The correlation between selected gene signatures with the tumor immune microenvironment.
Supplemental material

Supplemental Material

Download MS Word (3.8 MB)

Supplemental Material

Download MS Excel (228.5 KB)

Data availability statement

The original contributions presented in the study are included in the article/Supplementary Table S1, further inquiries can be directed to the corresponding author. All the scripts used in this study could be obtained by request to the corresponding author.