Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 38
1.
Hum Vaccin Immunother ; 20(1): 2350101, 2024 Dec 31.
Article En | MEDLINE | ID: mdl-38738709

Clinical guidelines have recently advised combination therapy involving immunotherapy (IO) and tyrosine kinase inhibitors (TKI) as the first-line therapy approach for advanced renal cell carcinoma (RCC). Nevertheless, there is currently no available biomarker that can effectively distinguish the progression-free survival (PFS). RNA-sequencing and immunohistochemistry were conducted on our cohort of metastatic RCC patients, namely ZS-MRCC, who received combination therapy consisting of IO and TKI. We further applied RNA-sequencing, immunohistochemistry, and flow cytometry to examine the immune cell infiltration and functionality inside the tumor microenvironment of high-risk localized RCC samples. SPP1 expression was significantly higher in non-responders to IO-TKI therapy. Elevated levels of SPP1 were associated with poor PFS in both the ZS-MRCC cohort (HR = 2.73, p = .018) and validated in the JAVELIN Renal 101 cohort (HR = 1.61, p = .004). By multivariate Cox analysis, SPP1 was identified as a significant independent prognosticator. Furthermore, there existed a negative correlation between elevated levels of SPP1 and the presence of GZMB+CD8+ T cells (Spearman's ρ= -0.48, p < .001). Conversely, SPP1 expression is associated with T cell exhaustion markers. A significant increase in the abundance of Tregs was observed in tumors with high levels of SPP1. Additionally, a machine-learning-based model was constructed to predict the benefit of IO-TKI treatment. High SPP1 is associated with therapeutic resistance and unfavorable PFS in IO-TKI therapy. SPP1 expression have also been observed to be indicative of malfunction and exhaustion in T cells. Increased SPP1 expression has the potential to serve as a potential biomarker for treatment selection of metastatic RCC.


Carcinoma, Renal Cell , Immunotherapy , Kidney Neoplasms , Osteopontin , Protein Kinase Inhibitors , Humans , Carcinoma, Renal Cell/therapy , Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Kidney Neoplasms/therapy , Kidney Neoplasms/pathology , Male , Female , Immunotherapy/methods , Middle Aged , Protein Kinase Inhibitors/therapeutic use , Osteopontin/metabolism , Osteopontin/genetics , Aged , Tumor Microenvironment/immunology , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Prognosis , Treatment Outcome , Adult , CD8-Positive T-Lymphocytes/immunology , Cohort Studies , Combined Modality Therapy
2.
Discov Oncol ; 15(1): 86, 2024 Mar 22.
Article En | MEDLINE | ID: mdl-38519766

BACKGROUND: Immunotherapy (IO) plus tyrosine kinase inhibitor (TKI) therapy is the first-line recommendation for advanced renal cell carcinoma (RCC), but no biomarker has been approved for it. Annexin A2 (ANXA2) can induce immune escape in tumors. METHODS: Two independent cohorts of advanced RCC treated by IO + TKI were utilized for survival analysis (ZS-MRCC, n = 45; Javelin-101, n = 726). ANXA2 expression was determined by RNA-sequencing. The impact of ANXA2 on the tumor microenvironment was assessed by RNA-sequencing, flow cytometry and immunohistochemistry in two localized RCC datasets (ZS-HRRCC, n = 40; TCGA-KIRC, n = 530). RESULTS: ANXA2 was upregulated in non-responders of IO + TKI therapy (p = 0.027). High-ANXA2 group showed poor progression-free survival (PFS) in both the ZS-MRCC cohort (HR, 2.348; 95% CI 1.084-5.085; P = 0.025) and the Javelin-101 cohort (HR, 1.472; 95% CI 1.043-2.077; P = 0.027). Multivariate Cox regression determined ANXA2 as an independent prognostic factor (HR, 2.619; 95% CI 1.194-5.746; P = 0.016). High-ANXA2 was correlated with decreased proportion of granzyme B+ CD8+ T cells (Spearman's ρ = - 0.40, P = 0.01), and increased TIM-3+ (Spearman's ρ = 0.43, P < 0.001) and CTLA4+ (Spearman's ρ = 0.49, P < 0.001) tumor-infiltrating lymphocytes. A random forest (RF) score was further build by integrating ANXA2 and immune genes, which stratified patients who would benefit from IO + TKI therapy (low-RF score, IO + TKI vs TKI, HR = 0.453, 95% CI 0.328-0.626; high-RF score, IO + TKI vs TKI, HR = 0.877, 95% CI 0.661-1.165; interaction P = 0.003). CONCLUSIONS: Upregulated ANXA2 was associated with poor PFS and therapeutic resistance in RCC treated by IO + TKI therapy, and related with T cell exhaustion. The integrated RF score could stratify patients who would benefit from IO + TKI therapy.

3.
Cancer Med ; 13(7): e7113, 2024 Apr.
Article En | MEDLINE | ID: mdl-38545824

BACKGROUND: In renal cell carcinoma (RCC), no clinically available biomarker has been utilized for checkpoint inhibitor immunotherapy (IO) + tyrosine kinase inhibitor (TKI) combinations. Galectin-1 overexpression is found in tumors, with potential immune-regulating roles. METHODS: RNA-sequencing was performed in two cohorts of RCC treated with IO/TKI combination therapy (ZS-MRCC, JAVELIN-101). Immunohistochemistry and flow cytometry were performed to investigate immune cell infiltration and function in the tumor microenvironment of RCC. The RECIST criteria were used to define response and progression-free survival (PFS). RESULTS: Galectin-1 expression was elevated in RCC with higher stage (p < 0.001) and grade (p < 0.001). Galectin-1 expression was also elevated in non-responders of IO/TKI therapy (p = 0.047). High galectin-1 was related with shorter PFS in both ZS-MRCC cohort (p = 0.036) and JAVELIN-101 cohort (p = 0.005). Multivariate Cox analysis defined galectin-1 as an independent factor for PFS (HR 2.505; 95% CI 1.116-5.622; p = 0.026). In the tumor microenvironment, high galectin-1 was related with decreased GZMB+CD8+ T cells (Speraman's ρ = -0.31, p = 0.05), and increased PD1 + CD8+ T cells (Speraman's ρ = 0.40, p = 0.01). Besides, elevated number of regulatory T cells (p = 0.039) and fibroblasts (p = 0.011) was also found in high galectin-1 tumors. Finally, a random-forest score (RFscore) was built for predicting IO/TKI benefit. IO/TKI therapy showed benefit only in low-RFscore patients (HR 0.489, 95% CI 0.358-0.669, p < 0.001), rather than high-RFscore patients (HR 0.875, 95% CI 0.658-1.163, p = 0.357). CONCLUSIONS: High galectin-1 indicated therapeutic resistance and shorter PFS of IO/TKI therapy. High galectin-1 also indicated CD8+ T cell dysfunction. High galectin-1 could be applied for patient selection of IO/TKI therapy in RCC.


Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Galectin 1/genetics , Galectin 1/therapeutic use , Immune Checkpoint Inhibitors/therapeutic use , Protein-Tyrosine Kinases , Prognosis , Protein Kinase Inhibitors/therapeutic use , Kidney Neoplasms/pathology , Tumor Microenvironment
4.
BMC Urol ; 24(1): 8, 2024 Jan 03.
Article En | MEDLINE | ID: mdl-38172737

BACKGROUND: Checkpoint inhibitor immunotherapy plus tyrosine kinase inhibitor (IO/TKI) have been recently recommended as standard first-line therapy for advanced renal cell carcinoma, while no clinical-available biomarker has been applied. This study aimed to investigate the associations between RUNX3 pathway signature and IO/TKI benefits in renal cell carcinoma (RCC). METHODS: Two IO/TKI cohorts (ZS-MRCC, JAVELIN-101) and one high-risk localized RCC cohort (ZS-HRRCC) were included. All samples were evaluated by RNA-sequencing, and RUNX Family Transcription Factor 3 (RUNX3) pathway were determined by single sample gene set enrichment analysis. Flow cytometry were applied for immune cell infiltration and function. RESULTS: RUNX3 signature was elevated in RCC samples, compared non-tumor tissues (P < 0.001). High-RUNX3 signature was associated with shorter progression-free survival (PFS) in both IO/TKI cohorts (ZS-MRCC cohort, P = 0.025; JAVELIN-101 cohort, P = 0.019). RUNX3 signature also predicted IO/TKI benefit in advanced RCC, compared with TKI monotherapy (interaction p = 0.027). RUNX3 signature was associated with decreased number of GZMB + CD8 + T cells (Spearman's ρ=-0.42, P = 0.006), and increased number of PD1 + CD8 + T cells (Spearman's ρ = 0.29, P = 0.072). Moreover, the integration of RUNX3 signature and GZMB expression showed predictive potential for TKI/IO (log-rank P < 0.001). In addition, the predictive value of RUNX3 signature for IO/TKI benefit was restricted in SETD2-wild type patients (log-rank P < 0.001). Finally, a risk score was established by random forest for IO/TKI benefit, showing remarkable predictive potency (Log-rank P < 0.001). CONCLUSIONS: RUNX3 pathway signature could be a potential predictive biomarker for IO/TKI treatment in advanced RCC, for both prognosis and treatment selection between IO/TKI and TKI monotherapy.


Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/pathology , Immune Checkpoint Inhibitors/therapeutic use , Protein-Tyrosine Kinases , Protein Kinase Inhibitors/therapeutic use , Kidney Neoplasms/pathology , Biomarkers
5.
Neoplasia ; 43: 100919, 2023 09.
Article En | MEDLINE | ID: mdl-37517099

BACKGROUND: Immunotherapy plus tyrosine kinase inhibitor (IO-TKI) has become the first-line management for metastatic renal cell carcinoma (RCC), despite the absence of biomarkers. Recently, pyrroline-5-carboxylate reductase 1 (PYCR1) and proline metabolism have been reported regulatory roles in the anti-tumor response. METHODS: There were three cohorts enrolled: two from our institution (ZS-MRCC and ZS-HRRCC) and one from a clinical trial (JAVELIN-101). The PYCR1expression in each sample was evaluated by RNA sequencing. Flow cytometry and immunohistochemistry were performed to assess immune infiltration. Single-cell RNA-seq (scRNA-seq) data was used for cluster analysis of T cells and macrophages. Primary endpoints were set as response and progression-free survival (PFS). RESULTS: Patients in the low-PYCR1 group had greater objective response rate (52.2% vs 18.2%) and longer PFS in both cohorts (ZS-MRCC cohort, P=0.01, HR=2.80; JAVELIN-101 cohort, P<0.001, HR=1.85). In responders, PYCR1 expression was decreased (P<0.05). In the high PYCR1 group, CD8+ T cells exhibited an exhausted phenotype with decreased GZMB (Spearman's ρ=-0.36, P=0.02). scRNA-seq revealed tissue-resident memory T (Trm) (P<0.05) and tissue-resident macrophage (P<0.01) were decreased in samples with high PYCR1 expression. A machine learning score was further built by random forest, involving PYCR1 and Trm markers. Only in the subgroup with the lower RFscore did IO+TKI show a favorable outcome, compared to TKI monotherapy. CONCLUSIONS: Immunosuppression and IO+TKI resistance were correlated with high PYCR1 expression. T cell exhaustion and dysfunction were also related with the expression of PYCR1. PYCR1 has the potential to be employed as a biomarker to discriminate between IO+TKI and TKI monotherapy as the optimal patient treatment strategy.


Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , CD8-Positive T-Lymphocytes , Immunotherapy , Kidney Neoplasms/drug therapy , Prognosis , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/genetics , delta-1-Pyrroline-5-Carboxylate Reductase
6.
Scand J Immunol ; 98(4): e13304, 2023 Oct.
Article En | MEDLINE | ID: mdl-37294700

Checkpoint inhibitor immunotherapy plus tyrosine kinase inhibitor (IO/TKI) has become the first-line treatment for metastatic renal cell carcinoma (RCC), despite the lack of biomarkers. Cyclin-dependent kinase 6 (CDK6) has shown a regulatory role in antitumour response. The study enrolled two cohorts of metastatic RCC treated by IO/TKI (Zhongshan Hospital [ZS]-MRCC, n = 45; JAVELIN-101, n = 726) and two cohorts of localized RCC (ZS-HRRCC, n = 40; TCGA-KIRC, n = 530). CDK6 was evaluated by RNA-sequencing. Progression-free survival (PFS) was the primary endpoint. The prognostic role of CDK6 was evaluated by survival analysis. The correlation between CDK6 and tumour microenvironment was assessed by immunohistochemistry and flow cytometry. The high-CDK6 group displayed a lower response rate (13.6%) than the low-CDK6 group (56.5%) (P = .002). High-CDK6 was associated with poor PFS in both the ZS-MRCC cohort (high-CDK6, median PFS 6.4 months; low-CDK6, median PFS not reached; P = .010) and JAVELIN-101 cohort (high-CDK6, median PFS 10.0 months; low-CDK6, median PFS 13.3 month; P = .033). High-CDK6 was associated with increased PD1+ CD8+ T cells (Spearman's ρ = .47, P < .001) and decreased Granzyme B+ CD8+ T cells (Spearman's ρ = -.35, P = .030). Finally, a random forest score (RFscore) was built by integrating CDK6 and immunologic genes, which was associated with survival benefits of IO/TKI (RFscore-low, TKI vs IO/TKI, HR = 2.47, 95% CI 1.82-3.35, P < .001; RFscore-high, TKI vs IO/TKI, HR = 0.99, 95% CI 0.75-1.32, P = .963). Elevated CDK6 expression indicated resistance and poor PFS under IO/TKI therapy, which was related to exhausted CD8+ T cells. Integrated RFscore could evaluate the benefits of IO/TKI.


Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/pathology , Prognosis , Kidney Neoplasms/drug therapy , Kidney Neoplasms/pathology , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Protein-Tyrosine Kinases , CD8-Positive T-Lymphocytes/pathology , Cyclin-Dependent Kinase 6/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Retrospective Studies , Tumor Microenvironment
7.
Cancer Res Treat ; 55(4): 1321-1336, 2023 Oct.
Article En | MEDLINE | ID: mdl-37024096

PURPOSE: Immunotherapy (IO) plus tyrosine kinase inhibitor (TKI) has become the first-line treatment for advanced renal cell carcinoma, despite the lack of prognostic biomarkers. Cyclin-dependent kinase 5 (CDK5) affects the tumor microenvironment, which may influence the efficacy of TKI+IO. MATERIALS AND METHODS: Two cohorts from our center (Zhongshan Metastatic Renal Cell Carcinoma [ZS-MRCC] cohort, Zhongshan High-risk Localized Renal Cell Carcinoma [ZS-HRRCC] cohort) and one cohort from a clinical trial (JAVELIN-101) were enrolled. The expression of CDK5 of each sample was determined by RNA sequencing. Immune infiltration and T cell function were evaluated by flow cytometry and immunohistochemistry. Response and progression-free survival (PFS) were set as primary endpoints. RESULTS: Patients of low CDK5 expression showed higher objective response rate (60.0% vs. 23.3%) and longer PFS in both cohorts (ZS-MRCC cohort, p=0.014; JAVELIN-101 cohort, p=0.040). CDK5 expression was enhanced in non-responders (p < 0.05). In the ZS-HRRCC cohort, CDK5 was associated with decreased tumor-infiltrating CD8+ T cells, which was proved by immunohistochemistry (p < 0.05) and flow cytometry (Spearman's ρ=-0.49, p < 0.001). In the high CDK5 subgroup, CD8+ T cells revealed a dysfunction phenotype with decreased granzyme B, and more regulatory T cells were identified. A predictive score was further constructed by random forest, involving CDK5 and T cell exhaustion features. The RFscore was also validated in both cohorts. By utilizing the model, more patients might be distinguished from the overall cohort. Additionally, only in the low RFscore did TKI+IO outperform TKI monotherapy. CONCLUSION: High-CDK5 expression was associated with immunosuppression and TKI+IO resistance. RFscore based on CDK5 may be utilized as a biomarker to determine the optimal treatment strategy.


Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , Kidney Neoplasms/drug therapy , Kidney Neoplasms/genetics , Protein-Tyrosine Kinases , Cyclin-Dependent Kinase 5/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Immunotherapy , Tumor Microenvironment
8.
Cancer Med ; 12(9): 10512-10525, 2023 05.
Article En | MEDLINE | ID: mdl-37031459

BACKGROUND: Immunotherapy (IO) plus tyrosine kinase inhibitor (TKI) emerged as standard first-line therapy for advanced renal cell carcinoma (RCC). The heme Oxygenase 1 (HMOX1) pathway is involved in tumor development and treatment resistance, which may affect the efficacy of TKI + IO. METHODS: Two cohorts from our center (ZS-MRCC, ZS-HRRCC), one cohort from clinical trial (JAVELIN Renal 101) and the Cancer Genome Atlas (TCGA-KIRC) were enrolled. HMOX1 pathway signatures were determined for each sample by RNA-sequencing and gene set enrichment analysis. Immune infiltration was evaluated by flow cytometry. Response and progression-free survival (PFS) were set as primary endpoints. RESULTS: Patients of low-HMOX1 signature showed higher objective response rate (43.5% vs. 27.3%) in ZS-MRCC cohort and longer PFS in both cohorts (ZS-MRCC cohort, p = 0.019; JAVELIN-101 cohort, p = 0.036). Patients in the high-HMOX1 signature arm also showed greater clinical benefit from TKI + IO, rather than TKI monotherapy (p < 0.001). In high-HMOX1 signature RCC tissues, CD8+ T cells showed a dysfunctional phenotype with decreased GZMB expression (Spearman's ρ = -0.32, p = 0.045). A risk score based on HMOX1 signature was further constructed by random forest approach, involving HMOX1 signature and immunologic features. In patients with a low risk level, TKI + IO combination therapy demonstrated longer PFS than TKI monotherapy (p < 0.001), however in individuals with a high risk score group, these two regimens did not give different advantages. CONCLUSIONS: Our study identified the HMOX1 pathway signature was a potential prognostic factor of progression-free survival for TKI + IO combination therapy in the advanced RCC in different cohort, especially in first-line management of mRCC in the Javelin 101 cohort. Moreover, HMOX1 signature was associated with T-cell function in tumor environment.


Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , CD8-Positive T-Lymphocytes/pathology , Heme Oxygenase-1/genetics , Heme Oxygenase-1/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Kidney Neoplasms/drug therapy , Kidney Neoplasms/genetics , Immunotherapy
9.
Immunogenetics ; 75(2): 133-143, 2023 04.
Article En | MEDLINE | ID: mdl-36515717

Immunotherapy plus tyrosine kinase inhibitor (IO-TKI) has become the standard first-line therapy for advanced renal cell carcinoma (RCC). However, the modest response rate of IO-TKI therapy and the absence of biomarkers limited the selection of treatment strategies for RCC patients. There were three cohorts enrolled: two from our facility (ZS-MRCC and ZS-HRRCC) and one from a clinical study (JAVELIN-101). By RNA sequencing, the expression of ADAM9 in each sample was measured. By flow cytometry and immunohistochemistry, immune infiltration and T cell function were examined. Primary outcomes were established as treatment response and progression-free survival (PFS). Patients with low-ADAM9 expression had a higher objective response rate (56.5% vs 13.6%, P = 0.01) and longer PFS in both cohorts. In the ZS-HRRCC cohort, the expression of ADAM9 was associated with increased tumor-infiltrating T cells, which was proved by immunohistochemistry (P < 0.05) and flow cytometry (Spearman's ρ = 0.42, P < 0.001). In the high-ADAM9 group, CD8+ and CD4+ T cells revealed an exhausted phenotype with decreased GZMB (Spearman's ρ = - 0.31, P = 0.05, and Spearman's ρ = - 0.49, P < 0.001, respectively), and fewer Macrophages were identified. A predictive RFscore was further constructed by random forest approach, involving ADAM9 and immunologic genes. Only in the subgroup with the lower RFscore did IO-TKI outperform TKI monotherapy. High-ADAM9 expression was associated with immunosuppression and IO-TKI resistance. Expression of ADAM9 was also associated with the exhaustion and dysfunction of T cells. ADAM9-based RFscore has the potential to be used as a biomarker to distinguish the optimal patient treatment methods between IO-TKI and TKI monotherapy.


Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , Kidney Neoplasms/drug therapy , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/therapeutic use , Immunotherapy/methods , Nephrectomy , Membrane Proteins/genetics , ADAM Proteins/genetics , ADAM Proteins/therapeutic use
10.
Urol Oncol ; 41(1): 51.e13-51.e23, 2023 01.
Article En | MEDLINE | ID: mdl-36328922

BACKGROUND: Latest guidelines recommended immunotherapy (IO) plus tyrosine kinase inhibitor (TKI) combination as standard first-line therapy in renal cell carcinoma (RCC), with no predictive biomarker being applied. Complement system shapes tumor microenvironment, which may influence TKI+IO benefit. METHODS: Two cohorts from our institute and 2 external cohorts were enrolled. RNA-sequencing was performed for each sample, and alternative complement pathway signature (ACPS) was defined by single sample gene set enrichment analysis. Immune infiltration and function were assessed by immunohistochemistry and flow cytometry. RESULTS: Under TKI+IO therapy, ACPS was elevated in non-responders (P<0.01), and high-ACPS predicted lower response rate and shorter progression-free survival (P=0.040). Moreover, TKI+IO, rather than TKI monotherapy, may benefit patients of low-ACPS combined with SETD2-wild type (HR=0.55, P<0.001). In RCC, ACPS was associated with increased tumor-infiltrating T cells (Spearman's ρ=0.50, P=0.001). However, in high-ACPS samples, CD8+ T cells revealed an exhausted phenotype with decreased GZMB (P<0.001) and increased PD1 (P=0.008) expression. Elevated PD1 expression in high-ACPS samples was confirmed by immunohistochemistry (P=0.046). Besides, macrophage infiltration was increased in high-ACPS samples (P=0.045), along with suppressive cytokines. CONCLUSIONS: Under TKI+IO, high-ACPS was linked to immunosuppression and treatment resistance. ACPS might be used as a biomarker for better treatment strategy between TKI+IO or TKI monotherapy.


Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , Complement Pathway, Alternative , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Immunotherapy , Kidney Neoplasms/drug therapy , Kidney Neoplasms/metabolism , Immunosuppression Therapy , Tumor Microenvironment
11.
Evol Bioinform Online ; 17: 11769343211049270, 2021.
Article En | MEDLINE | ID: mdl-34733102

We aimed to discover prognostic factors of muscle-invasive bladder cancer (MIBC) and investigate their relationship with immune therapies. Online data of MIBC were obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus database (GEO) database. Weighted gene co-expression network analysis (WGCNA) and univariate Cox analysis were applied to classify genes into different groups. Venn diagram was used to find the intersection of genes, and prognostic efficacy was proved by Kaplan-Meier analysis. Heatmap was utilized for differential analysis. Riskscore (RS) was calculated according to multivariate Cox analysis and evaluated by receiver operating characteristic curve (ROC). MIBC samples from TCGA and GEO were analyzed by WGCNA and univariate Cox analysis and intersected at 4 genes, CLK4, DEDD2, ENO1, and SYTL1. Higher SYTL1 and DEDD2 expressions were significantly correlated with high tumor grades. Riskscore based on genes showed great prognostic efficiency in predicting overall survival (OS), disease-specific survival (DSS), and progression-free interval (PFI) in TCGA dataset (P < .001). The area under the ROC curve (AUC) of RS reached 0.671 in predicting 1-year survival and 0.653 in 3-year survival. KEGG pathways enrichment filtered 5 enriched pathways. xCell analysis showed increased T cell CD4+ Th2 cell, macrophage, macrophage M1, and macrophage M2 infiltration in high RS samples (P < .001). In immune checkpoints analysis, PD-L1 expression was significantly higher in patients with high RS. We have, therefore, constructed RS as a convincing prognostic index for MIBC patients and found potential targeted pathways.

12.
Urol J ; 18(5): 564-572, 2021 Oct 04.
Article En | MEDLINE | ID: mdl-34606081

PURPOSE: Benign prostatic hyperplasia (BPH) significantly reduces the quality of life. However, the biological mechanisms of BPH development remain largely unknown. We aimed to investigate the essential genomic and immunogenic features in BPH. MATERIALS AND METHODS: Transcriptome profiling and clinical data of BPH and normal prostate samples were acquired from GEO datasets. The discovery sets were composed of GSE119195, GSE7307, GSE101486, while validation set was GSE132714. ESTIMATE and CIBERSORT was used to investigate the immunogenic features. Furthermore, transcriptional and weighted gene co-expression network analysis (WGCNA) was used for further analysis. RESULTS: BPH samples presented higher immune score. Meanwhile, CIBERSORT deconvolution revealed that BPH exists significantly abundant M2 Macrophages, follicular T helper cells, resting mast cells, and fewer plasma cells, activated CD4+ memory T cells and activated mast cells. WGCNA analysis also revealed significantly enriched immune-related modules in BPH. Transcriptomic analysis identified SOCS3, IL6, C3, IGF1, NOTCH1 and VCAN as key regulators of immunogenic phenotype in BPH. Moreover, we generated an immunological gene signature for BPH, which worked well in validation cohort. CONCLUSION: In our study, BPH samples exhibited a distinct immune infiltration pattern, represented by an immunological gene signature. This genomic-based assessment model reveals the potential transcriptomic patterns during BPH development.


Prostatic Hyperplasia , Gene Expression Profiling , Humans , Male , Memory T Cells , Prostatic Hyperplasia/genetics , Quality of Life
13.
Ann Clin Lab Sci ; 51(5): 597-608, 2021 Sep.
Article En | MEDLINE | ID: mdl-34686501

OBJECTIVE: Clear cell renal cell carcinoma (ccRCC) is the most common histological type of renal malignancies. Our aim was to find the prognostic crux of ccRCC. METHODS: The data for ccRCC was acquired from The Cancer Genome Atlas (TCGA) and the International Genome Consortium (ICGC) database. Weighted gene co-expression network analysis (WGCNA), differentially expressed genes (DEGs), and univariate Cox analysis were applied to classify the gene groups. A Venn diagram was used to find the intersection of the gene groups. The prognostic efficiency was proved by Kaplan-Meier analysis. Heatmap and volcano plots were utilized for differential analysis. The risk score (RS) was calculated based on the multivariate Cox analysis. RESULTS: The ccRCC samples were analyzed respectively via WGCNA, DEGs, and univariate Cox analysis based on five-year overall survival (OS) and intersected at two genes, ADGRF5 and EFNB2. The expressions of ADGRF5 and EFNB2 were negatively correlated with the OS and disease-specific survival (DSS) of the TCGA dataset, which was proved by Kaplan-Meier analysis (P<0.001). The RS was constructed and demonstrated great prognostic efficiency in predicting the OS, DSS, and progression-free interval (PFI) in TCGA (P<0.001) and the OS in ICGC (p=0.037). The area under the receiver operating characteristic (ROC) curve (AUC) of RS achieved 0.647 in the prediction of three-year survival and 0.714 for five-year survival. The KEGG pathway enrichment in high RS samples filtered five enriched pathways, in which CTNNB1 and LRP6 showed the best accordance with RS (P<0.001). xCell analysis revealed increased T cell CD4+ Th1 cell infiltration in high RS samples (P<0.001). PD-1 expression was higher in the high RS patients. CONCLUSIONS: We constructed RS as a convincing prognostic index for ccRCC patients and found potential scientific target pathways.


Biomarkers, Tumor/genetics , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/mortality , Kidney Neoplasms/genetics , Kidney Neoplasms/mortality , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Ephrin-B2/genetics , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Metabolic Networks and Pathways/genetics , Prognosis , Programmed Cell Death 1 Receptor/genetics , Proportional Hazards Models , ROC Curve , Receptors, G-Protein-Coupled/genetics , beta Catenin/genetics
15.
Oncol Lett ; 14(5): 5121-5128, 2017 Nov.
Article En | MEDLINE | ID: mdl-29142597

MicroRNAs (miRNAs) are small non-coding RNAs that affect various biological processes by altering the expression of a target gene. An miRNA microarray analysis has previously revealed a significant decrease in miR-193a-3p levels in prostate cancer tissues compared with that in their benign prostate hyperplasia counterparts. However, the role of miR-193a-3p has yet to be elucidated. In the present study, reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was used to evaluate the expression levels of miR-193a-3p in two human prostate cancer cell lines. Forced overexpression of miR-193a-3p was established by transfecting mimics into DU-145 and PC3 cell lines. Cell proliferation and the cell cycle were assessed using a cell viability assay, flow cytometry and a colony formation assay. In addition, the target gene of miR-193a-3p was determined by a luciferase assay, RT-qPCR and western blot analysis. The regulation of the cell cycle by miR-193a-3p was also evaluated by western blotting. The results demonstrated that miR-193a-3p expression levels were lower in prostate cancer cell lines as compared with the RWPE normal prostate epithelium cell line. Subsequent gain-of-function studies revealed that stable miR-193a-3p transfection inhibited cell viability, proliferation and colony formation, and induced G1 phase arrest in prostate cancer cells. A luciferase assay and western blot analysis identified cyclin D1 (CCND1) as a direct target gene of miR-193a-3p. In addition, the forced expression of CCND1 was able to counter the inhibitory effects of miR-193a-3p transfection in the prostate cancer cells. In summary, the results suggest that miR-193a-3p may inhibit the viability, proliferation and survival of prostate cancer cells by regulating the expression profile of CCND1, and that miR-193a-3p may be a novel therapeutic biomarker for prostate cancer.

16.
Aging Dis ; 8(5): 590-610, 2017 Oct.
Article En | MEDLINE | ID: mdl-28966804

Among age-related diseases, cardiovascular and cerebrovascular diseases are major causes of death. Vascular dysfunction is a key characteristic of these diseases wherein age is an independent and essential risk factor. The present work will review morphological alterations of aging vessels in-depth, which includes the discussion of age-related microvessel loss and changes to vasculature involving the capillary basement membrane, intima, media, and adventitia as well as the accompanying vascular dysfunctions arising from these alterations.

17.
Aging Dis ; 8(4): 486-505, 2017 Jul.
Article En | MEDLINE | ID: mdl-28840062

As with many age-related diseases including vascular dysfunction, age is considered an independent and crucial risk factor. Complicated alterations of structure and function in the vasculature are linked with aging hence, understanding the underlying mechanisms of age-induced vascular pathophysiological changes holds possibilities for developing clinical diagnostic methods and new therapeutic strategies. Here, we discuss the underlying molecular mediators that could be involved in vascular aging, e.g., the renin-angiotensin system and pro-inflammatory factors, metalloproteinases, calpain-1, monocyte chemoattractant protein-1 (MCP-1) and TGFß-1 as well as the potential roles of testosterone and estrogen. We then relate all of these to clinical manifestations such as vascular dementia and stroke in addition to reviewing the existing clinical measurements and potential interventions for age-related vascular dysfunction.

18.
Oncotarget ; 7(32): 51773-51783, 2016 Aug 09.
Article En | MEDLINE | ID: mdl-27429046

Despite the recent studies which have shown that microRNA (miRNA) negatively regulates gene expression by silencing the expression of target genes, here we reported the new evidence of microRNA-mediated gene activation by targeting specific promoter sites. We identified a miR-877-3p binding site on the promoter site of tumor suppressor gene p16 which alters frequently in bladder cancer. Enforced expression of miR-877-3p could increase the expression of p16, which inhibit the proliferation and tumorigenicity of bladder cancer through cell cycle G1-phase arrest. Further evidences confirmed that the correlation between p16 activation and miR-877-3p was due to the direct binding. These findings demonstrate the anti-tumor function of miR-877-3p in bladder cancer cells and reveal a new pattern of miRNA involved gene regulation.


Carcinoma, Transitional Cell/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , MicroRNAs/physiology , Urinary Bladder Neoplasms/genetics , Animals , Carcinoma, Transitional Cell/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Promoter Regions, Genetic , Up-Regulation , Urinary Bladder Neoplasms/pathology
19.
Mol Cells ; 38(2): 130-7, 2015.
Article En | MEDLINE | ID: mdl-25556372

MicroRNAs (miRNAs) are small, endogenous RNAs that play important gene-regulatory roles by binding to the imperfectly complementary sequences at the 3'-UTR of mRNAs and directing their gene expression. Here, we first discovered that miR-576-3p was down-regulated in human bladder cancer cell lines compared with the non-malignant cell line. To better characterize the role of miR-576-3p in bladder cancer cells, we over-expressed or down-regulated miR-576-3p in bladder cancer cells by transfecting with chemically synthesized mimic or inhibitor. The overexpression of miR-576-3p remarkably inhibited cell proliferation via G1-phase arrest, and decreased both mRNA and protein levels of cyclin D1 which played a key role in G1/S phase transition. The knock-down of miR-576-3p significantly promoted the proliferation of bladder cancer cells by accelerating the progression of cell cycle and increased the expression of cyclin D1. Moreover, the dual-luciferase reporter assays indicated that miR-576-3p could directly target cyclin D1 through binding its 3'-UTR. All the results demonstrated that miR-576-3p might be a novel suppressor of bladder cancer cell proliferation through targeting cyclin D1.


Cyclin D1/genetics , Cyclin D1/metabolism , MicroRNAs/genetics , Urinary Bladder Neoplasms/genetics , 3' Untranslated Regions , Cell Line, Tumor , Cell Proliferation , Down-Regulation , G1 Phase Cell Cycle Checkpoints , Gene Expression Regulation, Neoplastic , Humans , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology
20.
Mol Med Rep ; 11(2): 1004-8, 2015 Feb.
Article En | MEDLINE | ID: mdl-25351792

The mortality rate associated with prostate cancer is mainly due to metastases rather than primary organ­confined disease. Decreasing the incidence of metastasis is important in treating prostate cancer. 4',5,7­trihydroxyflavone (apigenin) has been demonstrated to be effective in inhibiting several types of cancer. The aim of this study was to investigate the effect and mechanism of apigenin on the movement of prostate cancer cells. In the present study, DU145 cells were treated with varying concentrations of apigenin for different time periods. Cell viability was evaluated using an MTT assay. Cell motility and invasiveness were assayed using wound healing assays and a Matrigel migration and invasion assay. Flow cytometric and western blot analyses were performed to examine the cell cycle and signaling pathways. The results demonstrated that apigenin suppressed the proliferation and inhibited the migration and invasive potential of the DU145 prostate cancer cells in a dose­ and time­dependent manner, which was associated with epithelial mesenchymal transition. These findings suggested that apigenin may be effective in treating human prostate cancer.


Apigenin/pharmacology , Cell Movement/drug effects , Epithelial-Mesenchymal Transition/drug effects , Prostatic Neoplasms/pathology , Cell Cycle/drug effects , Cell Line, Tumor/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Male , Neoplasm Invasiveness/prevention & control , Signal Transduction
...