Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
2.
Dis Markers ; 2022: 8724035, 2022.
Article in English | MEDLINE | ID: mdl-35548776

ABSTRACT

Background: In our previous research, we developed a 32-gene risk index model that may be utilized as a robust prognostic method for predicting prostate cancer (PCa) recurrence after surgery. Among the 32 genes, the Fifth Ewing Variant (FEV) gene was one of the top downregulated genes in relapsed PCa. However, current understanding of the FEV gene and its involvement in PCa is limited. Methods: FEV mRNA expression was analyzed and correlated to clinical outcomes in PCa patients who underwent prostatectomy at the Massachusetts General Hospital. Specimens from tissue microarray (TMA) including 102 prostate cancer patients were analysis for the expression of FEV. Meanwhile, FEV expression profiles were also assessed in PCa cell lines and in BPH-1 prostate epithelial cells using western blotting and quantitative reverse transcription-PCR (qRT-PCR). Furthermore, we transfected LNCaP and PC-3 cells with either an empty vector or full-length FEV gene and performed in vitro cell functional assays. The part FEV plays in tumor xenograft growth was also assessed in vivo. Results: Of the 191 patients included in this study base on the DASL dataset, 77 (40.3%) and 24 (13.6%), respectively, developed prostate-specific antigen (PSA) relapse and metastasis postradical prostatectomy. Significant FEV downregulation was observed in PCa patients showing PSA failure and metastasis. The protein expression of FEV was significantly negatively correlated with the Gleason score and pathological stage in prostate cancer tissues. Similarly, FEV expression significantly decreased in all PCa cell lines relative to BPH-1 (all P < 0.05). Functional assays revealed that FEV expression markedly inhibited PCa cell growth, migration, and invasion, which in turn significantly repressed the growth of tumor xenografts in vivo. Conclusion: The results of this study suggest an association between downregulated FEV expression and PSA relapse in PCa patients. In addition, FEV may act as a tumor suppressor in PCa.


Subject(s)
DNA-Binding Proteins , Prostatic Hyperplasia , Prostatic Neoplasms , Transcription Factors , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Male , Neoplasm Recurrence, Local/pathology , Prostate-Specific Antigen , Prostatectomy/methods , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery
3.
Mol Carcinog ; 59(8): 897-907, 2020 08.
Article in English | MEDLINE | ID: mdl-32319143

ABSTRACT

Lactate dehydrogenase isozyme (LDH) is a tetramer constituted of two isoforms, LDHA and LDHB, the expression of which is associated with cell metabolism and cancer progression. Our previous study reveals that CC-chemokine ligand-18 (CCL18) is involved in progression of prostate cancer (PCa).This study aims to investigate how CCL18 regulates LDH isoform expression, and therefore, contributes to PCa progression. The data revealed that the expression of LDHA was upregulated and LDHB was downregulated in PCa cells by CCL18 at both messenger RNA and protein levels. The depletion of CCR8 reduced the ability of CCL18 to promote the proliferation, migration, and lactate production of PCa cells. Depletion of a CCR8 regulated transcription factor, ARNT, significantly reduced the expression of LDHA. In addition, The Cancer Genome Atlas dataset analyses revealed a positive correlation between CCR8 and ARNT expression. Two dimension difference gel electrophoresis revealed that the LDHA/LDHB ratio was increased in the prostatic fluid of patients with PCa and PCa tissues. Furthermore, increased LDHA/LDHB ratio was associated with poor clinical outcomes of patients with PCa. Together, our results indicate that the CCR8 pathway programs LDH isoform expression in an ARNT dependent manner and that the ratio of LDHA/LDHB has the potential to serve as biomarkers for PCa diagnosis and prognosis.


Subject(s)
Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , Biomarkers, Tumor/metabolism , Chemokines, CC/metabolism , Gene Expression Regulation, Neoplastic , L-Lactate Dehydrogenase/metabolism , Prostatic Neoplasms/pathology , Receptors, CCR8/metabolism , Apoptosis , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Biomarkers, Tumor/genetics , Cell Proliferation , Chemokines, CC/genetics , Humans , Isoenzymes , L-Lactate Dehydrogenase/genetics , Male , Prognosis , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptors, CCR8/genetics , Survival Rate , Tumor Cells, Cultured
5.
Cell Signal ; 59: 152-162, 2019 07.
Article in English | MEDLINE | ID: mdl-30926388

ABSTRACT

Immunotherapy has made great breakthroughs in the field of cancer. However, the immunotherapeutic effect of prostate cancer is unsatisfactory. We found that the expression of TRIB1 was significantly correlated with the infiltration of CD163+ macrophages in prostate cancer. This study focused on the effects of TRIB1 on macrophage polarization in the immune microenvironment of prostate cancer. RNA sequencing analysis demonstrated that TRIB1 has significant effects on the regulation of the nuclear factor (NF)-κB signaling pathway and downstream cytokines. Flow cytometry and enzyme-linked immunosorbent assay were used to examine THP-1 cells cultured in conditioned medium from prostate cancer cells overexpressing TRIB1 and showed that overexpression of TRIB1 promoted the secretion of CXCL2 and interleukin (IL)8 by PC3 cells, which increased the secretion of IL12 by THP-1 cells as well as the expression of CD163 on THP-1 cells. IKB-zeta, regulated by TRIB1, was expressed in PC3 cells but was barely detectable in DU145 cells. The reductions in CXCL2 and IL8 by the inhibition of TRIB1 were rescued by the deletion of IKB-zeta. Here we showed that TRIB1 promoted the secretion of cytokines from prostate cancer cells and induced the differentiation of monocytes/macrophages into M2 macrophages.


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , Macrophages/immunology , Prostatic Neoplasms/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Tumor Microenvironment/immunology , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Chemokine CXCL2/immunology , Humans , Macrophage Activation , Macrophages/cytology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , NF-kappa B/immunology , PC-3 Cells , Protein Serine-Threonine Kinases/physiology , Receptors, Cell Surface/metabolism , THP-1 Cells
6.
Int J Cancer ; 144(12): 3099-3110, 2019 06 15.
Article in English | MEDLINE | ID: mdl-30537104

ABSTRACT

To investigate immune profile consisting of stromal PD-L1 expression, inhibitory or non-T-cell inflamed tumor microenvironment that may predict response to anti-PD-L1/PD-1 immunotherapy in prostate cancer, we validated the specificity of a PD-L1 monoclonal antibody (E1L3N) and identified PD-L1 specific expression in prostatic stromal nerve cells. PD-L1 expression was analyzed in 73 primary prostate cancers and 7 castration-resistant prostate cancers (CRPC) by immunohistochemistry (IHC) and resulting data from primary prostate cancers were correlated with tumor-associated lymphocytes (TALs), clinicopathological characteristics and clinical outcome. PD-L1 was expressed in the tumor cells in only one primary prostate cancer case and none of the CRPC. However, PD-L1 was frequently observed in the nerve branches in the tumor-associated stroma (69 of 73 cases, 94.5%), supported by colocalization with axonal marker PGP9.5. FoxP3-, CD3- and CD8-positive T lymphocytes were observed in 74.6% (47/63), 98.4% (62/63) and 100% (61/61) of the cases, respectively. The density of PD-L1+ tumor-associated nerves (TANs) was inversely correlated with that of CD8+ TALs. Higher density of PD-L1+ TANs was significantly associated with biochemical recurrence (BCR) in Kaplan-Meier survival analysis (p = 0.016). In both univariate and multivariate Cox analysis, the density of PD-L1+ TANs was independently prognostic of BCR. In conclusion, PD-L1 expression is rare in prostate tumor cells but prevalent in TANs and negatively correlated with CD8+ TALs. Neuro-immunological interaction may be a contribution to immune-suppressive microenvironment. Combinatorial treatment regimen designs to neural PD-L1 and TALs should be warranted in future clinical application of anti-PD-L1/PD-1 immunotherapy in prostate cancer.


Subject(s)
B7-H1 Antigen/biosynthesis , CD8-Positive T-Lymphocytes/immunology , Prostate/innervation , Prostatic Neoplasms, Castration-Resistant/immunology , Prostatic Neoplasms/immunology , B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/pathology , Humans , Immunohistochemistry , Male , Middle Aged , Prognosis , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , Prostatic Neoplasms, Castration-Resistant/pathology , Prostatic Neoplasms, Castration-Resistant/surgery , Tumor Microenvironment/immunology , Ubiquitin Thiolesterase
7.
Mol Cancer ; 16(1): 48, 2017 02 27.
Article in English | MEDLINE | ID: mdl-28241827

ABSTRACT

BACKGROUND: Even though aberrant expression of microRNA (miR)-30d has been reported in prostate cancer (PCa), its associations with cancer progression remain contradictory. The aim of this study was to investigate clinical significance, biological functions and underlying mechanisms of miR-30d deregulation in PCa. METHODS: Involvement of miR-30d deregulation in malignant phenotypes of PCa was demonstrated by clinical sample evaluation, and in vitro and in vivo experiments. The mechanisms underlying its regulatory effect on tumor angiogenesis were determined. RESULTS: miR-30d over-expression was observed in both PCa cells and clinical specimens. High-miR-30d was distinctly associated with high pre-operative PSA and Gleason score, advanced clinical and pathological stages, positive metastasis and biochemical recurrence (BCR), and reduced overall survival of PCa patients. Through gain- and loss-of-function experiments, we found that miR-30d promoted PCa cell proliferation, migration, invasion, and capillary tube formation of endothelial cells, as well as in vivo tumor growth and angiogenesis in a mouse model. Simulation of myosin phosphatase targeting subunit 1 (MYPT1), acting as a direct target of miR-30d, antagonized the effects induced by miR-30d up-regulation in PCa cells. Notably, miR-30d/MYPT1 combination was identified as an independent factor to predict BCR of PCa patients. Furthermore, miR-30d exerted its pro-angiogenesis function, at least in part, by inhibiting MYPT1, which in turn, increased phosphorylation levels of c-JUN and activated VEGFA-induced signaling cascade in endothelial cells. CONCLUSIONS: miR-30d and/or its target gene MYPT1 may serve as novel prognostic markers of PCa. miR-30d promotes tumor angiogenesis of PCa through MYPT1/c-JUN/VEGFA pathway.


Subject(s)
MicroRNAs/genetics , Myosin-Light-Chain Phosphatase/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Animals , Biomarkers, Tumor , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Disease Models, Animal , Disease Progression , Gene Expression Profiling , Gene Expression Regulation , Heterografts , Humans , Male , Mice , Myosin-Light-Chain Phosphatase/genetics , Prognosis , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , RNA Interference
8.
Int Urol Nephrol ; 49(5): 817-823, 2017 May.
Article in English | MEDLINE | ID: mdl-28213802

ABSTRACT

PURPOSE: Abnormal spindle microtubule assembly (ASPM) gene was known to be linked with poor clinical prognosis in various tumors. However, the clinical significance of ASPM in prostate cancer (PCa) has not yet been understood. The purpose of this study was to determine the association of ASPM with tumor progression and prognosis in PCa patients. METHODS: The expression of ASPM at protein level in human PCa and non-cancerous prostate tissue was detected by immunohistochemical analysis, which was further validated by using microarray-based dataset (NCBI GEO: GSE21032 and The Cancer Genome Atlas (TCGA) dataset) at mRNA level. Subsequently, the association of ASPM expression with the clinicopathological characteristics of patients with PCa was then statistically analyzed. RESULTS: Immunohistochemistry and dataset analyses revealed that ASPM expression was significantly increased in the PCa tissues with high Gleason score. Additionally, as showed by two datasets, ASPM expression was significantly high expressed in the PCa tissues when compared with the non-cancerous tissues, especially in advanced PCa pathological stage. The upregulation of ASPM mRNA expression in the PCa tissues significantly correlated with the presence of tumor metastasis, shorter overall survival and prostate-specific antigen (PSA) failure. Furthermore, both univariate and multivariate analyses showed that the upregulation of ASPM was a potential predictor of poor biochemical recurrence (BCR)-free survival. CONCLUSIONS: Our data suggest that ASPM may play an important role in the progression of PCa. More importantly, the increased expression of ASPM may potentially predict poor BCR-free survival in patients with PCa.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasm Recurrence, Local/genetics , Nerve Tissue Proteins/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Aged , Biopsy, Needle , Cohort Studies , Databases, Factual , Disease Progression , Disease-Free Survival , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Multivariate Analysis , Neoplasm Grading , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/pathology , Prognosis , Proportional Hazards Models , Prostatic Neoplasms/mortality , Survival Analysis , Up-Regulation
9.
Oncol Res Treat ; 40(1-2): 15-20, 2017.
Article in English | MEDLINE | ID: mdl-28118628

ABSTRACT

BACKGROUND: Novel molecular markers are important diagnostic tools for the assessment of cancer progression and evaluation of effectiveness of the treatment. SOX9, a key regulator of developmental processes, is overexpressed in various neoplasms, such as prostate, breast, and colorectal cancers. However, the utilization of SOX9 as a biomarker for other urological cancers has not yet been investigated. METHODS: In the present study, paired patient tissue microarrays were analyzed by immunohistochemistry, and the SOX9 protein expression was quantitated as immunoreactive scores in patients with renal cell carcinoma (RCC), bladder cancer (BCa), and penile cancer (PC). RESULTS: In comparison with normal tissues, SOX9 protein expression was significantly upregulated in RCC (p < 0.001) and BCa (p < 0.001), and significantly correlated with the advanced pathological grade (RCC: p = 0.023) and clinical stage (RCC: p = 0.022 and BCa: p = 0.046) of patients. Based on the mRNA level in the TCGA dataset, SOX9 was upregulated in RCC with gender (p = 0.027), advanced pathological grade (p = 0.003) and advanced clinical stage (p = 0.001). Kaplan-Meier survival curves revealed that RCC patients with high SOX9 levels had shorter survival (p < 0.001). Further, high SOX9 expression was an independent prognostic factor for RCC patients (hazard ratio 0.056, 95% confidence interval 0.607-1.184; p < 0.001). CONCLUSION: These findings suggest that SOX9 may play an important role in tumor progression of RCC and BCa and it may be used as a biomarker of this malignancy.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , Penile Neoplasms/pathology , SOX9 Transcription Factor/metabolism , Urinary Bladder Neoplasms/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Carcinoma, Renal Cell/metabolism , Child , Child, Preschool , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Kidney Neoplasms/metabolism , Male , Middle Aged , Neoplasm Staging , Penile Neoplasms/metabolism , Prognosis , RNA, Messenger/genetics , SOX9 Transcription Factor/genetics , Tissue Array Analysis , Urinary Bladder Neoplasms/metabolism , Young Adult
10.
Int J Oncol ; 48(4): 1650-8, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26891801

ABSTRACT

Increased expression of E2F1 has been reported to be associated with tumor growth and cell survival of prostate cancer (PCa). However, its roles and mechanisms on PCa have not been fully elucidated. The present study found that E2F1 overexpression in PCa tissues was significantly associated with high Gleason score (P=0.01) and advanced pathological stage (P=0.02). In addition, PCa patients with high E2F1 expression more frequently had shorter biochemical recurrence-free survival (P=0.047) than those with low E2F1 expression. Then, we confirmed that the knock-down of E2F1 expression was able to inhibit cell cycle progression, invasion and migration of PCa cell lines in vitro, along with tumor xenograft growth and epithelial-to-mesenchymal transition (EMT) in vivo. Moreover, we identified CD147 as a novel interaction partner for E2F1 through bio-informatic binding site prediction, combined with chromatin immunoprecipitation-PCR (ChIP-PCR) and western blot analysis. Taken together, our data delineate an as yet unrecognized function of E2F1 as enhancer of tumor invasion and migration of PCa via regulating the expression of CD147 in PCa. Importantly, E2F1 may function as a biomarker that can differentiate patients with biochemical recurrent and non-biochemical recurrent disease following radical prostatectomy, highlighting its potential as a therapeutic target.


Subject(s)
Basigin/metabolism , E2F1 Transcription Factor/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Animals , Basigin/genetics , Binding Sites , Cell Line, Tumor , Cell Movement , E2F1 Transcription Factor/chemistry , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Neoplasm Grading , Neoplasm Invasiveness , Prognosis , Prostatic Neoplasms/genetics , Survival Analysis
11.
Oncol Rep ; 33(5): 2648-54, 2015 May.
Article in English | MEDLINE | ID: mdl-25813864

ABSTRACT

Our previous study revealed the potential role of CD147 in human prostate cancer (PCa). Here, we investigated the CD147 promoter methylation status and the correlation with tumorigenicity in human PCa. CD147 mRNA and protein expression levels were both significantly higher in the 4 PCa cell lines, than in the 2 non-tumorigenic benign human prostatic epithelial cell lines (all P<0.01). We showed hypomethylation of promoter regions of CD147 in PCa cell lines with significant CD147 expression as compared to non-tumorigenic benign human prostatic epithelial cell lines slowly expressing CD147. Additionally, the treatment of methylated cell lines with 5-aza-2'-deoxycytidine increased CD147 expression significantly in low-expressing cell lines and also activated the expression of matrix metalloproteinase (MMP)-2, which may be one of the most important downstream targets of CD147. Furthermore, PCa tissues displayed decreased DNA methylation in the promoter region of CD147 compared to the corresponding non-cancerous prostate tissues, and methylation intensity correlated inversely with the CD147 mRNA levels. There was a significant negative correlation between CD147 mRNA levels and the number of methylated sites in PCa tissues (r=-0.467, P<0.01). In conclusion, our data offer convincing evidence for the first time that the DNA promoter hypomethylation of CD147 may be one of the regulatory mechanisms involved in the cancer-related overexpression of CD147 and may play a crucial role in the tumorigenesis of PCa.


Subject(s)
Basigin/genetics , DNA Methylation/genetics , Gene Expression Regulation, Neoplastic/genetics , Prostatic Neoplasms/genetics , Base Sequence , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Line, Tumor , Disease Progression , Humans , Male , Matrix Metalloproteinase 2/genetics , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Prostate/pathology , Prostatic Neoplasms/pathology , RNA, Messenger/genetics
12.
Biomed Res Int ; 2014: 230183, 2014.
Article in English | MEDLINE | ID: mdl-25197632

ABSTRACT

BACKGROUND AND AIM: CC chemokine ligand 18 (CCL18) promotes malignant behaviors of various human cancer types. However, its involvement in human prostate cancer has not been fully elucidated. The aim of this study was to investigate the role of CCL18 in PCa. METHODS: Expression of CCL18 at mRNA and protein levels was detected using real-time qRT-PCR and immunohistochemistry analysis. We analyzed the associations of CCL18 expression with clinical features of human PCa. The effects of PCa cell migration, invasion, and apoptosis were tested. The efficiency of CCL18 on prostate tumor growth was assessed in a subcutaneous xenograft model. RESULTS: CCL18 expression was upregulated (both P < 0.01) in PCa tissues compared with those in noncancerous prostate tissues. CCL18 upregulation was correlated with high Gleason score (P = 0.034) of patients with PCa. rCCL18 stimulation in PCa cells promoted cell migration and invasion but decreased DU145 cells apoptosis rate. Furthermore, subcutaneous homografts models showed the increased tumor growth and tumor vascularization with the CCL18 stimulation, and the expression of Ki67, PCNA, and CD31 in CCL18 stimulation mice was also significantly increased. CONCLUSIONS: Our data offer the convincing evidence that the upregulation of CCL18 may be involved in the malignant progression of PCa.


Subject(s)
Chemokines, CC/metabolism , Disease Progression , Prostatic Neoplasms/pathology , Aged , Allografts , Animals , Apoptosis/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Chemokines, CC/genetics , Gene Expression Regulation, Neoplastic , Humans , Male , Mice, Inbred C57BL , Middle Aged , Neoplasm Invasiveness , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/genetics , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction
13.
PLoS One ; 8(5): e63941, 2013.
Article in English | MEDLINE | ID: mdl-23737958

ABSTRACT

AIM: Early diagnosis of prostate cancer (PCa), which is a clinically heterogeneous-multifocal disease, is essential to improve the prognosis of patients. However, published PCa diagnostic markers share little overlap and are poorly validated using independent data. Therefore, we here developed an integrative proteomics and interaction network-based classifier by combining the differential protein expression with topological features of human protein interaction networks to enhance the ability of PCa diagnosis. METHODS AND RESULTS: By two-dimensional fluorescence difference gel electrophoresis (2D-DIGE) coupled with MS using PCa and adjacent benign tissues of prostate, a total of 60 proteins with the differential expression in PCa tissues were identified as the candidate markers. Then, their networks were analyzed by GeneGO Meta-Core software and three hub proteins (PTEN, SFPQ and HDAC1) were chosen. After that, a PCa diagnostic classifier was constructed by support vector machine (SVM) modeling based on the microarray gene expression data of the genes which encode the hub proteins mentioned above. Validations of diagnostic performance showed that this classifier had high predictive accuracy (85.96∼90.18%) and area under ROC curve (approximating 1.0). Furthermore, the clinical significance of PTEN, SFPQ and HDAC1 proteins in PCa was validated by both ELISA and immunohistochemistry analyses. More interestingly, PTEN protein was identified as an independent prognostic marker for biochemical recurrence-free survival in PCa patients according to the multivariate analysis by Cox Regression. CONCLUSIONS: Our data indicated that the integrative proteomics and interaction network-based classifier which combines the differential protein expression and topological features of human protein interaction network may be a powerful tool for the diagnosis of PCa. We also identified PTEN protein as a novel prognostic marker for biochemical recurrence-free survival in PCa patients.


Subject(s)
Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/metabolism , Protein Interaction Maps , Proteomics , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Histone Deacetylase 1/metabolism , Humans , Male , Middle Aged , PTB-Associated Splicing Factor , PTEN Phosphohydrolase/metabolism , RNA-Binding Proteins/metabolism , Reproducibility of Results
14.
Mol Cell Biochem ; 381(1-2): 51-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23666742

ABSTRACT

Suppressors of cytokine signaling (SOCS) proteins have been identified as negative feedback regulators of cytokine-mediated signaling in various tissues, and demonstrated to play critical roles in tumorigenesis and tumor development of different cancers. The involvement of SOCSs in human prostate cancer (PCa) has not been fully elucidated. Thus, the aim of this study is to investigate the expression patterns and the clinical significance of SOCSs in PCa. The expression changes of SOCSs at mRNA and protein levels in human PCa tissues compared with adjacent benign prostate tissues were, respectively, detected by using real-time quantitative reverse transcriptase-polymerase chain reaction (QRT-PCR) and immunohistochemistry analyses. The associations of SOCSs expression with clinicopathological features and clinical outcome of PCa patients were further statistically analyzed. Among SOCSs, both QRT-PCR and immunohistochemistry analyses found that SOCS2 expression was upregulated (at mRNA level: change ratio = 1.98, P = 0.031; at protein level: 5.12 ± 0.60 vs. 2.68 ± 0.37, P = 0.016) and SOCS6 expression was downregulated (at mRNA level: change ratio = -1.65, P = 0.008; at protein level: 3.03 ± 0.32 vs. 4.0.72 ± 0.39, P = 0.004) in PCa tissues compared with those in non-cancerous prostate tissues. In addition, the upregulation of SOCS2 in PCa tissues was correlated with the lower Gleason score (P < 0.001), the absence of metastasis (P < 0.001) and the negative PSA failure (P = 0.009); the downregulation of SOCS6 tended to be found in PCa tissues with the higher Gleason score (P = 0.016), the advanced pathological stage (P = 0.007), the positive metastasis (P = 0.020), and the positive PSA failure (P = 0.032). Furthermore, both univariate and multivariate analyses showed that the downregulation of SOCS2 was an independent predictor of shorter biochemical recurrence-free survival. Our data offer the convincing evidence for the first time that the dysregulation of SOCS2 and SOCS6 may be associated with the aggressive progression of PCa. SOCS2 may be potential markers for prognosis in PCa patients.


Subject(s)
Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Aged , Aged, 80 and over , Blotting, Western , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Prognosis , Prostate/metabolism , Prostate/pathology
15.
Mol Biol Rep ; 40(5): 3799-805, 2013 May.
Article in English | MEDLINE | ID: mdl-23283744

ABSTRACT

The ErbB3 binding protein 1 (Ebp1) represents a downstream effector of the ErbB signaling network and has been demonstrated to be a potent tumor suppressor in various human malignancies, however, its involvement in human bladder cancer is still unclear.To investigate the clinical significance and potential role of ErbB3 binding protein 1 (Ebp1) in bladder cancer. Ebp1 expression at protein and gene levels in 52 surgically removed bladder cancer specimens as well as 21 adjacent normal bladder specimens were respectively detected by immunohistochemistry and qRT-PCR. The association of Ebp1 protein expression with the clinicopathological features of bladder cancer was also statistically analyzed. Its roles in bladder cancer cell line were further evaluated. The expression level of Ebp1 protein and gene in bladder cancer tissues was significantly lower than that in adjacent normal bladder tissues (P < 0.01). When categorized into low vs. high expression, the down-regulation of Ebp1 protein was associated with the advanced pathologic stage (P = 0.036) and the high histologic grade (P = 0.001) of patients with bladder cancer. Moreover, following the transfection of Ebp1 in bladder cancer cells, not only cell proliferation and cell invasion decreased significantly, but also the cell cycle was blocked at G0/G1 stage. Our data suggest for the first time that the down-regulation of Ebp1 closely correlates with advanced clinicopathological characteristics of human bladder cancer. Furthermore, Ebp1 plays an important role in the bladder cancer cells' proliferation by regulating the cancer cell cycle from G0/G1 to S.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , RNA-Binding Proteins/metabolism , Urinary Bladder Neoplasms/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adenoviridae/genetics , Aged , Aged, 80 and over , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Disease Progression , Down-Regulation , Female , Gene Expression , Gene Expression Regulation, Neoplastic , Genetic Vectors/genetics , Humans , Male , Middle Aged , Neoplasm Staging , RNA-Binding Proteins/genetics , Transduction, Genetic , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...