1.Mismatch repair gene germline mutations in patients with prostate cancer.
Bangwei FANG ; Yu WEI ; Jian PAN ; Tingwei ZHANG ; Dingwei YE ; Yao ZHU
Journal of Zhejiang University. Medical sciences 2023;52(2):133-138
OBJECTIVES:
To investigate the prevalence of pathogenic germline mutations of mismatch repair (MMR) genes in prostate cancer patients and its relationship with clinicopathological characteristics.
METHODS:
Germline sequencing data of 855 prostate cancer patients admitted in Fudan University Shanghai Cancer Center from 2018 to 2022 were retrospectively analyzed. The pathogenicity of mutations was assessed according to the American College of Medical Genetics and Genomics (ACMG) standard guideline, Clinvar and Intervar databases. The clinicopathological characteristics and responses to castration treatment were compared among patients with MMR gene mutation (MMR+ group), patients with DNA damage repair (DDR) gene germline pathogenic mutation without MMR gene (DDR+MMR- group) and patients without DDR gene germline pathogenic mutation (DDR- group).
RESULTS:
Thirteen (1.52%) MMR+ patients were identified in 855 prostate cancer patients, including 1 case with MLH1 gene mutation, 6 cases with MSH2 gene mutation, 4 cases with MSH6 gene mutation and 2 cases with PMS2 gene mutation. 105 (11.9%) patients were identified as DDR gene positive (except MMR gene), and 737 (86.2%) patients were DDR gene negative. Compared with DDR- group, MMR+ group had lower age of onset (P<0.05) and initial prostate-specific antigen (PSA) (P<0.01), while no significant differences were found between the two groups in Gleason score and TMN staging (both P>0.05). The median time to castration resistance was 8 months (95%CI: 6 months-not achieved), 16 months (95%CI: 12-32 months) and 24 months (95%CI: 21-27 months) for MMR+ group, DDR+MMR- group and DDR- group, respectively. The time to castration resistance in MMR+ group was significantly shorter than that in DDR+MMR- group and DDR- group (both P<0.01), while there was no significant difference between DDR+MMR- group and DDR- group (P>0.05).
CONCLUSIONS
MMR gene mutation testing is recommended for prostate cancer patients with early onset, low initial PSA, metastasis or early resistance to castration therapy.
Male
;
Humans
;
Prostate-Specific Antigen/genetics*
;
Germ-Line Mutation
;
Retrospective Studies
;
DNA Mismatch Repair/genetics*
;
DNA-Binding Proteins/metabolism*
;
China
;
Prostatic Neoplasms/pathology*
2.LncRNA GAS5 enhances tumor stem cell-like medicated sensitivity of paclitaxel and inhibits epithelial-to-mesenchymal transition by targeting the miR-18a-5p/STK4 pathway in prostate cancer.
Ting-Ting LU ; Xia TAO ; Hua-Lei LI ; Ling GAI ; Hua HUANG ; Feng LI
Asian Journal of Andrology 2022;24(6):643-652
The onset of prostate cancer (PCa) is often hidden, and recurrence and metastasis are more likely to occur due to chemotherapy resistance. Herein, we identified downregulated long noncoding RNA (lncRNA) growth arrest-specific 5 (GAS5) in PCa that was associated with metastasis and paclitaxel resistance. GAS5 acted as a tumor suppressor in suppressing the proliferation and metastasis of paclitaxel-resistant PCa cells. GAS5 overexpression in vivo inhibited the tumor growth of xenografts and elevated PCa sensitivity to paclitaxel. Combination of GAS5 and paclitaxel treatment showed great potential in PCa treatment. Moreover, mechanistic analysis revealed a novel regulatory network of GAS5/miR-18a-5p/serine/threonine kinase 4 (STK4) that inhibits epithelial-to-mesenchymal transition (EMT) and enhances tumor stem cell-like-mediated sensitivity to paclitaxel in PCa. These findings provide a novel direction for the development of a potential adjunct to cancer chemotherapy that aims to improve the sensitivity of chemotherapy drugs in PCa.
Humans
;
Male
;
Cell Line, Tumor
;
Cell Proliferation/genetics*
;
Gene Expression Regulation, Neoplastic
;
MicroRNAs/metabolism*
;
Neoplastic Stem Cells
;
Paclitaxel/therapeutic use*
;
Prostatic Neoplasms/genetics*
;
Protein Serine-Threonine Kinases/metabolism*
;
RNA, Long Noncoding/metabolism*
;
Epithelial-Mesenchymal Transition
3.The transcription factor NF-kB1 regulates miR-195 expression in prostate cancer.
Chun-Hui LIU ; Ya-Li WANG ; Kai LU ; Lei ZHANG ; Guang-Yuan ZHANG ; Bin XU ; Shu-Qiu CHEN ; Ming CHEN
National Journal of Andrology 2020;26(10):875-880
Objective:
To investigate the regulatory effect of the transcription factor NF-kB1 on the expression of miR-195 in prostate cancer (PCa).
METHODS:
We analyzed the possibility of NF-kB1 binding to the miR-195 promoter and the expression of NF-kB1 in PCa using the JASPAR and Oncomine databases, respectively, and determined the expressions of NF-kB1 and miR-195 in PCa cells by real-time quantitative PCR after inhibiting the former by interfering RNA targeting NF-kB1. We detected the activity of the luciferase reporter gene after constructing its gene plasmid in the miR-195 promoter region and having it co-transfected with the NF-kB1 plasmid. Then we analyzed the correlation between the expressions of miR-195 and NF-kB1 in the prostate tissue.
RESULTS:
NF-kB1 was overexpressed in PCa. After inhibition of the expression of NF-kB1, that of miR-195 was increased in PC-3 and DU-145 cell lines, with a negative correlation between the NF-kB1 and miR-195 expressions in the PCa tissue. The results of luciferase reporter gene assay showed direct binding of NF-kB1 to the miR-195 promoter zone.
CONCLUSIONS
NF-kB1 regulates the expression of miR-195 in prostate cancer.
Cell Line, Tumor
;
Gene Expression Regulation, Neoplastic
;
Humans
;
Male
;
MicroRNAs/genetics*
;
NF-kappa B p50 Subunit/metabolism*
;
Promoter Regions, Genetic
;
Prostatic Neoplasms/genetics*
;
Transcription Factors/metabolism*
4.MTBP regulates migration and invasion of prostate cancer cells .
Zhuoyu XIAO ; Mingkun CHEN ; Jiankun YANG ; Cheng YANG ; Xianyuan LÜ ; Hu TIAN ; Cundong LIU
Journal of Southern Medical University 2019;39(1):6-12
OBJECTIVE:
To investigate the role of MTBP in regulating the migration and invasion of human prostate cancer cells.
METHODS:
The baseline expressions of MTBP in 3 different human prostate cancer cells lines (22RV1, DU145 and Lncap) were detected using Western blotting. The cells were transfected with a small interfering RNA (siRNA) for MTBP knockdown or MTBP plasmid for MTBP overexpression, and 48 h later, the cells were examined for MTBP expression with Western blotting; the changes in the migration abilities of the cells were evaluated using wound healing assay and Transwell assay, and the cell invasiveness was assessed using Matrigel Transwell assay. The expression of E-cadherin protein, a marker of epithelial mesenchymal transition (EMT), was detected using Western blotting.
RESULTS:
MTBP expression was the highest in DU145 cells followed by Lncap cells, and was the lowest in 22RV1 cells, indicating a positive correlation of MTBP expression with the level of malignancy of human prostate cancer cells. Transfection of the cells with siRNA or MTBP plasmids efficiently lowered or enhanced the expressions of MTBP in human prostate cancer cells. Wound healing assay showed that inhibition of MTBP expression decreased the migration ability of the prostate cancer cells, and MTBP overexpression significantly promoted the migration of the cells ( < 0.01). Transwell assay showed that MTBP knockdown significantly lowered the migration and invasion ability of the cells, while MTBP overexpression markedly increased the number of migrating and invading cells ( < 0.01); Western blotting results showed that MTBP knockdown increased the expression of E-cadherin protein, and MTBP overexpression decreased E-cadherin expression in the prostate cancer cells.
CONCLUSIONS
MTBP overexpression promotes the migration and invasion of human prostate cancer cells possibly relation to the induction of EMT.
Antigens, CD
;
metabolism
;
Cadherins
;
metabolism
;
Carrier Proteins
;
genetics
;
metabolism
;
Cell Line, Tumor
;
Cell Movement
;
Epithelial-Mesenchymal Transition
;
Gene Expression Regulation, Neoplastic
;
Gene Knockdown Techniques
;
Humans
;
Male
;
Neoplasm Invasiveness
;
Prostatic Neoplasms
;
metabolism
;
pathology
;
RNA, Small Interfering
;
Transfection
5.Establishment of enzalutamide-resistant human prostate cancer cell lines and screening of lncRNA and mRNA expression profiles.
Han GUAN ; Zhi-Xin LING ; Fang FANG ; Li-Kai MAO ; Zong-Hao YOU ; Can WANG ; Shu-Qiu CHEN ; Bin XU ; Ming CHEN
National Journal of Andrology 2018;24(2):116-121
Objective:
To establish enzalutamide-resistant human prostate cancer cell lines and screen out the lncRNA and mRNA expression profiles associated with enzalutamide resistance.
METHODS:
Human prostate cancer cell lines LNCAP and C4-2B were cultured with 10 μmol/L enzalutamide for 6 months in vitro for the establishment of enzalutamide-resistant subclones LNCAP-ENZA and C4-2B-ENZA. The IC50 value and enzalutamide resistance index of each cell line were examined by MTT assay, the expressions of enzalutamide-related genes FL-AR, AR-V7 and HnRNPA1 were determined by Western blot, and the lncRNA and mRNA differential expressions of C4-2B and C4-2B-ENZA were detected by high-throughout lncRNA microarray.
RESULTS:
Compared with LNCAP and C4-2B, the IC50 values of enzalutamide-resistant subclones LNCAP-ENZA (60.83 μmol/L) and C4-2B-ENZA (88.32 μmol/L) were increased significantly (P < 0.05) and the enzalutamide-resistance indexes of the LNCAP-ENZA and C4-2B-ENZA cells were 4.94 and 4.67, respectively. The expressions of AR-V7 and HnRNPA1 were markedly up-regulated in the LNCAP-ENZA and C4-2B-ENZA cells as compared with those in the LNCAP and C4-2B cells, but that of FL-AR showed no significant change. A total of 1 440 lncRNAs and 1 236 mRNAs were identified as differentially expressed in the C4-2B-ENZA cells.
CONCLUSIONS
Enzalutamide -resistant human prostate cancer cell subclones LNCAP-ENZA and C4-2B-ENZA were successfully established and enzalutamide resistance-associated lncRNA and mRNA were identified, which may provide some molecular evidence for the management of enzalutamide-resistant human prostate cancer.
Cell Line, Tumor
;
drug effects
;
Drug Resistance, Neoplasm
;
Humans
;
Male
;
Phenylthiohydantoin
;
analogs & derivatives
;
pharmacology
;
Prostatic Neoplasms
;
drug therapy
;
genetics
;
pathology
;
RNA, Long Noncoding
;
metabolism
;
RNA, Messenger
;
metabolism
;
RNA, Neoplasm
;
metabolism
;
Receptors, Androgen
6.The transcription factor ZEB1 promotes an aggressive phenotype in prostate cancer cell lines.
Octavio ORELLANA-SERRADELL ; Daniela HERRERA ; Enrique A CASTELLON ; Hector R CONTRERAS
Asian Journal of Andrology 2018;20(3):294-299
It has been reported that one of the factors that promotes tumoral progression is the abnormal activation of the epithelial-mesenchymal transition program. This process is associated with tumoral cells acquiring invasive and malignant properties and has the transcription factor zinc finger E-box-binding homeobox 1 (ZEB1) as one of its main activators. However, the role of ZEB1 in promoting malignancy in prostate cancer (PCa) is still unclear. Here, we report that ZEB1 expression correlates with Gleason score in PCa samples and that expression of ZEB1 regulates epithelial-mesenchymal transition and malignant characteristics in PCa cell lines. The results showed that ZEB1 expression is higher in samples of higher malignancy and that overexpression of ZEB1 was able to induce epithelial-mesenchymal transition by upregulating the mesenchymal marker Vimentin and downregulating the epithelial marker E-Cadherin. On the contrary, ZEB1 silencing repressed Vimentin expression and upregulated E-Cadherin. ZEB1 expression conferred enhanced motility and invasiveness and a higher colony formation capacity to 22Rv1 cells whereas DU145 cells with ZEB1 silencing showed a decrease in those same properties. The results showed that ZEB1 could be a key promoter of tumoral progression toward advanced stages of PCa.
Cadherins/metabolism*
;
Cell Line, Tumor
;
Cell Movement/genetics*
;
Epithelial-Mesenchymal Transition/genetics*
;
Gene Expression Regulation, Neoplastic
;
Gene Silencing
;
Humans
;
Male
;
Neoplasm Grading
;
Neoplasm Invasiveness/genetics*
;
Prostatic Neoplasms/pathology*
;
Vimentin/metabolism*
;
Zinc Finger E-box-Binding Homeobox 1/metabolism*
7.Phosphoglycerate mutase 1 knockdown inhibits prostate cancer cell growth, migration, and invasion.
Yao-An WEN ; Bo-Wei ZHOU ; Dao-Jun LV ; Fang-Peng SHU ; Xian-Lu SONG ; Bin HUANG ; Chong WANG ; Shan-Chao ZHAO
Asian Journal of Andrology 2018;20(2):178-183
Phosphoglycerate mutase 1 (PGAM1) is upregulated in many cancer types and involved in cell proliferation, migration, invasion, and apoptosis. However, the relationship between PGAM1 and prostate cancer is poorly understood. The present study investigated the changes in PGAM1 expression in prostate cancer tissues compared with normal prostate tissues and examined the cellular function of PGAM1 and its relationship with clinicopathological variables. Immunohistochemistry and Western blotting revealed that PGAM1 expression was upregulated in prostate cancer tissues and cell lines. PGAM1 expression was associated with Gleason score (P = 0.01) and T-stage (P = 0.009). Knockdown of PGAM1 by siRNA in PC-3 and 22Rv1 prostate cancer cell lines inhibited cell proliferation, migration, and invasion and enhanced cancer cell apoptosis. In a nude mouse xenograft model, PGAM1 knockdown markedly suppressed tumor growth. Deletion of PGAM1 resulted in decreased expression of Bcl-2, enhanced expression of Bax, caspases-3 and inhibition of MMP-2 and MMP-9 expression. Our results indicate that PGAM1 may play an important role in prostate cancer progression and aggressiveness, and that it might be a valuable marker of poor prognosis and a potential therapeutic target for prostate cancer.
Animals
;
Apoptosis/genetics*
;
Caspase 3/metabolism*
;
Cell Line, Tumor
;
Cell Movement/genetics*
;
Cell Proliferation/genetics*
;
Gene Deletion
;
Gene Knockdown Techniques
;
Humans
;
Male
;
Matrix Metalloproteinase 2/metabolism*
;
Matrix Metalloproteinase 9/metabolism*
;
Mice
;
Mice, Nude
;
Neoplasm Invasiveness/genetics*
;
Neoplasm Transplantation
;
PC-3 Cells
;
Phosphoglycerate Mutase/genetics*
;
Prostatic Neoplasms/pathology*
;
Proto-Oncogene Proteins c-bcl-2/metabolism*
;
RNA, Small Interfering
;
Transplantation, Heterologous
;
bcl-2-Associated X Protein/metabolism*
8.Expression of AXL enhances docetaxel-resistance of prostate cancer cells.
Jian-Zhong LIN ; Jia-Geng ZHU ; Hong-Fei WU ; Jiu-Ming LI ; Wei DE ; Zeng-Jun WANG
National Journal of Andrology 2017;23(4):302-308
Objective:
To explore the effect of the AXL expression on the chemosensitivity of prostate cancer PC-3 and DU145 cells to docetaxel and possible mechanisms.
METHODS:
Using Western blot, we examined the expressions of the AXL protein, p-AXL and Gas6 in the docetaxel-resistant PC-3 (PC-3-DR) and DU145 (DU145-DR) cells stimulated with gradually increased concentrations of docetaxel. We transfected the PC-3 and DU145 cells with negative NC ShRNA and AXL-ShRNA, respectively, which were confirmed to be effective, detected the proliferation, apoptosis and cycle distribution of the cells by CCK8, MTT and flow cytometry after treated with the AXL-inhibitor MP470 and/or docetaxel, and determined the expression of the ABCB1 protein in the PC-3-DR and DU145-DR cells after intervention with the AXL-inhibitor R428 and/or docetaxel.
RESULTS:
The expression of the AXL protein in the PC-3 and DU145 cells was significantly increased after docetaxel treatment (P <0.05). The expressions AXL and p-AXL were remarkably higher (P <0.05) while that of Gas6 markedly lower (P <0.05) in the PC-3 and DU145 than in the PC-3-DR and DU145-DR cells. The inhibitory effect of docetaxel on the proliferation and its enhancing effect on the apoptosis of the PC-3 and DU145 cells were significantly decreased at 48 hours after AXL transfection (P <0.05). MP470 obviously suppressed the growth and promoted the apoptosis of the PC-3-DR and DU145-DR cells, with a higher percentage of the cells in the G2/M phase when combined with docetaxel than used alone (P <0.05). R428 markedly reduced the expression of ABCB1 in the PC-3-DR and DU145-DR cells, even more significantly in combination with docetaxel than used alone (P <0.05).
CONCLUSIONS
The elevated expression of AXL enhances the docetaxel-resistance of PC-3 and DU145 prostate cancer cells and AXL intervention improves their chemosensitivity to docetaxel, which may be associated with the increased cell apoptosis in the G2/M phase and decreased expression of ABCB1.
ATP Binding Cassette Transporter, Subfamily B, Member 1
;
metabolism
;
Antineoplastic Agents
;
pharmacology
;
Apoptosis
;
drug effects
;
Cell Count
;
Cell Cycle
;
Cell Line, Tumor
;
Cell Proliferation
;
drug effects
;
Docetaxel
;
Drug Resistance, Neoplasm
;
Humans
;
Intercellular Signaling Peptides and Proteins
;
metabolism
;
Male
;
Prostatic Neoplasms
;
drug therapy
;
metabolism
;
pathology
;
Proto-Oncogene Proteins
;
drug effects
;
genetics
;
metabolism
;
Pyrimidines
;
pharmacology
;
RNA, Small Interfering
;
Receptor Protein-Tyrosine Kinases
;
drug effects
;
genetics
;
metabolism
;
Taxoids
;
pharmacology
9.Expression of long non-coding RNA H19 in prostate cancer and its effect on the proliferation and glycometabolism of human prostate cancer cells.
Shi-Cheng SUN ; Hu ZHAO ; Rui LIU ; Bi-Liang WANG ; Yi-Qing LIU ; Yan ZHAO ; Zhen-Duo SHI
National Journal of Andrology 2017;23(2):120-124
Objective:
To study the expression of long noncoding RNA (lncRNA) H19 in human prostate cancer tissue and its effect on the glycometabolism and growth of human prostate cancer cells.
METHODS:
Realtime quantitative RTPCR (qRTPCR) was employed to detect the expression of lncRNA H19 in human prostate tissues from 20 patients with prostate cancer (10 cases of highGleason score prostate cancer [HGPC] and 10 cases of lowGleason score prostate cancer [LGPC]) and another 5 with benign prostatic hyperplasia (BPH). After transfection of H19 siRNA into the DU145 and PC3 prostate cancer cells, the growth of the cells and the H19 expression in the cells were determined by MTT and qRTPCR respectively, and the changes in the glycometabolism of the prostate cancer cells were analyzed by measuring the contents of glucose and lactate in the culture medium. Nontransfected and transfected negative vectors were used as blank and negative controls respectively.
RESULTS
The relative expression of H19 was significantly increased in both the HGPC and LGPC tissues (0.725±0.385 and 2.086±0.542) as compared with that in the BPH tissue (0.210±0.068) (P< 0.01), even higher in the HGPC than in the LGPC tissue (P< 0.01). After transfection of H19 siRNA, the expressions of H19 were remarkably decreased in the DU145 and PC3 prostate cancer cells in comparison with those in the blank control and negative control groups (P< 0.01), and so were the proliferation of and the glucose and lactate levels in the DU145 and PC3 cells (P< 0.01).
Cell Line, Tumor
;
Cell Proliferation
;
Gene Expression Regulation, Neoplastic
;
Glucose
;
metabolism
;
Humans
;
Lactic Acid
;
metabolism
;
Male
;
Prostate
;
metabolism
;
Prostatic Hyperplasia
;
metabolism
;
Prostatic Neoplasms
;
metabolism
;
pathology
;
RNA, Long Noncoding
;
genetics
;
metabolism
;
RNA, Small Interfering
;
Transfection
10.Effect of silencing the VDR gene on the migration and invasion of prostate cancer cells.
Yuan-Dong ZHANG ; Hui ZHAO ; Kang-Jian LI ; Run-Yun GUAN
National Journal of Andrology 2017;23(11):969-974
Objective:
To investigate the effect of small interfering RNA silencing the vitamin D receptor (VDR) on the biological behavior of prostate cancer PC-3 cells.
METHODS:
We constructed the VDR-shRNA lentiviral vector and determined the mRNA and protein expressions of VDR by RT-PCR and Western blot. Using scratch wound healing and Transwell chamber assays, we detected the changes in the migration and invasiveness of the PC-3 cells after silencing VDR.
RESULTS:
The VDR-shRNA plasmid significantly interfered the VDR expression and successfully screened the cell lines with stable VDR-shRNA interference. The rate of scratch wound healing was markedly lower in the VDR interference group than in the blank control and LV3 negative control groups (59% vs 73.6% and 77.8%, P <0.05), but with no statistically significant difference between the latter two (P >0.05), and so was the count of permeable cells (P <0.05), but with no significant difference between the latter two groups, either (P >0.05). The migration ability and invasiveness of the VDR-treated cells were remarkably decreased as compared with those of the control cells.
CONCLUSIONS
Down-regulated expression of the VDR gene may reduce the migration and invasiveness of prostate cancer cells.
Cell Line, Tumor
;
Cell Movement
;
genetics
;
Cell Proliferation
;
Down-Regulation
;
Gene Silencing
;
Humans
;
Lentivirus
;
Male
;
Neoplasm Invasiveness
;
genetics
;
Plasmids
;
Prostatic Neoplasms
;
genetics
;
pathology
;
RNA, Messenger
;
metabolism
;
RNA, Small Interfering
;
Receptors, Calcitriol
;
genetics
;
metabolism
;
Transfection
;
Wound Healing
;
genetics

Result Analysis
Print
Save
E-mail