Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 192
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Biol Chem ; 299(1): 102720, 2023 01.
Article in English | MEDLINE | ID: mdl-36410440

ABSTRACT

Cancer cells, including those of prostate cancer (PCa), often hijack intrinsic cell signaling to reprogram their metabolism. Part of this reprogramming includes the activation of de novo synthesis of fatty acids that not only serve as building blocks for membrane synthesis but also as energy sources for cell proliferation. However, how de novo fatty acid synthesis contributes to PCa progression is still poorly understood. Herein, by mining public datasets, we discovered that the expression of acetyl-CoA carboxylase alpha (ACACA), which encodes acetyl-CoA carboxylase 1 (ACC1), was highly expressed in human PCa. In addition, patients with high ACACA expression had a short disease-free survival time. We also reported that depletion of ACACA reduced de novo fatty acid synthesis and PI3K/AKT signaling in the human castration-resistant PCa (CRPC) cell lines DU145 and PC3. Furthermore, depletion of ACACA downregulates mitochondrial beta-oxidation, resulting in mitochondrial dysfunction, a reduction in ATP production, an imbalanced NADP+/NADPhydrogen(H) ratio, increased reactive oxygen species, and therefore apoptosis. Reduced exogenous fatty acids by depleting lipid or lowering serum supplementation exacerbated both shRNA depletion and pharmacological inhibition of ACACA-induced apoptosis in vitro. Collectively, our results suggest that inhibition of ectopic ACACA, together with suppression of exogenous fatty acid uptake, can be a novel strategy for treating currently incurable CRPC.


Subject(s)
Acetyl-CoA Carboxylase , Fatty Acids , Mitochondria , Prostatic Neoplasms, Castration-Resistant , Humans , Male , Acetyl-CoA Carboxylase/metabolism , Fatty Acids/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Phosphatidylinositol 3-Kinases/genetics , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Cell Line, Tumor
2.
Nucleic Acids Res ; 50(D1): D1139-D1146, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34500460

ABSTRACT

MicroRNAs (miRNAs), which play critical roles in gene regulatory networks, have emerged as promising diagnostic and prognostic biomarkers for human cancer. In particular, circulating miRNAs that are secreted into circulation exist in remarkably stable forms, and have enormous potential to be leveraged as non-invasive biomarkers for early cancer detection. Novel and user-friendly tools are desperately needed to facilitate data mining of the vast amount of miRNA expression data from The Cancer Genome Atlas (TCGA) and large-scale circulating miRNA profiling studies. To fill this void, we developed CancerMIRNome, a comprehensive database for the interactive analysis and visualization of miRNA expression profiles based on 10 554 samples from 33 TCGA projects and 28 633 samples from 40 public circulating miRNome datasets. A series of cutting-edge bioinformatics tools and machine learning algorithms have been packaged in CancerMIRNome, allowing for the pan-cancer analysis of a miRNA of interest across multiple cancer types and the comprehensive analysis of miRNome profiles to identify dysregulated miRNAs and develop diagnostic or prognostic signatures. The data analysis and visualization modules will greatly facilitate the exploit of the valuable resources and promote translational application of miRNA biomarkers in cancer. The CancerMIRNome database is publicly available at http://bioinfo.jialab-ucr.org/CancerMIRNome.


Subject(s)
Biomarkers, Tumor/genetics , Databases, Genetic , MicroRNAs/genetics , Neoplasms/genetics , Biomarkers, Tumor/classification , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/genetics , Humans , MicroRNAs/classification , Neoplasms/classification
3.
Article in English | MEDLINE | ID: mdl-38581330

ABSTRACT

Objective: Metabolism, a basic need and biochemical process for cell survival and proliferation, is closely connected with the pathogenesis and progression of prostate cancer. Methods: A four-gene signature construct that includes CKM (CKM), CD38, Enoyl Coenzyme A(EHHADH), and Arginase 2(ARG2) was created by bioinformatics. Finally, hub genes were validated by IHC and in vitro experiments. Results: The results showed the AUCs of the logistic regression and neural networks diagnostic model for the diagnosis of two subtypes were 0.920 and 0.936, respectively. The risk score demonstrated by univariable and multivariable Cox analysis is an independent predictive component of the prognostic signature for DFS. According to immunohistochemical analyses, ARG2 and CD38 expression levels were considerably under-expressed, but CKM and EHHADH expression levels were significantly overexpressed. Furthermore, The expression of ARG2 was significantly down-regulated in the late Gleason score. Finally, we found that ARG2 is lowly expressed in prostate cancer cells. Furthermore, based on the effect of ARG2 on the malignant phenotype of PCa in vitro, we also found that ARG2 may be a tumor suppressor that plays an important role in inhibiting proliferation, migration, and invasion. Conclusions: These findings suggest that ARG2 has been tentatively identified as a new target for research into how PCa develops in metabolism and for the development of innovative targeted treatments.

4.
Hum Genet ; 2023 Sep 27.
Article in English | MEDLINE | ID: mdl-37758909

ABSTRACT

Nonmutational epigenetic reprogramming is a crucial mechanism contributing to the pronounced heterogeneity of prostate cancer (PCa). Among these mechanisms, N6-methyladenosine (m6A)-modified long non-coding RNAs (lncRNAs) have emerged as key players. However, the precise roles of m6A-modified lncRNAs in PCa remain to be elucidated. In this study, methylated RNA immunoprecipitation sequencing (MeRIP-seq) was conducted on primary and metastatic PCa samples, leading to the identification of 21 lncRNAs exhibiting differential methylation and expression patterns. We further established a PCa prognostic signature, named m6A-modified lncRNA score (mLs), based on 9 differential methylated lncRNAs in 4 multicenter cohorts. The high mLs score cohort exhibited a tendency for earlier biochemical recurrence (BCR) compared to the low mLs score cohort. Remarkably, the predictive performance of the mLs score surpassed that of five previously reported lncRNA-based signatures. Functional enrichment analysis underscored a negative correlation between the mLs score and lipid metabolism. Additionally, through MeRIP-qPCR, we pinpointed a hub gene, MIR210HG, which was validated through in vitro and in vivo experiments. These findings collectively illuminate the landscape of m6A-methylated lncRNAs in PCa tissue via MeRIP-seq and harness this information to prognosticate PCa outcomes using the mLs score. Furthermore, our study validates, both experimentally and mechanistically, the facilitative role of MIR210HG in driving PCa progression.

5.
Small ; 19(2): e2205024, 2023 01.
Article in English | MEDLINE | ID: mdl-36398604

ABSTRACT

Mono-chemotherapy has significant side effects and unsatisfactory efficacy, limiting its clinical application. Therefore, a combination of multiple treatments is becoming more common in oncotherapy. Chemotherapy combined with the induction of ferroptosis is a potential new oncotherapy. Furthermore, polymeric nanoparticles (NPs) can improve the antitumor efficacy and decrease the toxicity of drugs. Herein, a polymeric NP, mPEG-b-PPLGFc@Dox, is synthesized to decrease the toxicity of doxorubicin (Dox) and enhance the efficacy of chemotherapy by combining it with the induction of ferroptosis. First, mPEG-b-PPLGFc@Dox is oxidized by endogenous H2 O2 and releases Dox, which leads to an increase of H2 O2 by breaking the redox balance. The Fe(II) group of ferrocene converts H2 O2 into ·OH, inducing subsequent ferroptosis. Furthermore, glutathione peroxidase 4, a biomarker of ferroptosis, is suppressed and the lipid peroxidation level is elevated in cells incubated with mPEG-b-PPLGFc@Dox compared to those treated with Dox alone, indicating ferroptosis induction by mPEG-b-PPLGFc@Dox. In vivo, the antitumor efficacy of mPEG-b-PPLGFc@Dox is higher than that of free Dox. Moreover, the loss of body weight in mice treated mPEG-b-PPLGFc@Dox is lower than in those treated with free Dox, indicating that mPEG-b-PPLGFc@Dox is less toxic than free Dox. In conclusion, mPEG-b-PPLGFc@Dox not only has higher antitumor efficacy but it reduces the damage to normal tissue.


Subject(s)
Ferroptosis , Nanoparticles , Mice , Animals , Metallocenes , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Polyethylene Glycols , Polymers
6.
Brief Bioinform ; 22(3)2021 05 20.
Article in English | MEDLINE | ID: mdl-32898860

ABSTRACT

Prognostic tests using expression profiles of several dozen genes help provide treatment choices for prostate cancer (PCa). However, these tests require improvement to meet the clinical need for resolving overtreatment, which continues to be a pervasive problem in PCa management. Genomic selection (GS) methodology, which utilizes whole-genome markers to predict agronomic traits, was adopted in this study for PCa prognosis. We leveraged The Cancer Genome Atlas (TCGA) database to evaluate the prediction performance of six GS methods and seven omics data combinations, which showed that the Best Linear Unbiased Prediction (BLUP) model outperformed the other methods regarding predictability and computational efficiency. Leveraging the BLUP-HAT method, an accelerated version of BLUP, we demonstrated that using expression data of a large number of disease-relevant genes and with an integration of other omics data (i.e. miRNAs) significantly increased outcome predictability when compared with panels consisting of a small number of genes. Finally, we developed a novel stepwise forward selection BLUP-HAT method to facilitate searching multiomics data for predictor variables with prognostic potential. The new method was applied to the TCGA data to derive mRNA and miRNA expression signatures for predicting relapse-free survival of PCa, which were validated in six independent cohorts. This is a transdisciplinary adoption of the highly efficient BLUP-HAT method and its derived algorithms to analyze multiomics data for PCa prognosis. The results demonstrated the efficacy and robustness of the new methodology in developing prognostic models in PCa, suggesting a potential utility in managing other types of cancer.


Subject(s)
Algorithms , Biomarkers, Tumor/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Genomics/methods , Prostatic Neoplasms/genetics , Aged , Humans , Kaplan-Meier Estimate , Male , MicroRNAs/genetics , Middle Aged , Models, Genetic , Neoplasm Staging , Phenotype , Prognosis , Prostatectomy/methods , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery
7.
J Transl Med ; 21(1): 884, 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-38057852

ABSTRACT

BACKGROUND: Advanced prostate cancer (PCa) will develop into castration-resistant prostate cancer (CRPC) and lead to poor prognosis. As the primary subtype of CRPC, CRPC-AR accounts for the major induction of PCa heterogeneity. CRPC-AR is mainly driven by 25 transcription factors (TFs), which we speculate may be the key factors driving PCa toward CRPC. Therefore, it is necessary to clarify the key regulator and its molecular mechanism mediating PCa progression. METHODS: Firstly, we downloaded transcriptomic data and clinical information from TCGA-PRAD. The characteristic gene cluster was identified by PPI clustering, GO enrichment, co-expression correlation and clinical feature analyses for 25 TFs. Then, the effects of 25 TFs expression on prognosis of PCa patients was analyzed using univariate Cox regression, and the target gene was identified. The expression properties of the target gene in PCa tissues were verified using tissue microarray. Meanwhile, the related mechanistic pathway of the target gene was mined based on its function. Next, the target gene was silenced by small interfering RNAs (siRNAs) for cellular function and mechanistic pathway validation. Finally, CIBERSORT algorithm was used to analyze the infiltration levels of 22 immune cells in PCa patients with low and high expression of target gene, and validated by assaying the expression of related immunomodulatory factor. RESULTS: We found that HOX family existed independently in 25 TFs, among which HOXC10, HOXC12 and HOXC13 had unique clinical features and the PCa patients with high HOXC13 expression had the worst prognosis. In addition, HOXC13 was highly expressed in tumor tissues and correlated with Gleason score and pathological grade. In vitro experiments demonstrated that silencing HOXC13 inhibited 22RV1 and DU145 cell function by inducing cellular DNA damage and activating cGAS/STING/IRF3 pathway. Immune infiltration analysis revealed that high HOXC13 expression suppressed infiltration of γδ T cells and plasma cells and recruited M2 macrophages. Consistent with these results, silencing HOXC13 up-regulated the transcriptional expression of IFN-ß, CCL2, CCL5 and CXCL10. CONCLUSION: HOXC13 regulates PCa progression by mediating the DNA damage-induced cGAS/STING/IRF3 pathway and remodels TIME through regulation of the transcription of the immune factors IFN-ß, CCL2, CCL5 and CXCL10.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Male , Humans , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Gene Expression Regulation, Neoplastic , Transcription Factors/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , DNA Damage , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism
8.
Mol Biol Evol ; 37(12): 3684-3698, 2020 12 16.
Article in English | MEDLINE | ID: mdl-32668004

ABSTRACT

Compared with genomic data of individual markers, haplotype data provide higher resolution for DNA variants, advancing our knowledge in genetics and evolution. Although many computational and experimental phasing methods have been developed for analyzing diploid genomes, it remains challenging to reconstruct chromosome-scale haplotypes at low cost, which constrains the utility of this valuable genetic resource. Gamete cells, the natural packaging of haploid complements, are ideal materials for phasing entire chromosomes because the majority of the haplotypic allele combinations has been preserved. Therefore, compared with the current diploid-based phasing methods, using haploid genomic data of single gametes may substantially reduce the complexity in inferring the donor's chromosomal haplotypes. In this study, we developed the first easy-to-use R package, Hapi, for inferring chromosome-length haplotypes of individual diploid genomes with only a few gametes. Hapi outperformed other phasing methods when analyzing both simulated and real single gamete cell sequencing data sets. The results also suggested that chromosome-scale haplotypes may be inferred by using as few as three gametes, which has pushed the boundary to its possible limit. The single gamete cell sequencing technology allied with the cost-effective Hapi method will make large-scale haplotype-based genetic studies feasible and affordable, promoting the use of haplotype data in a wide range of research.


Subject(s)
Genetic Techniques , Germ Cells , Haplotypes , Software , Chromosomes , Humans , Recombination, Genetic , Zea mays
9.
Cancer Sci ; 112(10): 4365-4376, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34252262

ABSTRACT

A substantial proportion of prostatic adenocarcinoma (PRAD) patients experience biochemical failure (BCF) after radical prostatectomy (RP). The immune microenvironment plays a vital role in carcinogenesis and the development of PRAD. This study aimed to identify a novel immune-related gene (IRG)-based signature for risk stratification and prognosis of BCF in PRAD. Weighted gene coexpression network analysis was carried out to identify a BCF-related module in a discovery cohort of patients who underwent RP at the Massachusetts General Hospital. The median follow-up time was 70.32 months. Random forest and multivariate stepwise Cox regression analyses were used to identify an IRG-based signature from the specific module. Risk plot analyses, Kaplan-Meier curves, receiver operating characteristic curves, univariate and multivariate Cox regression analyses, stratified analysis, and Harrell's concordance index were used to assess the prognostic value and predictive accuracy of the IRG-based signature in the internal discovery cohort; The Cancer Genome Atlas database was used as a validation cohort. Tumor immune estimation resource database analysis and CIBERSORT algorithm were used to assess the immunophenotype of PRAD. A novel IRG-based signature was identified from the specific module. Five IRGs (BUB1B, NDN, NID1, COL4A6, and FLRT2) were verified as components of the risk signature. The IRG-based signature showed good prognostic value and predictive accuracy in both the discovery and validation cohorts. Infiltrations of various immune cells were significantly different between low-risk and high-risk groups in PRAD. We identified a novel IRG-based signature that could function as an index for assessing tumor immune status and risk stratification in PRAD.


Subject(s)
Adenocarcinoma/genetics , Gene Regulatory Networks , HLA Antigens/genetics , Prostatic Neoplasms/genetics , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Adenocarcinoma/surgery , Cell Cycle Proteins/genetics , Cohort Studies , Collagen Type IV/genetics , Follow-Up Studies , Gene Expression Profiling , Genetic Markers , Humans , Immunity, Cellular , Immunophenotyping , Kaplan-Meier Estimate , Male , Membrane Glycoproteins/genetics , Middle Aged , Neoplasm Grading , Neoplasm Staging , Predictive Value of Tests , Prognosis , Prostate-Specific Antigen/blood , Prostatectomy , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , Protein Serine-Threonine Kinases/genetics , ROC Curve , Regression Analysis , Risk Assessment , Treatment Failure , Tumor Microenvironment/immunology , Tumor Suppressor Proteins/genetics
10.
J Pak Med Assoc ; 71(2(A)): 473-478, 2021 02.
Article in English | MEDLINE | ID: mdl-33819231

ABSTRACT

OBJECTIVE: To compare the expression levels of Defective In Cullin Neddylation 1 Domain Containing 1 oncogene in prostate cancer tissues and normal prostate tissues, to explore its effect on cancerous cells, and to investigate its underlying mechanisms on such cells in vitro. METHODS: The cross-sectional study was conducted at Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics from Jan 03,2017 to Nov 05,2018, and comprised prostate tissue samples on which immunohistochemistry was used to detect the expression of Defective In Cullin Neddylation 1 Domain Containing 1 oncogene. Short hairpin ribonucleic acid expression plasmid targeting the oncogene was constructed and transferred into prostate cance cell line DU145. The roles of the oncogene in prostate cancer progression were confirmed in vitro. The expression of vimentin and epithelial cadherin influenced by the oncogene were detected using Western blot. Data was analysed using SPSS 24. RESULTS: Of the 80 samples, 3(3.75%) were normal prostate tissues, 7(8.75%) adjacent normal prostate tissues, 20(25%) hyperplasia, and 50(62.5%) prostate cancer tissues. Defective In Cullin Neddylation 1 Domain Containing 1 oncogene expression in prostate cancerous tissues was significantly associated with high Gleason score (p<0.001), metastasis (p<0.05) and pathological stage (p<0.001). The oncogene was found to be an independent prognostic factor for disease-free survival of prostate cancer patients (p=0.0108). In vitro analysis confirmed the tumour promotive role of the oncogene through cell proliferation, invasion and migration assays. Its expression was closely correlated with aggressive progression and poor prognosis in prostate cancer patients (p<0.05). Vimentin and epithelial cadherin were affected by the oncogene. CONCLUSIONS: Defective In Cullin Neddylation 1 Domain Containing 1 oncogene highly expressed in DU145 and the prostate cancer tissues, which correlated with prognosis.


Subject(s)
Prostatic Neoplasms , Cell Line , Cell Proliferation , Cross-Sectional Studies , Gene Expression Regulation, Neoplastic , Humans , Male , Prognosis , Prostatic Neoplasms/genetics
11.
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
12.
BMC Cancer ; 20(1): 289, 2020 Apr 06.
Article in English | MEDLINE | ID: mdl-32252682

ABSTRACT

BACKGROUND: Solute Carrier Family 6 Member 1 (SLC6A1) has been identified as a cancer-promoting gene in various human cancers, such as clear cell renal cell carcinoma and ovarian cancer. However, its roles in prostate cancer (PCa) has not been fully elucidated. The aim of this study was to investigate the expression and clinical significance of SLC6A1 in PCa tissues and its effect on drug resistance to docetaxel in PCa. METHODS: Expression patterns of SLC6A1 protein in PCa tissues were examined by immunohistochemistry based on Tissue microarray. Associations of SLC6A1 protein expression with various clinicopathological features and patients' prognosis of PCa were also statistically evaluated based on TCGA data. Roles of SLC6A1 deregulation in prostate carcinogenesis and drug resistance was further determined in vitro and in vivo experiments. RESULTS: Based on TCGA Dataset, SLC6A1 expression was markedly higher in patients with high Gleason score, advanced clinical stage and positive biochemical recurrence than those with control features (all P < 0.05). Both unvariate and multivariate analyses demonstrated that SLC6A1 expression was significantly associated with biochemical recurrence-free survival in PCa patients. In addition, enforced expression of SLC6A1 effectively promoted cell proliferation, migration and invasion of PCa cells in vitro. Moreover, the inhibition of SLC6A1 suppressed the tumor growth in vivo. Additionally, immunohistochemical notches of PCNA and MMP-9 in the low-expression cluster were pointedly lower compared to those of NC group. Finally, the cell viability revealed that the overexpression of SLC6A1 obviously promoted the PCa cell resistant to docetaxel (DTX), and the transplanted tumor in the overexpression group had no significant reduction compared with the untreated group. CONCLUSIONS: Our data suggest that SLC6A1 overexpression may be associated with aggressive progression and short biochemical recurrence-free survival of PCa, and may be related to the resistance to docetaxel therapy.


Subject(s)
Biomarkers, Tumor/metabolism , Docetaxel/pharmacology , Drug Resistance, Neoplasm , GABA Plasma Membrane Transport Proteins/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Prostatic Neoplasms/pathology , Aged , Animals , Antineoplastic Agents/pharmacology , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cell Proliferation , Computational Biology/methods , Databases, Genetic/statistics & numerical data , Disease Progression , GABA Plasma Membrane Transport Proteins/genetics , Humans , Male , Mice , Mice, Nude , Prognosis , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Survival Rate , Xenograft Model Antitumor Assays
13.
Clin Invest Med ; 43(3): E49-59, 2020 09 24.
Article in English | MEDLINE | ID: mdl-32971585

ABSTRACT

PURPOSE: To investigate the clinical relevance and biological function of the kinesin super-family protein 4A (KIF4A) expression in prostate cancer (PCa). METHODS: We examined 1) the relationship between the expression of KIF4A and clinico-pathological characteristics of PCa patients using a tissue microarray and the Cancer Genome Atlas database, 2) the prognostic value of KIF4A expression in patients using Kaplan-Meier plots and 3) the functions of KIF4A in LNCaP and DU145 cells, such as cell proliferation, cell cycle and cell apoptosis. RESULTS: Compared with normal prostate, the mRNA and protein expressions of KIF4A were up-regulated in PCa. The up-regulation expression rates of KIF4A in PCa were significantly related to the Gleason score (P.


Subject(s)
Gene Expression Regulation, Neoplastic , Prostatic Neoplasms , Biomarkers , Humans , Kinesins/genetics , Kinesins/metabolism , Male , Prognosis , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/genetics
15.
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
16.
Bioinformatics ; 34(14): 2515-2517, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29509844

ABSTRACT

Motivation: The large-scale multidimensional omics data in the Genomic Data Commons (GDC) provides opportunities to investigate the crosstalk among different RNA species and their regulatory mechanisms in cancers. Easy-to-use bioinformatics pipelines are needed to facilitate such studies. Results: We have developed a user-friendly R/Bioconductor package, named GDCRNATools, for downloading, organizing and analyzing RNA data in GDC with an emphasis on deciphering the lncRNA-mRNA related competing endogenous RNAs regulatory network in cancers. Many widely used bioinformatics tools and databases are utilized in our package. Users can easily pack preferred downstream analysis pipelines or integrate their own pipelines into the workflow. Interactive shiny web apps built in GDCRNATools greatly improve visualization of results from the analysis. Availability and implementation: GDCRNATools is an R/Bioconductor package that is freely available at Bioconductor (http://bioconductor.org/packages/devel/bioc/html/GDCRNATools.html). Detailed instructions, manual and example code are also available in Github (https://github.com/Jialab-UCR/GDCRNATools).


Subject(s)
Computational Biology/methods , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , RNA, Messenger/metabolism , Software , Gene Regulatory Networks , Humans , Neoplasms/genetics , Neoplasms/metabolism
17.
Epidemiology ; 30(3): 458-465, 2019 05.
Article in English | MEDLINE | ID: mdl-30601243

ABSTRACT

BACKGROUND: Few studies have modeled smoking histories by combining smoking intensity and duration to show what profile of smoking behavior is associated with highest risk of bladder cancer. This study aims to provide insight into the association between smoking exposure history and bladder cancer risk by modeling both smoking intensity and duration in a pooled analysis. METHODS: We used data from 15 case-control studies included in the bladder cancer epidemiology and nutritional determinants study, including a total of 6,874 cases and 17,727 controls. To jointly interpret the effects of intensity and duration of smoking, we modeled excess odds ratios per pack-year by intensity continuously to estimate the risk difference between smokers with long duration/low intensity and short duration/high intensity. RESULTS: The pattern observed from the pooled excess odds ratios model indicated that for a fixed number of pack-years, smoking for a longer duration at lower intensity was more deleterious for bladder cancer risk than smoking more cigarettes/day for a shorter duration. We observed similar patterns within individual study samples. CONCLUSIONS: This pooled analysis shows that long duration/low intensity smoking is associated with a greater increase in bladder cancer risk than short duration/high intensity smoking within equal pack-year categories, thus confirming studies in other smoking-related cancers and demonstrating that reducing exposure history to a single metric such as pack-years was too restrictive.


Subject(s)
Models, Biological , Smoking/epidemiology , Smoking/psychology , Urinary Bladder Neoplasms/epidemiology , Case-Control Studies , Female , Humans , Male , Risk Factors , Time Factors
18.
Clin Nephrol ; 91(4): 211-221, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30802203

ABSTRACT

OBJECTIVE: The current meta-analysis was performed to evaluate the safety and efficacy of retroperitoneoscopic renal pedicle ligation of lymphatic disconnection (RRPLD) compared with open surgery (OS) in the treatment of chyluria. MATERIALS AND METHODS: Relevant studies were retrieved from MEDLINE, EMBASE, -SCOPUS, the Cochrane library and two Chinese literature database resources (Wanfang and CNKI) in March 2016. All eligible studies comparing RRPLD with OS for chyluria were included in this study. The main outcome including operative time, blood loss, postoperative (PO) intestinal recovery time, PO drainage duration, PO hospital stay, PO time of returning to work, PO bed time, and complications as well as rate of recurrence for RRPLD and OS were pooled using the Revman software. RESULTS: Twelve studies with a total of 620 patients were included in this meta-analysis. Of these patients, 365 and 255 had undergone renal pedicle lymphatic ligation via RRPLD and OS, respectively. There were significant reductions in operative time, PO intestinal recovery time, PO drainage duration, PO hospital stay, PO time of returning to work, and possible reductions in intraoperative blood loss intraoperative and PO complications for RRPLD compared to OS. However, other outcome variables, such as PO time in bed and PO recurrence, were not found to be statistically significant for either group. CONCLUSION: Compared with OS, RRPLD has several advantages such as shorter operative time, less intraoperative blood loss, and lower incidence of complications. Thus, it may be an efficacious and safe therapeutic modality for chyluria.


Subject(s)
Chyle , Kidney/surgery , Laparoscopy , Lymphatic Vessels/surgery , Blood Loss, Surgical , Humans , Intestines/physiology , Laparoscopy/adverse effects , Length of Stay , Ligation , Operative Time , Postoperative Complications/etiology , Recovery of Function , Recurrence , Retroperitoneal Space , Urine , Urologic Surgical Procedures/adverse effects , Urologic Surgical Procedures/methods
19.
J Cell Biochem ; 119(9): 7245-7255, 2018 09.
Article in English | MEDLINE | ID: mdl-29797600

ABSTRACT

AT-rich interaction domain 4A (ARID4A) and AT-rich interaction domain 4B (ARID4B), which are both the AT-rich interaction domain (ARID) family, have been reported to be oncogene or tumor suppressor gene in various human malignances, but there is no involvement about their functions in prostate cancer (PCa). Our previous study has reported that microRNA-30d (miR-30d) expression can predicted poor clinical prognosis in PCa, however, the underlying mechanisms of miR-30d have not been fully described. The aim of our study is to investigate the expression relevance between miR-30d and ARID4A or ARID4B, and examine the clinical significance and biological function of ARID4A and AIRD4B in PCa. In this study, both ARID4A and ARID4B were identified as the target genes of miR-30d. In addition, the mRNA expression of miR-30d in PCa tissues were significantly negative correlated with ARID4A (Pearson correlation coefficient = -0.313, P = 0.001) and ARID4B (Pearson correlation coefficient = -0.349, P < 0.001), while there was a positive correlation between ARID4A and ARID4B (Pearson correlation coefficient = 0.865, P < 0.001). Moreover, both ARID4A and ARID4B were significantly downregulated in PCa tissues with high Gleason scores (P = 0.005, P = 0.033), PSA failure (P = 0.012, P = 0.05) and short biochemical recurrent-free survival (P = 0.033, P = 0.031). Furthermore, the knockout expression of ARID4A and ARID4B promoted PCa cell proliferation, migration and invasion in vitro. In conclusion, our results indicated that ARID4A and ARID4B may serve as tumor suppressor in PCa progression, suggesting that they might be the potential therapeutic targets in prostate cancer.


Subject(s)
Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Retinoblastoma-Binding Protein 1/genetics , Retinoblastoma-Binding Protein 1/metabolism , Aged , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cohort Studies , Disease Progression , Down-Regulation , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Genes, Tumor Suppressor , Humans , Kaplan-Meier Estimate , Male , Neoplasm Invasiveness , Statistics, Nonparametric
SELECTION OF CITATIONS
SEARCH DETAIL