Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
2.
Transl Androl Urol ; 11(2): 139-148, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35280663

ABSTRACT

Background: Quick and accurate identification of urinary calculi patients with positive urinary cultures is critical to the choice of the treatment strategy. Predictive models based on machine learning algorithms provide a new way to solve this problem. This study aims to determine the predictive value of machine learning algorithms using a urine culture predictive model based on patients with urinary calculi. Methods: Data were collected from four clinical centers in the period of June 2016, to May 2019. 2,054 cases were included in the study. The dataset was randomly split into ratios of 5:5, 6:4, and 7:3 for model construction and validation. Predictive models of urine culture outcomes were constructed and validated by logistic regression, random forest, adaboost, and gradient boosting decision tree (GBDT) models. Each ratio's construction and verification were repeated five times independently for cross-validation. The Matthews correlation coefficient (MMC), F1-score, receiver operating characteristic (ROC) curve with the area under curve (AUC) was used to evaluate the performance of each prediction model. The additive net reclassification index (NRI) and absolute NRI were used to assess the predictive capabilities of the models. Results: Four prediction models of urinary culture results in patients with urinary calculi were constructed. The mean AUCs of the logistic regression, random forest, adaboost, and GBDT models were 0.761 (95% CI: 0.753-0.770), 0.790 (95% CI: 0.782-0.798), 0.779 (95% CI: 0.766-0.791), and 0.831 (95% CI: 0.823-0.840), respectively. Moreover, the average MMC and F1-score of GBDT model was 0.460 and 0.588, which was improved compared to logistic regression model of 0.335 and 0.501. The additive NRI and absolute NRI of the GBDT and logistic regression models were 0.124 (95% CI: 0.106-0.142) and 0.065 (95% CI: 0.060-0.069), respectively. Conclusions: Our results indicate that machine learning algorithms may be useful tools for urine culture outcome prediction in patients with urinary calculi because they exhibit superior performance compared with the logistic regression model.

3.
Int J Oncol ; 60(4)2022 Apr.
Article in English | MEDLINE | ID: mdl-35191514

ABSTRACT

Subsequently to the publication of the above article, the authors have realized that, on p. 390, the data selected for the siRNA­1 and siRNA­2 experiments for the ACHN and 786-O cell lines concerning both the invasion and the migration assays in Fig. 4B were selected inappropriately. Furthermore, after having inspected the published version of Fig. 5, the authors have realized that, for the immunofluorescence experiments shown in Fig. 5D, the first 'Merged' pictures for the first two columns of the ACHN cell line were accidentally published in the wrong order. The corrected versions of Figs. 4, and 5, including all the correct data for Figs. 4B and 5D, are shown on the next three pages. The authors confirm that these data continue to support the main conclusions presented in their paper, and are grateful to the Editor of International Journal of Oncology for granting them this opportunity to publish a Corrigendum. They also apologize to the readership for any inconvenience caused. [International Journal of Oncology 53: 384­394, 2018; DOI: 10.3892/ijo.2018.4395].

4.
Cell Mol Life Sci ; 77(6): 1135-1152, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31321444

ABSTRACT

ZNFX1 anti-sense RNA 1 (ZFAS1) has been indicated in the tumorigenesis of various human cancers. However, the role of ZFAS1 in prostate cancer (PCa) progression and the underlying mechanisms remain incompletely understood. In the present study, we discovered that ZFAS1 is upregulated in PCa and that ZFAS1 overexpression predicted poor clinical outcomes. ZFAS1 overexpression notably promoted the proliferation, invasion, and epithelial-mesenchymal transition of PCa cells. Furthermore, we not only discovered that miR-27a/15a/16 are targeted by ZFAS1, which binds to their miRNA-response elements, but also revealed their tumor suppressor roles in PCa. We also identified that the Hippo pathway transducer YAP1, as well as its cooperator, TEAD1, are common downstream targets of miR-27a/15a/16. In addition, H3K9 demethylase KDM3A was found to be another target gene of miR-27a. Importantly, YAP1, TEAD1, and KDM3A all act as strong c-Myc inducers in an androgen-independent manner. Taken together, we suggest a regulatory network in which ZFAS1 is capable of enhancing c-Myc expression by inducing the expression of YAP1, TEAD1, and KDM3A through crosstalk with their upstream miRNAs, thereby globally promoting prostate cancer tumorigenesis.


Subject(s)
Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins c-myc/genetics , RNA, Long Noncoding/genetics , Animals , Cell Line, Tumor , Gene Regulatory Networks , Humans , Male , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics
5.
Cancer Gene Ther ; 27(6): 412-423, 2020 06.
Article in English | MEDLINE | ID: mdl-31186514

ABSTRACT

Minichromosome maintenance 3 (MCM3) protein has been widely studied due to its essential role in DNA replication. In addition, it is overexpressed in several human tumor types. However, the role of this protein in renal cell carcinoma (RCC) is not widely known. In this study, we demonstrated that polo-like kinase 1 (PLK1)-mediated MCM3 phosphorylation regulates proliferation and apoptosis in RCC. Our results confirm that PLK1 and phospho-MCM3 (p-MCM3) are highly expressed in renal cell carcinoma. The expression of PLK1 is closely related to the clinical characteristics of renal cell carcinoma. They play important roles in the proliferation and apoptosis of RCC. In vitro, after overexpression of PLK1 or MCM3, the proliferation of RCC cells was significantly enhanced and cell apoptosis was inhibited, while after knockout, the proliferation of RCC cells was weakened and cell apoptosis was promoted. In addition, Mn2+-Phos-tag SDS-PAGE, western blotting, and immunofluorescence were utilized to determine that MCM3 is a physiological substrate of PLK1, which is phosphorylated on serine 112 (Ser112) in a PLK1-dependent manner. PLK1-mediated MCM3 phosphorylation promotes RCC cell cycle proliferation and suppresses apoptosis in vitro. Moreover, we found that PLK1-mediated MCM3 phosphorylation induced cellular proliferation and decreased apoptosis, as well as tumor growth in mice. Overall, we conclude that PLK1-mediated MCM3 phosphorylation is a novel mechanism to regulate RCC proliferation and apoptosis.


Subject(s)
Carcinoma, Renal Cell/metabolism , Cell Cycle Proteins/metabolism , Kidney Neoplasms/metabolism , Minichromosome Maintenance Complex Component 3/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Adult , Aged , Animals , Apoptosis/physiology , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Proliferation/physiology , Female , Heterografts , Humans , Kidney Neoplasms/pathology , Male , Mice , Mice, Nude , Middle Aged , Phosphorylation , Survival Analysis , Transfection , Up-Regulation , Polo-Like Kinase 1
6.
Cancer Gene Ther ; 27(3-4): 189-202, 2020 04.
Article in English | MEDLINE | ID: mdl-30723269

ABSTRACT

Accumulating evidence shows that circular RNAs (circRNAs) function as microRNA sponges that regulate gene expression in the progression of human cancers. However, the roles of circRNAs and functional miRNA sponges in bladder cancer (BC) remain largely unknown. In the present study, we applied bioinformatics methods and hypothesised that miR-146b may target the 3'-untranslated region (UTR) of CARMA3 mRNA and circINTS4 may serve as a sponge for miR-146b in BC tumorigenesis. Expression of circINTS4 was significantly increased in miR-146b-downregulated BC tissues and cell lines compared to adjacent normal tissues. Furthermore, circINTS4 was found to control multiple pathological processes, including cell proliferation and migration, the cell cycle and apoptosis. Regarding the mechanism, circINTS4 directly bound to miR-146b to inhibit its activity of targeting the 3'-UTR of CARMA3 mRNA. In addition, circINTS4 could activate the NF-kB signalling pathway and suppress the P38 MAPK signalling pathway in a CARMA3-mediated manner in BC cells. In summary, the circINTS4/miR-146b/CARMA3 axis might serve as a promising therapeutic target for BC intervention.


Subject(s)
CARD Signaling Adaptor Proteins/genetics , Carcinogenesis/genetics , MicroRNAs/metabolism , RNA, Circular/metabolism , Urinary Bladder Neoplasms/genetics , 3' Untranslated Regions/genetics , Aged , Animals , Apoptosis/genetics , Cell Cycle/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cystectomy , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Humans , MAP Kinase Signaling System/genetics , Male , Mice , Middle Aged , NF-kappa B/metabolism , Urinary Bladder/pathology , Urinary Bladder/surgery , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/surgery , Xenograft Model Antitumor Assays
7.
Mol Oncol ; 13(10): 2079-2097, 2019 10.
Article in English | MEDLINE | ID: mdl-31294899

ABSTRACT

DAPK, a transcriptional target of the p53 protein, has long been characterized as a tumor suppressor that acts as a negative regulator in multiple cellular processes. However, increasing studies have suggested that the role of DAPK may vary depending on cell type and cellular context. Thus far, the expression and function of DAPK in clear cell renal cell carcinoma (ccRCC) remain ambiguous. Since ccRCC behaves in an atypical way with respect to p53, whether the p53-DAPK axis functions normally in ccRCC is also an intriguing question. Here, tissue specimens from 61 ccRCC patients were examined for DAPK expression. Functional studies regarding apoptosis, growth, and migration were used to determine the role of DAPK in renal cancer cells. The validity of the p53-DAPK axis in ccRCC was also determined. Our study identified DAPK as a negative regulator of ccRCC, and its expression was reduced in certain subgroups. However, the p53-DAPK axis was disrupted due to upregulation of miR-34a-5p under stressed conditions. miR-34a-5p was identified as a novel repressor of DAPK acting downstream of p53. Inhibition of miR-34a-5p can correct the p53-DAPK axis disruption by upregulating DAPK protein and may have potential to be used as a therapeutic target to improve outcomes for ccRCC patients.


Subject(s)
Carcinoma, Renal Cell/genetics , Kidney Neoplasms/genetics , MicroRNAs/genetics , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Aged , Animals , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Death-Associated Protein Kinases/metabolism , Disease Progression , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Male , Mice, Inbred BALB C , Middle Aged
8.
Cancer Cell Int ; 19: 128, 2019.
Article in English | MEDLINE | ID: mdl-31114448

ABSTRACT

BACKGROUND: Cumulative evidences demonstrated the aberrant overexpression of Small Nucleolar RNA Host Gene 12 (SNHG12) in diverse human cancer. However, the expression status and involvement of SNHG12 in renal cell carcinoma is still elusive. METHODS: The expression of SNHG12 was determined by q-PCR. The transcriptional regulation was interrogated by luciferase reporter assay. Cell viability was measured with CCK-8 kit. The anchorage-independent was evaluated by soft agar assay. Cell apoptosis was analyzed by Annexin V/7-AAD double staining. The migration and invasion were determined by trans-well assay and wound scratch closure. The in vivo tumor growth was monitored in xenograft mice model. Protein expression was quantified by immunoblotting. RESULTS: SNHG12 was aberrantly up-regulated in renal carcinoma both in vivo and in vitro. High expression of SNHG12 associated with poor prognosis. Deficiency of SNHG12 significantly suppressed cell viability, anchorage-independent growth and induced apoptosis. In addition, SNHG12 silencing inhibited migrative and invasive in vitro and xenograft tumor growth in vivo. Mechanistically, SNHG12 modulated HIF1α expression via competing with miR-199a-5p, which consequently contributed to its oncogenic potential. MiR-199a-5p inhibition severely compromised SNHG12 silencing-elicited tumor repressive effects. CONCLUSION: Our data uncovered a crucial role of SNHG12-miR-199a-5p-HIF1α axis in human renal cancer.

9.
Onco Targets Ther ; 12: 2713-2727, 2019.
Article in English | MEDLINE | ID: mdl-31043790

ABSTRACT

BACKGROUND: The non-receptor tyrosine kinase Fyn-related kinase (FRK) has been reported to affect cell proliferation in several cancer types. However, its effect on the proliferation of clear cell renal cell carcinoma (ccRCC) remains largely unknown. PURPOSE: The objective of this study was to investigate the expression pattern and function of FRK in ccRCC. We further determined how FRK interacted with other molecules to regulate ccRCC proliferation. PATIENTS AND METHODS: The expression of FRK in ccRCC samples and paired normal renal tissues from 30 patients were analyzed by immunoblotting, immunohistochemistry and quantitative PCR. Then the role of FRK in ccRCC proliferation was analyzed by Cell Counting Kit-8, colony formation assay and EdU incorporation assay. In addition, the miRNA targeting FRK was predicted through a bioinformatic approach and validated by quantitative PCR, immunoblotting and luciferase reporter assay. Finally, the underlying mechanism of FRK regulation of ccRCC proliferation was also determined. RESULTS: Low expression of FRK was detected in ccRCC samples and predicted poor survival for ccRCC patients. FRK inhibited the proliferation of ccRCC cells via phosphorylating downstream PTEN. miR-19 was identified as a novel suppressor of FRK in renal cancer cells and it promoted the proliferation of ccRCC by inhibiting the FRK-PTEN axis. CONCLUSION: Our results unravel a new regulatory mechanism involved in ccRCC proliferation and may be useful in the identification of therapeutic targets for ccRCC.

10.
Oncol Rep ; 41(4): 2440-2452, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30816525

ABSTRACT

Adrenocortical carcinoma (ACC) is a rare disease associated with a poor prognosis. Furthermore, the underlying molecular mechanism of carcinogenesis is poorly understood, and prognostic prediction of ACC has low accuracy. In the present study, a bioinformatics approach was used to investigate the molecular mechanisms and prognosis of ACC. Samples of adrenal tumors were collected from patients undergoing adrenalectomy at the Department of Urology, the First Hospital of China Medical University. The analyzed gene datasets were downloaded from the Gene Expression Omnibus and The Cancer Genome Atlas (TCGA) database. Following this, the differentially expressed genes (DEGs) were included in Gene Ontology enrichment, and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway, protein­protein interaction network and survival analyses. MTT colorimetric assays, colony formation assays and 5­ethynyl­20­deoxyuridine incorporation assays were also conducted to evaluate ACC cell proliferation. The identified DEGs included 20 downregulated genes and 51 upregulated genes, which were highly associated with the cell cycle, organelle fission, chromosome segregation, cell division and spindle stability. The top 14 hub genes were subsequently confirmed by reverse transcription­quantitative polymerase chain reaction in ACC and adrenocortical adenoma samples. It was identified that the nuclear division cycle 80, cyclin B2 and topoisomerase 2­α may serve important roles in adrenocortical tumor development. Furthermore, these three genes predicted overall survival and recurrence­free survival in patients with ACC from the TCGA cohort. The findings identified three novel genes that have important roles in carcinogenesis and in the prognostic prediction of ACC.


Subject(s)
Adrenal Cortex Neoplasms/pathology , Adrenocortical Carcinoma/pathology , Cyclin B2/metabolism , DNA Topoisomerases, Type II/metabolism , Neoplasm Recurrence, Local/diagnosis , Nuclear Proteins/metabolism , Poly-ADP-Ribose Binding Proteins/metabolism , Adrenal Cortex/pathology , Adrenal Cortex/surgery , Adrenal Cortex Neoplasms/genetics , Adrenal Cortex Neoplasms/mortality , Adrenalectomy , Adrenocortical Carcinoma/genetics , Adrenocortical Carcinoma/mortality , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Proliferation/genetics , Computational Biology , Cyclin B2/genetics , Cytoskeletal Proteins , DNA Topoisomerases, Type II/genetics , Databases, Genetic/statistics & numerical data , Datasets as Topic , Disease-Free Survival , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Nuclear Proteins/genetics , Poly-ADP-Ribose Binding Proteins/genetics , Prognosis , Protein Interaction Maps/genetics , RNA, Small Interfering/metabolism , Up-Regulation
11.
J Exp Clin Cancer Res ; 38(1): 111, 2019 Mar 01.
Article in English | MEDLINE | ID: mdl-30823924

ABSTRACT

BACKGROUND: The effect of competing endogenous RNA (ceRNA) can regulate gene expression by competitively binding microRNAs. Fascin-1 (FSCN1) plays an important role in the regulation of cellular migration and invasion during tumor progression, but how its regulatory mechanism works through the ceRNA effect is still unclear in bladder cancer (BLCA). METHODS: The role of fascin-1, miR-200b, and ZEB1-AS1 in BLCA was investigated in vitro and in vivo. The interaction between fascin-1, miR-200b, and ZEB1-AS1 was identified using bioinformatics analysis, luciferase activity assays, RNA-binding protein immunoprecipitation (RIP), quantitative PCR, and western blotting. Loss (or gain)-of-function experiments were performed to investigate the biological roles of miR-200b and ZEB1-AS1 on migration, invasion, proliferation, cell apoptosis, and cell cycle. RESULTS: ZEB1-AS1 functions as a competing endogenous RNA in BLCA to regulate the expression of fascin-1 through miR-200b. Moreover, the oncogenic long non-coding RNA ZEB1-AS1 was highly expressed in BLCA and positively correlated with high tumor grade, high TNM stage, and reduced survival of patients with BLCA. Moreover, ZEB1-AS1 downregulated the expression of miR-200b, promoted migration, invasion, and proliferation, and inhibited apoptosis in BLCA. Furthermore, we found TGF-ß1 induced migration and invasion in BLCA by regulating the ZEB1-AS1/miR-200b/FSCN1 axis. CONCLUSION: The observations in this study identify an important regulatory mechanism of fascin-1 in BLCA, and the TGF-ß1/ZEB1-AS1/miR-200b/FSCN1 axis may serve as a potential target for cancer therapeutic purposes.


Subject(s)
Carrier Proteins/genetics , Gene Expression Regulation, Neoplastic/genetics , MicroRNAs/genetics , Microfilament Proteins/genetics , RNA, Long Noncoding/genetics , Urinary Bladder Neoplasms/pathology , Adult , Aged , Animals , Cell Line, Tumor , Cell Movement/physiology , Female , Heterografts , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Invasiveness/genetics , Signal Transduction/genetics , Transforming Growth Factor beta1/metabolism , Urinary Bladder Neoplasms/genetics
12.
J Cell Mol Med ; 23(3): 2064-2076, 2019 03.
Article in English | MEDLINE | ID: mdl-30592142

ABSTRACT

Urinary bladder neoplasm is one of the most common cancers worldwide. Cancer stem cells (CSCs) have been proven to be an important cause of cancer progression and poor prognosis. In the present study, we established bladder CSCs and identified the crucial differentially expressed genes (DEGs) between these cells and parental bladder cancer cells. Analyses of bioinformatics data and clinical samples from local hospitals showed that stearoyl CoA desaturase-1 (SCD) was the key factor among the DEGs. A significant correlation between SCD gene expression and poor prognosis among patients with bladder cancer was observed in our data. Loss-of-function experiments further revealed that the SCD inhibitor A939572 and SCD gene interference reduced cell proliferation and invasion. The above data suggest that SCD may serve as a novel marker for the prediction of tumour progression and poor prognosis in patients with bladder cancer.


Subject(s)
Stearoyl-CoA Desaturase/metabolism , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Proliferation/physiology , Disease Progression , Female , Humans , Mice , Mice, Nude , Neoplastic Stem Cells/pathology , Prognosis
13.
Am J Cancer Res ; 8(8): 1414-1426, 2018.
Article in English | MEDLINE | ID: mdl-30210913

ABSTRACT

Several studies have shown that low expression of hZIP1 is closely associated with many human cancers, including clear cell renal cell carcinoma (ccRCC). In this study, we aimed to explore the potential mechanism responsible for hZIP1 silencing and revealed a novel regulatory pathway in the pathogenesis of ccRCC. Here, miR-223 was predicted and experimentally validated to be a regulator of hZIP1, and its expression was negatively correlated with the mRNA levels of hZIP1 in primary tumors. Upregulation of hZIP1 inhibited cell proliferation, cell cycle progression, and invasion and induced apoptosis, while inhibition of miR-223 showed the opposite effect on cellular processes. Moreover, GAS5 interacted with miR-223 and was markedly downregulated in tumors. Knockdown of GAS5 partially reversed the effect of the miR-223 inhibitor on cell proliferation, cell cycle distribution, apoptosis and invasion. In addition, GAS5 acted as a molecular sponge to positively regulate the mRNA and protein levels of hZIP1 via regulating miR-223. The tumorigenicity of ccRCC cells was enhanced by silencing GAS5 but diminished by overexpression of hZIP1 in vivo. Clinically, the low expression of hZIP1 was significantly correlated with advanced clinical stage and Fuhrman stage. Downregulation of GAS5 indicated tumor progression and recurrence and was independently associated with disease-free survival of patients. Taken together, our results suggest that GAS5 may act as a competing endogenous RNA (ceRNA) to regulate hZIP1 by sponging miR-223 in the progression of ccRCC and that targeting the GAS5/miR-223/hZIP1 axis may serve as a therapeutic strategy for patients.

14.
Proteomics ; 18(19): e1800001, 2018 10.
Article in English | MEDLINE | ID: mdl-29882248

ABSTRACT

In light of the increasing incidence of renal cell carcinoma (RCC), its molecular mechanisms have been comprehensively explored in numerous recent studies. However, few studies focus on the influence of multi-factor interactions during the occurrence and development of RCC. This study aims to investigate the quantitative global proteome and the changes in lysine succinylation in related proteins, seeking to facilitate a better understanding of the molecular mechanisms underlying RCC. LC-MS/MS combined with bioinformatics analysis are used to quantitatively detect the perspectives at the global protein level. IP and WB analysis were conducted to further verify the alternations of related proteins and lysine succinylation. A total of 3,217 proteins and 1,238 lysine succinylation sites are quantified in RCC tissues, and 668 differentially expressed proteins and 161 differentially expressed lysine succinylation sites are identified. Besides, expressions of PGK1 and PKM2 at protein and lysine, succinylation levels are significantly altered in RCC tissues. Bioinformatics analysis indicates that the glycolysis pathway is a potential mechanism of RCC progression and lysine succinylation may plays a potential role in energy metabolism. These results can provide a new direction for exploring the molecular mechanism of RCC tumorigenesis.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Renal Cell/metabolism , Energy Metabolism , Kidney/metabolism , Lysine/metabolism , Proteome/metabolism , Succinic Acid/metabolism , Biomarkers, Tumor/analysis , Carcinoma, Renal Cell/pathology , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Protein Processing, Post-Translational , Proteome/analysis
15.
Med Sci Monit ; 24: 3405-3414, 2018 May 23.
Article in English | MEDLINE | ID: mdl-29790485

ABSTRACT

BACKGROUND Cancer immunotherapy is the use of the immune system to treat cancer. After years of research, there have been a significant number of publications in this field. We analyzed the literature and performed a hotspot analysis to identify important areas of future scientific research. MATERIAL AND METHODS Articles (2945) related to cancer immunotherapy published in the past 3 years were selected as the research sample. BICOMB software was then used to retrieve the high-frequency words and construct a text/co-word matrix. Next, gCLUTO software was used to analyze the matrix by double-clustering and visual analysis, in a strategy of hotspot identification. RESULTS We constructed a text and co-word matrix composed of 40 high-frequency words and 2945 articles and generated a hotspot "peak map" based on double-clustering analysis. The strategic coordinates were set by use of a co-word matrix and clustering analysis. The distribution of organs or disease and the subclass of cancer immunotherapy were analyzed. CONCLUSIONS In this study, we classified the hot-spots of "tumor immunotherapy" into 6 categories and 8 aspects. Calculation and analysis revealed that the field of tumor immunotherapy shows a slight trend of polarization, and the immune checkpoint inhibitor PD1 blocker shows the greatest potential for future development.


Subject(s)
Bibliometrics , Immunotherapy , Neoplasms/immunology , Neoplasms/therapy , Cluster Analysis , Humans , Medical Subject Headings , Publications , Surveys and Questionnaires
16.
Int J Oncol ; 53(1): 384-394, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29750422

ABSTRACT

Aberrant expression of long noncoding RNAs (lncRNAs) is associated with cancer tumorigenesis and progression. It has been suggested that lncRNAs may be potential clinical diagnostic and prognostic biomarkers, and therapeutic targets. In the present study, the expression levels of small nucleolar RNA host gene 15 (SNHG15) were significantly upregulated in renal cell carcinoma (RCC) tissues and cell lines compared with in adjacent tissues and a proximal tubule epithelial cell line, as determined by reverse transcription­quantitative polymerase chain reaction. Subsequently, knockdown of SNHG15 expression with small interfering RNA inhibited RCC proliferation, invasion and migration, was determined by western blotting and Transwell assays. Furthermore, the present study suggested that SNHG15 may be involved in the nuclear factor­κB signaling pathway, induce the epithelial­mesenchymal transition process, and promote RCC invasion and migration.


Subject(s)
Carcinoma, Renal Cell/genetics , Cell Proliferation/genetics , RNA, Long Noncoding/genetics , Transcription Factor RelA/genetics , Aged , Carcinoma, Renal Cell/pathology , Cell Movement/genetics , Disease Progression , Disease-Free Survival , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Male , Middle Aged , NF-kappa B/genetics , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Staging , Signal Transduction/genetics
17.
Biochem Biophys Res Commun ; 501(4): 913-919, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29772238

ABSTRACT

Transforming growth factor-ß1 (TGF-ß1) plays a crucial role in the signaling network that controls cellular invasion and motility capability during tumor development. To investigate whether fascin1 plays a crucial role in TGF-ß1-facilitated invasion and migration of kidney cancer cells (KCC), real-time PCR and western blotting were used to test the fascin1 expression after TGF-ß1 treatment (10 ng/ml) in 769-P and OSRC cells. Fascin1 was silenced using the small interfering RNA (siRNA) technique. Cytoskeleton staining was used to test the change of Cytoskeleton. Cell migration and invasion changes were measured by wound-healing and Transwell assay. The results indicate that mRNA and protein levels of fascin1 were dramatically increased after treatment with 10 ng/ml TGF-ß1 in 769-P and OSRC cells. TGF-ß1 promoted the occurrence of EMT (Epithelial-Mesenchymal Transition) and the invasive and migratory capabilities of the two cell lines after treatment with 10 ng/ml TGF-ß1. In addition, fascin1 siRNA dramatically attenuated the invasiveness and migration induced by TGF-ß1. Furthermore, we identified that specific inhibitors of ERK and JNK signaling pathways, FR180204 and SP600125, can suppress TGF-ß1-induced fascin1 expression. In conclusion, these results reveal that fascin1 is an important mediator of TGF-ß1-induced invasion and migration of KCC through ERK and JNK signal pathways.


Subject(s)
Carrier Proteins/genetics , Cell Movement/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , MAP Kinase Signaling System/drug effects , Microfilament Proteins/genetics , Transforming Growth Factor beta1/pharmacology , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Movement/genetics , Epithelial-Mesenchymal Transition/drug effects , Gene Knockdown Techniques , Humans , Kidney Neoplasms/enzymology , Microfilament Proteins/metabolism , Neoplasm Invasiveness , RNA, Small Interfering/metabolism
18.
Int J Mol Med ; 41(3): 1765-1773, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29328435

ABSTRACT

miR­214 has been reported to be downregulated in several cancer types, such as bladder cancer. However, its involvement in apoptosis and chemoresistance has not been investigated. The present study aimed to clarify the biological function of miR­214 and potential mechanisms in chemoresistance of bladder cancer cells. Reverse transcription­quantitative polymerase chain reaction demonstrated that miR­214 was downregulated in bladder cancer tissues compared with the level in normal tissues. miR­214 was downregulated in bladder cancer cell lines compared with the level in the normal cell line SV­HUC­1. miR­214 mimics were transfected into T24 and J82 cell lines to restore its expression. The results indicated that miR­214 mimic inhibited proliferation and invasion in these cell lines. In addition, miR­214 mimic reduced cisplatin resistance in T24 and J82 cells, indicated by the inhibition of cell viability and upregulation of cell apoptosis. Western blotting demonstrated that miR­214 mimic was able to upregulate cleaved caspase­3 and cleaved poly (ADP­ribose) polymerase (PARP), while downregulate caspase­3 and PARP expression, and AKT phosphorylation. Using prediction software, it was revealed that the netrin­1 oncoprotein is on the target list of miR­214. miR­214 also downregulated netrin­1 protein and mRNA expression levels in the T24 and J82 cell lines. Luciferase reporter assays demonstrated that netrin­1 acted as a direct target of miR­214. A negative correlation between netrin­1 and miR­214 expression in bladder cancer tissues was also observed. In addition, cisplatin treatment could induce netrin­1 protein expression in bladder cancer cells and miR­214 mimic partly blocked this phenomenon. Netrin­1 plasmid transfection inhibited cisplatin­induced apoptosis, upregulated AKT phosphorylation, and downregulated caspase­3 and PARP cleavage. Netrin­1 was restored in cells transfected with miR­214 mimic using plasmid transfection. Netrin­1 transfection restored AKT phosphorylation and blocked caspase/PARP cleavage in the T24 and J82 cell lines. In conclusion, the present study demonstrated that miR­214 is downregulated in bladder cancer tissues and cell lines. miR­214 reduces chemoresistance by targeting netrin­1 in bladder cancer cell lines.


Subject(s)
Cisplatin/pharmacology , Drug Resistance, Neoplasm/genetics , MicroRNAs/metabolism , Netrin-1/metabolism , Urinary Bladder Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation/drug effects , Down-Regulation/genetics , Drug Resistance, Neoplasm/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , MicroRNAs/genetics , Middle Aged , Neoplasm Invasiveness , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism
19.
Eur Urol ; 73(3): 385-391, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29137830

ABSTRACT

BACKGROUND: Recent large high-quality trials have questioned the clinical effectiveness of medical expulsive therapy using tamsulosin for ureteral stones. OBJECTIVE: To evaluate the efficacy and safety of tamsulosin for distal ureteral stones compared with placebo. DESIGN, SETTING, AND PARTICIPANTS: We conducted a double-blind, placebo-controlled study of 3296 patients with distal ureteral stones, across 30 centers, to evaluate the efficacy and safety of tamsulosin. INTERVENTION: Participants were randomly assigned (1:1) into tamsulosin (0.4mg) or placebo groups for 4 wk. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: The primary end point of analysis was the overall stone expulsion rate, defined as stone expulsion, confirmed by negative findings on computed tomography, over a 28-d surveillance period. Secondary end points included time to stone expulsion, use of analgesics, and incidence of adverse events. RESULTS AND LIMITATIONS: Among 3450 patients randomized between September 1, 2011, and August 31, 2013, 3296 (96%) were included in the primary analysis. Tamsulosin benefits from a higher stone expulsion rate than the placebo (86% vs 79%; p<0.001) for distal ureteral stones. Subgroup analysis identified a specific benefit of tamsulosin for the treatment of large distal ureteral stones (>5mm). Considering the secondary end points, tamsulosin-treated patients reported a shorter time to expulsion (p<0.001), required lower use of analgesics compared with placebo (p<0.001), and significantly relieved renal colic (p<0.001). No differences in the incidence of adverse events were identified between the two groups. CONCLUSIONS: Our data suggest that tamsulosin use benefits distal ureteral stones in facilitating stone passage and relieving renal colic. Subgroup analyses find that tamsulosin provides a superior expulsion rate for stones >5mm, but no effect for stones ≤5mm. PATIENT SUMMARY: In this report, we looked at the efficacy and safety of tamsulosin for the treatment of distal ureteral stones. We find that tamsulosin significantly facilitates the passage of distal ureteral stones and relieves renal colic.

20.
BMC Cancer ; 17(1): 861, 2017 Dec 16.
Article in English | MEDLINE | ID: mdl-29246203

ABSTRACT

BACKGROUND: The significance of PLK1 (polo-like kinase 1) has become increasingly essential as both a biomarker and a target for cancer treatment. Here, we aimed to determine the downstream genes of PLK1 and their effects on the carcinogenesis and progression of bladder cancer. METHODS: Specific siRNA was utilized to silence the target gene expression. The cell proliferation, invasion and migration of bladder cancer cells by MTT assay, BrdU assay and transwell assay. The differential expression genes were identified using Affymetrix HTA2.0 Array. The KEGG, GO and STRING analysis were used to analyze the signaling pathway and protein-protein interaction. Spearman analysis was used to analyze the correlation between protein and protein, between protein and clincopathologic characteristics. RESULTS: PLK1 siRNA hindered the proliferation, invasion and migration of bladder cancer cells, as determined by the MTT, BrdU and transwell assays. A total of 561 differentially expressed genes were identified using an Affymetrix HTA2.0 Array in PLK1 knockdown T24 cells. According to KEGG, GO and STRING analysis, five key genes (BUB1B, CCNB1, CDC25A, FBXO5, NDC80) were determined to be involved in cell proliferation, invasion and migration. PLK1 knockdown decreased BUB1B, CCNB1, CDC25A and NDC80 expressions but increased FBXO5 expression. BUB1B, CCNB1, CDC25A and NDC80 were positively correlated with cell proliferation, invasion, migration and PLK1 expression in tissues, but FBXO5 was negatively correlated with each of those factors. The results showed that the five genes expressions were significantly correlation with the PLK1 expression in normal bladder tissues and bladder cancer tissues. Four of them (BUB1B, CCNB1, CDC25A, NDC80) were obviously positive correlations with pT stage and metastasis. But FBXO5 was negative correlated with pT stage and metastasis. Furthermore, significant correlations were found between CCNB1 or CDC25A or NDC80 and histological grade; between BUB1B or NDC80 and recurrence. CONCLUSION: Five downstream genes of PLK1 were associated with the regulation of cell proliferation, invasion and migration in bladder cancer. Furthermore, these genes may play important roles in bladder cancer and become important biomarkers and targets for cancer treatment.


Subject(s)
Carcinogenesis/genetics , Cell Cycle Proteins/metabolism , Gene Expression Regulation, Neoplastic , Neoplasm Recurrence, Local/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Urinary Bladder Neoplasms/genetics , Apoptosis/genetics , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Disease Progression , Gene Expression Profiling/methods , Gene Knockdown Techniques , Humans , Neoplasm Invasiveness/genetics , Oligonucleotide Array Sequence Analysis , Protein Interaction Maps/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/metabolism , Signal Transduction/genetics , Software , Urinary Bladder/pathology , Urinary Bladder Neoplasms/pathology , Polo-Like Kinase 1
SELECTION OF CITATIONS
SEARCH DETAIL