Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Front Oncol ; 13: 1130680, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37207146

RESUMO

Background: For metastatic prostate cancer (mPCa), radical prostatectomy (RP) and radiation therapy (RT) may improve overall survival (OS) and cancer-specific survival (CSS). Compared with RT, RP shows significant advantages in improving patient outcomes. External beam radiation therapy (EBRT) even slightly elevates CSM with no statistical difference in OS compared with no local treatment (NLT). Objective: To evaluate OS and CSS after local treatment (LT) (including RP and RT) versus NLT in mPCa. Design, setting, and participants Within the Surveillance, Epidemiology and End Results (SEER) database (2000-2018), 20098 patients with metastatic prostate cancer were selected in this study, of which 19433 patients had no local treatment, 377 patients with radical prostate treatment, and 288 patients with RT. Outcome measurements and statistical analysis: Multivariable competing risks regression analysis after propensity score matching (PSM) was used to calculate CSM. Multivariable Cox regression analysis was used to identify the risk factors. Kaplan-Meier methods were used to calculate OS. Results and limitations: A total of 20098 patients were included: NLT (n = 19433), RP (n=377) and RT (n=288). In a competing risk regression analysis after PSM (ratio 1:1), RP resulted in a significantly lower CSM (hazard ratio [HR] 0.36, 95% confidence interval [CI] 0.29-0.45) than NLT, while RT showed a slightly lower CSM (HR 0.77, 95% CI 0.63-0.95). In a competing risk regression analysis after PSM (ratio 1:1), RP led to a lower CSM (HR 0.56, 95% CI 0.41-0.76) versus RT. As for all-cause mortality (ACM), RP (HR 0.37, 95% CI 0.31-0.45) and RT (HR 0.66, 95% CI 0.56-0.79). also showed a downward trend. In terms of OS, RP and RT significantly improved the survival probability compared with NLT, with the effect of RP being more pronounced. Obviously, older age, Gleason scores ≥8, AJCC T3-T4 stage, AJCC N1, AJCC M1b-M1c were all associated with higher CSM (P <0.05). The same results held true for ACM. The limitation of this article is that it is not possible to assess the effect of differences in systemic therapy on CSM in mPCa patients and clinical trials are needed to verify the results. Conclusions: For patients with mPCa, both RP and RT are beneficial to patients, and the efficacy of RP is better than RT from the perspective of CSM and ACM. Older age, higher gleason scores and the more advanced AJCC TNM stage all put patients at higher risk of dying. Patient summary: A large population-based cancer database showed that in addition to first-line therapy (hormonal treatment), RP and radiotherapy can also benefit patients with mPCa.

2.
Aging Dis ; 14(5): 1757-1774, 2023 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-37196108

RESUMO

Neuroendocrine prostate cancer (NEPC) is a lethal subtype of prostate cancer. It is characterized by the loss of androgen receptor (AR) signaling in neuroendocrine transdifferentiation, and finally, resistance to AR-targeted therapy. With the application of a new generation of potent AR inhibitors, the incidence of NEPC is gradually increasing. The molecular mechanism of neuroendocrine differentiation (NED) after androgen deprivation therapy (ADT) remains largely unclear. In this study, using NEPC-related genome sequencing database analyses, we screened RACGAP1, a common differentially expressed gene. We investigated RACGAP1 expression in clinical prostate cancer specimens by IHC. Regulated pathways were examined by Western blotting, qRT-PCR, luciferase reporter, chromatin immunoprecipitation, and immunoprecipitation assays. The corresponding function of RACGAP1 in prostate cancer was analyzed by CCK-8 and Transwell assays. The changes of neuroendocrine markers and AR expression in C4-2-R and C4-2B-R cells were detected in vitro. We confirmed that RACGAP1 contributed to NE transdifferentiation of prostate cancer. Patients with high tumor RACGAP1 expression had shorter relapse-free survival time. The expression of RACGAP1 was induced by E2F1. RACGAP1 promoted neuroendocrine transdifferentiation of prostate cancer by stabilizing EZH2 expression in the ubiquitin-proteasome pathway. Moreover, overexpression of RACGAP1 promoted enzalutamide resistance of castration-resistant prostate cancer (CRPC) cells. Our results showed that the upregulation of RACGAP1 by E2F1 increased EZH2 expression, which drove NEPC progression. This study explored the molecular mechanism of NED and may provide novel methods and ideas for targeted therapy of NEPC.

4.
Front Oncol ; 11: 706883, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34631536

RESUMO

OBJECTIVE: Tumor metabolism has always been the focus of cancer research. SLC16A1, as a key factor in catalysis of monocarboxylate transport across the plasma membrane, has been found to be associated with the occurrence and metastasis of a variety of cancers, but its prognostic significance and mechanism in different tumors are still unclear. METHODS: Based on the gene expression matrix and clinical information of human cancer tissues acquired from TCGA and GTEX databases, the differential expression of SLC16A1 in different tumors and normal tissues was analyzed. To confirm the association between its expression, the mutation of MMRS gene, and the expression level of DNMTs. Univariate Cox regression was applied to analyze the association between SLC16A1 expression and patient prognosis. The effect of SLC16A1 expression on patient survival was examined by Kaplan Meier analysis. GSEA was used to identify related signaling pathways. RESULTS: The expression of SLC16A1 was differentially expressed in most tumors, especially in the urinary tract where it is commonly highly expressed, and differential expression of SLC16A1 in different clinical stages. SLC16A1 expression was significantly positively correlated with MMRS gene mutation and DNMTS expression. Moreover, high SLC16A1 expression was associated with poorer overall survival (OS) and progression-free survival (PFS) in urological cancers. In particular, the results of the enrichment analysis showed that SLC16A1 was associated with processes such as cell adhesion and many signaling pathways affecting cell cycle were significantly enriched in the group with high-expressed SLC16A1. CONCLUSION: SLC16A1 expression was upregulated in urological cancer. SLC16A1 may promote tumor development by regulating the epigenetic process of urological cancer and demonstrated a great potential as a prognostic biomarker of urological cancer patients.

5.
Front Oncol ; 11: 651921, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33869056

RESUMO

Clear cell renal cell carcinoma (ccRCC) is the most frequent and lethal subtype, which has high risk of metastasis or recurrence, accounting for 75-83% of renal cell carcinoma (RCC). Zrt- and Irt-like proteins (ZIP) family members (SLC39A1-14) function to pass zinc into the cytoplasm for many critical biological processes when cellular zinc is depleted. However, the functional analysis of individual ZIP family genes in ccRCC is not clarified. This study aimed to investigate whether ZIP family genes are related to the clinicopathological features and survival of ccRCC patients, and to identify the function of key gene of ZIP family in ccRCC in vitro. Through bioinformatics analysis of tumor databases, SLC39A8 was identified as a key gene of ZIP family in ccRCC, which could be used as an effective indicator for diagnosing ccRCC and judging its prognosis. With the progression of tumor, the expression of SLC39A8 decreased progressively. The prognosis of patients with low expression of SLC39A8 is significantly worse. Furthermore, we found that overexpression of SLC39A8 or treatment with low concentration of zinc chloride could effectively inhibit the proliferation, migration and invasion of ccRCC cells. Moreover, the inhibition effect of SLC39A8 overexpression could be enhanced by low concentration zinc supplement. Therefore, this study provides a novel understanding for the role of SLC39A8/zinc in the regulation of ccRCC progression. These findings provide a new direction and target for progressive ccRCC drug development and combination therapy strategies.

6.
Theranostics ; 11(2): 841-860, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33391508

RESUMO

Current endocrine therapy for prostate cancer (PCa) mainly inhibits androgen/androgen receptor (AR) signaling. However, due to increased intratumoural androgen synthesis and AR variation, PCa progresses to castration-resistant prostate cancer (CRPC), which ultimately becomes resistant to endocrine therapy. A search for new therapeutic perspectives is urgently needed. Methods: By screening lipid metabolism-related gene sets and bioinformatics analysis in prostate cancer database, we identified the key lipid metabolism-related genes in PCa. Bisulfite genomic Sequence Polymerase Chain Reaction (PCR) (BSP) and Methylation-Specific Polymerase Chain Reaction (PCR) (MSP) were preformed to detect the promoter methylation of ACSS3. Gene expression was analyzed by qRT-PCR, Western blotting, IHC and co-IP. The function of ACSS3 in PCa was measured by CCK-8, Transwell assays. LC/MS, Oil Red O assays and TG and cholesterol measurement assays were to detect the levels of TG and cholesterol in cells. Resistance to Enzalutamide in C4-2 ENZR cells was examined in a xenograft tumorigenesis model in vivo. Results: We found that acyl-CoA synthetase short chain family member 3 (ACSS3) was downregulated and predicted a poor prognosis in PCa. Loss of ACSS3 expression was due to gene promoter methylation. Restoration of ACSS3 expression in PCa cells significantly reduced LD deposits, thus promoting apoptosis by increasing endoplasmic reticulum (ER) stress, and decreasing de novo intratumoral androgen synthesis, inhibiting CRPC progression and reversing Enzalutamide resistance. Mechanistic investigations demonstrated that ACSS3 reduced LD deposits by regulating the stability of the LD coat protein perilipin 3 (PLIN3). Conclusions: Our study demonstrated that ACSS3 represses prostate cancer progression through downregulating lipid droplet-associated protein PLIN3.


Assuntos
Biomarcadores Tumorais/metabolismo , Coenzima A Ligases/metabolismo , Regulação Neoplásica da Expressão Gênica , Gotículas Lipídicas/metabolismo , Perilipina-3/antagonistas & inibidores , Perilipina-3/metabolismo , Neoplasias da Próstata/patologia , Animais , Apoptose , Biomarcadores Tumorais/genética , Proliferação de Células , Coenzima A Ligases/genética , Resistencia a Medicamentos Antineoplásicos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Perilipina-3/genética , Prognóstico , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Aging (Albany NY) ; 13(2): 1842-1858, 2020 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-33201837

RESUMO

We investigated the prognostic significance of Death-Associated Protein Kinase 1 (DAPK1) and its role in sunitinib resistance in clear cell renal cell carcinoma (ccRCC). DAPK1 mRNA levels were significantly lower in tumor tissues than normal kidney tissues in TCGA-KIRC dataset (n=428). Both overall survival and disease-free survival were significantly shorter in ccRCC patients with low DAPK1 expression than those with high DAPK1 expression. Receiver operating characteristic curve analysis showed that low DAPK1 expression correlated with poor prognosis in ccRCC patients. Multivariate analysis confirmed that DAPK1 expression was an independent prognostic indicator in ccRCC. Gene set enrichment analysis showed that low DAPK1 expression correlates with upregulation of pathways related to metastasis, drug resistance, hypoxia and invasiveness in ccRCC patients. Sunitinib-resistant ccRCC cells show significantly lower DAPK1 mRNA and protein levels than sunitinib-sensitive ccRCC cells. DAPK1 overexpression enhances apoptosis in sunitinib-resistant ccRCC cells via the ATF6-dependent ER stress pathway. Xenograft tumors derived from DAPK1-overxpressing ccRCC cells were significantly smaller than the controls in nude mice. Our finding demonstrates that low DAPK1 expression is an independent prognostic indicator that correlates with ccRCC progression and sunitinib resistance.


Assuntos
Antineoplásicos/uso terapêutico , Carcinoma de Células Renais/genética , Proteínas Quinases Associadas com Morte Celular/genética , Resistencia a Medicamentos Antineoplásicos/genética , Neoplasias Renais/genética , Sunitinibe/uso terapêutico , Animais , Apoptose , Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , Proteínas Quinases Associadas com Morte Celular/metabolismo , Progressão da Doença , Estresse do Retículo Endoplasmático , Feminino , Humanos , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Invasividade Neoplásica , Metástase Neoplásica , Estadiamento de Neoplasias , Transplante de Neoplasias , Prognóstico , RNA Mensageiro/metabolismo , Hipóxia Tumoral/genética
8.
Aging (Albany NY) ; 12(17): 17459-17479, 2020 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-32921632

RESUMO

Patients with advanced renal cell carcinoma who are resistant to sunitinib currently have limited clinical options for treatment. Therefore, it is necessary to explore the biological basis of sunitinib resistance and to uncover new targets for the intervention of sunitinib resistance. In this study, we identified that LINC00160 was associated with sunitinib resistance in renal cell carcinoma. Resistant tumor cells highly expressed LINC00160 to recruit transcriptional factor TFAP2A, which bound to SAA1 promoter regions and activated its expression. On one hand, SAA1 linked to ABCB1 protein, which facilitated sunitinib cellular efflux and diminished drug accumulation. On the other hand, SAA1 stimulated JAK-STAT signaling pathways, which countered cellular survival inhibition from drug. All these regulatory networks were well organized and collaborated, thus promoting sunitinib resistance in renal cell carcinoma. LINC00160 mediates sunitinib resistance in renal cell carcinoma via SAA1 that is implicated in STAT3 activation and compound transportation, which offers an opportunity for targeted intervention and molecular therapies in the future.

9.
Crit Rev Eukaryot Gene Expr ; 30(2): 153-168, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32558494

RESUMO

Prostate cancer (PCa) is one of the most common malignancies affecting men's health worldwide. The aim of this study is to identify key genes and their regulatory networks and evaluate the usefulness of these genes on diagnosis of and prognosis for prostate cancer. The gene expression microarray dataset GSE55945 was downloaded for analysis. The differentially expressed genes (DEGs) were accessed with RStudio. Gene ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed using the database for annotation, visualization and integrated discovery (DAVID) database. A protein-protein interaction network was carried out using STRING. The survival and diagnostic analysis of hub genes were conducted using the cancer genome atlas (TCGA) data. Finally, we identified 387 DEGs. GO and KEGG analyses reveled that the DEGs in PCa were mainly enriched in the bone morphogenetic protein (BMP) signaling pathway and cytochrome P450. Among 15 hub genes, we found that only a different expression level of MYH11 affected patient survival. And further gene set enrichment analysis (GSEA) showed that low expression of MYH11 was associated with the cell cycle, DNA replication, TGF-P1 signal pathway, and PCa. In conclusion, we identified 387 DEGs that may be involved in core pathways such as the BMP pathway and cytochrome P450, which may contribute to the progression of PCa. In addition, hub gene MYH11 has the potential to be a novel biomarker for diagnosing and determining the prognosis for PCa.


Assuntos
Biomarcadores Tumorais/genética , Cadeias Pesadas de Miosina/genética , Prognóstico , Neoplasias da Próstata/genética , Intervalo Livre de Doença , Regulação Neoplásica da Expressão Gênica/genética , Ontologia Genética , Redes Reguladoras de Genes/genética , Humanos , Masculino , Neoplasias da Próstata/patologia , Mapas de Interação de Proteínas/genética , Transcriptoma
10.
J Cancer ; 11(7): 1712-1726, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32194783

RESUMO

Purpose: To investigate the potential mechanisms contributing to metastasis of clear cell renal cell carcinoma (ccRCC), screen the hub genes, associated pathways of metastatic ccRCC and identify potential biomarkers. Methods: The ccRCC metastasis gene expression profile GSE47352 was employed to analyze the differentially expressed genes (DEGs). DAVID was performed to assess Gene ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses. The protein-protein interaction (PPI) network and modules were constructed. The function pathway, prognostic and diagnostic analysis of these hub genes was picked out to estimate their potential effects on metastasis of ccRCC. Results: A total of 873 DEGs were identified (503 upregulated genes and 370 downregulated genes). Meanwhile, top 20 hub genes were displayed. GO analysis showed that the top 20 hub genes were enriched in regulation of phosphatidylinositol 3-kinase signaling, positive regulation of DNA replication, protein autophosphorylation, protein tyrosine kinase activity, etc. KEGG analysis indicated these hub genes were enriched in the Ras signaling pathway, PI3K-Akt signaling pathway, HIF-1 signaling pathway, Pathways in cancer, etc. The GO and KEGG enrichment analyses for the hub genes disclosed important biological features of metastatic ccRCC. PPI network showed the interaction of top 20 hub genes. Gene Set Enrichment Analysis (GSEA) revealed that some of the hub genes was associated with metastasis, epithelial mesenchymal transition (EMT), hypoxia cancer and adipogenesis of ccRCC. Some top hub genes were distinctive and new discoveries compared with that of the existing associated researches. Conclusions: Our analysis uncovered that changes in signal pathways such as Ras signaling pathway, PI3K-Akt signaling pathway, etc. may be the main signatures of metastatic ccRCC. We identified several candidate biomarkers related with overall survival (OS) and disease-free survival (DFS) of ccRCC patients. Accordingly, they might be novel therapeutic targets and used as potential biomarkers for diagnosis, prognosis of ccRCC.

11.
Mol Ther ; 28(5): 1299-1313, 2020 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-32209435

RESUMO

Recently, cytoophidium, a nonmembrane-bound intracellular polymeric structure, has been shown to exist in various organisms, including tumor tissues, but its function and mechanism have not yet been examined. Examination of cytoophidia-assembled gene inosine monophosphate dehydrogenase (IMPDH) and cytidine triphosphate synthetase (CTPS) mRNA levels showed that only IMPDH1 levels were significantly higher in the clear cell renal cell carcinoma (ccRCC). IMPDH1 was positively correlated with the metastasis-related gene Y-box binding protein 1 (YB-1) and served as an independent prognostic factor in ccRCC. Kaplan-Meier analysis indicated that patients with tumors that expressed high IMPDH1 levels had a shorter overall survival (OS) and disease-free survival (DFS). Furthermore, detection of cytoophidia by immunofluorescence staining in ccRCC tissues showed that IMPDH1-assembled cytoophidia are positively associated with tumor metastasis. Mechanistically, IMPDH1 and YB-1 formed an autoregulatory positive feedback loop: IMPDH1 maintained YB-1 protein stabilization; YB-1 induced IMPDH1 expression by binding to the IMPDH1 promoter motif. Functionally, IMPDH1-assembled cytoophidia physically interacted with YB-1 and translocated YB-1 into the cell nucleus, thus correlating with ccRCC metastasis. Our findings provide the first solid theoretical rationale for targeting the IMPDH1/YB-1 axis to improve metastatic renal cancer treatment.


Assuntos
Carcinoma de Células Renais/metabolismo , Retroalimentação Fisiológica , IMP Desidrogenase/metabolismo , Neoplasias Renais/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/secundário , Proteína 1 de Ligação a Y-Box/metabolismo , Animais , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Intervalo Livre de Doença , Feminino , Humanos , IMP Desidrogenase/genética , Estimativa de Kaplan-Meier , Neoplasias Renais/patologia , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Plasmídeos/genética , RNA Mensageiro/metabolismo , Transfecção , Carga Tumoral/genética , Ensaios Antitumorais Modelo de Xenoenxerto , Proteína 1 de Ligação a Y-Box/genética
12.
Aging (Albany NY) ; 12(2): 1808-1827, 2020 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-32003757

RESUMO

Clear cell renal cell carcinoma (ccRCC) is one of the most common malignancies and lacks reliable biomarkers for diagnosis and prognosis, which results in high incidence and mortality rates of ccRCC. In this study, ISG20, HJURP, and FOXM1 were identified as hub genes via weighted gene co-expression network analysis (WGCNA) and Cox regression analysis. Samples validation showed that only ISG20 was up-regulated in ccRCC. Therefore, ISG20 was selected for further study. High ISG20 expression was associated with poor overall survival and disease-free survival. Furthermore, the expression of ISG20 could effectively differentiate ccRCC from normal tissues and was positively correlated to clinical stages. Functional experiments proved that knockdown of ISG20 expression could obviously inhibit cell growth, migration, and invasion in ccRCC cells. To find the potential mechanisms of ISG20, gene set enrichment analysis (GSEA) was performed and revealed that high expression of ISG20 was significantly involved in metastasis and cell cycle pathways. In addition, we found that ISG20 could regulate the expression of MMP9 and CCND1. In conclusion, these findings suggested that ISG20 promoted cell proliferation and metastasis via regulating MMP9/CCND1 expression and might serve as a potential biomarker and therapeutic target in ccRCC.


Assuntos
Biomarcadores Tumorais , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/patologia , Exorribonucleases/genética , Neoplasias Renais/genética , Neoplasias Renais/patologia , Oncogenes , Biologia Computacional , Progressão da Doença , Proteína Semelhante a ELAV 2/genética , Proteína Semelhante a ELAV 2/metabolismo , Exorribonucleases/metabolismo , Perfilação da Expressão Gênica , Humanos , Modelos Biológicos , Gradação de Tumores , Estadiamento de Neoplasias , Prognóstico , Modelos de Riscos Proporcionais , Transcriptoma
13.
J Cell Physiol ; 235(2): 1235-1246, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31267540

RESUMO

Prostate cancer (PCa) is the second leading cause of death among American men. Increasing evidence has shown that long noncoding RNAs (lncRNAs) play important roles in tumorigenesis of PCa. In this study, we explored the biological functions of small nucleolar RNA host gene 12 (SNHG12) and investigated the interaction between miR-133b and SNHG12 in the progression of PCa. Data was downloaded from The Cancer Genome Atlas and Human Cancer Metastasis Database, and clinicopathological characteristics were analyzed with relapse-free survival rate. We detected SNHG12 expression level in PCa cells and tissues, and then analyzed its clinical significance, which revealed that SNHG12 has the potent to predict prognosis of PCa. Bioinformatic analysis revealed that SNHG12 was closely related to the progression of PCa and could target candidate microRNA (miR-133b). After transfecting SNHG12 silencing plasmid and miR-133b mimic/sponge, biological function assays were conducted and results illustrated that SNHG12 associated with miR-133b exerted biological effects on cancer cell growth, migration, and invasion. Direct interactions between miR-133b and SNHG12 have been found and SNHG12 acts as an oncogene to promote tumorigenesis of PCa by sponging tumor suppressor gene miR-133b.


Assuntos
Carcinogênese , MicroRNAs/metabolismo , Neoplasias da Próstata/metabolismo , RNA Longo não Codificante/metabolismo , Biomarcadores Tumorais , Linhagem Celular Tumoral , Movimento Celular , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Invasividade Neoplásica , Prognóstico , Neoplasias da Próstata/genética , RNA Longo não Codificante/genética , Regulação para Cima
14.
Clin Cancer Res ; 26(6): 1516-1528, 2020 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-31796514

RESUMO

PURPOSE: Emerging evidence indicates that castration-resistant prostate cancer (CRPC) is often driven by constitutively active androgen receptor (AR) or its V7 splice variant (AR-V7) and commonly becomes resistant to endocrine therapy. The aim of this work is to evaluate the function of a kinesin protein, KIF4A, in regulating AR/AR-V7 in prostate cancer endocrine therapy resistance. EXPERIMENTAL DESIGN: We examined KIF4A expression in clinical prostate cancer specimens by IHC. Regulated pathways were investigated by qRT-PCR, immunoblot analysis, immunoprecipitation, and luciferase reporter and chromatin immunoprecipitation (ChIP) assays. A series of functional analyses were conducted in cell lines and xenograft models. RESULTS: Examination of the KIF4A protein and mRNA levels in patients with prostate cancer showed that increased expression of KIF4A was positively correlated with androgen receptor (AR) levels. Patients with lower tumor KIF4A expression had improved overall survival and disease-free survival. Mechanistically, KIF4A and AR form an auto-regulatory positive feedback loop in prostate cancer: KIF4A binds AR and AR-V7 and prevents CHIP-mediated AR and AR-V7 degradation; AR binds the promoter region of KIF4A and activates its transcription. KIF4A promotes castration-sensitive and castration-resistant prostate cancer cell growth through AR- and AR-V7-dependent signaling. Furthermore, KIF4A expression is upregulated in enzalutamide-resistant prostate cancer cells, and KIF4A knockdown effectively reverses enzalutamide resistance and enhances the sensitivity of CRPC cells to endocrine therapy. CONCLUSIONS: These findings indicate that KIF4A plays an important role in the progression of CRPC and serves as a crucial determinant of the resistance of CRPC to endocrine therapy.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Regulação Neoplásica da Expressão Gênica , Cinesinas/antagonistas & inibidores , Feniltioidantoína/análogos & derivados , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Receptores Androgênicos/química , Idoso , Idoso de 80 Anos ou mais , Antagonistas de Receptores de Andrógenos/farmacologia , Animais , Benzamidas , Linhagem Celular Tumoral , Proliferação de Células , Bases de Dados Genéticas/estatística & dados numéricos , Humanos , Cinesinas/metabolismo , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Nitrilas , Feniltioidantoína/farmacologia , Neoplasias de Próstata Resistentes à Castração/genética , Neoplasias de Próstata Resistentes à Castração/metabolismo , Neoplasias de Próstata Resistentes à Castração/patologia , Receptores Androgênicos/metabolismo , Taxa de Sobrevida , Ensaios Antitumorais Modelo de Xenoenxerto
15.
J Cancer ; 10(25): 6384-6394, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31772671

RESUMO

Renal cell carcinoma (RCC) is one of the most common malignancies in the urinary system, among which the proportion of clear cell RCC (ccRCC) is over 80%. This study aims to explore potential signaling pathways and key biomarkers that drive RCC progression. The RCC GEO Datasets GSE15641 was featured to screen differentially expressed genes (DEGs). The pathway enrichment and functional annotation of differentially expressed genes were analyzed using the Kyoto Encyclopedia of Genes and Genomes (KEGG) and the Gene Ontology (GO). We screened Hub genes from DEGs using protein-protein interaction (PPI) networks and Cytoscape software. The survival and diagnostic analysis of these hub genes was performed to evaluate their potential prognostic and diagnostic value for ccRCC. In GSE15641 dataset, 598 DEGs were captured according to screening criteria (406 up-regulated genes and 192 down-regulated genes). Meanwhile, 15 hub genes were screened out from DEGs using PPI and Cytoscape. Kaplan Meier and ROC curve analysis identified three potential prognostic and diagnostic biomarkers (TGFB1, TIMP1 and VIM) for ccRCC from 15 hub genes. Gene set enrichment analysis (GSEA) revealed that these three dysregulated genes are mainly enriched in primary immunodeficiency, ECM receptor interaction, cytokine receptor interaction and natural killer cell-mediated cytotoxicity pathway. In summary, our findings discovered pivotal gene expression signatures and signaling pathways in the progression of ccRCC. TGFB1, TIMP1 and VIM might contribute to the progression of ccRCC, which could have potential as biomarkers or therapeutic targets for ccRCC.

16.
J Pineal Res ; 67(4): e12607, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31469445

RESUMO

Metabolic adaptations are emerging hallmarks of cancer progression and cellular transformation. Clear cell renal cell carcinoma (ccRCC) is a metabolic disease defined histologically by lipid accumulation and lipid storage, which promote tumor cell survival; however, the significance of eliminating the lipid remains unclear. Here, we demonstrate that melatonin activates transcriptional coactivator peroxisome proliferator-activated receptor gamma coactivator 1A (PGC1A) and uncoupling protein 1 (UCP1)-dependent lipid autophagy and a lipid browning program to elicit a catabolic state called "tumor slimming," thus suppressing tumor progression. Metabolic coregulator data analysis revealed that PGC1A expression was decreased in ccRCC tissues versus normal tissues, and poor patient outcome was associated with lower expression of PGC1A in The Cancer Genome Atlas (TCGA-KIRC). PGC1A was downregulated in ccRCC and associated with disease progression. Restoration of PGC1A expression by melatonin in ccRCC cells significantly repressed tumor progression and eliminated the abnormal lipid deposits. Furthermore, a phenomenon called "tumor slimming" was observed, in which tumor cell volume was reduced and lipid droplets transformed into tiny pieces. Additional studies indicated that melatonin promoted "tumor slimming" and suppressed ccRCC progression through PGC1A/UCP1-mediated autophagy and lipid browning. During this process, autophagy and lipid browning eliminate lipid deposits without providing energy. These studies demonstrate that the novel "tumor slimming" pathway mediated by melatonin/PGC1A/UCP1 exhibits prognostic potential in ccRCC, thus revealing the significance of monitoring and manipulating this pathway for cancer therapy.


Assuntos
Morte Celular Autofágica/efeitos dos fármacos , Metabolismo dos Lipídeos/efeitos dos fármacos , Melatonina/farmacologia , Proteínas de Neoplasias/metabolismo , Neoplasias , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Proteína Desacopladora 1/metabolismo , Animais , Linhagem Celular Tumoral , Humanos , Gotículas Lipídicas/metabolismo , Gotículas Lipídicas/patologia , Camundongos , Proteínas de Neoplasias/genética , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Proteína Desacopladora 1/genética , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Int J Oncol ; 55(3): 645-656, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31364727

RESUMO

As one of the most commonly reported malignancies of the urinary system, clear cell renal cell carcinoma (ccRCC) is an advanced metastatic tumor with high mortality rates. The Rac family small GTPase 2 (RAC2) is a member of the Rho GTPases. Although Rho GTPases play an important role in numerous different types of tumor, whether they have functions in ccRCC remains uncertain. The present study utilized bioinformatics analyses in order to compare the expression levels of RAC2 in ccRCC tumors vs. adjacent tissues, and assessed the association between RAC2 expression and clinicopathological parameters. Furthermore, reverse transcription­quantitative PCR, western blotting and immunohistochemistry assays were performed to validate RAC2 expression levels in human ccRCC tissues and cell lines. Functional experiments were also conducted in order to identify the roles of RAC2 in vitro. The results revealed that RAC2 was upregulated in ccRCC tissues and cell lines. In addition, elevated expression levels of RAC2 were significantly associated with a poor overall survival (P=0.0061), higher Tumor­Node­Metastasis stage and worse G grade. Receiver operating characteristic analysis indicated that high expression levels of RAC2 could be a diagnostic index for ccRCC (area under the curve, 0.9095; P<0.0001). Furthermore, knockdown of RAC2 in vitro attenuated the proliferation, migration and invasion of renal carcinoma cells. In conclusion, the results of the present study demonstrated that RAC2 may act as a promising prognostic and diagnostic biomarker of ccRCC, and could be considered as a potential therapeutic target for treating ccRCC.


Assuntos
Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Carcinoma de Células Renais/patologia , Neoplasias Renais/patologia , Proteínas rac de Ligação ao GTP/genética , Proteínas rac de Ligação ao GTP/metabolismo , Adulto , Idoso , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Progressão da Doença , Detecção Precoce de Câncer , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Renais/genética , Neoplasias Renais/metabolismo , Masculino , Pessoa de Meia-Idade , Gradação de Tumores , Estadiamento de Neoplasias , Prognóstico , Regulação para Cima , Adulto Jovem , Proteína RAC2 de Ligação ao GTP
18.
Biomed Pharmacother ; 118: 109264, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31390578

RESUMO

BACKGROUND: Clear cell renal cell carcinoma (ccRCC) is one of the most common malignancies in urinary system. However, there are still no reliable biomarkers for the diagnosis and prognosis of ccRCC. In this study, we aimed to screen candidate biomarkers and potential therapeutic targets for ccRCC. METHODS: Differentially expressed genes (DEGs) were screened using NetworkAnalyst. Protein-protein interaction (PPI) network and weighted gene co-expression network analysis (WGCNA) were utilized to identify hub genes. Then, we assessed the prognostic and diagnostic values of hub genes to screen candidate biomarkers. Gene Set Enrichment Analysis (GSEA) was applied to reveal potential mechanisms of candidate biomarkers in ccRCC. Oncomine database and The Human Protein Atlas were used to verify the expression of candidate biomarkers online. In addition, qRT-PCR, Enzyme linked immunosorbent assay (ELISA) and Immunohistochemistry (IHC) assays were performed to validate the expression level of candidate biomarkers in ccRCC cells and tissues. RESULTS: A total of 771 genes were identified as DEGs. GO function analysis showed that DEGs were mostly enriched in excretion, apical part of cell and monovalent inorganic cation transmembrane transporter activity. KEGG pathway analysis demonstrated that DEGs were mostly involved in Neuroactive ligand-receptor interaction. After utilizing PPI network and WGCNA, nine genes (IFNG, CXCR3, PMCH, CD2, FASLG, CXCL13, CD8A, CD3D and GZMA) were identified as the hub genes. Moreover, survival analysis exhibited that high expression of CXCL13 predicted poor survival in both overall survival (OS) and disease free survival (DFS). The ROC curves indicated that CXCL13 could distinguish ccRCC samples from normal kidney samples. High expression of CXCL13 group was mostly associated with RB and MEL18 pathways by GSEA. Furthermore, qRT-PCR, ELISA and IHC results showed that the expression of CXCL13 was elevated in ccRCC. CONCLUSIONS: Our study illustrated that CXCL13 had good diagnostic and prognostic value, which may become a candidate biomarker and therapeutic target for ccRCC.


Assuntos
Biomarcadores Tumorais/metabolismo , Carcinoma de Células Renais/metabolismo , Quimiocina CXCL13/metabolismo , Biologia Computacional , Neoplasias Renais/metabolismo , Carcinoma de Células Renais/genética , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Ontologia Genética , Redes Reguladoras de Genes , Humanos , Estimativa de Kaplan-Meier , Neoplasias Renais/genética , Gradação de Tumores , Estadiamento de Neoplasias , Mapas de Interação de Proteínas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Curva ROC , Reprodutibilidade dos Testes
19.
Adv Sci (Weinh) ; 6(10): 1801862, 2019 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-31131187

RESUMO

Emerging evidence has highlighted the important role of abnormal lipid accumulation in cancer development and progression, but the mechanism for this phenomenon remains unclear. Here, it is demonstrated that phospholipase C-like 1/uncoupling protein 1 (PLCL1)/(UCP1)-mediated lipid browning promotes tumor cell "slimming" and represses tumor progression. By screening three independent lipid metabolism-related gene sets in clear cell renal cell carcinoma (ccRCC) and analyzing the TCGA database, it is found that PLCL1 predicted a poor prognosis and was downregulated in ccRCC. Restoration of PLCL1 expression in ccRCC cells significantly represses tumor progression and reduces abnormal lipid accumulation. Additionally, a phenomenon called tumor cell "slimming," in which tumor cell volume is reduced and lipid droplets are transformed into tiny pieces, is observed. Further studies show that PLCL1 promotes tumor cell "slimming" and represses tumor progression through UCP1-mediated lipid browning, which consumes lipids without producing ATP energy. Mechanistic investigations demonstrate that PLCL1 improves the protein stability of UCP1 by influencing the level of protein ubiquitination. Collectively, the data indicate that lipid browning mediated by PLCL1/UCP1 promotes tumor cell "slimming" and consumes abnormal lipid accumulation, which represses the progression of ccRCC. Tumor cell "slimming" offers a promising new concept and treatment modality against tumor development and progression.

20.
J Cancer ; 10(6): 1398-1408, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31031850

RESUMO

Purpose: The present study aims to explore the potential mechanisms contributing to prostate cancer (PCa), screen the hub genes, and identify potential biomarkers and correlated pathways of PCa progression. Methods: The PCa gene expression profile GSE3325 was operated to analyze the differentially expressed genes (DEGs). DAVID was used to evaluate Gene ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses. A protein-protein interaction (PPI) network was constructed to visualize interactions of the hub genes. The prognostic and diagnostic analysis of these hub genes was carried out to evaluate their potential effects on PCa. Results: A total of 847 DEGs were identified (427 upregulated genes and 420 downregulated genes). Meanwhile, top15 hub genes were showed. GO analysis displayed that the DEGs were mainly enriched in cell cycle, DNA damage response, signal transduction by p53 class mediator resulting in cell cycle arrest and proteinaceous extracellular matrix. KEGG analysis indicated the DEGs were enriched in the p53 signaling pathway and cell cycle pathway. The GO and KEGG enrichment analyses for the DEGs disclosed important biological features of PCa. PPI network showed the interaction of top 15 hub genes. Gene Set Enrichment Analysis (GSEA) revealed that some of the hub genes were associated with biochemical recurrence (BCR) and metastasis of PCa. Some top hub genes were distinctive and new discoveries compared with that of the existing associated researches. Conclusions: Our analysis revealed that the changes of cell cycle and p53 signaling pathway are two major signatures of PCa. CENPA, KIF20A and CDCA8 might promote the tumorigenesis and progression of PCa, especially in BCR and metastasis, which could be novel therapeutic targets and biomarkers for diagnosis, prognosis of PCa.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...