Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Prostate ; 83(15): 1446-1457, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37545197

RESUMEN

BACKGROUND: Androgen deprivation therapy (ADT) is one of the main treatment modalities for prostate cancer (PCa); however, almost all patients treated with ADT eventually progress into castration-resistant PCa (CRPC). Although second-generation androgen receptor (AR) antagonists, such as enzalutamide, have been approved for CRPC treatment, AR signaling in CRPC cells is reactivated through multiple mechanisms, resulting in resistance to treatment and tumor progression with a very poor prognosis. The present study aimed to explore the anticancer effect of a treatment combining AR antagonist enzalutamide with AR degrader IU1 on PCa cells. METHODS: The joint effects of enzalutamide and IU1 on PCa cell proliferation and apoptosis and associated cell signaling were evaluated in vitro. Mechanistically, the ubiquitination level and half-life of AR were examined under the combination treatment. The binding of IU1 and enzalutamide to AR was further verified using cellular thermal shift analysis and isothermal dose-response curve fingerprinting. RESULTS: The combination of IU1 and three AR antagonists showed synergistic effects in different prostate cell lines. IU1 and enzalutamide synergistically promoted the degradation of AR and AR-V7 proteins, as well as suppressed the expression levels of AR and AR-V7 downstream target genes at the transcriptional and protein levels. The combination also synergistically blocked the PCa cell cycle and promoted apoptosis in PCa cell lines. Mechanistically, the combination promoted increased levels of AR ubiquitination. In CRPC cell lines and in the presence of increased androgen concentrations, enzalutamide was still able to bind AR competitively with androgens, reducing the stability of AR and thus promoting the degradation effect of IU1 on AR, synergistically producing an inhibitory effect on PCa cells. CONCLUSION: Taken together, our findings suggest that the combination of AR degrader and enzalutamide potentially represents a new therapeutic strategy for CRPC.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración , Masculino , Humanos , Neoplasias de la Próstata Resistentes a la Castración/patología , Andrógenos/metabolismo , Antagonistas de Andrógenos/uso terapéutico , Receptores Androgénicos/metabolismo , Benzamidas/uso terapéutico , Nitrilos/uso terapéutico , Antagonistas de Receptores Androgénicos/farmacología , Línea Celular Tumoral , Resistencia a Antineoplásicos
2.
Mol Med ; 29(1): 7, 2023 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-36647005

RESUMEN

BACKGROUND: Bladder cancer (BCa) is the most common malignant tumor of the urinary system, with transitional cell carcinoma (TCC) being the predominant type. EP300 encodes a lysine acetyltransferase that regulates a large subset of genes by acetylating histones and non-histone proteins. We previously identified several bladder cancer-associated mutations in EP300 using high-throughput sequencing; however, the functional consequences of these mutations remain unclear. METHODS: Bladder cancer cells T24 and TCC-SUP were infected with shEP300 lentiviruses to generate stable EP300 knockdown cell lines. The expression levels of EP300, p16 and p21 were detected by real-time PCR and western blots. The transcriptional activity of p16 and p21 were detected by dual luciferase assay. Cell proliferation assay, flow cytometric analyses of cell cycle, invasion assay and xenograft tumor model were used to measure the effect of EP300-R1627W mutation in bladder cancer. Immunoprecipitation was used to explore the relationship between EP300-R1627W mutation and p53. Structural analysis was used to detect the structure of EP300-R1627W protein compared to EP300-wt protein. RESULTS: we screened the mutations of EP300 and found that the EP300-R1627W mutation significantly impairs EP300 transactivation activity. Notably, we demonstrated that the R1627W mutation impairs EP300 acetyltransferase activity, potentially by interfering with substrate binding. Finally, we show that EP300-R1627W is more aggressive in growth and invasion in vitro and in vivo compared to cells expressing EP300-wt. We also found that the EP300-R1627W mutation occurs frequently in seven different types of cancers. CONCLUSION: In summary, our work defines a driver role of EP300-R1627W in bladder cancer development and progression.


Asunto(s)
Neoplasias de la Vejiga Urinaria , Humanos , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/metabolismo , Mutación , Histonas , Ciclo Celular , Proteína p300 Asociada a E1A/genética , Proteína p300 Asociada a E1A/metabolismo
3.
Biochem Biophys Res Commun ; 655: 35-43, 2023 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-36924677

RESUMEN

Histone methyltransferase KMT2D plays a critical role as a human oncogene in prostate cancer (PCa). Dysregulated inflammatory responses and cytokine signaling are implicated in cancer progression. Furthermore, interleukin 6 (IL-6) is a pleiotropic cytokine that contributes to PCa progression; however, the association between KMT2D and IL-6 in PCa remains unclear. PCa cell proliferative potential, migratory potential, and apoptosis in vitro were determined using cell counting kit-8 (CCK-8), EdU incorporation, wound healing, and apoptosis assays. Proliferation and migratory potential were impaired and apoptosis was induced in PCa cells cultured with the conditioned medium from KMT2D-depleted cells. Cytokine array analysis showed that IL-6 was the most affected cytokine in the conditioned media. KMT2D knockdown significantly downregulated the expression of IL-6 in PCa cells. What's more, proliferation and migration were also impaired and apoptosis was also induced by silencing IL-6R expression. Immunohistochemistry (IHC) and quantitative reverse transcription-polymerase chain reaction (qRT-PCR) were performed to validate the positive correlation between KMT2D and IL-6 in PCa tissue samples. Chromatin immunoprecipitation (ChIP)-PCR demonstrated that KMT2D and H3K4me1 occupied IL-6 enhancer regions and therefore, directly regulated IL-6 expression. The present study revealed that the KMT2D knockdown suppressed prostate cancer progression through the downregulation of paracrine IL-6 signaling. These results suggest that KMT2D could be regarded as a potential new target for PCa therapy.


Asunto(s)
Interleucina-6 , Neoplasias de la Próstata , Humanos , Masculino , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Histona Metiltransferasas/metabolismo , Interleucina-6/metabolismo , Próstata/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo
4.
Pharmazie ; 77(2): 59-66, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35209965

RESUMEN

Background: Patients with prostate cancer often develop resistance to androgen deprivation therapy, a condition called castration-resistant prostate cancer (CRPC). Enzalutamide (MDV3100) can prolong the survival of patients with CRPC after chemotherapy, but ∼50% of patients eventually relapse and develop resistance to MDV3100. Thus, it is necessary to explore new treatment methods to improve the therapeutic effect of MDV3100. Tyrosine kinases play an essential role in the pathogenesis of CRPC. Methods: MTT assay was used to detect the inhibitory effects of MDV3100 and tyrosine kinase inhibitor on prostate cancer cells. CompuSyn version 1.0 was used to calculate the combination index (CI) values using the Chou-Talalay method. Clone formation and EdU assay were used to detect the effect of afatinib combined with MDV3100 on the proliferation of 22Rv1 cells. RT-qPCR and Western blot were used to explore the mechanism of drug combination. The aim of the present study was to determine the effects of several tyrosine kinase inhibitors (TKIs) when used in combination with MDV3100 in vitro. Results: The results demonstrated that TKIs combined with MDV3100 exerted a synergistic effect on a variety of PCa cells. Afatinib combined with MDV3100 could suppress the proliferation and migration of 22RV1 cells, as well as cause their cell cycle arrest and apoptosis. Mechanistically, afatinib effectively reduced the protein expression levels of HER2 and HER3 and inhibited EGFR phosphorylation, thereby enhancing the effect of MDV3100 and suppressing CRPC. Conclusions: These findings suggested that afatinib treatment improved the effect of MDV3100 on 22RV1 cells, highlighting this drug as a potential therapeutic strategy for patients with CRPC.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración , Afatinib/farmacología , Afatinib/uso terapéutico , Antagonistas de Andrógenos , Benzamidas , Línea Celular Tumoral , Proliferación Celular , Resistencia a Antineoplásicos , Humanos , Masculino , Recurrencia Local de Neoplasia , Nitrilos/farmacología , Feniltiohidantoína , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/patología , Receptores Androgénicos/metabolismo , Receptores Androgénicos/uso terapéutico
5.
Prostate ; 81(12): 893-901, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34184786

RESUMEN

The androgen receptor (AR) plays an important role in the progression of prostate cancer and is the most important therapeutic target. However, androgen deprivation therapy will finally lead patients to progress to castration-resistant prostate cancer (CRPC). Here, we confirmed that GAS5, a long noncoding RNA, could interact and suppress AR transactivation in CRPC C4-2 cells. Knockdown GAS5 by short hairpin RNA would enhance the transcription of AR via promote AR recruitment to the promoter of its downstream target genes. Functionally, GAS5 overexpression inhibits cell proliferation partially through inhibiting AR transactivation in C4-2 cells. Moreover, knocking down GAS5 protects C4-2 cells from the docetaxel-induced cell apoptosis. In return, the suppressed AR was found to downregulate the GAS5 expression, which forms a feedback loop resulted in AR high transcription activity in CRPC. Collectively, our findings revealed the important role of GAS5 in AR axis activity regulation and CRPC progression. Targeting GAS5 to intervene the feedback loop might be a new potential therapeutic approach for the patients at CRPC stage.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/fisiología , Neoplasias de la Próstata/metabolismo , ARN Largo no Codificante/metabolismo , Receptores Androgénicos/metabolismo , Línea Celular Tumoral , Células HEK293 , Humanos , Masculino , Neoplasias de la Próstata/genética , Unión Proteica/fisiología , ARN Largo no Codificante/genética , Receptores Androgénicos/genética
6.
BMC Cancer ; 19(1): 204, 2019 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-30845932

RESUMEN

BACKGROUND: Heterogeneity in bladder cancer results in variable clinical outcomes, posing challenges for clinical management of this malignancy. Recent studies suggest both tumor suppressive and oncogenic role of PPARγ in bladder cancer. The fuction of PPARγ signaling pathway in modulating carcinogenesis is controversial. METHODS: The expression of PPARγ and association with overall survival were analyzed in patients from two cohorts. The effect of PPARγ activation on cell proliferation, cell cycle, and cell apoptosis were determined with the agonists (rosiglitazone and pioglitazone), the inverse agonist (T0070907), and the antagonist (GW9662) in Umuc-3 and 5637 bladder cancer cells. The correlation of PPARγ activation with PI3K-Akt pathway was evaluated with RNA sequencing data from the TCGA cases and 30 human bladder cancer cell lines. The effect of PPARγ activation on tumor growth was validated with subcutaneous tumor models in vivo. The effect of PPARγ activation on PI3K-Akt signaling transduction was determined with multiple assays including immunohistochemistry, flow cytometry, proteomic array, and western blotting. RESULTS: We showed that PPARγ was a favorable prognostic factor in patients with bladder cancer. PPARγ activation by rosiglitazone and pioglitazone markedly induced cell cycle G2 arrest and apoptosis in bladder cancer cells, which resulted in inhibition of cell proliferation in vitro and suppression of tumor growth in vivo. The underlying mechanism involved marked inhibition of PI3K-Akt pathway. CONCLUSIONS: This study reported the tumor-suppressive effect of PPARγ agonists in bladder cancer, suggesting that transactivation of PPARγ could be served as a potential strategy for the chemoprevention and therapeutic treatment of bladder cancer.


Asunto(s)
PPAR gamma/agonistas , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Neoplasias de la Vejiga Urinaria/metabolismo , Animales , Apoptosis/efectos de los fármacos , Biomarcadores , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Humanos , Inmunohistoquímica , Masculino , Ratones , Modelos Biológicos , Pronóstico , Rosiglitazona/farmacología , Transcriptoma , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/mortalidad , Ensayos Antitumor por Modelo de Xenoinjerto
7.
J Sci Food Agric ; 96(13): 4492-8, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26858163

RESUMEN

BACKGROUND: Biluochun is a typical non-fermented tea and is also famous for its unique aroma in China. Few studies have been performed to evaluate the effect of the manufacturing process on the formation and content of its aroma. RESULTS: The volatile components were extracted at different manufacturing process steps of Biluochun green tea using fully automated headspace solid-phase microextraction (HS-SPME) and further characterised by gas chromatography-mass spectrometry (GC-MS). Among 67 volatile components collected, the fractions of linalool oxides, ß-ionone, phenylacetaldehyde, aldehydes, ketones, and nitrogen compounds were increased while alcohols and hydrocarbons declined during the manufacturing process. The aroma compounds decreased the most during the drying steps. CONCLUSION: We identified a number of significantly changed components that can be used as markers and quality control during the producing process of Biluochun. The drying step played a major role in the aroma formation of green tea products and should be the most important step for quality control. © 2016 Society of Chemical Industry.


Asunto(s)
Camellia sinensis/química , Manipulación de Alimentos , Calidad de los Alimentos , Hojas de la Planta/química , Brotes de la Planta/química , Té/química , Compuestos Orgánicos Volátiles/análisis , Métodos Analíticos de la Preparación de la Muestra , Automatización de Laboratorios , Biomarcadores/análisis , Camellia sinensis/crecimiento & desarrollo , Camellia sinensis/metabolismo , China , Inspección de Alimentos/métodos , Liofilización , Cromatografía de Gases y Espectrometría de Masas , Calor , Odorantes , Hojas de la Planta/crecimiento & desarrollo , Hojas de la Planta/metabolismo , Brotes de la Planta/crecimiento & desarrollo , Brotes de la Planta/metabolismo , Control de Calidad , Microextracción en Fase Sólida , Terpenos/análisis , Terpenos/química , Terpenos/metabolismo , Compuestos Orgánicos Volátiles/química , Compuestos Orgánicos Volátiles/metabolismo , Volatilización
8.
Comput Biol Chem ; 110: 108082, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38663187

RESUMEN

PURPOSE: Clear cell renal cell carcinoma (ccRCC) frequently progresses to advanced stages due to tumor thrombus (TTs) formation. In this study, we aimed to investigate the role of coagulation-related pathway activation in the progression of ccRCC. METHODS: Consensus clustering was used to identify coagulation-related molecular clusters of ccRCC patients from The Cancer Genome Atlas Program (TCGA) database. The function of coagulation and its correlation with the immune microenvironment were investigated. Protein-protein interactions and differential expression analysis were used to identify the key gene, which was verified by external experiments. The coagulation-associated risk score was constructed by cox proportional hazards regression. RESULTS: Notable disparities were detected in immune characteristics, prognostic differentiation and drug sensitivity between two coagulation-related clusters. Through the integration of clinical stage significance and protein-protein interactions, the key gene MMP9 was screened and it was significantly correlated with CD4+T cells, CD8+T cells and Treg cells. A coagulation-related risk score prognostic model was developed in the Cancer Genome Atlas (TCGA) cohort for risk stratification and prognosis prediction. The prognostic predictive values of the coagulation-related risk score were further authenticated in both TCGA-KIRC and E-MTAB-1980 cohorts. CONCLUSION: There is an obvious correlation between the coagulation and the tumor microenvironment in ccRCC. As a key coagulation-related gene, MMP9 may promote the progression of renal cell carcinoma by influencing immune infiltration of CD8+T cells and Treg cells. Additionally, the risk score could be used as a durable prognostic biomarker, which could assist in clinical decision making for ccRCC patients.


Asunto(s)
Biomarcadores de Tumor , Carcinoma de Células Renales , Neoplasias Renales , Microambiente Tumoral , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Humanos , Microambiente Tumoral/genética , Neoplasias Renales/genética , Neoplasias Renales/patología , Biomarcadores de Tumor/genética , Pronóstico , Coagulación Sanguínea/genética , Metaloproteinasa 9 de la Matriz/genética
9.
Micromachines (Basel) ; 14(7)2023 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-37512793

RESUMEN

This manuscript presents an ultra-low-power analog multiplier-divider compatible with digital code words, which is applicable to the integrated structure of resistive random-access memory (RRAM)-based computing-in-memory (CIM) macros. Current multiplication and division are accomplished by a current-mirror-based structure. Compared with digital dividers to achieve higher precision and operation speed, analog dividers present the advantages of a reduced power consumption and a simple circuit structure in lower precision operations, thus improving the energy efficiency. Designed and fabricated in a 55 nm CMOS process, the proposed work is capable of achieving 8-bit precision for analog current multiplication and division operations. Measurement results show that the signal delay is 1 µs when performing 8-bit operation, with a bandwidth of 1.4 MHz. The power consumption is less than 6.15 µW with a 1.2 V supply voltage. The proposed multiplier-divider can increase the operation capacity by dividing the input current and digital code while reducing the power consumption and complexity required by division, which can be further utilized in real-time operation of edge computing devices.

10.
Int Urol Nephrol ; 55(3): 579-588, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36472798

RESUMEN

PURPOSE: We aim to compare the long-term oncologic outcomes, including overall survival (OS), cancer-specific survival (CSS), and bladder cancer recurrence (BCR) among patients with ureter carcinoma who received nephroureterectomy (RNU) or partial ureterectomy (PU). METHODS: We performed a retrospective cohort study using the Surveillance, Epidemiology, and End Results database between 2004 and 2015 of patients with ureter carcinoma who underwent RNU or PU. Propensity score matching (PSM) was applied to balance the baseline data. The Kaplan-Meier method with subgroup analysis was conducted to verify the effect of the two surgery types. Fine-Gray competing risk regression estimated the cumulative incidence of BCR. RESULTS: A total of 2509 patients were involved; 665 (26.5%) patients underwent PU, and 1844 (73.5%) patients underwent RNU. Patients who underwent PU experienced a similar OS and CSS compared with those who underwent RNU in both PSM cohorts (HR [hazard ratio], 1.07 (0.93-1.23); P = 0.37; HR, 1.10 (0.91-1.31); P = 0.32, respectively), adjust model (HR, 0.99 (0.88-1.11); P = 0.87; HR, 1.05 (0.90-1.20); P = 0.55, respectively), and the subgroup analysis. For BCR, the patients who underwent PU were associated with a similar risk of developing BCR compared with those that received RNU, according to the univariate competing risk model (P = 0.47), adjust model (HR, 1.00 (0.73-1.37); P = 1), and subgroup analysis. CONCLUSION: RNU did not confer a distinct survival advantage compared with PU, which supports the role of PU in treating patients with ureter carcinomas.


Asunto(s)
Carcinoma de Células Transicionales , Uréter , Neoplasias Ureterales , Neoplasias de la Vejiga Urinaria , Humanos , Uréter/cirugía , Nefroureterectomía , Estudios Retrospectivos , Nefrectomía/métodos , Carcinoma de Células Transicionales/cirugía , Neoplasias Ureterales/cirugía , Neoplasias de la Vejiga Urinaria/patología
11.
Transl Cancer Res ; 11(8): 2607-2621, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-36093518

RESUMEN

Background: Prostate cancer (PCa) is the most common type of cancer in men. Destruction of or blocking lipid metabolism impairs the growth, proliferation, and survival of tumor cells. Recent studies on hepatic steatosis suggest that kinase tethers histone-lysine N-methyltransferase 2D (KMT2D) to peroxisome proliferator-activated receptor gamma (PPARγ), transactivating its target genes. Here, to determine a therapeutic approach that may interfere with PCa lipid metabolism, the interaction mechanism of KMT2D and PPARγ was verified in PCa. Methods: Molecular techniques and bioinformatics analysis were used to explore the relationship between KMT2D and lipid metabolism pathways in PCa. Moreover, the changes of lipid droplets were detected by oil red O staining and BODIPY staining. Molecular techniques were used to investigate the effect of KMT2D on PPARγ signaling in PCa cells. Co-immunoprecipitation (Co-IP) and DNA pull-down verified the mechanism of interaction between KMT2D and PPARγ. Results: KMT2D knockdown reduced the lipid droplet content in PC-3 and DU-145 cells and downregulated the expression of lipid metabolic genes. Low-dose rosiglitazone (ROSI) effectively activated the PPARγ pathway to promote lipid droplet synthesis and cell proliferation and migration. However, ROSI could not function effectively after KMT2D knockdown. Both co-IP and DNA pull-down analyses showed that KMT2D and PPARγ could be tethered to regulate the expression of PPARγ target genes. Conclusions: In PCa, KMT2D interacted with PPARγ, which directly participated in the regulation of lipid metabolism-related genes and affected lipid synthesis. Therefore, inhibiting the interaction between KMT2D and PPARγ is a potential therapeutic strategy.

12.
Cell Death Discov ; 8(1): 471, 2022 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-36446767

RESUMEN

The androgen receptor (AR) plays an essential role in prostate cancer progression and is a key target for prostate cancer treatment. However, patients with prostate cancer undergoing androgen deprivation therapy eventually experience biochemical relapse, with hormone-sensitive prostate cancer progressing into castration-resistant prostate cancer (CRPC). The widespread application of secondary antiandrogens, such as enzalutamide, indicates that targeting AR remains the most efficient method for CRPC treatment. Unfortunately, neither can block AR signaling thoroughly, leading to AR reactivation within several months. Here, we report an approach for suppressing reactivated AR signaling in the CRPC stage. A combination of the protein phosphatase 1 subunit α (PP1α)-specific inhibitor tautomycin and enzalutamide synergistically inhibited cell proliferation and AR signaling in LNCaP and C4-2 cells, as well as in AR variant-positive 22RV1 cells. Our results revealed that enzalutamide competed with residual androgens in CRPC, enhancing tautomycin-mediated AR degradation. In addition, the remaining competitive inhibitory role of enzalutamide on AR facilitated tautomycin-induced AR degradation in 22RV1 cells, further decreasing ARv7 levels via a full-length AR/ARv7 interaction. Taken together, our findings suggest that the combination of tautomycin and enzalutamide could achieve a more comprehensive inhibition of AR signaling in CRPC. AR degraders combined with AR antagonists may represent a new therapeutic strategy for CRPC.

13.
Cancer Gene Ther ; 29(5): 573-584, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-33888871

RESUMEN

Clear cell renal cell carcinoma (ccRCC) is the most common type of renal cancer affecting many people worldwide. Although the 5-year survival rate is 65% in localized disease, after metastasis, the survival rate is <10%. Emerging evidence has shown that microRNAs (miRNAs) play a crucial regulatory role in the progression of ccRCC. Here, we show that miR-335, an anti-onco-miRNA, is downregulation in tumor tissue and inhibited ccRCC cell proliferation, invasion, and migration. Our studies further identify the H3K9me1/2 histone demethylase KDM3A as a new miR-335-regulated gene. We show that KDM3A is overexpressed in ccRCC, and its upregulation contributes to the carcinogenesis and metastasis of ccRCC. Moreover, with the overexpression of KDM3A, YAP1 was increased and identified as a direct downstream target of KDM3A. Enrichment of KDM3A demethylase on YAP1 promoter was confirmed by CHIP-qPCR and YAP1 was also found involved in the cell growth and metastasis inhibitory of miR-335. Together, our study establishes a new miR-335/KDM3A/YAP1 regulation axis, which provided new insight and potential targeting of the metastasized ccRCC.


Asunto(s)
Carcinoma de Células Renales , Histona Demetilasas con Dominio de Jumonji , Neoplasias Renales , MicroARNs , Proteínas Señalizadoras YAP , Carcinogénesis/genética , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Proliferación Celular/genética , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Humanos , Histona Demetilasas con Dominio de Jumonji/genética , Histona Demetilasas con Dominio de Jumonji/metabolismo , Neoplasias Renales/patología , MicroARNs/genética , MicroARNs/metabolismo , Proteínas Señalizadoras YAP/genética , Proteínas Señalizadoras YAP/metabolismo
14.
Cell Death Dis ; 13(9): 754, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-36050295

RESUMEN

Ivermectin is a widely used antiparasitic drug and shows promising anticancer activity in various cancer types. Although multiple signaling pathways modulated by ivermectin have been identified in tumor cells, few studies have focused on the exact target of ivermectin. Herein, we report the pharmacological effects and targets of ivermectin in prostate cancer. Ivermectin caused G0/G1 cell cycle arrest, induced cell apoptosis and DNA damage, and decreased androgen receptor (AR) signaling in prostate cancer cells. Further in vivo analysis showed ivermectin could suppress 22RV1 xenograft progression. Using integrated omics profiling, including RNA-seq and thermal proteome profiling, the forkhead box protein A1 (FOXA1) and non-homologous end joining (NHEJ) repair executer Ku70/Ku80 were strongly suggested as direct targets of ivermectin in prostate cancer. The interaction of ivermectin and FOXA1 reduced the chromatin accessibility of AR signaling and the G0/G1 cell cycle regulator E2F1, leading to cell proliferation inhibition. The interaction of ivermectin and Ku70/Ku80 impaired the NHEJ repair ability. Cooperating with the downregulation of homologous recombination repair ability after AR signaling inhibition, ivermectin increased intracellular DNA double-strand breaks and finally triggered cell death. Our findings demonstrate the anticancer effect of ivermectin in prostate cancer, indicating that its use may be a new therapeutic approach for prostate cancer.


Asunto(s)
Factor Nuclear 3-alfa del Hepatocito , Ivermectina , Autoantígeno Ku , Neoplasias de la Próstata , Antígenos Nucleares/genética , Antígenos Nucleares/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , Reparación del ADN , Factor Nuclear 3-alfa del Hepatocito/efectos de los fármacos , Factor Nuclear 3-alfa del Hepatocito/metabolismo , Humanos , Ivermectina/farmacología , Ivermectina/uso terapéutico , Autoantígeno Ku/efectos de los fármacos , Autoantígeno Ku/metabolismo , Masculino , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo
15.
J Clin Invest ; 131(4)2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33332287

RESUMEN

Nuclear localization of the androgen receptor (AR) is necessary for its activation as a transcription factor. Defining the mechanisms regulating AR nuclear localization in androgen-sensitive cells and how these mechanisms are dysregulated in castration-resistant prostate cancer (CRPC) cells is fundamentally important and clinically relevant. According to the classical model of AR intracellular trafficking, androgens induce AR nuclear import and androgen withdrawal causes AR nuclear export. The present study has led to an updated model that AR could be imported in the absence of androgens, ubiquitinated, and degraded in the nucleus. Androgen withdrawal caused nuclear AR degradation, but not export. In comparison with their parental androgen-sensitive LNCaP prostate cancer cells, castration-resistant C4-2 cells exhibited reduced nuclear AR polyubiquitination and increased nuclear AR level. We previously identified 3-(4-chlorophenyl)-6,7-dihydro-5H-pyrrolo[1,2-a]imidazole (CPPI) in a high-throughput screen for its inhibition of androgen-independent AR nuclear localization in CRPC cells. The current study shows that CPPI is a competitive AR antagonist capable of enhancing AR interaction with its E3 ligase MDM2 and degradation of AR in the nuclei of CRPC cells. Also, CPPI blocked androgen-independent AR nuclear import. Overall, these findings suggest the feasibility of targeting androgen-independent AR nuclear import and stabilization, two necessary steps leading to AR nuclear localization and activation in CRPC cells, with small molecule inhibitors.


Asunto(s)
Antagonistas de Receptores Androgénicos/farmacología , Núcleo Celular/metabolismo , Sistemas de Liberación de Medicamentos , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Receptores Androgénicos/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Transporte Activo de Núcleo Celular/genética , Antagonistas de Receptores Androgénicos/síntesis química , Antagonistas de Receptores Androgénicos/química , Animales , Células COS , Línea Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/patología , Chlorocebus aethiops , Células HEK293 , Humanos , Masculino , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/patología , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Receptores Androgénicos/genética , Ubiquitinación/efectos de los fármacos , Ubiquitinación/genética
16.
Aging (Albany NY) ; 12(13): 12771-12782, 2020 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-32657763

RESUMEN

Prostatic cancer stem-like cells (PCSLCs) play an essential role in PCa development. Accumulating evidence suggests that androgen deprivation therapy (ADT) or chemotherapy using docetaxel could expand the population of PCSLCs. Therefore, understanding the underlying mechanisms responsible for PCSLCs expansion has broadly scientific interest. Here, our results revealed that lncRNA HOTAIR could increase PCSLCs population via activating STAT3 signaling. Mechanistically, HOTAIR functioned as miR-590-5p sponge and prevented it from targeting the 3'UTR of IL-10, one upstream molecule of STAT3 signaling, leading to IL-10 upregulation and STAT3 activation. We also found that HOTAIR was required and sufficient to cause Docetaxel resistance (DocR) in C4-2 PCa cells. Moreover, our in vivo animal study also confirmed that Du145-HOTAIR mice had a faster tumor growth rate and a poorer survival rate compared to control cohorts. Our data build compelling rationale to target HOTAIR for the depletion of PCSLCs and alleviation of Docetaxel resistance.


Asunto(s)
Docetaxel/farmacología , Resistencia a Antineoplásicos/fisiología , Células Madre Neoplásicas/efectos de los fármacos , ARN Largo no Codificante/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Línea Celular Tumoral , Células Cultivadas , Xenoinjertos , Humanos , Masculino , Ratones , Ratones Desnudos , Células Madre Neoplásicas/metabolismo , Neoplasias de la Próstata/patología
17.
Cancer Manag Res ; 12: 4411-4427, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32606936

RESUMEN

BACKGROUND: Elongation factor for RNA polymerase II 2 (ELL2) was reported as a putative tumor suppressor in the prostate. ELL2 is frequently down-regulated in prostatic adenocarcinoma specimens, and loss of ELL2 induced murine prostatic intraepithelial neoplasia and enhanced AR-positive prostate cancer cell proliferation. However, the ELL2 gene appears to be amplified in AR-negative neuroendocrine prostate tumors, suggesting a potential oncogenic role for ELL2 in AR-negative prostate cancer cells. In this study, we explored the potential function of ELL2 in PC-3 and DU145, two AR-negative prostate cancer cell lines. MATERIALS AND METHODS: The role of ELL2 in PC-3 and DU145 cells was studied using siRNA-mediated ELL2 knockdown. Genes regulated by ELL2 knockdown in PC-3 cells were identified and analyzed using RNA-Seq and bioinformatics. The expression of representative genes was confirmed by Western blot and/or quantitative PCR. Cell growth was determined by BrdU, MTT and colony formation assays. Cell death was analyzed by 7-AAD/Annexin V staining and trypan blue exclusion staining. Cell cycle was determined by PI staining and flow cytometry. RESULTS: ELL2 knockdown inhibited the proliferation of PC-3 and DU145 cells. RNA-Seq analysis showed an enrichment in genes associated with cell death and survival following ELL2 knockdown. The interferon-γ pathway was identified as the top canonical pathway comprising of 55.6% of the genes regulated by ELL2. ELL2 knockdown induced an increase in STAT1 and IRF1 mRNA and an induction of total STAT1 and phosphorylated STAT1 protein. Inhibition of cell proliferation by ELL2 knockdown was partly abrogated by STAT1 knockdown. ELL2 knockdown inhibited colony formation and induced apoptosis in both PC-3 and DU145 cells. Furthermore, knockdown of ELL2 caused S-phase cell cycle arrest, inhibition of CDK2 phosphorylation and cyclin D1 expression, and increased expression of cyclin E. CONCLUSION: ELL2 knockdown in PC-3 and DU145 cells induced S-phase cell cycle arrest and profound apoptosis, which was accompanied by the induction of genes associated with cell death and survival pathways. These observations suggest that ELL2 is a potential oncogenic protein required for survival and proliferation in AR-negative prostate cancer cells.

18.
Asian J Androl ; 22(2): 162-168, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31134918

RESUMEN

Prostate cancer (PCa) exhibits epidemiological and molecular heterogeneity. Despite extensive studies of its phenotypic and genetic properties in Western populations, its molecular basis is not clear in Chinese patients. To determine critical molecular characteristics and explore correlations between genomic markers and clinical parameters in Chinese populations, we applied an integrative genetic/transcriptomic assay that combines targeted next-generation sequencing and quantitative real-time PCR (qRT-PCR) on samples from 46 Chinese patients with PCa. Lysine (K)-specific methyltransferase 2D (KMT2D), zinc finger homeobox 3 (ZFHX3), A-kinase anchoring protein 9 (AKAP9), and GLI family zinc finger 1 (GLI1) were frequently mutated in our cohort. Moreover, a clinicopathological analysis showed that RB transcriptional corepressor 1 (RB1) deletion was common in patients with a high risk of disease progression. Remarkably, four genomic events, MYC proto-oncogene (MYC) amplification, RB1 deletion, APC regulator of WNT signaling pathway (APC) mutation or deletion, and cyclin-dependent kinase 12 (CDK12) mutation, were correlated with poor disease-free survival. In addition, a close link between KMT2D expression and the androgen receptor (AR) signaling pathway was observed both in our cohort and in The Cancer Genome Atlas Prostate Adenocarcinoma (TCGA-PRAD) data. In summary, our results demonstrate the feasibility and benefits of integrative molecular characterization of PCa samples in disease pathology research and personalized medicine.


Asunto(s)
Mutación , Neoplasias de la Próstata/genética , Receptores Androgénicos/genética , Proteínas de Anclaje a la Quinasa A/genética , Adulto , Anciano , Biomarcadores de Tumor/genética , China , Proteínas del Citoesqueleto/genética , Proteínas de Unión al ADN/genética , Amplificación de Genes , Secuenciación de Nucleótidos de Alto Rendimiento , Proteínas de Homeodominio/genética , Humanos , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/genética , Neoplasias de la Próstata/patología , Proto-Oncogenes Mas , Transducción de Señal/genética , Proteína con Dedos de Zinc GLI1/genética
19.
Epigenetics ; 14(12): 1194-1208, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31232159

RESUMEN

Histone methyltransferase KMT2D has diverse functions and distinct mechanisms in different cancers. Although we have previously found KMT2D serves as an oncogene that promotes tumor growth and metastasis in prostate cancer (PCa), the functions and mechanisms of KMT2D are complicated and most remain undefined. Here, the function of KMT2D regarding DNA damage in PCa and the underlying mechanisms of KMT2D in epigenetic regulation were explored in a series of studies. Knockdown of KMT2D sensitized cells to DNA damage through the disturbance of antioxidative gene expression and increased levels of intracellular reactive oxygen species, which led to cell apoptosis and senescence. The loss of KMT2D reduced the abundance of enhancer activity markers H3K4me1 and H3K27ac, which blocked the DNA binding of FOXO3, a critical mediator of the cellular response to oxidative stress, and suppressed antioxidative gene transcription. Moreover, KMT2D deletion in PCa cells also increased their sensitivity to genotoxic anticancer drugs and a PARP inhibitor, which suggested that lower levels of KMT2D may mediate the response of PCa to particular treatments. These findings further highlighted the important role of KMT2D in PCa progression and suggested that targeting KMT2D might be therapeutically beneficial for advanced PCa treatment.


Asunto(s)
Daño del ADN , Proteínas de Unión al ADN/genética , Proteínas de Neoplasias/genética , Estrés Oxidativo , Neoplasias de la Próstata/genética , Apoptosis , Línea Celular Tumoral , Resistencia a Antineoplásicos , Elementos de Facilitación Genéticos , Epigénesis Genética , Proteína Forkhead Box O3/genética , Proteína Forkhead Box O3/metabolismo , Código de Histonas , Humanos , Masculino , Neoplasias de la Próstata/metabolismo , Unión Proteica , Especies Reactivas de Oxígeno/metabolismo
20.
Nan Fang Yi Ke Da Xue Xue Bao ; 38(5): 606-611, 2018 May 20.
Artículo en Zh | MEDLINE | ID: mdl-29891460

RESUMEN

OBJECTIVE: To analyze the advantages of spatial measurement of anatomical parameters in a 3D model in surgical planning for laparoscopic partial nephrectomy (LPN). METHODS: From February, 2016 to October, 2017, 37 patients diagnosed with T1 renal mass underwent LPN based on 3D reconstruction after enhanced CT scanning using the Uromedix-3D system (group A), and another 38 patients received LPN with conventional CT planning (group B). The anatomical parameters were measured in the reconstructed 3D model and the demographic data, surgical outcome and postoperative data were compared between the two groups. RESULTS: In group A, the average time for 3D model reconstruction was (29.3∓9.7) min; the length, width and depth of the renal defect in 3D model were 3.2∓1.1 cm, 2.6∓0.9 cm and 1.7∓0.7 cm, respectively; The distance of the tumor from the collecting system was 3.8∓2.2 mm; The mean R.E.N.A.L score of the patients was 7∓1.5, and 3 patients had accessory renal artery and 2 had early branching of the renal artery. LPNs were completed via the retroperitoneal approach in all the 75 patients without conversion to open or total nephrectomy. Group A and group B showed significant differences in warm ischemic time (26.7∓6.4 vs 31.9∓7.0 min), tumor-excision time (8.4∓2.6 vs 10.4∓2.8 min), renal defect suture time (18.3∓3.9 vs 21.5∓3.4 min), 24-h volume of retroperitoneal drainage (88.6∓40.2 vs 134.3∓58.3 mL) and 48-h volume of retroperitoneal drainage (127.9∓54.5 vs 198.1∓86.3 mL), but not in the demographic data, operation time, intraoperative blood loss or postoperative hospital stay. CONCLUSIONS: 3D reconstruction of the renal masses can be completed efficiently and accurately using this system. Compared with conventional CT-based measurement, 3D spatial measurement of the anatomical structures helps to increase the precision in the performance of LPN and reduce the warm ischemia time.


Asunto(s)
Imagenología Tridimensional/métodos , Neoplasias Renales/diagnóstico por imagen , Neoplasias Renales/cirugía , Laparoscopía/métodos , Nefrectomía/métodos , Tomografía Computarizada por Rayos X/métodos , Humanos , Neoplasias Renales/patología , Estudios Retrospectivos , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA