Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Nanobiotechnology ; 22(1): 236, 2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38724995

RESUMO

Increased proinflammatory cytokines and infiltration of inflammatory cells in the stroma are important pathological features of type IIIA chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS-A), and the interaction between stromal cells and other cells in the inflammatory microenvironment is closely related to the inflammatory process of CP/CPPS-A. However, the interaction between stromal and epithelial cells remains unclear. In this study, inflammatory prostate epithelial cells (PECs) released miR-203a-3p-rich exosomes and facilitated prostate stromal cells (PSCs) inflammation by upregulating MCP-1 expression. Mechanistically, DUSP5 was identified as a novel target gene of miR-203a-3p and regulated PSCs inflammation through the ERK1/2/MCP-1 signaling pathway. Meanwhile, the effect of exosomes derived from prostatic fluids of CP/CPPS-A patients was consistent with that of exosomes derived from inflammatory PECs. Importantly, we demonstrated that miR-203a-3p antagomirs-loaded exosomes derived from PECs targeted the prostate and alleviated prostatitis by inhibiting the DUSP5-ERK1/2 pathway. Collectively, our findings provide new insights into underlying the interaction between PECs and PSCs in CP/CPPS-A, providing a promising therapeutic strategy for CP/CPPS-A.


Assuntos
Células Epiteliais , Exossomos , MicroRNAs , Prostatite , Células Estromais , Masculino , Exossomos/metabolismo , Prostatite/genética , Prostatite/patologia , Prostatite/metabolismo , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Células Estromais/metabolismo , Células Estromais/patologia , Animais , Fosfatases de Especificidade Dupla/genética , Fosfatases de Especificidade Dupla/metabolismo , Próstata/patologia , Próstata/metabolismo , Dor Pélvica , Inflamação/genética , Inflamação/patologia , Camundongos , Sistema de Sinalização das MAP Quinases
2.
J Transl Med ; 22(1): 178, 2024 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-38369471

RESUMO

BACKGROUND: Castration-resistant prostate cancer (CRPC) is refractory to hormone treatment, and the underlying mechanism has not been fully elucidated. This study aimed to clarify the role and mechanism of Human antigen R (HuR) as a therapeutic target for CRPC progression. METHODS: HuR was knocked out by Cas9 or inhibited by the HuR-specific inhibitor KH-3 in CRPC cell lines and in a mouse xenograft model. The effects of HuR inhibition on tumour cell behaviors and signal transduction were examined by proliferation, transwell, and tumour xenograft assays. Posttranscriptional regulation of BCAT1 by HuR was determined by half-life and RIP assays. RESULTS: HuR knockout attenuated the proliferation, migration, and invasion of PC3 and DU145 cells in vitro and inhibited tumour progression in vivo. Moreover, BCAT1 was a direct target gene of HuR and mediated the oncogenic effect of HuR on CRPC. Mechanistically, HuR directly interacted with BCAT1 mRNA and upregulated BCAT1 expression by increasing the stability and translation of BCAT1, which activated ERK5 signalling. Additionally, the HuR-specific inhibitor KH-3 attenuated CRPC progression by disrupting the HuR-BCAT1 interaction. CONCLUSIONS: We confirmed that the HuR/BCAT1 axis plays a crucial role in CRPC progression and suggest that inhibiting the HuR/BCAT1 axis is a promising therapeutic approach for suppressing CRPC progression.


Assuntos
Neoplasias de Próstata Resistentes à Castração , Masculino , Humanos , Animais , Camundongos , Neoplasias de Próstata Resistentes à Castração/genética , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Neoplasias de Próstata Resistentes à Castração/patologia , Linhagem Celular Tumoral , Transdução de Sinais , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Transaminases/genética
3.
DNA Cell Biol ; 40(11): 1445-1455, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34767732

RESUMO

Purpose: Metastatic prostate cancer (PCa) has become a major obstacle in the treatment of PCa. The study's purpose is to find biomarkers of tumor metastasis by proteomics and enzyme-linked immunosorbent assay (ELISA), and to design related experiments to study its role in the progress and metastasis of PCa. Method: We analyzed serum from primary PCa stage and metastatic stage of 12 patients to find metastatic PCa serum protein biomarkers using isobaric tags for relative and absolute quantitation (iTRAQ). An effective diagnostic model based on validated biomarkers using logistic regression was established. In vivo and in vitro biological behavior experiments (wound healing, CCK8, and Transwell tests) were carried out after obtaining the biomarkers. Related mechanism has been studied, which may be associated with metastatic PCa. Result: Actin gamma 1 (ACTG1) is a potential biomarker in the metastasis of PCa. Bioinformatics and related experiments show that ACTG1 is high-expressed in PCa tissues and cells. In vivo and in vitro experiments illustrated that the ability of proliferation, migration, and invasion of PCa cells was significantly inhibited after the knockdown of ACTG1 expression. Surprisingly, ERK protein expression was downregulated after ACTG1 knockdown. At the same time, the expression of epithelial-mesenchymal transition-related markers in PCa cells decrease after treated with ERK1/2 inhibitor, which indicating that ACTG1 may affect the metastatic ability of PCa cells through MAPK/ERK signaling pathway. Conclusion: ACTG1 is a marker of metastasis PCa. It mediates cell proliferation and may regulate the metastasis of PCa through MAPK/ERK signaling pathway, which provides a useful theoretical basis for exploring the treatment of PCa.


Assuntos
Actinas/genética , Neoplasias da Próstata/genética , Actinas/metabolismo , Biomarcadores Tumorais/sangue , Biomarcadores Tumorais/genética , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , China , Transição Epitelial-Mesenquimal/genética , Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Sistema de Sinalização das MAP Quinases , Masculino , Proteína Quinase 3 Ativada por Mitógeno , Metástase Neoplásica/genética , Neoplasias da Próstata/metabolismo , Transdução de Sinais/efeitos dos fármacos
4.
Oncol Rep ; 45(5)2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-34105734

RESUMO

The aim of the present study was to explore and verify the potential mechanism of seminoma progression. Data on 132 RNA­seq and 156 methylation sites from stage II/III and I seminoma specimens were downloaded from The Cancer Genome Atlas database. An initial filter of |fold­change| >2 and false discovery rate <0.05 were used to identify differentially expressed genes (DEGs) which were associated with differential methylation site genes; these genes were considered potential candidates for further investigation by survival analysis. Potassium voltage­gated channel subfamily C member 1 (KCNC1) expression was verified in seminoma human tissues and three seminoma cell lines. The invasive, proliferative and apoptotic abilities of the human testicular tumor Ntera­2 and normal human testis Hs1.Tes cell lines were assessed following aberrant KCNC1 expression. KCNC1 was identified as a DEG, in which hypermethylation inhibited its expression and it was associated with poor overall survival in patients with seminoma. The present results demonstrated that KCNC1 is negatively correlated with methylation. Due to the abnormal expression of KCNC1 in seminoma cells, it was suggested that KCNC1 could be used as a diagnostic indicator and therapeutic target for the progression of seminoma.


Assuntos
Metilação de DNA , Seminoma/genética , Canais de Potássio Shaw/genética , Neoplasias Testiculares/genética , Adulto , Apoptose/genética , Proliferação de Células/genética , Técnicas de Inativação de Genes , Humanos , Imuno-Histoquímica , Masculino , Invasividade Neoplásica , Metástase Neoplásica , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Seminoma/metabolismo , Seminoma/mortalidade , Seminoma/patologia , Canais de Potássio Shaw/biossíntese , Taxa de Sobrevida , Neoplasias Testiculares/metabolismo , Neoplasias Testiculares/mortalidade , Neoplasias Testiculares/patologia , Transfecção
5.
Gene ; 744: 144608, 2020 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-32234541

RESUMO

Prostate cancer (PCa) is the third most common malignancy worldwide. Novel and effective therapeutic targets are needed for PCa. The purpose of this study was to discover novel therapeutic targets for PCa by performing advanced analysis on PCa RNA sequencing (RNAseq) data from The Cancer Genome Atlas (TCGA). Weighted correlation-network analysis (WGCNA) was performed on the RNAseq data of tumor samples, and the module most relevant to the Gleason score was identified. Combining differential gene-expression analysis and survival analysis, we narrowed down potential therapeutic target genes and found that PKMYT1 might be one. Subsequently, functional studies (i.e., cell-proliferation assays, cell cycle analysis, and colony-formation assays) demonstrated that knockdown of PKMYT1 significantly inhibited the growth of PCa cells. Further investigation illustrated that PKMYT1 promoted the growth of PCa cells through targeting CCNB1 and CCNE1 expression. In addition, fostamatinib, an inhibitor of PKMYT1, effectively inhibited the proliferation of PCa cells. Taken together, our results suggest that PKMYT1 is a gene associated with malignancy of PCa and is a novel therapeutic target.


Assuntos
Neoplasias da Próstata/enzimologia , Aminopiridinas , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Ciclina B1/genética , Ciclina B1/metabolismo , Ciclina E/genética , Ciclina E/metabolismo , Humanos , Masculino , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/metabolismo , Morfolinas , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Oxazinas/uso terapêutico , Prognóstico , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo , Piridinas/uso terapêutico , Pirimidinas
6.
Exp Cell Res ; 385(1): 111669, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31605696

RESUMO

PURPOSE: The main objective of our study was to explore changes in the expression levels of differentially expressed genes associated with prostate cancer progression and to design a series of experiments to verify the function of differentially expressed genes. METHOD: The transcriptome datas of 499 cases of prostate cancer patients was downloaded from TCGA database. Differential genes associated with Gleason score were selected and filtered out by p < 0.05 and spearman coefficient >0.3. KEGG signaling pathway was enriched by differentially expressed genes, and TTK was selected as the research object. The expression of TTK was tested in prostate cancer tissues and prostate cancer cell lines. The changes of biological behavior of prostate cancer cell lines were verified after TTK was knocked out by siRNA and tumorigenic effect of TTK was verified by shRNA in vivo experiments. RESULT: The expression of TTK was positively correlated with Gleason score of prostate cancer, and the expression of protein and mRNA in metastatic prostate cancer cell lines was higher than that in non-metastatic prostate cancer cell lines. Vitro biological experiments showed that TTK gene knockout could inhibit the proliferation, invasion and migration of PC3 and DU145 cells, and promote cell apoptosis. In vivo experiments showed that TTK knockout inhibited tumorigenesis in mice. It was found that the expression of CDK2 and CCNE1 decreased after TTK was knocked out. CONCLUSION: Our results suggest that TTK is a gene associated with malignancy of PCa and could be a novel therapeutic target for clinical application.


Assuntos
Proteínas de Ciclo Celular/genética , Proliferação de Células/genética , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Tirosina Quinases/genética , Animais , Apoptose/genética , Carcinogênese/genética , Carcinogênese/patologia , Linhagem Celular Tumoral , Quinase 2 Dependente de Ciclina/genética , Progressão da Doença , Humanos , Masculino , Camundongos , Gradação de Tumores , Proteínas Oncogênicas/genética , Células PC-3 , Próstata/patologia , RNA Interferente Pequeno/genética , Transdução de Sinais/genética , Transcriptoma/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA