Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
BMC Urol ; 17(1): 14, 2017 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-28193277

RESUMO

BACKGROUND: To evaluate anti-prostate cancer effects of a chimeric tumor-targeted killer protein. METHODS: We established a novel fusion gene, immunocasp-3, composed of NH2-terminal leader sequence fused with an anti-prostate-specific membrane antigen (PSMA) antibody (J591), the furin cleavage sequences of diphtheria toxin (Fdt), and the reverse coding sequences of the large and small subunits of caspase-3 (revcaspase-3). The expressing level of the immunocasp-3 gene was evaluated by using the reverse transcription-PCR (RT-PCR) and western blot analysis. Cell viability assay and cytotoxicity assay were used to evaluate its anti-tumor effects in vitro. Apoptosis was confirmed by electron microscopy and Annexin V-FITC staining. The antitumor effects of immunocasp-3 were assessed in nude mice xenograft models containing PSMA-overexpressing LNCaP cells. RESULTS: This study shows that the immunocasp-3 proteins selectively recognized and induced apoptotic death in PSMA-overexpressing LNCaP cells in vitro, where apoptotic cells were present in 15.3% of the cells transfected with the immunocasp-3 expression vector at 48 h after the transfection, in contrast to 5.5% in the control cells. Moreover, LNCaP cells were significantly killed under the condition of the co-culture of the immunocasp-3-secreting Jurkat cells and more than 50% of the LNCaP cells died when the two cell lines were co-cultured within 5 days. In addition, The expression of immunocasp-3 also significantly suppressed tumor growth and greatly prolonged the animal survival rate in vivo. CONCLUSION: A novel fusion gene, immunocasp-3, may represent a viable approach to treating PSMA-positive prostate cancer.


Assuntos
Adenocarcinoma/terapia , Antígenos de Superfície , Terapia Genética , Glutamato Carboxipeptidase II , Neoplasias da Próstata/terapia , Adenocarcinoma/patologia , Animais , Antígenos de Superfície/genética , Fusão Gênica Artificial , Caspase 3/genética , Terapia Genética/métodos , Glutamato Carboxipeptidase II/genética , Humanos , Imunotoxinas/genética , Masculino , Camundongos Nus , Neoplasias da Próstata/patologia , Células Tumorais Cultivadas
2.
PeerJ ; 11: e16314, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38047026

RESUMO

Background: Poly (ADP-ribose) polymerase (PARP) inhibitors (PARPi), have gained approval for treating patients with castration-resistant prostate cancer (CRPC). Maternally expressed gene 3 (MEG3), a long non-coding RNA (lncRNA), plays a role in inhibiting tumorigenesis through regulating DNA repair genes. This study aimed to investigate the association between the anti-prostate cancer (PCa) effect of niraparib, a representative PARPi, and MEG3 expression, as well as explore the downstream pathway involved. Methods: The levels of MEG3, miR-181-5p, GATA binding protein 6 (GATA6) in clinical samples from PCa patients were accessed by RT-qPCR. PC3 cells were treated with niraparib, and the expression of MEG3, miR-181-5p, GATA6 expression was tested. PC3 cell proliferation, migration, and invasion were tested by CCK-8, wound healing, and Transwell assays, respectively. The bindings between miR-181-5p and MEG3/GATA6 were determined by dual-luciferase reporter gene assay. Furthermore, rescue experiments were conducted to investigate the underlying mechanism of MEG3/miR-181-5p/GATA6 axis in PCa progression. Additionally, mice were injected with PC3 cells transfected with sh-MEG3 and treated with niraparib, and the xenograft tumor growth was observed. Results: MEG3 and GATA6 were upregulated and miR-181-5p was downregulated in PCa patients. Niraparib treatment substantially upregulated MEG3 and GATA6, and downregulated miR-181-5p expression in PCa cells. Niraparib effectively restrained PC3 cell proliferation, migration, and invasion. MiR-181-5p targeted to MEG3, and the inhibitory effects of MEG3 overexpression on PC3 cell proliferation and metastasis were abrogated by miR-181-5p overexpression. Moreover, GATA6 was identified as a target of miR-181-5p, and GATA6 silencing abolished the inhibitory effects of miR-181-5p inhibition on PC3 cell proliferation and metastasis. Besides, MEG3 silencing could abrogate niraparib-mediated tumor growth inhibition in mice. Conclusions: Niraparib restrains prostate cancer cell proliferation and metastasis and tumor growth in mice by regulating the lncRNA MEG3/miR-181-5p/GATA6 pathway.


Assuntos
MicroRNAs , Hiperplasia Prostática , Neoplasias da Próstata , RNA Longo não Codificante , Masculino , Humanos , Camundongos , Animais , RNA Longo não Codificante/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , MicroRNAs/genética , Neoplasias da Próstata/tratamento farmacológico , Fator de Transcrição GATA6/genética
3.
Am J Transl Res ; 15(2): 1502-1509, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36915765

RESUMO

OBJECTIVE: This study aims to establish and validate a predictive model for bone metastasis in prostate cancer patients based on multiple immune inflammatory parameters. METHODS: In this retrospective study, 162 prostate cancer patients who met the inclusion criteria were selected by Urology Surgery, Shaanxi Provincial People's Hospital. Based on the medical record number of patients and the random number table method, 40 patients were randomly included in a validation group, and the rest were in a modeling group. The patients in the modeling group were divided into a metastatic group (n=67) and a non-metastatic group (n=55) according to the whole-body bone imaging results. RESULTS: The predictive model was established based on the results of Logistics regression analysis: Logit (P) = -5.341 + 0.930*total Gleason score + 1.426*total prostate specific antigen + 0.836*neutrophil-lymphocyte ratio + 0.896*platelet lymphocyte ratio + 0.641*lymphocyte/monocyte ratio + 0.750*albumin/globulin ratio. ROC analysis showed that the areas under the curve of the predictive model for bone metastasis in the modeling and validation groups were 0.896 and 0.870, respectively. Hosmer-Lemeshow test showed that P=0.253, indicating a high degree of the fitting. External verification results showed that the C-index for predicting prostate cancer bone metastasis in the predictive model established in this study was 0.760 (95% CI: 0.670-0.851). CONCLUSION: The bone metastasis predictive model based on the multiple immune inflammatory parameters (neutrophil-lymphocyte ratio, platelet lymphocyte ratio, lymphocyte/monocyte ratio and albumin/globulin ratio) in prostate cancer patients can reasonably predict the occurrence of bone metastasis and is well worth clinical application.

4.
PLoS One ; 5(4): e10147, 2010 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-20418948

RESUMO

BACKGROUND: Previous work has shown reduced expression levels of let-7 in lung tumors. But little is known about the expression or mechanisms of let-7a in prostate cancer. In this study, we used in vitro and in vivo approaches to investigate whether E2F2 and CCND2 are direct targets of let-7a, and if let-7a acts as a tumor suppressor in prostate cancer by down-regulating E2F2 and CCND2. METHODOLOGY/PRINCIPAL: Findings Real-time RT-PCR demonstrated that decreased levels of let-7a are present in resected prostate cancer samples and prostate cancer cell lines. Cellular proliferation was inhibited in PC3 cells and LNCaP cells after transfection with let-7a. Cell cycle analysis showed that let-7a induced cell cycle arrest at the G1/S phase. A dual-luciferase reporter assay demonstrated that the 3'UTR of E2F2 and CCND2 were directly bound to let-7a and western blotting analysis further indicated that let-7a down-regulated the expression of E2F2 and CCND2. Our xenograft models of prostate cancer confirmed the capability of let-7a to inhibit prostate tumor development in vivo. CONCLUSIONS/SIGNIFICANCE: These findings help to unravel the anti-proliferative mechanisms of let-7a in prostate cancer. Let-7a may also be novel therapeutic candidate for prostate cancer given its ability to induce cell-cycle arrest and inhibit cell growth, especially in hormone-refractory prostate cancer.


Assuntos
Proliferação de Células/efeitos dos fármacos , Ciclina D2/metabolismo , Fator de Transcrição E2F2/metabolismo , MicroRNAs/farmacologia , Neoplasias da Próstata/patologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Ciclina D2/genética , Regulação para Baixo/efeitos dos fármacos , Fator de Transcrição E2F2/genética , Humanos , Masculino , MicroRNAs/genética , Transfecção
5.
Cancer Lett ; 276(1): 88-94, 2009 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-19081179

RESUMO

Our previous studies indicated a direct correlation with loss of CIAPIN1 and carcinogenesis of tumor in human gastric cancer. Here we presented that the expression of CIAPIN1 was absent or significantly decreased in 102 cases of clear cell renal cell carcinoma (CCRCC) tissues (P<0.05). Up-regulating CIAPIN1 by adenoviral vectors exhibited significant inhibition of CCRCC-derived cell growth in vitro and in vivo with G1 cell cycle arrest. Simultaneously, CIAPIN1-induced growth suppression was found partially to regulate various proteins, including inhibition of cyclinD1, cyclinE, cdk2, cdk4, p-Rb and VEGF, but up-regulation of p27Kip1 and Rb.


Assuntos
Carcinoma de Células Renais/metabolismo , Proliferação de Células , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neoplasias Renais/metabolismo , Biomarcadores Tumorais/análise , Western Blotting , Carcinoma de Células Renais/genética , Expressão Gênica , Humanos , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intracelular/genética , Neoplasias Renais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA