Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Biochem Mol Toxicol ; 37(8): e23399, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37345681

RESUMO

Metabolic abnormalities and uncontrolled angiogenesis are two vital features of malignant tumors. Although fibroblast growth factor 6 (FGF6) is known to promote the proliferation and migration of bladder cancer (BC) cells, its influences on aerobic glycolysis and angiogenesis in BC remain unclear. Gene expression at messenger RNA and protein levels were examined by reverse transcription-quantitative polymerase chain reaction and Western blot analyses, respectively. Lactate production and glucose uptake in BC cells were evaluated by performing aerobic glycolysis assays. A vasculogenic mimicry assay was executed for assessing the angiogenesis of BC cells. The viability, migration, and angiogenesis of human umbilical vein endothelial cells (HUVECs) cocultured with supernatants of BC cells were detected using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, wound healing assay, and tube formation assay. It was found that FGF6 displayed a high level in BC cell lines. Silencing of FGF6 reduced the levels of lactate production, glucose uptake, and the expression of angiogenic factors and glycolytic enzymes in BC cells, which also inhibited the viability and migration of HUVECs. In addition, FGF6 depletion or aerobic glycolysis inhibitor 2-deoxy-d-glucose treatment decreased the total branching length and intersection number of both BC cells and HUVECs. Moreover, glucose or lactate treatment reversed FGF6-induced suppression of cell viability, migration, tube formation, and vasculogenic mimicry. The activation of the phosphatidylinositol-3-kinase (PI3K)/protein kinase B (Akt) and mitogen-activated protein kinase (MAPK) signaling pathways was blocked by silenced FGF6. Furthermore, PI3K/Akt inhibitor (LY2940002) and p38-MAPK inhibitor (SB203580) inhibited the levels of aerobic glycolysis-related proteins. In conclusion, FGF6 knockdown suppressed aerobic glycolysis, thereby inhibiting angiogenesis in BC via regulation of the PI3K/Akt and MAPK signaling pathways.


Assuntos
Proteínas Proto-Oncogênicas c-akt , Neoplasias da Bexiga Urinária , Humanos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais , Células Endoteliais da Veia Umbilical Humana/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Glicólise , Glucose/metabolismo , Lactatos , Proliferação de Células , Movimento Celular
2.
J Biochem Mol Toxicol ; 37(8): e23389, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37300450

RESUMO

It has been reported ursolic acid (UA), one of the naturally abundant pentacyclic triterpenes, possesses a wide range of biological activities including anti-inflammatory, anti-atherosclerotic, and anticancer properties. Renal cell carcinoma (RCC) is a severe malignancy due to its asymptomatically spreading ability. Our study aimed to investigate the role and molecular mechanism of UA in RCC. RCC cell proliferation, migration, invasion, and angiogenesis were assessed using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, Transwell, and tube formation assays. Xenograft tumor models were established to confirm the role of UA and long noncoding RNA ASMTL antisense RNA 1 (ASMTL-AS1) in vivo. Expression levels of ASMTL-AS1 and vascular endothelial growth factor (VEGF) were measured using reverse transcriptase quantitative polymerase chain reaction and western blot analysis. The interaction probabilities of ASMTL-AS1 or VEGF with RNA-binding protein human antigen R (HuR) were verified by RNA immunoprecipitation experiment. The half-life period of messenger RNA (mRNA) was determined using actinomycin D. UA inhibited RCC cell growth in vivo and tumorigenesis in vitro. ASMTL-AS1 was highly expressed in RCC cell lines. Of note, UA downregulated ASMTL-AS1 expression, and overexpressed ASMTL-AS1 reversed the UA-induced suppression on RCC cell migration, invasion, and tube formation. Additionally, ASMTL-AS1 bound to HuR to maintain the stability of VEGF mRNA. Rescue experiments showed that the suppressed malignancy of RCC cells mediated by ASMTL-AS1 knockdown was counteracted by overexpression of VEGF. Moreover, silenced ASMTL-AS1 inhibited RCC tumor growth and metastasis in vivo. The obtained data suggest UA as a promising therapeutic agent to attenuate the development of RCC via regulation of the targeted molecules.


Assuntos
Carcinoma de Células Renais , Neoplasias Renais , MicroRNAs , RNA Longo não Codificante , Humanos , Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , RNA Longo não Codificante/genética , Linhagem Celular Tumoral , MicroRNAs/genética , Proliferação de Células/genética , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/metabolismo , RNA Mensageiro , Regulação Neoplásica da Expressão Gênica , Movimento Celular/genética , Ácido Ursólico
3.
Hum Exp Toxicol ; 40(12_suppl): S434-S446, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34591706

RESUMO

BACKGROUND: Bladder cancer (BCa) is a common genitourinary malignancy with higher incidence in males. Long intergenic non-protein coding RNA 265 (LINC00265) is identified as an oncogene in many malignancies, while its role in BCa development remains unknown. PURPOSE: To explore the functions and mechanism of LINC00265 in BCa. RESEARCH DESIGN: Reverse transcription quantitative polymerase chain reaction was performed to examine LINC00265 expression in BCa cells. Cell counting kit-8 assays, colony formation assays, TdT-mediated dUTP Nick-End Labeling assays, and Transwell assays were conducted to examine BCa cell viability, proliferation, apoptosis, and migration. Luciferase reporter assays and RNA immunoprecipitation assays were carried out to explore the binding capacity between miR-4677-3p and messenger RNA fibroblast growth factor 6 (FGF6) (or LINC00265). Xenograft tumor model was established to explore the role of LINC00265 in vivo. RESULTS: LINC00265 was highly expressed in BCa cells. LINC00265 knockdown inhibited xenograft tumor growth and BCa cell viability, proliferation and migration while enhancing cell apoptosis. Moreover, LINC00265 interacted with miR-4677-3p to upregulate the expression of FGF6. FGF6 overexpression reversed the suppressive effect of LINC00265 knockdown on malignant phenotypes of BCa cells. CONCLUSIONS: LINC00265 promotes the viability, proliferation, and migration of BCa cells by binding with miR-4677-3p to upregulate FGF6 expression.


Assuntos
Sobrevivência Celular , Fator 6 de Crescimento de Fibroblastos , Regulação Neoplásica da Expressão Gênica , RNA Longo não Codificante , Animais , Humanos , Masculino , Camundongos , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Sobrevivência Celular/fisiologia , Fator 6 de Crescimento de Fibroblastos/genética , Fator 6 de Crescimento de Fibroblastos/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Camundongos Nus , Neoplasias Experimentais , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo
4.
Cancer Manag Res ; 12: 11701-11712, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33235500

RESUMO

BACKGROUND: Prostate cancer (PCa) is a major contributor to reduce the life quality of males. Circular RNAs were frequently reported to be associated with cancers. In the case of radiotherapy to PCa, the role of circ_0062020 was still inconclusive, which was further explored in this study. METHODS: Quantitative real-time polymerase chain reaction (qRT-PCR) was used to determine the expression of circ_0062020, miR-615-5p and thyroid hormone receptor interactor 13 (TRIP13) in PCa tissues and cells, as well as in normal tissues and cell. Meanwhile, the proliferation of PCa cells was evaluated by clone formation assay and cell counting kit 8 (CCK8) assay. Moreover, the metastasis of PCa cells was assessed by transwell and wound healing assays. Furthermore, the apoptosis of PCa cells was determined by flow cytometry assay. Besides, dual-luciferase reporter system was applied to verify the correlation between miR-615-5p and circ_0062020 or TRIP13, which was predicted by online tool CircRNA interactome or TargetScan. In addition, the protein expression of TRIP13 was measured by Western blot in PCa tissues and cells and normal tissues and cells. Finally, xenograft tumor assay was performed to further confirming the function of circ_0062020 in PCa in vivo. RESULTS: Circ_0062020 and TRIP13 were upregulated, while miR-615-5p was downregulated in PCa tissues and cells. Circ_0062020 knockdown or miR-615-5p overexpression inhibited the proliferation and metastasis, and promoted apoptosis, which could be reversed by miR-615-5p inhibitor or pc-TRIP13 in ionizing radiation (IR)-treated PCa cells. As expected, circ_0062020 sponged miR-615-5p to regulate TRIP13 expression in PCa cells. Circ_0062020 knockdown also suppressed PCa tumor growth in vivo. CONCLUSION: Circ_0062020 suppressed the radiosensitivity by miR-615-5p/TRIP13 axis in PCa cells, which might provide insights into the radiotherapy for PCa.

5.
Onco Targets Ther ; 12: 3363-3372, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31118688

RESUMO

Objective: Circular RNA is a type of endogenous RNA molecule with a stable closed-loop structure which is ubiquitous in mammals. Circular RNA HIPK3 (circHIPK3) is highly expressed in hepatocellular carcinoma and promotes the growth of hepatoma cells. However, its role in prostate cancer (PCa) has not been reported. This study aims to explore whether circHIPK3 could affect the proliferative and invasive potentials of PCa cells by regulating miRNA-338-3p. Methods: Expression levels of circHIPK3 and miRNA-338-3p in PCa tissues and cells were determined by RT-qPCR. The regulatory effects of circHIPK3 and miRNA-338-3p on proliferative and invasive potentials of PCa cells were evaluated by CCK-8 and transwell assay, respectively. We verified the binding between miRNA-338-3p and ADAM17, as well as miRNA-338-3p and circHIPK3 through dual-luciferase reporter gene assay. Rescue experiments were conducted to clarify whether circHIPK3 affected the proliferative and invasive potentials of PCa cells by regulating miRNA-338-3p. Results: Expression level of circHIPK3 in PCa tissues was remarkably higher than that of paracancerous tissues. Knockdown of circHIPK3 inhibited the proliferative and invasive rates of PC-3 and DU145 cells. Dual-luciferase reporter gene assay indicated that circHIPK3 could bind to miRNA-338-3p. Moreover, miRNA-338-3p expression was downregulated in PCa tissues. miRNA-338-3p expression was negatively correlated with lymph node metastasis and distant metastasis. miRNA-338-3p overexpression markedly reduced proliferative and invasive abilities of PC-3 and DU145 cells. Furthermore, ADAM17 was confirmed to be the target gene of miRNA-338-3p. Overexpression of ADAM17 enhanced proliferative and invasive abilities of PC-3 and DU145 cells. Finally, rescue experiments indicated that miRNA-338-3p knockdown in PC-3 and DU145 cells partially reversed the regulatory effects of circHIPK3 on proliferative and invasive potentials. Conclusion: Overexpression of circHIPK3 promotes the proliferative and invasive potentials of PCa cells through sponging miRNA-338-3p to regulate ADAM17 expression, thus accelerating the malignant progression of PCa.

6.
Medicine (Baltimore) ; 97(34): e11994, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30142837

RESUMO

RATIONALE: Inflammatory myofibroblastic tumor (IMT) is uncommon, coexistence of IMTs in the kidney and abdominal wall are more uncommon. PATIENT CONCERNS AND DIAGNOSIS: We report a 74-year-old female who presented with 6 months history of left flank pain and approximately 5 kg weight loss that were diagnosed as renal cell carcinoma and locally metastatic abdominal wall tumor. INTERVENTIONS AND OUTCOMES: A left radical nephrectomy and excision of the abdominal wall tumor were done. The pathologic result was IMTs. After follow-up for 66 months, the patient showed no signs of tumor recurrence. LESSONS: Coexistence of IMTs in the kidney and abdominal wall is extremely rare and is often diagnosed as malignancy. Therefore, IMTs should be considered in the diagnosis of the patient with both kidney and abdominal wall tumors.


Assuntos
Neoplasias Abdominais/diagnóstico , Carcinoma de Células Renais/diagnóstico , Neoplasias Renais/diagnóstico , Miofibromatose/diagnóstico , Parede Abdominal/patologia , Idoso , Diagnóstico Diferencial , Feminino , Humanos , Rim/patologia
7.
Exp Ther Med ; 14(4): 3221-3228, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28912872

RESUMO

Lenvatinib is an oral, multi-targeted tyrosine kinase inhibitor of vascular endothelial growth factor receptors 1-3, fibroblast growth factor receptors 1-4, platelet-derived growth factor receptor ß, RET and KIT. Cellular immunotherapy has the potential to be a highly targeted treatment, with low toxicity to normal tissues and a high capacity to eradicate tumor tissue. The present study assessed the safety, maximum tolerated dose (MTD) and preliminary antitumor activity of lenvatinib and cellular immunotherapy in a murine model of renal cell carcinoma (RCC). The present study used a therapeutic dose of 0.12 mg lenvatinib and/or 104 rat uterine cancer adenocarcinoma (RuCa)-sensitized lymphocytes administered once daily continuously in 7-day cycles. Tumor regression was observed in mice with RCC following treatment with lenvatinib and 104 RuCa-sensitized lymphocytes. MTD was established as once daily administration of 0.18 mg lenvatinib and 106 RuCa-sensitized lymphocytes. The most common treatment-related adverse effects observed were fatigue (40%), mucosal inflammation (30%), proteinuria, diarrhea, vomiting, hypertension and nausea (all 40%). Combination therapy using lenvatinib and cellular immunotherapy enhanced the antitumor effect induced by single treatments and prolonged the survival of mice with RCC compared with either of the single treatments. Treatment with lenvatinib (0.12 mg) combined with 104 RuCa-sensitized lymphocytes was associated with manageable toxicity consistent with individual agents. Further evaluation of this combination therapy in mice with advanced RCC is required. In conclusion, cellular immunotherapy and oncolytic therapy for cancer may be improved by the synergistic effects of lenvatinib and sensitized lymphocytes. In the present study, the inherent antineoplastic and immune stimulatory properties of the two agents were enhanced when used in combination, which may provide a basis for clinical treatment of patients with RCC.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...