Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biotechnol Lett ; 42(12): 2537-2549, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33009634

RESUMO

OBJECTIVES: Esophageal cancer is one of the malignant tumor with poor survival. The 5-year survival rate of esophageal cancer patients remains poor due to limited therapeutic options and the development of drug-resistance. Recent evidence suggests that long non-coding RNAs (lncRNAs) are involved in occurrence and development of tumor, however, the molecular mechanisms of lncRNA taurine-upregulated gene 1 (TUG1) in esophageal cancer remain unknown. RESULTS: TUG1 was overexpressed in esophageal cancer tissues and cells. The knockdown of TUG1 repressed proliferation and invasion, while promoted apoptosis of esophageal cancer cells by negatively regulating miR-1294 expression. Furthermore, PLK1 was a target mRNA of miR-1294 in esophageal cancer cells. Therefore, the effects of PLK1 silencing on proliferation, apoptosis, and invasion of esophageal cancer cells could be overturned by silencing miR-1294. Additionally, TUG1 silencing inhibited growth of tumor cells in vivo. CONCLUSIONS: TUG1 was found as oncogenic gene in esophageal cancer. Mechanically, TUG1 attributed to esophageal cancer process by regulating miR-1294/ PLK1 axis.


Assuntos
Proteínas de Ciclo Celular/genética , Neoplasias Esofágicas/genética , MicroRNAs/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/genética , RNA Longo não Codificante/genética , Apoptose/genética , Biomarcadores Tumorais/genética , Carcinogênese/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Progressão da Doença , Neoplasias Esofágicas/patologia , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Masculino , Ativação Transcricional/genética , Quinase 1 Polo-Like
2.
Biomater Sci ; 8(17): 4767-4778, 2020 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-32724941

RESUMO

Multidrug resistance (MDR) remains a serious impediment to successful tumor chemotherapy. Despite considerable efforts to address MDR, limited approaches have been successful in the clinic to date. Here, we have developed pH/redox cascade-sensitive multiscale nanoparticles (DMA-NPs) with size- and charge-changeable properties for the efficient delivery of a non-P-glycoprotein substrate anticancer drug (podophyllotoxin, PPT) to combat MDR. DMA-NPs are composed of a charge-reversible polymer (PEG-PAH-DMA) shell and a redox-sensitive small-sized dendrimeric PPT-prodrug (PAMAM-ss-PPT) core. The PEG-PAH-DMA polymer shell on DMA-NPs maintains a negative charge in a normal environment, which reverts to a positive charge in a mildly acidic tumor environment (pH 6.5), leading to the release of positive PAMAM-ss-PPT via electrostatic repulsion. PAMAM-ss-PPT completely releases PPT under elevated intracellular glutathione (GSH) conditions in tumors. Several properties facilitate the hierarchical transport of DMA-NPs across multiple drug resistance pathological obstacles, including long blood circulation times, significant accumulation in tumors, deep tumor permeation, cancer cell internalization, and rapid and complete drug release. Experimental evaluations, both in vitro and in vivo, collectively indicate that nanomedicines can effectively penetrate xenografted A549 paclitaxel-resistant lung cancer cells and inhibit tumor proliferation with negligible toxicity. The current study presents a novel nanoparticle-based therapeutic strategy aimed at overcoming MDR.


Assuntos
Sistemas de Liberação de Medicamentos , Nanopartículas , Resistência a Múltiplos Medicamentos , Concentração de Íons de Hidrogênio , Oxirredução
3.
Oncol Lett ; 20(1): 751-757, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32566001

RESUMO

miR-203 has been indicated to be a tumor suppressor in esophageal cancer, however, the underlying molecular mechanisms by which it functions are not fully understood. The present study aimed to investigate the molecular mechanisms underlying the regulatory activities of microRNA (miR)-203 in esophageal cancer. The miR-203 mimic/inhibitor, Mitogen-Activated Protein Kinase Kinase Kinase 1 (MAP3K1) overexpression plasmid and MAP3K1 small interfering (si)RNA were transfected into TE-1 cells. miR-203 and MAP3K1 mRNA expression were detected via reverse transcription-quantitative PCR analysis, while MAP3K1 protein expression was detected via western blot analysis. Dual-luciferase reporter assay was used to determine whether MAP3K1 was a direct target of miR-203. Cell proliferation and invasion abilities were assessed via MTT and Matrigel assays, respectively. Cell apoptosis was analyzed via flow cytometry, Caspase 8/3 Assay kits and western blot analysis. The results demonstrated that MAP3K1 was a direct target of miR-203. Overexpression of MAP3K1 reversed the suppressed cell proliferation and invasion abilities induced by miR-203 mimic, as well as the inhibitory effect of miR-203 mimic on cell apoptosis. Furthermore, MAP3K1 siRNA weakened the effect of miR-203 inhibitor on cell proliferation, apoptosis and invasion.

4.
J Exp Clin Cancer Res ; 38(1): 369, 2019 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-31438989

RESUMO

BACKGROUND: Esophageal squamous cell carcinoma (ESCC) is one of the most common cancers worldwide, which lacks effective biomarkers for prognosis. Therefore, it is urgent to explore new potential molecular markers to discriminate patients with poorer survival in ESCC. METHODS: Bioinformatics analysis, qRT-PCR, and western blot were applied to investigate S1PR1 expression. CCK-8 assay, colony formation assay, flow cytometry dual staining assay, and immunofluorescence were performed to examine cell proliferation ability and apoptosis rate. Mouse xenograft model of TE-13 cells was established to confirm the roles of S1PR1 in vivo. Gene set enrichment analysis (GSEA) was used to investigate the downstream signaling pathways related to S1PR1 functions. Co-IP was performed to verify the direct binding of S1PR1 and STAT3. Western blot was applied to determine the phosphorylation level of STAT3. Immunohistochemistry was conducted to identify protein expression of S1PR1 and p- STAT3 in tumor tissues. RESULTS: In the present study, we found that S1PR1 expression was higher in ESCC patients and was a potential biomarker for poor prognosis. Silencing S1PR1 expression inhibited proliferation, and increased apoptosis of ESCC cells, while overexpression of S1PR1 had opposite effects. Mechanistically, S1PR1 played the roles of promoting proliferation and attenuating apoptosis through directly activating p-STAT3. Furthermore, in vivo experiments verified this mechanism. CONCLUSION: Our findings indicated that S1PR1 enhanced proliferation and inhibited apoptosis of ESCC cells by activating STAT3 signaling pathway. S1PR1 may serve as a prognostic biomarker for clinical applications.


Assuntos
Apoptose , Biomarcadores Tumorais/metabolismo , Proliferação de Células , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago/patologia , Fator de Transcrição STAT3/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo , Animais , Biomarcadores Tumorais/genética , Movimento Celular , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas do Esôfago/genética , Carcinoma de Células Escamosas do Esôfago/metabolismo , Feminino , Seguimentos , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Prognóstico , Fator de Transcrição STAT3/genética , Transdução de Sinais , Receptores de Esfingosina-1-Fosfato/genética , Taxa de Sobrevida , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Pathol Res Pract ; 215(9): 152526, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31324391

RESUMO

Increasing evidence indicates that microRNAs may play important roles in tumor development and may take part in different processes in different cancers. miR-429 is known as a cancer suppressor or oncogene that is dysregulated in different malignancies, including esophageal squamous cell carcinoma (ESCC). However, the effect of miR-429 in ESCC has not been fully explored. The purpose of this study was to investigate the functions of miR-429 in ESCC. qRT-PCR assays were performed to detect miR-429 expression in ESCC tissues and cell lines. To assess the effects of miR-429 on ESCC cells, wound healing and transwell assays were used. Luciferase reporter and western blot assays were employed to determine whether Slug is a major target of miR-429.Our results showed that the expression levels of miR-429 in ESCC tissues and cells were lower than in normal esophageal epithelial tissues and cells. Furthermore, overexpression of endogenous miR-429 inhibited the migration and invasion of ESCC cell lines. In addition, Luciferase reporter and western blot assays provided evidence that miR-429 can bind to the 3' untranslated regions of Slug to regulate its expression and that of downstream epithelial-to-mesenchymal transition (EMT) markers. We found that Slug serves as a major target of miR-429. miR-429 plays a vital role in ESCC progression and represents a new therapeutic target for ESCC.


Assuntos
Movimento Celular , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago/patologia , Regulação Neoplásica da Expressão Gênica/genética , MicroRNAs/metabolismo , Fatores de Transcrição da Família Snail/biossíntese , Movimento Celular/genética , Transição Epitelial-Mesenquimal/genética , Neoplasias Esofágicas/genética , Carcinoma de Células Escamosas do Esôfago/genética , Humanos , MicroRNAs/genética , Invasividade Neoplásica/genética
6.
J Environ Manage ; 197: 331-337, 2017 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-28402915

RESUMO

A composite material consisting of nanoscale zerovalent iron particles supported on herb-residue biochar (nZVI/BC) was synthesized and used for treatment of Cr(VI)-contaminated water. The effects of initial pH, chromium concentration, contact time, and competition with coexisting anions and natural organic matter (NOM) were also investigated. nZVI/BC was characterized by X-ray diffraction (XRD), transmission electron microscopy (TEM), and scanning electron microscopy analysis (SEM), and the Brunauer-Emmett-Teller surface area was measured. TEM and X-ray photoelectron spectroscopy (XPS) analysis before and after reaction with Cr(VI) showed that reduction and coprecipitation occurred during hexavalent chromium adsorption. The removal of Cr(VI) was highly pH-dependent and the adsorption kinetics data agreed well with the pseudo-second-order model. The presence of SO42- and humic acid promoted Cr(VI) removal at both low and high concentrations, while the HCO3- inhibited the reaction. These results prove that nZVI/BC can be an effective reagent for removal of Cr(VI) from solutions.


Assuntos
Carvão Vegetal , Cromo , Poluentes Químicos da Água , Ferro , Água
7.
Oncotarget ; 8(8): 13312-13319, 2017 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-28076324

RESUMO

Oxaliplatin (L-OHP) is standard treatment for colorectal cancer. However, resistance to L-OHP often leads to treatment failure or cancer relapse. Understanding of the mechanism underlying L-OHP resistance is important to overcome the resistance and improve colorectal cancer treatment. This study aimed to identify new proteins that mediates L-OHP resistance in colorectal cancer and elucidate their mode of function. HT-29 cells were exposed to gradually increased concentration of L-OHP to select L-OHP resistant HT-29/L-OHP cell line. Proteomic analysis of HT-29 and HT-29/L-OHP cells were performed to identify differentially expressed proteins, including Poly(C)-binding protein 1 (PCBP1). PCBP1 expression level in 20 cases of L-OHP sensitive patients and 20 cases of L-OHP refractory patients was analyzed by immunohistochemistry. Chemoresistance and Akt activation in HT-29 and HT-29/L-OHP cells were analyzed by MTT assay and Western blot analysis. We identified 37 proteins showing differential expression in HT-29/L-OHP and HT-29 cells. In particular, PCBP1 protein level increased 15.6 fold in HT-29/L-OHP cells compared to HT-29 cells. Knockdown of PCBP1 sensitized HT-29/L-OHP and HT-29 cells to L-OHP, while overexpression of PCBP1 increased L-OHP resistance in HT-29 cells. In addition, PCBP1 expression was significantly higher in tumor samples from L-OHP refractory patients than in those from L-OHP responsive patients. Furthermore, we found that knockdown of PCBP1 inhibited the activation of Akt in HT-29/L-OHP and HT-29 cells. In conclusion, our findings suggest that PCBP1 is a molecular marker of L-OHP resistance in colorectal cancer and a promising target for colorectal cancer therapy.


Assuntos
Neoplasias Colorretais/patologia , Resistencia a Medicamentos Antineoplásicos/fisiologia , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Antineoplásicos/farmacologia , Western Blotting , Neoplasias Colorretais/metabolismo , Proteínas de Ligação a DNA , Eletroforese em Gel Bidimensional , Técnicas de Silenciamento de Genes , Células HT29 , Humanos , Imuno-Histoquímica , Compostos Organoplatínicos/farmacologia , Oxaliplatina , Proteínas de Ligação a RNA , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
8.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 21(1): 78-81, 2013 Feb.
Artigo em Chinês | MEDLINE | ID: mdl-23484696

RESUMO

This study was aimed to investigate the effect of homoharringtonine (HHT) on K562 cell proliferation, apoptosis and expression of BCL-2 and NF-κB proteins. The cells proliferation was assayed with MTT method, the cell apoptosis, cell cycle and BCL-2 expression were analyzed with flow cytometry, NF-κB protein expression was detected with Western blot. The results showed that HHT concentration-dependently inhibited proliferation of K562 cells, the IC50 at 48 h was 43.89 ng/ml. Treated with HHT 10 ng/ml for 48 h, K562 cell apoptosis significantly increased, cell cycle was blocked at G0/G1, the expression level of BCL-2 and NF-κB proteins was lower than that in control group (P < 0.05). It is concluded that HHT may inhibit the proliferation of K562 cells, and down-regulating expression levels of BCL-2 and NF-κB may be one of its anti-CML mechanisms.


Assuntos
Harringtoninas/farmacologia , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Citometria de Fluxo , Mepesuccinato de Omacetaxina , Humanos , Células K562
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...