Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
PLoS Pathog ; 17(9): e1009901, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34506605

RESUMO

Neddylation, an important type of post-translational modification, has been implicated in innate and adapted immunity. But the role of neddylation in innate immune response against RNA viruses remains elusive. Here we report that neddylation promotes RNA virus-induced type I IFN production, especially IFN-α. More importantly, myeloid deficiency of UBA3 or NEDD8 renders mice less resistant to RNA virus infection. Neddylation is essential for RNA virus-triggered activation of Ifna gene promoters. Further exploration has revealed that mammalian IRF7undergoes neddylation, which is enhanced after RNA virus infection. Even though neddylation blockade does not hinder RNA virus-triggered IRF7 expression, IRF7 mutant defective in neddylation exhibits reduced ability to activate Ifna gene promoters. Neddylation blockade impedes RNA virus-induced IRF7 nuclear translocation without hindering its phosphorylation and dimerization with IRF3. By contrast, IRF7 mutant defective in neddylation shows enhanced dimerization with IRF5, an Ifna repressor when interacting with IRF7. In conclusion, our data demonstrate that myeloid neddylation contributes to host anti-viral innate immunity through targeting IRF7 and promoting its transcriptional activity.


Assuntos
Imunidade Inata/imunologia , Fator Regulador 7 de Interferon/imunologia , Células Mieloides/imunologia , Infecções por Vírus de RNA/imunologia , Vírus de RNA/imunologia , Animais , Fator Regulador 7 de Interferon/biossíntese , Camundongos , Células Mieloides/metabolismo , Proteína NEDD8/deficiência , Processamento de Proteína Pós-Traducional , Ubiquitinas/deficiência
2.
World J Surg Oncol ; 20(1): 322, 2022 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-36171617

RESUMO

BACKGROUND: Soft tissue sarcoma (STS) is a malignant tumor of highly heterogeneous mesenchymal origin. STS has a biological pattern and clinical transformation with localized invasive growth and is susceptible to hematogenous metastasis. Local therapeutic strategies may treat recurrent and oligometastatic STS, including surgery and radiation therapy. This study aimed to evaluate the safety and efficacy of stereotactic body radiotherapy (SBRT) for recurrent and oligometastatic STS. METHODS: We retrospectively analyzed 37 recurrent and oligometastatic STS patients with 58 lesions treated with SBRT from 2009 to 2019 at our institution. Oligometastatic is defined as metastatic lesions less than or equal to 3. The primary endpoint was local control (LC); secondary endpoints were survival and toxicity. RESULTS: The median follow-up was 21.0 months (3.0 to 125.0 months). Among 37 patients, 18 were recurrent patients, and 19 were oligometastatic patients. Median LC was 25.0 months (95% CI 20.0-45.0). The 1-, 2-, and 3-year LC rates were 80.2%, 58.3%, and 46.6%, respectively. Median overall survival (OS) was 24.0 months (95% CI 13.0-28.0), and the survival rates after SBRT were 71.5%, 40.0%, and 29.1% at 1, 2, and 3-year, respectively. Median progression-free survival (PFS) was 10.0 months (95% CI 8.0-15.0 months), PFS rate after SBRT was 43.6%, 26.8%, and 18.4% at 1, 2, and 3 years, respectively. Late grade 3 radiation dermatitis was observed in one patient (2.7%). Using univariate and multivariate COX analysis, better OS, PFS, and LC were obtained in the histologic grade 1(G1) group, and tumor size and a number of lesions influenced LC. CONCLUSIONS: SBRT is a safe and effective treatment for patients with recurrent and oligometastatic STS. Histological grade influences local control and survival. SBRT may be a promising treatment option for recurrent and oligometastatic STS.


Assuntos
Radiocirurgia , Sarcoma , Neoplasias de Tecidos Moles , Humanos , Metástase Neoplásica , Recidiva Local de Neoplasia/radioterapia , Intervalo Livre de Progressão , Radiocirurgia/efeitos adversos , Estudos Retrospectivos , Sarcoma/patologia , Sarcoma/radioterapia , Neoplasias de Tecidos Moles/patologia , Neoplasias de Tecidos Moles/radioterapia , Resultado do Tratamento
3.
Exp Ther Med ; 26(6): 547, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37928508

RESUMO

[This retracts the article DOI: 10.3892/etm.2020.8540.].

4.
Exp Ther Med ; 19(4): 3081-3089, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32256796

RESUMO

Esophageal squamous cell carcinoma (ESCC) is a common cancer in China and has a high mortality rate. MicroRNAs (miRs) are a family of post-transcriptional regulators, which negatively regulate target gene expression. miR-613 has been revealed to be a diagnostic and prognostic biomarker in ESCC. However, the role of miR-613 in ESCC remains unclear. In the present study, miR-613 expression was identified to be reduced in tumor tissues in comparison with corresponding adjacent normal tissues. TargetScan and a dual-luciferase reporter assay verified glucose-6-phosphate dehydrogenase (G6PD) as a direct target of miR-613. In contrast with miR-613, G6PD expression was increased in tumor tissues compared with matched healthy tissues. Furthermore, overexpression of miR-613 inhibited cell migration and invasion of Eca109 cells compared with controls, while G6PD overexpression reversed the inhibition induced by miR-613, as determined by wound healing and Transwell assays. In addition, miR-613 overexpression decreased the mRNA and protein expression of G6PD, matrix metalloproteinase (MMP)2 and MMP9, and reduced the phosphorylation of signal transducer and activator of transcription 3 (STAT3) compared with controls, while G6PD reversed the effects of miR-613. However, miR-613 and G6PD did not affect the expression of STAT3. In conclusion, the aforementioned results suggest that miR-613 targets G6PD to suppress ESCC cell migration and invasion through reduced MMP2 and MMP9 expression and inactivation of the STAT3 signaling pathway. Thus, the present study may provide a new molecular foundation for treatment of ESCC.

5.
Am J Transl Res ; 12(11): 7223-7235, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33312362

RESUMO

OBJECTIVE: LMNB2 is a protein that belongs to the RAB family. It is correlated with the tumorigenesis and development of several human cancers. The effect of LMNB2 on esophageal cancer (EC) has not yet been reported. The previous study showed that lncRNA ROR could promote the proliferation of EC. The current study aimed at exploring the correlation between ROR with LMNB2 and the role of ROR and LMNB2 in proliferation and migration of EC. METHODS: This study performed dual luciferase reporter assay to evaluate the binding between miR-145 and ROR as well as miR-145 and LMNB2. Gene expression in EC tissues and cells were detected using quantitative real-time PCR (qRT-PCR) assay. The effect of ROR or miR-145 on LMNB2 expression was detected using western blot (WB) assay. Cells proliferation was detected by CCK8 and clone formation assay. Transwell and wound healing assay were carried out to determine the cells migration. Mouse xenograft assay was performed to detect the effect of LMNB2 on tumor growth in vivo. RESULTS: This study demonstrated that miR-145 directly targets ROR and LMNB. ROR and LMNB2 were up-regulated and miR-145 was down-regulated in EC tissues and cells. The proliferation and migration of EC cells were promoted by overexpression of of ROR or LMNB2. MiR-145 was capable of reversing the effect of ROR. The results also determined that down-regulation of LMNB2 had inhibitory effects and up-regulation of LMNB2 had catalytic effects on tumor growth in vivo. CONCLUSION: LMNB2 which is regulated by ROR and miR-145 was highly expressed in EC and promoted the proliferation and migration of EC in vitro and in vivo. The study suggests that ROR and LMNB2 could be potentially the therapeutic targets of EC.

6.
Drug Deliv ; 24(1): 10-19, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28155336

RESUMO

Multidrug resistance (MDR) is the major underlying cause of the low 5-year survival rate of esophageal carcinoma. In this study, we developed a novel microemulsion system (SD-ME) co-loaded with docetaxel (DTX) and Schizandrin A, a potent chemotherapeutic agent and a potential drug resistance modulator, respectively. In the physicochemical characterization studies, SD-ME displayed a well-defined spherical shape and size (56.62 ± 4.16 nm), a narrow polydispersity index (PDI, 0.132 ± 0.002), and a negative surface charge (-19.81 ± 3.11 mv). In the cellular uptake studies, SD-ME with a DTX concentration of 30 µg/mL exhibited a 3.9-fold enhancement of DTX internalization in DTX-resistant EC109 (EC109/DDR) cells in comparison to that observed for EC109 cells, and the mechanisms were associated with reducing P-gp expression and inhibiting P-gp ATPease. The half-maximal inhibitory concentrations (IC50) of DTX and SD-ME against EC109/DDR cells were 40.57 ± 0.39 and 3.59 ± 0.06 µg/mL, respectively. Likewise, the apoptotic rate of EC109/DDR treated with SD-ME increased up to 20-fold compared to that observed with free DTX. In anticancer efficacy studies in vivo, SD-ME markedly retarded the tumor growth of nude mice bearing EC109/DDR tumor xenografts compared with D-ME and free DTX throughout the duration of study. Consequently, mice treated with SD-ME had the highest survival rate (37.5%) during the observation period (70 days). In addition, there were no apparent side effects after the administration of SD-ME. Overall, our study provides evidence for SD-ME as an effective drug delivery system for enhanced MDR tumor treatment.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Carcinoma de Células Escamosas/tratamento farmacológico , Ciclo-Octanos/administração & dosagem , Resistência a Múltiplos Medicamentos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Neoplasias Esofágicas/tratamento farmacológico , Lignanas/administração & dosagem , Compostos Policíclicos/administração & dosagem , Taxoides/administração & dosagem , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/antagonistas & inibidores , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/química , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Apoptose/efeitos dos fármacos , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Ciclo-Octanos/química , Ciclo-Octanos/farmacocinética , Docetaxel , Relação Dose-Resposta a Droga , Combinação de Medicamentos , Composição de Medicamentos , Emulsões , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patologia , Humanos , Concentração Inibidora 50 , Lignanas/química , Lignanas/farmacocinética , Camundongos Endogâmicos BALB C , Camundongos Nus , Tamanho da Partícula , Compostos Policíclicos/química , Compostos Policíclicos/farmacocinética , Solubilidade , Propriedades de Superfície , Taxoides/química , Taxoides/farmacocinética , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA