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.
Cytometry A ; 103(9): 723-731, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37276218

RESUMO

Breast cancer is the most common cancer, and triple-negative breast cancer (TNBC) has the highest metastasis and mortality rate among all breast cancer subtypes. Rujifang is a traditional Chinese medicine formula with many years of clinical application in breast cancer treatment. Here, we aim to investigate the effects of Rujifang on circulating tumor cell (CTC) dynamics and the tumor microenvironment in a ZsGreen/luciferase double-labeled TNBC orthotopic model. We report that the number of CTCs monitored by in vivo flow cytometry (IVFC) strongly correlates with disease progression. Rujifang treatment decreased the number of CTCs and suppressed the distant metastasis of TNBC. Moreover, immunofluorescence analysis revealed that Rujifang treatment could affect the tumor microenvironment by downregulating Kindlin-1, which has been reported to promote metastasis of TNBC. Our study provides evidence of the anti-metastatic effect of Rujifang against TNBC in an animal model using fluorescent cell lines. The results suggest the potential therapeutic value of Rujifang as an anti-metastatic drug, however, further clinical trials are needed to validate these findings in humans.


Assuntos
Células Neoplásicas Circulantes , Neoplasias de Mama Triplo Negativas , Animais , Humanos , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/patologia , Citometria de Fluxo , Células Neoplásicas Circulantes/patologia , Linhagem Celular Tumoral , Proliferação de Células , Microambiente Tumoral
2.
Bioengineered ; 12(2): 11369-11375, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34872438

RESUMO

Long noncoding RNAs (lncRNAs) Colorectal Neoplasia Differentially Expressed (CRNDE) and taurine-upregulated gene 1 (TUG1) play similar roles in sepsis, indicating the existence of the crosstalk between them. Sepsis is a major cause of injuries in heart, which are related to high mortality rates. This study was therefore carried out to analyze the potential crosstalk between CRNDE and TUG1 in sepsis, with a focus on sepsis-induced cell apoptosis in heart. Expression of CRNDE and TUG1 was analyzed with RT-qPCR. Correlations between them were analyzed by Pearson's correlation coefficient. CRNDE and TUG1 were overexpressed in cardiomyocytes to determine the relationship between them. The roles of CRNDE and TUG1 in regulating the apoptosis of cardiomyocytes were explored by cell apoptosis assay. We found that both CRNDE and TUG1 were downregulated in sepsis. In cardiomyocytes, LPS treatment resulted in the downregulation of CRNDE and TUG1. Overexpression of CRNDE and TUG1 in cardiomyocytes increased the expression levels of each other. Under lipopolysaccharide (LPS) treatment, decreased apoptosis rates of cardiomyocytes were observed after CRNDE and TUG1 overexpression. CRNDE and TUG1 co-overexpression showed a stronger effect. In conclusion, CRNDE and TUG1 are downregulated in sepsis and they positively regulate each other to suppress the apoptosis of cardiomyocytes.


Assuntos
Apoptose/genética , Regulação para Baixo/genética , Miócitos Cardíacos/patologia , RNA Longo não Codificante/genética , Sepse/genética , Adulto , Idoso , Feminino , Humanos , Lipopolissacarídeos , Masculino , Pessoa de Meia-Idade , Miócitos Cardíacos/metabolismo , RNA Longo não Codificante/metabolismo
3.
J Int Med Res ; 49(8): 3000605211037495, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34407684

RESUMO

OBJECTIVE: This study aimed to clarify the mechanism by which the long non-coding RNA cancer susceptibility candidate 9 (CASC9) alleviates sepsis-related acute kidney injury (S-AKI). METHODS: A lipopolysaccharide (LPS)-induced AKI model was established to simulate S-AKI. HK-2 human renal tubular epithelial cells were treated with LPS to establish an in vitro model, and mice were intraperitoneally injected with LPS to generate an in vivo model. Subsequently, the mRNA expression of inflammatory and antioxidant factors was validated by quantitative reverse transcription polymerase chain reaction (RT-qPCR). Reactive oxygen species (ROS) production was assessed using an assay kit. Apoptosis was detected by western blotting and fluorescence-activated cell sorting. RESULTS: CASC9 was significantly downregulated in the LPS-induced AKI model. CASC9 attenuated cell inflammation and apoptosis and enhanced the antioxidant capacity of cells. Regarding the mechanism, miR-424-5p was identified as the downstream target of CASC9, and the interaction between CASC9 and miR-424-5p promoted thioredoxin-interacting protein (TXNIP) expression. CONCLUSIONS: CASC9 alleviates LPS-induced AKI in vivo and in vitro, and CASC9 directly targets miR-424-5p and further promotes the expression of TXNIP. We have provided a possible reference strategy for the treatment of S-AKI.


Assuntos
Injúria Renal Aguda , MicroRNAs , RNA Longo não Codificante , Sepse , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/genética , Animais , Proteínas de Transporte , Humanos , Lipopolissacarídeos/toxicidade , Camundongos , MicroRNAs/genética , RNA Longo não Codificante/genética , Sepse/induzido quimicamente , Sepse/genética , Tiorredoxinas
4.
Cell Cycle ; 18(16): 1948-1964, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31234706

RESUMO

Recently, MicroRNA-98 (miR-98) works as a biomarker in some diseases, such as lung cancer, Schizophrenia, and breast cancer, but there still lack of studies on the function of miR-98 during sepsis. Thus, our study is conducted to figure out the function of miR-98 for the regulation of cardiac dysfunction, liver and lung injury in sepsis mice. Cecum ligation and puncture was used to establish the sepsis mice model. Next, miR-Con and agomiR-98 were injected into the tail vein of mice 48 h after modeling. Then, expression of miR-98, HMGA2, NF-κB, inflammatory factors, apoptosis-related proteins in myocardial, liver and lung tissues of septic mice were determined. Moreover, other indices that were associated with cardiac dysfunction, liver and lung injury in septic mice were detected. Finally, bioinformatics analysis and luciferase activity assay were utilized to validate the binding site between miR-98 and HMGA2. miR-98 was poorly expressed, while HMGA2, NF-κB pathway-related proteins were highly expressed in myocardial, liver, and lung tissues of mice with sepsis. Upregulated miR-98 inhibited HMGA2, NF-κB, TNF-α, IL-6, Bcl-2 and increased IL-10, Cleaved caspase-3 and Bax expression in myocardial, liver, and lung tissues of septic mice. Upregulation of miR-98 decreased LVEDP, CTn-I, BNP, ALT, AST, TBIL, LDH, and PaCO2 while increased +dp/dt max, -dp/dt max, pH and PaO2 in sepsis mice. miR-98 was a direct target gene of HMGA2. Our study provides evidence that miR-98 protects sepsis mice from cardiac dysfunction, liver and lung injury by negatively mediating HMGA2 via the inhibition of the NF-κB signaling pathway.


Assuntos
Proteína HMGA2/metabolismo , Fígado/metabolismo , Pulmão/metabolismo , MicroRNAs/metabolismo , Miócitos Cardíacos/metabolismo , NF-kappa B/metabolismo , Sepse/metabolismo , Animais , Apoptose/genética , Modelos Animais de Doenças , Células HEK293 , Proteína HMGA2/genética , Humanos , Lesão Pulmonar/diagnóstico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Sepse/patologia , Transdução de Sinais/genética , Transfecção
5.
Exp Ther Med ; 2(5): 969-975, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22977607

RESUMO

Previous studies have shown that both pemetrexed, a cytotoxic drug, and erlotinib, an epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI), inhibit the cell growth of pancreatic cancer cells. However, whether they exert a synergistic antitumor effect on pancreatic cancer cells remains unknown. The present study aimed to assess the synergistic effect of erlotinib in combination with pemetrexed using different sequential administration schedules on the proliferation of human pancreatic cancer BXPC-3 and PANC-1 cells and to probe its cellular mechanism. The EGFR and K-ras gene mutation status was examined by quantitative PCR high-resolution melting (qPCR-HRM) analysis. BXPC-3 and PANC-1 cells were incubated with pemetrexed and erlotinib using different administration schedules. MTT assay was used to determine cytotoxicity, and cell cycle distribution was determined by flow cytometry. The expression and phosphorylation of EGFR, HER3, AKT and MET were determined using Western blotting. Both pemetrexed and erlotinib inhibited the proliferation of BXPC-3 and PANC-1 cells in a dose- and time-dependent manner in vitro. Synergistic effects on cell proliferation were observed when pemetrexed was used in combination with erlotinib. The degree of the synergistic effects depended on the administration sequence, which was most obvious when erlotinib was sequentially administered at 24-h interval following pemetrexed. Cell cycle studies revealed that pemetrexed induced S arrest and erlotinib induced G(0)/G(1) arrest. The sequential administration of erlotinib following pemetrexed induced S arrest. Western blot analyses showed that pemetrexed increased and erlotinib decreased the phosphorylation of EGFR, HER3 and AKT, respectively. However, both pemetrexed and erlotinib exerted no significant effects on the phosphorylation of c-MET. The phosphorylation of EGFR, HER3 and AKT was significantly suppressed by scheduled incubation with pemetrexed followed by erlotinib, but not by concomitant or sequential incubation with erlotinib followed by pemetrexed. In summary, our results demonstrated that the combined use of erlotinib and pemetrexed exhibited a strong synergism in BXPC-3 and PANC-1 cells. The inhibitory effects were strongest after sequential administration of pemetrexed followed by erlotinib. The synergistic effects may be related to activation of the EGFR/HER3/AKT pathway induced by pemetrexed.

6.
Nan Fang Yi Ke Da Xue Xue Bao ; 30(1): 149-52, 2010 Jan.
Artigo em Chinês | MEDLINE | ID: mdl-20118008

RESUMO

OBJECTIVE: To investigate the sequence-dependent effect of combined use of gemcitabine and pemetrexed on the proliferation of human pancreatic carcinoma cell lines BXPC-3 and PANC-1 in vitro and explore the cellular mechanism. METHODS: MTT assay was used to determine the proliferation of the two cells after addition of the two drugs in different sequences, and the cell cycle changes were analyzed by flow cytometry. RESULTS: Both gemcitabine (10(-7)-10 mg/ml) and pemetrexed (10(-7)-10 mg/ml) significantly inhibited the proliferation of BXPC-3 and PANC-1 cells in a dose- and time-dependent manner. The effect of combined administration of gemcitabine and pemetrexed on the cell proliferation varied with the order of the drug delivery, and addition of gemcitabine 24 h after pemetrexed administration produced a significant enhancement of the inhibitory effect as compared with simultaneous drug administration (P<0.05) or the administration of the two drugs in a reverse order (P<0.05). Compared with the control group, combined administration of gemcitabine and pemetrexed caused obvious cell cycle arrest at G1 and S phases (P<0.05). Simultaneous administration of the two drugs resulted in significantly reduced G2-phase cells (P<0.05); addition of gemcitabine prior to pemetrexed caused cell cycle arrest in G1 phase (P<0.05), while the reverse caused cell cycle in S phase (P<0.05). CONCLUSION: Both gemcitabine and pemetrexed can inhibit the proliferation of BXPC-3 and PANC-1 cells, and their synergetic effect depends on the sequence of their administration. The sequential administration of pemetrexed followed by gemcitabine produces significant synergetic effects against the cell proliferation, which might not be associated with their influence of the cell cycle.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Desoxicitidina/análogos & derivados , Glutamatos/farmacologia , Guanina/análogos & derivados , Neoplasias Pancreáticas/patologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Desoxicitidina/farmacologia , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Guanina/farmacologia , Humanos , Pemetrexede , Gencitabina
7.
Appetite ; 52(1): 8-14, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18657581

RESUMO

The validity of the Child Feeding Questionnaire (CFQ), one of the measures used to assess parental child feeding practices and attitudes, has been confirmed in American populations. We used confirmatory factor analysis to test the validity and factor structure of the translated version of the CFQ among parents of Japanese elementary school children. The structural equation modelling software Linear Structural Relationships (LISREL) was applied to explore the validity of the translated CFQ to examine child feeding behaviours and attitudes in a sample of 920 parents of Japanese elementary schoolchildren from schools in Koshu City in Yamanashi prefecture (grades 4-6), Japan. The confirmatory factor analysis suggested that after dropping the items with a low factor loading and adding three error covariances between items, the 7-factor model displayed acceptable fit and most items loaded as expected. Of the 24 direct factor-item correlations, 22 were greater than 0.50. Our study confirmed the validity of the translated CFQ to assess child feeding practices and attitudes among parents of elementary schoolchildren brought up in a Japanese eating culture.


Assuntos
Dieta , Inquéritos e Questionários , Antropometria , Índice de Massa Corporal , Criança , Fenômenos Fisiológicos da Nutrição Infantil , Dieta/psicologia , Feminino , Conhecimentos, Atitudes e Prática em Saúde , Humanos , Japão/epidemiologia , Masculino , Modelos Estatísticos , Obesidade/epidemiologia , Reprodutibilidade dos Testes , Instituições Acadêmicas , Software
8.
Zhongguo Fei Ai Za Zhi ; 11(3): 339-44, 2008 Jun 20.
Artigo em Chinês | MEDLINE | ID: mdl-20731930

RESUMO

BACKGROUND: Previous clinical trials showed that there was no clinical benefit in the treatment of advanced non-small cell lung cancer when chemotherapy combined with gefitinib. The present study aims to assess the sequential administration of paclitaxel and gefitinib on the cell proliferation of lung adenocarcinoma cell SPC-A1 and to explore its mechanism by observing their effects on the cell cycle. METHODS: The expression of EGFR mRNA and EGFR protein were examined by RT-PCR and western blotting respectively. MTT was used to measure the cell proliferation of SPC-A1 cells. Cell cycle was detected by flow cytometry. RESULTS: Both EGFR mRNA and EGFR protein were overexpressed in SPC-A1 cells. From 1*10(-14) M to 1*10(-6) M, both paclitaxel and gefitinib inhibited the cell proliferation of SPC-A1 cells in a dose-dependent and time-dependent manner in vitro . The effects of paclitaxel in combination with gefitinib on cell proliferation depended on the sequence. No significant additive effects on cell proliferation was found when they were used simultaneously or gefitinib was added before paclitaxel. However, sequential administration of gefitinib following paclitaxel can remarkably enhanced the effect of paclitaxel on the cell proliferation of SPC-A1 cells. Cell cycle studies showed that paclitaxel and gefitinib induced G2/M and G0/G1 arrest respectively. The G0/G1 arrest was observed when paclitaxel and gefitinib was used simultaneously or gefitinib was added before paclitaxel. In contrast, sequential administration of gefitinib following paclitaxel induced G2/M arrest. CONCLUSIONS: Both paclitaxel and gefitinib inhibits the cell proliferation of SPC-A1 cells. The additive effects on cell proliferation are sequential-dependent. The concomitant and the sequential treatment of gefitinib followed by paclitaxel exert no significant additive effects on the cell proliferation and resulted in the accumulation of cells in G0/G1 phase, which may decrease the effectiveness of paclitaxel in subsequent cycles. The additive effected on the cell proliferation are observed only when gefitinib is sequentially administrated following paclitaxel, which results in the G2/M arrest. The increase in G2/M phase suggests that cell cycle effects might not explain the observed additive effects.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...