Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Publication year range
1.
J Int Med Res ; 49(8): 3000605211037495, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34407684

ABSTRACT

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.


Subject(s)
Acute Kidney Injury , MicroRNAs , RNA, Long Noncoding , Sepsis , Acute Kidney Injury/chemically induced , Acute Kidney Injury/genetics , Animals , Carrier Proteins , Humans , Lipopolysaccharides/toxicity , Mice , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Sepsis/chemically induced , Sepsis/genetics , Thioredoxins
2.
Exp Ther Med ; 2(5): 969-975, 2011 Sep.
Article in English | MEDLINE | ID: mdl-22977607

ABSTRACT

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.

3.
Nan Fang Yi Ke Da Xue Xue Bao ; 30(1): 149-52, 2010 Jan.
Article in Chinese | MEDLINE | ID: mdl-20118008

ABSTRACT

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.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Cell Proliferation/drug effects , Deoxycytidine/analogs & derivatives , Glutamates/pharmacology , Guanine/analogs & derivatives , Pancreatic Neoplasms/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Deoxycytidine/pharmacology , Dose-Response Relationship, Drug , Drug Synergism , Guanine/pharmacology , Humans , Pemetrexed , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL
...