Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add filters








Year range
1.
Chinese Journal of Pathophysiology ; (12): 2195-2201, 2017.
Article in Chinese | WPRIM | ID: wpr-663027

ABSTRACT

AIM:To investigate the effect of dihydroartemisinin ( DHA) adjuvant treatment on enhancing the antitumor effect of 5-fluorouracil (5-FU) against gastric cancer .METHODS:The gastric cancer BGC-823 cells were di-vided into control group , DHA group, 5-FU group, 5-FU+DHA group and 5-FU+DHA+SIRT1 plasmid group.The via-bility of BGC-823 cells treated with DHA and 5-FU was measured by MTT assay .The expression of SIRT1 and NADPH ox-idase, activation of caspase-9 and caspase-3, and phosphorylation of ASK1 and JNK in the BGC-823 cells treated with DHA and 5-FU were determined by Western blot .The production of ROS and the apoptosis of the BGC-823 cells treated with DHA and 5-FU were analyzed by flow cytometry .RESULTS:Dihydroartemisinin significantly inhibited the expression of SIRT1 and increased NADPH oxidase protein level (P<0.05).DHA increased the sensitivity of BGC-823 cells to 5-FU, thus decreasing the IC50 of 5-FU to the gastric cancer cells.However, transfection with SIRT1 plasmid decreased the cytotoxicity of DHA and 5-FU co-treatment to the BGC-823 cells.DHA promoted the production of ROS and phosphoryla-tion of ASK1 and JNK induced by 5-FU in the BGC-823 cells ( P<0.05 ) .However , ROS scavenger N-acetylcysteine ( NAC) or JNK specific inhibitor SP600125 inhibited the cell death and activation of caspase-9 and caspase-3 induced by DHA and 5-FU co-treatment (P<0.05).In addition, NAC significantly inhibited the phosphorylation of JNK in the BGC-823 cells co-treated with DHA and 5-FU.However, treatment with SP600125 did not influence the ROS production in the BGC-823 cells, indicating that JNK was the downstream target of ROS pathway .CONCLUSION: Combination of DHA with 5-FU induces caspase-dependent apoptosis in gastric cancer cells through the SIRT 1/NADPH oxidase/ROS/JNK sig-naling pathway .

2.
Chinese Journal of Nephrology ; (12): 371-377, 2017.
Article in Chinese | WPRIM | ID: wpr-619642

ABSTRACT

Objective To investigate the effect of Sirt1 gene knockout on chronic kidney disease induced by 5/6 nephrectomy in mice and vascular endothelial growth factor (VEGF)/fetal liver kinase-1 (Flk-1) signaling pathway.Methods Twenty four male Sirt1 +/+ and Sirt1 +/-mice wererandomly divided into four groups:Sirt1+/+ mice with sham-operation (WT-Sham,n=6),Sirt1+/-mice with sham-operation (KO-Sham,n=6),Sirt1 +/+ mice with 5/6 nephrectomy (WT-Nx,n=6) and Sirt1 +/-mice with 5/6 nephrectomy (KO-Nx,n=6).Proteinuria was determined by urine collection from 8:00 to 8:00 the next day at 20 weeks.Serum creatinine (Scr),urea nitrogen (BUN) and the renal pathological changes were measured after 20 weeks.Expressions of Sirt1,collagen Ⅰ and transforming growth factor β(TGF-β) were used to analyze the changes of renal fibrosis by immunohistochemistry staining.Real-time PCR and Western blotting were used to measure the mRNA and protein expressions of Sirt1,fibronectin,collagen Ⅰ,VEGF and Flk-1 in kidney.Results Sirt1 expressed in glomernlar endothelial cells,podocytes,mesangial cells and renal tubular epithelial cells in Sirt1 +/+ mice,while Sirt1 expression intensity was significantly reduced in Sirt1 +/-mice.Compared with the WT-Sham group,WT-Nx group had increased proteinuria,BUN,Scr,glomernlar sclerosis index and tubulointerstitial fibrosis index at 12 weeks after operation (all P < 0.01),and KO-Nx group had exacerbated the above up-regulations (all P < 0.01).Compared with those in WT-Sham group,the expressions of fibronectin,collagen Ⅰ and TGF-β were up-regulated in WT-Nx group (all P < 0.01),and were significantly augmented in KO-Nx group (all P < 0.01).Compared with those in WT-Sham group,renal mRNA and protein expressions of VEGF and Flk-1 were decreased in WT-Nx group,and KO-Nx group aggravated their down-regulation (all P < 0.01).Conclusions Sirt1 gene knockout can increase proteinuria and Scr,and aggravate renal pathology and renal fibrosis in 5/6 nephrectomized mice,which is associated with the inhibition of VEGF/Flk-1 signaling pathway.It is suggested that Sirt1 may be a potential therapeutic target of chronic kidney disease.

3.
Cancer Research and Treatment ; : 345-354, 2016.
Article in English | WPRIM | ID: wpr-64171

ABSTRACT

PURPOSE: We previously reported that forkhead transcription factors of the O class 1 (FOXO1) expression in gastric cancer (GC) was associated with angiogenesis-related molecules. However, there is little experimental evidence for the direct role of FOXO1 in GC. In the present study, we investigated the effect of FOXO1 on the tumorigenesis and angiogenesis in GC and its relationship with SIRT1. MATERIALS AND METHODS: Stable GC cell lines (SNU-638 and SNU-601) infected with a lentivirus containing FOXO1 shRNA were established for animal studies as well as cell culture experiments. We used xenograft tumors in nude mice to evaluate the effect of FOXO1 silencing on tumor growth and angiogenesis. In addition, we examined the association between FOXO1 and SIRT1 by immunohistochemical tissue array analysis of 471 human GC specimens and Western blot analysis of xenografted tumor tissues. RESULTS: In cell culture, FOXO1 silencing enhanced hypoxia inducible factor-1alpha (HIF-1alpha) expression and GC cell growth under hypoxic conditions, but not under normoxic conditions. The xenograft study showed that FOXO1 downregulation enhanced tumor growth, microvessel areas, HIF-1alpha activation and vascular endothelial growth factor (VEGF) expression. In addition, inactivated FOXO1 expression was associated with SIRT1 expression in human GC tissues and xenograft tumor tissues. CONCLUSION: Our results indicate that FOXO1 inhibits GC growth and angiogenesis under hypoxic conditions via inactivation of the HIF-1alpha-VEGF pathway, possibly in association with SIRT1. Thus, development of treatment modalities aiming at this pathway might be useful for treating GC.


Subject(s)
Animals , Humans , Mice , Angiogenesis Modulating Agents , Hypoxia , Blotting, Western , Carcinogenesis , Cell Culture Techniques , Cell Line , Down-Regulation , Forkhead Transcription Factors , Heterografts , Lentivirus , Mice, Nude , Microvessels , RNA, Small Interfering , Stomach Neoplasms , Tissue Array Analysis , Transcription Factors , Vascular Endothelial Growth Factor A
4.
Korean Journal of Pathology ; : 523-531, 2012.
Article in English | WPRIM | ID: wpr-155867

ABSTRACT

BACKGROUND: Sirtuin 1 (SIRT1) and deleted in breast cancer 1 (DBC1) are known as tumor suppressor or promoter genes. This may be due to their diverse functions and interaction with other proteins. Gastric adenocarcinoma is one of the most common malignancies, but little is known about its carcinogenesis. Therefore, we investigated the association of immunohistochemical expression of SIRT1, DBC1, p53, and beta-catenin and their variable clinicopathological characteristics. METHODS: We obtained samples from 452 patients who underwent gastrectomy. Tissue microarray blocks were constructed and immonohistochemical staining was performed. RESULTS: Expression of DBC1 and SIRT1 was associated with lower histologic grade, intestinal type of Lauren classification, and lower pT (p<0.001) and pN stage (DBC1, p=0.002; SIRT1, p<0.001). Association between absence of lymphatic invasion, and SIRT1 (p=0.001) and DBC1 (p=0.004) was observed. Cytoplasmic beta-catenin expression was associated with lower histologic grade, pT, pN, tumor-node-metastasis (TNM) stage, DBC1 (p<0.001), and SIRT1 (p=0.001). Expression of SIRT1 and DBC1 was not associated with p53 (p=0.063 and p=0.060). DBC1 was an independent good prognostic factor in multivariate analysis (p=0.012). CONCLUSIONS: SIRC1 and DBC1 can be considered to be good prognostic factors in gastric adenocarcinoma.


Subject(s)
Humans , Adenocarcinoma , beta Catenin , Breast Neoplasms , Cytoplasm , Gastrectomy , Multivariate Analysis , Proteins , Sirtuin 1 , Stomach , Tumor Suppressor Proteins
5.
Experimental & Molecular Medicine ; : 8-13, 2007.
Article in English | WPRIM | ID: wpr-37560

ABSTRACT

Human SIRT1 controls various physiological responses including cell fate, stress, and aging, through deacetylation of its specific substrate protein. In processing DNA damage signaling, SIRT1 attenuates a cellular apoptotic response by deacetylation of p53 tumor suppressor. The present study shows that, upon exposure to radiation, SIRT1 could enhance DNA repair capacity and deacetylation of repair protein Ku70. Ectopically over-expressed SIRT1 resulted in the increase of repair of DNA strand breakages produced by radiation. On the other hand, repression of endogenous SIRT1 expression by SIRT1 siRNA led to the decrease of this repair activity, indicating that SIRT1 can regulate DNA repair capacity of cells with DNA strand breaks. In addition, we found that SIRT1 physically complexed with repair protein Ku70, leading to subsequent deacetylation. The dominant-negative SIRT1, a catalytically inactive form, did not induce deacetylation of Ku70 protein as well as increase of DNA repair capacity. These observations suggest that SIRT1 modulates DNA repair activity, which could be regulated by the acetylation status of repair protein Ku70 following DNA damage.


Subject(s)
Humans , Sirtuins/genetics , RNA, Small Interfering/genetics , DNA-Binding Proteins/metabolism , DNA Repair/genetics , DNA/genetics , Cell Line , Antigens, Nuclear/metabolism , Acetylation
SELECTION OF CITATIONS
SEARCH DETAIL