首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 46 毫秒
1.
AIM:To observe the effects of shikonin on the apoptosis and autophagy of human cervical cancer HeLa cells, and to explore the possible role of PI3K/Akt/mTOR signaling pathway in these processes. METHODS:The HeLa cells were treated with shikonin, and the cell viability was detected by CCK-8 assay. The apoptosis was detected by Annexin V/PI double staining. The autophagosome was observed by transfection with GFP-LC3 into the HeLa cells. After the treatment with shikonin combined with autophagy inhibitor 3-methyladenine (3-MA) or apoptosis inhibitor Z-DEVD-FMK, the protein levels of autophagy-and apoptosis-related molecules microtuble-associated protein 1 light chain 3 (LC3) and cleaved caspase-3 in the HeLa cells were determined by Western blot. The protein levels of phosphorylated PI3K (p-PI3K), phosphorylated Akt (p-Akt) and phosphorylated mTOR (p-mTOR) were also determined by Western blot. RESULTS:Shikonin significantly inhibited the viability of HeLa cells (P<0.05). Compared with control group, shikonin significantly induced apoptosis of HeLa cells (P<0.05). The results of GFP-LC3 plasmid transfection analysis showed that green dot-like congregate autophagosomes appeared in the cytoplasm of the HeLa cells after shikonin treatment, while the autophagosomes were rarely observed in control group. Compared with shikonin group, LC3-Ⅱ/LC3-I was significantly decreased and cleaved caspase-3 was significantly increased in shikonin+3-MA group (P<0.05). Compared with shikonin group, LC3-Ⅱ/LC3-I was significantly increased and cleaved caspase-3 was significantly decreased in shikonin+Z-DEVD-FMK group (P<0.05). Compared with control group, shikonin significantly decreased the protein levels of p-PI3K, p-Akt and p-mTOR (P<0.05). CONCLUSION:The apoptosis and autophagy of the HeLa cells are induced by shikonin, these two processes are complementary. The mechanism may be related to inhibition of PI3K/Akt/mTOR signaling pathway.  相似文献   

2.
AIM:To investigate whether honokiol induces the autophagy of human lung cancer A549 cells and to explore its mechanism. METHODS:The A549 cells were cultured in vitro, and treated with honokiol at different concentrations (0, 10, 20, 40, 60 and 80 μmol/L) for 48 h. MTT assay was performed to analyze the effect of honokiol on the viability of the A549 cells. The formation of autophagosome was observed by staining with acridine orange under fluorescence microscope. The protein levels of autophagy-associated protein LC3, mTOR and p-mTOR in the A549 cells treated with honokiol, or combined with autophagy inhibitor 3-methyladenine (3-MA) were determined by Western blot. RESULTS:Honokiol significantly inhibited the viability of A549 cells in a dose-dependent manner (P<0.05). The number of the intracellular acidic autophageic vacuoles with bright red fluorescence was significantly increased after honokiol treatment. The protein level of LC3-Ⅱ/LC3-I in the A549 cells was significantly enhanced after honokiol (40 μmol/L) treatment, and the ratio of LC3-Ⅱ/LC3-I was significantly decreased by treatment with 3-MA (P<0.05). Furthermore, treatment with honokiol (40 μmol/L) in the A549 cells for 48 h also resulted in significant down-regulation of phosphorylated form of mTOR (P<0.01), while the total protein level was not changed. CONCLUSION:Honokiol significantly inhibits the growth of lung cancer A549 cells and induces the autophagy, which may be correlated with inhibition of mTOR signaling pathway.  相似文献   

3.
AIM: To investigate the autophagy of human ovarian cancer SKOV3 cells induced by cepharanthine and to explore its mechanism. METHODS: The effect of cepharanthine on the viability of ovarian cancer SKOV3 cells was measured by CCK-8 assay. The SKOV3 cells were treated with cepharanthine, and then the formation of autophagosome was observed with acridine orange staining under fluorescence microscope. The protein levels of LC3, AKT, p-AKT, mTOR, p-mTOR and GAPDH in the SKOV3 cells treated with cepharanthine were determined by Western blot.RESULTS: Cepharanthine significantly inhibited the viability of ovarian cancer SKOV3 cells in a dose-dependent manner (P<0.05). The number of the intracellular acidic autophagosomes with bright red fluorescence was significantly increased after cepharanthine treatment in the SKOV3 cells. The expression of LC3-Ⅱ in SKOV3 cells was significantly enhanced after cepharanthine treatment. Furthermore, treatment with cepharanthine in the SKOV3 cells also resulted in a significant down-regulation of phosphorylated form of AKT and mTOR (P<0.01), while the total protein level was not changed. Combination of cepharanthine and 3-methyladenine resulted in a substantial decrease in the cell viability compared with using cepharanthine alone.CONCLUSION: Cepharanthine significantly inhibits the growth of human ovarian cancer SKOV3 cells and induces the autophagy, which may be correlated with down-regulation of PI3K/AKT/mTOR signaling pathway.  相似文献   

4.
AIM: To investigate the effect of low-intensity ultrasound combined with microbubble contrast agent on autophagic death of thyroid cancer cells, and to analyze the mechanism of autophagy activation and its effect on cell viability. METHODS: Human thyroid cancer cell line TPC1 was treated with low-intensity ultrasound at 20 kHz frequency and 80 mW intensity combined with microbubbles. The cell death and viability were analyzed by Live/Dead assay and CCK-8 assay 60, 120 and 240 s after the treatment. The protein levels of microtubule-associated protein 1 light chain 3-Ⅱ (LC3-Ⅱ), autophagy-related protein 5 (ATG5) and SQSTM1/P62 were determined by Western blot. The number of intracellular autophagosomes was measured by the methods of monodansylcadaverine (MDC) staining, green fluorescent protein (GFP)-LC3 transfection and transmission electron microscopy. The level of reactive oxygen species (ROS) was mea-sured and the effect of ROS on autophagy activation was evaluated by N-acetyl-L-cysteine (NAC) treatment. The effect of ATG5 siRNA transfection on autophagy was analyzed for determining the role of autophagic death. RESULTS: Low-intensity ultrasound combined with microbubbles significantly promoted TPC1 cell death and inhibited TPC1 cell viability (P<0.05) in a time-dependent manner. Compared with low-intensity ultrasound group and microbubble group, ultrasound combined with microbubbles significantly increased the protein levels of LC3-Ⅱ and ATG5, but inhibited the protein level of P62 (P<0.05). The results of MDC staining, GFP-LC3 transfection and transmission electron microscopy showed that ultrasound combined with microbubbles significantly increased the number of autophagosomes in the TPC1 cells. Compared with low-intensity ultrasound group and microbubble group, ultrasound combined with microbubbles increased the level of ROS, while NAC significantly reduced the protein level of LC3-Ⅱ (P<0.05). Thansfection with ATG5 siRNA inhibited the autophagy, significantly decreased the percentage of cell death and increased cell viability (P<0.05). CONCLUSION: Low-intensity ultrasound combined with microbubbles promotes the autophagic cell death by increasing the level of ROS in thyroid cancer cells, leading to death of thyroid cancer cells.  相似文献   

5.
AIM:To explore whether receptor-interacting protein 2 (Rip2) induces autophagy and its under-lying mechanisms in human pancreatic cancer cell line Panc-1. METHODS:The empty plasmid pEGFP-C2 or recombinant plasmid pEGFP-Rip2 was transfected into the Panc-1 cells by jetPRIME reagent. The untreated cells served as control group. The protein levels of Rip2, autophagy-related molecules (beclin-1 and LC3-Ⅱ), and phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) pathway-related proteins were determined by Western blot 48 h after transfection. The morphological changes of the autophagosomes were observed by transmission electron microscopy. RESULTS:The protein level of Rip2 was significantly increased in the Panc-1 cells transfected with pEGFP-Rip2 plasmid. The protein expression of beclin-1 and LC3-Ⅱ in pEGFP-Rip2 group was higher than that in control group and pEGFP-C2 group (all P<0.01). An increased number of autophagosomes was observed under transmission electron microscope in pEGFP-Rip2 group as compared with control group and pEGFP-C2 group. Furthermore, the protein levels of p-mTOR and p-AKT in pEGFP-Rip2 group were lower than those in control group and pEGFP-C2 group (all P<0.01), while no significant difference of the total mTOR and AKT protein levels was found among the 3 groups. CONCLUSION:Rip2 induces autophagy in the Panc-1 cells and its mechanism may be related to inhibiting the activation of PI3K/Akt/mTOR pathway.  相似文献   

6.
AIM: To investigate the role of autophagy in inhibition of human lung cancer PC9 cell proliferation by ursolic acid (UA) as well as the underlying mechanism. METHODS: MTT assay and Trypan blue exclusion test were performed to analyze the effect of UA on the proliferation of PC9 cells. The PC9 cells were treated with UA, and autophagy was observed under fluorescence microscope through acridine orange staining. The expression of autophagy-associated proteins LC3 and ATG5 in the PC9 cells were detected by Western blot. The effect of UA, 3-methyladenine (3-MA) 3-MA or their combination on the cell viability was measured by MTT assay. RESULTS: The viability of PC9 cells was significantly inhibited by UA (P<0.05 or P<0.01). The number of bright red fluorescence positive cells was significantly increased after treatment with UA. The protein expression of LC3-Ⅱ and ATG5 was significantly up-regulated compared with control group (P<0.01). Furthermore, combination of UA and 3-MA resulted in a substantial decrease in cell viability compared with using UA alone (P<0.01). CONCLUSION: UA inhibits the proliferation and induces the autophagy of the PC9 cells, in which autophagy plays a protective role. The inhibition of autophagy significantly promotes the death of the PC9 cells induced by UA.  相似文献   

7.
AIM:To study the effects of maternal expressed gene 3 (MEG3) and adenosine on the autophagy and proliferation of human hepatocellular carcinoma HepG2 cells, and to explore the possible mechanisms of autophagy and the effect on the proliferation of HepG2 cells induced by MEG3 and adenosine. METHODS:HepG2 cells were cultured according to the conventional cultural method, and divided into control group, MEG3 group (the cells were transfected with MEG3 lentivirus), adenosine group (the cells were treated with 1 mmol/L adenosine) and MEG3+adenosine group (the cells were treated with 1 mmol/L adenosine and MEG3 lentivirus). The protein expression of LC3-Ⅱ, LC3-Ⅰ and mammalian target of rapamycin (mTOR) was determined by Western blot. MDC staining was used to observe the number of autophagosomes. The cell viability was measured by CCK-8 assay, and the number of viable cells were counted by automated cell counter. RESULTS:Compared with control group, LC3-Ⅱ expression and the ratio of LC3-Ⅱ/LC3-Ⅰ were decreased, mTOR expression was increased (P<0.05), the viability of HepG2 cells was decreased, and the number of autophagosomes were reduced in MEG3 group and adenosine group. In MEG3+adenosine group, LC3-Ⅱ expression and the ratio of LC3-Ⅱ/LC3-Ⅰ were decreased significantly (P<0.01), mTOR expression was increased significantly (P<0.01), and the viability and autophagosomes of HepG2 cells were reduced markedly as compared with MEG3 group and adenosine group. After treated with MEG3 and adenosine for 24~72 h, the viable HepG2 cells reduced significantly in MEG3 group and adenosine group (P<0.01), especially in MEG3+adenosine group (P<0.01). CONCLUSION:MEG3 overexpression and low concentration of adenosine activate the mTOR pathway, and inhibit the autophagy and proli-feration of HepG2 cells. MEG3 enhances the effect of adenosine on HepG2 cells.  相似文献   

8.
AIM:To observe whether autophagy occurs in curcumin-induced human acute myeloid leukemia KG1a cells in the presence of chemotherapeutic drug cytarabine and the possible mechanism. METHODS:KG1a cells were cultured in vitro. The ultrastructural changes of the cells were observed under transmission electron microscope. Autophagy was detected by acridine orange staining. The cell viability was measured by MTT assay. The cell cycle distribution was analyzed by flow cytometry. The expression of autophagy-related molecules beclin-1 and LC3 at mRNA and protein le-vels was determined by RT-qPCR and Western blot. RESULTS:Curcumin dose-dependently inhibited the viability of KG1a cells (P<0.05). The growth inhibition rate in combination group was significantly higher than that in single reagent group and control group (P<0.01). Electron microscopical observation showed that curcumin induced the occurrence of autophagosomes, and cytarabine increased curcumin-induced autophagosomes. Acridine orange staining showed that the combined treatment with cytarabine increased the autophagy induced by curcumin, and the number of autophagic acid vesicles and cells containing autophagic acid vesicles were increased. Curcumin blocked the cell cycle in the G0/G1 phase. The mRNA expression levels of beclin-1 and LC3 in combination group were significantly higher than those in single reagent group and control group(P<0.01). The results of Western blot showed that the protein expression of beclin-1 was significantly up-regulated in combination group (P<0.05), and the ratio of LC3-Ⅱ/LC3-I was higher than that in control group (P<0.01). CONCLUSION:Curcumin inhibits the viability of KG1a cells and induces autophagy. Cytarabine promotes autophagy, which is superior to curcumin alone. It may be related to the up-regulation of beclin-1 and LC3-Ⅱ by the two reagents.  相似文献   

9.
AIM: To investigate the effects of marrow stromal cell line HS-5 on human lung adenocarcinoma A549 cells in the tumor microenvironment. METHODS: The effects of HS-5 cell-conditioned medium (HS-5-CM) on the viability and migration ability of A549 cells were detected by MTT assay and wound-healing assay. After treatment with HS-5-CM, the expression of CX3C chemokine receptor 1 (CX3CR1) at mRNA level in the A549 cells was examined by qPCR. The protein levels of p-ERK and ERK in the A549 cells treated with MAPK/ERK pathway inhibitor U0126 were observed by Western blot, the migration ability of the A549 cells was measured by wound-healing assay, and the protein expression of CX3CR1 was determined by Western blot. RESULTS: HS-5-CM promoted the viability and migration ability of the A549 cells (P<0.01). The expression of CX3CR1 at mRNA level in the A549 cells was increased after treatment with HS-5-CM. MAPK/ERK inhibitor U0126 inhibited the activation of MAPK/ERK signaling pathway (P<0.01), and reduced the migration ability (P<0.01) and the expression of CX3CR1 (P<0.05) in the A549 cells. CONCLUSION: HS-5-CM significantly promotes the A549 cell viability and migration ability. Activation of MAPK/ERK signaling pathway and the expression of CX3CR1 may play a important role in this process.  相似文献   

10.
AIM: To explore the anticancer function of Shp2 in lung adenocarcinoma A549 cells and the related molecular mechanisms. METHODS: The viability and proliferation of A549 cells treated with Shp2 specific inhibitor Phps-1 or cisplatin (DDP) were measured by CCK-8 assay and EdU assay. Annexin V-FITC/PI double staining was applied to detect apoptotic rate of A549 cells with different interventions. The protein levels of caspase-3-17p, Bcl-2, Bax, p-STAT3/STAT3 and p-ERK/ERK were determined by Western blot. RESULTS: Compared with control group, Phps-1 at the concentration of 20 μmol/L significantly increased the viability of A549 cells after 24 h of treatment (P<0.05). Meanwhile, the proliferation rate of A549 cells in Phps-1 20 μmol/L group was significant increased compared with control group (P<0.05). The apoptotic rate of A549 cells in DDP treatment group decreased from 13.01%±2.62% to 3.67%±0.93% after adding Phps-1 (P<0.05). Phps-1 down-regulated the protein levels of caspase-3-17p, Bax and p-ERK, but up-regulated p-STAT3.CONCLUSION: Shp2 is a tumor suppressor in A549 cells, which may be associated with the activation of STAT3 signal pathway.  相似文献   

11.
AIM: To evaluate the effects of angiotensinⅡ (AngⅡ) on autophagy induction in vascular endothelial cells. METHODS: Human vascular endothelial EA.hy926 cells were used in the study. Intracellular reactive oxygen species (ROS) levels were detected by a microplate reader after the cells were treated with AngⅡ (10-7 mol/L) or AngⅡ combined with antioxidant N-acetyl-L-cysteine (NAC,50 μmol/L) for 24 h. The protein levels of LC3-Ⅱ was detected by Western blotting after the cells were stimulated by different concentrations (10-8, 10-7, 10-6 mol/L) of AngⅡ for 24 h or by AngⅡ (10-7mol/L) for different time (0 h, 6 h, 12 h, 24 h, 36 h). The number of autophagosomes was evaluated by fluorescence microscopy after stained with acridine orange. Similarly, the protein level of LC3-Ⅱ and the number of autophagosomes were detected after treated with AngⅡ(10-7mol/L), AngⅡ combined with autophagy inhibitor 3-methyladenine (3-MA) at concentration of 2 mmol/L or AngⅡ combined with NAC at concentration of 50 μmol/L. RESULTS: Intracellular ROS level and LC3-Ⅱprotein level were significantly increased (P<0.05) after the cells were treated with AngⅡ, accompanied by the significant increase in the number of autophagosomes. AngⅡ-induced autophagy (as showed both in LC3-Ⅱprotein level and autophagosomes) was dramatically down-regulated by the treatment with 3-MA or NAC in EA.hy926 cells (P<0.05). CONCLUSION: AngⅡ induces autophagy through elevating ROS levels in EA.hy926 cells.  相似文献   

12.
AIM: To study the expression of microRNA (miRNA)-181a in different human lung adenocarcinoma cell lines, and to investigate the effect of miRNA-181a on cell function and its mechanism in human lung adenocarcinoma drug resistant cell A549/DDP. METHODS: Real-time PCR was used to detect the expression of miRNA-181a in BEAS-2B cells, A549 cells and A549/DDP cells. The A549/DDP cells were transfected with pGenesil-miRNA-181a eukaryotic expression plasmid. At the same time, the untransfection group and negative transfection group were also set up. The expression of miRNA-181a, cell viability, cell growth inhibition and apoptosis rate during cis-diamminedichloroplatinum (DDP) treatment, cell cycle, cell invasion, the protein expression of miRNA-181a target genes bcl-2 and p53 in the A549/DDP cells were determined by real-time fluorescence quantitative PCR, MTT assay, flow cytometry, Transwell method and Western blot, respectivly. RESULTS: The expression of miRNA-181a in A549 cells and A549/DDP cells was significantly lower than that in BEAS-2B cells, and the lowest expression level was observed in A549/DDP cells (P<0.05). The expression of miRNA-181a in A549/DDP cells was significantly increased after transfection with pGenesil-miRNA-181a (P<0.05). The cell viability, cell cycle and invasion ability of the A549/DDP cells were inhibited after miRNA-181a transfection (P<0.05). The cell growth inhibition rate and apoptotic rate of the A549/DDP cells were increased (P<0.05). The expression of Bcl-2 was reduced, but the expression of P53 was increased after transfection with miRNA-181a in A549/DDP cells (P<0.05). CONCLUSION: miRNA-181a may be correlated with the development of human lung adenocarcinoma. miRNA-181a can serve as a new target for treatment of lung cancer.  相似文献   

13.
AIM To investigate the effect of tetrandrine on the autophagy of human ovarian serous cystadenocarcinoma SKOV3 cells, and to explore its molecular mechanism. METHODS The SKOV3 cells were treated with various concentrations of tetrandrine, and the cell viability was measured by MTT assay. The formation of autophagolysosomes was observed by acridine orange staining under fluorescence microscope. The protein levels of LC3, mTOR, p-mTOR, Akt and p-Akt in the SKOV3 cells were determined by Western blot. The viability of the SKOV3 cells treated with tetrandrine alone or combined with autophagy inhibitor 3-methyladenine (3-MA) were measured by MTT assay. RESULTS Tetrandrine significantly inhibited the viability of SKOV3 cells in a dose-dependent manner (P<0.01). The results of acridine orange fluorescence staining showed that the number of intracellular autophagolysosomes with bright red fluorescence in the SKOV3 cells was significantly increased after tetrandrine treatment, while the autophagolysosomes were rarely observed in control group. The protein levels of LC3-II and P62 in the SKOV3 cells were significantly increased after tetrandrine treatment (P<0.01). Furthermore, treatment with tetrandrine resulted in significant down-regulation of phosphorylated form of mTOR and AKT in the SKOV3 cells (P<0.01), while total mTOR and AKT protein levels were not changed. Finally, combination of tetrandrine and 3-MA significantly decreased the cell viability compared with using tetrandrine alone (P<0.01). CONCLUSION The autophagy of human ovarian cancer SKOV3 cells were induced by tetrandrine and the molecular mechanism may be related to inhibition of PI3K/Akt/mTOR signaling pathway.  相似文献   

14.
AIM: To construct a conditionally replicating adenovirus vector activated by CXCR4 promoter and to evaluate its ability of lysing the lung cancer cells specifically. METHODS: Human CXCR4-E1A gene amplified by PCR was cloned into the shuttle plasmid pDC316-GFP to construct the recombinant shuttle plasmid pDC316-CXCR4-GFP. The recombinat shuttle plasmid and adenovirus genomic plasmid pBHG-lox-E1, 3Cre were transfected into 293 cells to construct the recombinant adenovirus CRAd-CXCR4-GFP. PCR was used to detect the target gene fragments, and the viral titer was determined. A549 cells with the highest mRNA expression of CXCR4 were screened out from 5 kinds of lung cancer cell lines by real-time PCR. CXCR4 promoter activity and adenovirus replication numbers were detected in A549 cells after transfection of CRAd-CXCR4-GFP and Ad-NULL. CRAd-CXCR4-GFP and Ad-NULL were transfected into A549 cells and 16HBE cells, the apoptotic rates were detected by flow cytometry and the viability was analyzed by CCK-8 assay. RESULTS: The recombinant plasmid pDC316-CXCR4-GFP was constructed successfully. Green fluorescence was observed in 293 cells under fluorescent microscope after co-transfection of pDC316-CXCR4-GFP and pBHG-lox-E1, 3Cre at 11 d. Green fluorescence was observed in 293 cells after infection of amplified 3rd generational adenovirus. PCR showed that the purpose gene was successfully integrated in recombinant adenovirus genome. The virus in the supernatant reached a titer of 1×1013 PFU/L. The mRNA expression of E1A and E4 in the A549 cells after transfection of CRAd-CXCR4-GFP was markedly increased compared with Ad-NULL group. Compared with Ad-NULL group and empty control group, the apoptotic rate and the viability of A549 cells in CRAd-CXCR4-GFP group had no significant difference in the first 4 d, the apoptotic rate increased significantly at 5 d, and the cell viability declined significantly at 5 d, but the apoptotic rate and the viability of 16HBE cells in each group had no significant difference within 5 days. CONCLUSION: The conditionally replicating adenovirus vector CRAd-CXCR4-GFP has been successfully constructed, which has the ability of lysing lung cancer cells specifically.  相似文献   

15.
AIM: To investigate the effects of cordycepin on the proliferation and migration abilities of gallbladder cancer cell line SNU-308 and its molecular mechanism. METHODS: The viability of SNU-308 cells treated with cordycepin at different concentrations was measured by MTT assay and the colony formation ability was also detected. The effect of cordycepin on apoptosis was analyzed by flow cytometry with Annexin V/PI double staining. The protein levels of apoptosis and autophagy markers, and the phosphorylation level of Akt, ERK1/2 and Ezrin were evaluated by Western blot. Immunofluorescence staining was also used to analyze the expression level of LC3 after cordycepin treatment. Wound healing assay and Transwell assay were performed to evaluate the migration ability of the SNU-308 cells after cordycepin treatment. Wound healing assay was also used to evaluate the effects of Akt inhibitor, ERK1/2 inhibitor and Ezrin knockdown on the changes of migration ability. RESULTS: Cordycepin significantly inhibited the viability and the ability of colony formation of gallbladder cancer cells (P<0.05). Induction of apoptosis by cordycepin were revealed by flow cytometry (P<0.05). The protein expression of Bcl-2 was down-regulated, while the protein levels of Bax, cytochrome C (Cyto C), Fas, FasL and cleaved caspase-3 were increased and the autophagy marker beclin 1 and the ratio of LC3-Ⅱ/I were upregulated by Western blot analysis (P<0.05). LC3 accumulation in the cytoplasm after cordycepin treatment was demonstrated by immunofluorescence staining. Cordycepin treatment resulted in the inhibition of cell migration were detected by Transwell assay and wound healing assay (P<0.05). The protein levels of p-Akt, p-ERK1/2 and p-Ezrin were down-regulated after cordycepin treatment (P<0.05). Besides, Ezrin knockdown, Akti-1/2 and GDC-0994 all resulted in the inhibition of migration ability (P<0.05). CONCLUSION: Cordycepin induces apoptosis and autophagy to inhibit gallbladder can-cer cell proliferation and migration by regulating ERK1/2, Ezrin and Akt signaling pathways.  相似文献   

16.
AIM To study the effect of pterostilbene (PTE) on autophagy and SIRT1-FoxO signaling pathway in cervical cancer cells. METHODS Human cervical cancer HeLa cells were used as the study objects. CCK-8 assay and flow cytometry were used to determine the effects of PTE at different concentrations on the viability and apoptotic rate of HeLa cells. The mRNA expression of SIRT1 and FoxO in the cells was detected by qPCR. The number of autophagosomes in the cells was observed under transmission electron microscope.The protein levels of SIRT1, FoxO, LC3-Ⅰ,LC3-Ⅱ, p62, Bax and Bcl-2 in the cells were determined by Western blot. RESULTS After treatment with PTE for 24 and 48 h, the viability inhibitory rate of the HeLa cells was increased with increasing PTE concentration. Compared with 0 μmol/L PTE, after 15, 30 and 60 μmol/L PTE treatment, the apoptotic rate of the HeLa cells, the number of autophagosomes in the cells, and the protein levels of Bax, LC3-Ⅱ/LC3-Ⅰ, SIRT1 and FoxO were increased (P<0.05), while the protein levels of Bcl-2 and p62 were decreased (P<0.05) in a concentration-dependent manner. CONCLUSION PTE may induce autophagy and apoptosis of HeLa cells by activating SIRT1-FoxO signaling pathway, and inhibit HeLa cell viability.  相似文献   

17.
AIM: To explore whether NOD8 inhibits autophagy in human pancreatic cancer cells and its underlying mechanisms, and to investigate the effect of apoptosis on the autophagy regulated by NOD8. METHODS: The empty plasmid pEGFP-C2 and recombinant plasmid pEGFP-NOD8 were transfected into the Panc-1 cells using JetPRIME reagent.The untransfected cells served as control group. The protein levels of NOD8, autophagy-related proteins beclin-1 and LC3-II, and phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) pathway-related proteins Akt, p-Akt, mTOR and p-mTOR were determined by Western blot 48 h after transfection. Meanwhile, the number of LC3 spots was quantified by immunofluorescence staining. Furthermore, after a broad caspase inhibitor Z-VAD-FMK was applied to NOD8-over-expressing cells, the protein expression levels of beclin-1 and LC3-II were detected by Western blot and the number of LC3 spots was observed by immunofluorescence staining. RESULTS: The protein level of NOD8 in pEGFP-NOD8 group was significantly higher than that in control group and pEGFP-C2 group (P<0.01). The protein expression of beclin-1 and LC3-II, and the number of LC3 spots in pEGFP-NOD8 group were significantly decreased as compared with control group and pEGFP-C2 group. Moreover, the protein levels of p-AKT and p-mTOR in pEGFP-NOD8 group were higher than those in control group and pEGFP-C2 group, while no significant difference of mTOR and AKT protein expression was found among these 3 groups. Furthermore, the protein levels of beclin-1 and LC3-II, and the number of LC3 spots in pEGFP-NOD8+Z-VAD-FMK group were significantly increased compared with pEGFP-NOD8 group. CONCLUSION: NOD8 inhibits autophagy in the Panc-1 cells and its mechanism may be related to the activation of PI3K/Akt/mTOR pathways. Apoptosis enhances the inhibitory effect of NOD8 on autophagy.  相似文献   

18.
AIM: To investigate the relationship between morphological changes of autophagy and apoptosis in the PC12 cells induced by oxygen-glucose deprivation and reoxygenation. METHODS: The PC12 cells were randomly divided into normal control group, oxygen-glucose deprivation and reoxygenation group, autophagy inhibitor group and autophagy activator group. The cells in oxygen-glucose deprivation and reoxygenation group, autophagy inhibitor group and autophagy activator group were exposed to reoxygenation (12 h) after 3 h of oxygen-glucose deprivation, and autophagy inhibitor 3-methyladenine and autophagy activator rapamycin were added into the cells at the same time. Using transmission electron microscope and monodansylcadaverine fluorescence staining, the morphological changes of autophagosome were observed. The apoptosis of the PC12 cells were analyzed by flow cytometry with Annexin V-FITC/PI staining and TUNEL method. RESULTS: Compared with normal control group, the numbers of autophagosomes and the apoptotic rates increased in oxygen-glucose deprivation and reoxygenation group (P<0.05). Compared with oxygen-glucose deprivation and reoxygenation group, the numbers of autophagosomes decreased obviously (P<0.05) and the apoptotic rates increased markedly in autophagy inhibitor group (P<0.05). The numbers of autophagosomes increased obviously (P<0.05), the apoptotic rates decreased markedly (P<0.05), the autophagosomes became bigger in size, and autolysosomes was also found in autophagy activator group. CONCLUSION: Oxygen-glucose deprivation and reoxygenation induce autophagy in PC12 cells, and autophagy inhibits cell apoptosis to play a protective role.  相似文献   

19.
LIANG Min  CHEN Xin 《园艺学报》2016,32(12):2239-2244
AIM: To investigate the effects of ursolic acid (UA) on the migration and invasion of human lung cancer cell line A549, and to explore its mechanism. METHODS: The cell viability was detected by MTT assay. The expression of miRNA-133a was detected in the A549 cells treated with UA by real-time PCR. The miRNA-133a mimics and inhibitor were transfected into the A549 cells, and the transfection efficiency was analyzed by real-time PCR. The cell migratory and invasive abilities were determined by wound healing and Transwell methods, respectively. RESULTS: The viability of the human lung cancer A549 cells was significantly inhibited by UA in a dose-dependent manner (P<0.05). IC50 of UA (24 h) for lung cancer A549 cells was 31.04 μmol/L. UA treatment significantly inhibited the migratory and invasive abilities of A549 cells in a concentration-dependent manner, accompanied by significantly elevation of miRNA-133a expression. The mimics and inhibitor of miRNA-133a significantly upregulated and downregulated the expression of miRNA-133a in the transfected A549 cells, respectively. In addition, the viability of the A549 cells was decreased extremely after tansfected with the miRNA-133a mimics (P<0.01), so did the results of the cell migration and invasion test. The A549 cells tansfected with the miRNA-133a inhibitor showed an opposite changes of the cell viability, migration and invasion. CONCLUSION: UA inhibited the viability, migration and invasion of lung cancer A549 cells by elevating the expression of miRNA-133a.  相似文献   

20.
AIM:To investigate whether ellagic acid (EA) attenuates hypoxic-ischemic encephalopathy (HIE) by down-regulating autophagy. METHODS:In vivo, Sprague-Dawley rats (n=17) were randomly divided into 3 groups:5 rats for sham group, 6 rats for HIE group and 6 rats for HIE+EA pretreatment group. The rats in HIE+EA pretreatment group were treated with EA (10 mg/kg, 10 mL/kg, suspended in corn oil, ig). After 24 h of operation, the rats from each group were sacrificed and their brains were collected. TTC staining and HE staining were used to define the infarct areas and brain structure. The autophagy-related proteins beclin-1, P62, LC3-Ⅱ/-I and Atg5 in the cortex in each group were compared by Western blot. In vitro, PC12 cells were divided into 3 groups:control group, CoCl2 group and CoCl2+EA pretreatment group. CoCl2 at 800 μmol/L was added to the PC12 cells to induce an anoxic environment. The PC12 cells were pretreated with EA at 8 μmol/L and the cell viability was measured by CCK-8 assay. The production of reactive oxidative species (ROS) in the cells was detected by flow cytometry with DCFH-DA staining. MDC staining and TMRE staining were applied to reflect the extent of autophagy and the state of apoptosis, respectively. The autophagy-related proteins in PC12 cells were also investigated. RESULTS:In HIE group, 7-day-old rats were given the operations and the their large infarct areas in the hemisphere were observed by TTC staining. HE staining displayed the injured hemispheres which contained few neurons, and exhibited edema status and serious structural damage. EA pretreatment decreased the infarct area and alleviated the damage to hemisphere with more visible neurons, compared with HIE group. Compared with sham group, the levels of autophagy-related proteins Atg5, beclin-1 and LC3-Ⅱ/-I in the cortex were increased (P<0.01), and P62 protein expression was decreased (P<0.01) in HIE group. Compared with HIE group, the protein expression of Atg5, beclin-1 and LC3-Ⅱ/-I was decreased (P<0.01) and P62 protein expression was increased in HIE+EA pretreatment group (P<0.01). In vitro, compared with CoCl2 group, the PC12 cells in CoCl2+EA pretreatment group showed a lower ROS level. Moreover, the cells in CoCl2+EA pretreatment group exhibited higher mitochondrial membrane potential than that in CoCl2 group. MDC staining in CoCl2 group showed high value of fluorescence and increased number of autophagosomes. EA pretreatment reduced the number of autophagosomes and the extent of autophagy to protect PC12 cells. Furthermore, the protein levels of Atg5, beclin-1 and LC3-Ⅱ/-I in CoCl2 group were higher (P<0.01), and the protein expression of P62 was lower (P<0.01) than those in control group. In CoCl2+EA pretreatment group, the protein levels of Atg5, beclin-1 and LC3-Ⅱ/-I were decreased (P<0.01) and the protein expression of P62 was increased as compared with CoCl2 group (P<0.01). CONCLUSION:EA pretreatment attenuates autophagy to protect the neurons against HIE injury.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号