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 31 毫秒
1.
2.
3.
4.
含关木通血清对人肾小管上皮细胞增殖周期的影响   总被引:4,自引:0,他引:4  
目的 了解含关木通血清对人肾小管上皮细胞增殖周期的影响. 方法 以体外培养的人类肾小管近端上皮细胞系(HK-2)为研究对象,用低、中、高剂量关木通灌胃家兔后,制备不同浓度含药血清,分别作用于人肾小管上皮细胞,并与空白血清比较,采用流式细胞仪检测细胞增殖周期的变化. 结果 对照组与关木通低剂量组细胞增殖周期分布正常,含低剂量关木通血清组细胞的G0/G1期未见明显阻滞;含关木通中、高剂量血清组细胞的G0/G1期发生明显阻滞. 结论 中、高剂量关木通的肾毒性机制与细胞周期阻滞有一定的关系.  相似文献   

5.
6.
We screened a library of 11,000 small molecular weight chemicals, looking for compounds that affect cell viability. We have identified 2-amino-N-quinoline-8-yl-benzenesulfonamide (QBS) as a potent cytotoxic compound that induces cell cycle arrest and apoptosis. Treatment of Jurkat T cells with QBS increased the levels of cyclin B1 as well as phosphorylated-cdc2, which was accompanied by reduced activity of cdc2 kinase, suggesting that QBS may induce cell cycle arrest at G2 phase. Structural analogues of QBS also exhibited similar effects on cell cycle progression and cell viability. Long-term treatment with QBS resulted in DNA fragmentation, cytochrome C release, and PARP cleavage, and an increase in the number of subdiploidy cells, indicative of cellular apoptosis. Moreover, QBS-induced apoptosis was blocked by z-VAD-fmk, a pan-caspase inhibitor. These results suggest that QBS is a novel and potent compound that induces G2 arrest and subsequent apoptosis, implicating it as a putative candidate for chemotherapy.  相似文献   

7.
Jeng JH  Kuo MY  Lee PH  Wang YJ  Lee MY  Lee JJ  Lin BR  Tai TF  Chang MC 《Toxicology》2006,223(3):235-247
Butyrate is a metabolite produced by oral and colonic microorganism. Butyrate has been shown to reduce colon cancer, whereas its role in oral carcinogenesis is not clear. Butyrate concentration in dental plaque and saliva ranged from 0.2 to 16 mM. In this study, we found that sodium butyrate inhibited the growth of SAS tongue cancer cells by 32% and 53% at concentrations of 1 and 2mM, respectively. Low concentrations of sodium butyrate (1-8mM) induced G0/G1 cell cycle arrest of SAS cells, whereas concentrations of 4-16 mM elicited G2/M arrest and a slight increase in apoptotic cell populations. These events were concomitant with induction of intracellular reactive oxygen species (ROS) production. An elevation in p21 mRNA and protein level was noted in SAS cells by sodium butyrate. On the contrary, a decline of cyclin Bl, cdc2 and cdc25C mRNA and protein expression in SAS cells was found after exposure to sodium butyrate. In addition, no evident increase in cdc2 inhibitory phosphorylation was found in sodium butyrate-treated SAS cancer cells. Inclusion of N-acetyl-l-cysteine (NAC) (3mM), catalase (1000 U/ml) and dimethylthiourea (DMT, 5mM), and also SOD (500 U/ml) attenuated the sodium butyrate-induced ROS production in SAS cells. However, they were not able to prevent the cell cycle arrest, apoptosis and growth inhibition in SAS cells induced by 1, 2 and 16 mM of sodium butyrate. These results indicate that sodium butyrate is toxic and inhibits the tongue cancer cell growth via induction of cell cycle arrest and apoptosis. Sodium butyrate mediates these events by mechanisms additional to ROS production.  相似文献   

8.
目的进一步研究七叶皂苷肾细胞毒性的氧化应激机制及谷胱甘肽(GSH)对七叶皂苷毒性的保护作用。方法以人肾近曲小管上皮细胞(HK-2)为模型,荧光探针法检测七叶皂苷钠(SA)0,10,15,20和25μmol·L-1处理2h后细胞内活性氧(ROS)含量变化;分别用N-乙酰-L-半胱氨酸(NAC)1,5和10mmol·L-1,丁硫氨酸亚砜亚胺(BSO)0.5和2.5mmol·L-1预处理HK-2细胞4h,然后加入SA20μmol·L-1作用24h,DTNB法测定预处理后及加入SA后细胞内GSH的含量;MTT法测定经NAC5mmol·L-1或BSO2.5mmol·L-1预处理及未经预处理的HK-2细胞与SA10,15,20,25,30和40μmol·L-1作用24h后的细胞存活率,并计算IC50值。结果 SA15,20和25μmol·L-1作用2h后,HK-2细胞内ROS含量显著高于正常对照组(P<0.01)。与正常对照组1×106个细胞内的GSH含量(5.1±1.2)nmol相比,BSO2.5mmol·L-1及NAC10mmol·L-1预处理后1×106个细胞内的GSH含量2.8±0.8和(2.7±2.3)nmol均显著降低(P<0.05),其他预处理组GSH含量无显著变化。除NAC10mmol·L-1外,各预处理组与SA20μmol·L-1作用24h后GSH含量显著降低(P<0.01)。与未经预处理的SA20,25和30μmol·L-1细胞存活率〔(83±5)%,(69±5)%和(54±6)%〕相比,经BSO2.5mmol·L-1预处理后,对应的细胞存活率显著降低,分别为〔(69±6)%,(40±13)%和(25±15)%〕(P<0.05),NAC预处理对细胞存活率无显著影响;未经预处理及NAC和BSO预处理的SA的IC50分别为31.3±1.7,23.6±2.7和(34.2±1.5)μmol·L-1。结论七叶皂苷通过氧化应激途径发挥肾细胞毒性,细胞内谷胱甘肽可对其氧化损伤起到保护作用。  相似文献   

9.
This work was designed to further study the mechanism by which sulforaphane (SFN) exerts a renoprotective effect against cisplatin (CIS)-induced damage. It was evaluated whether SFN attenuates the CIS-induced mitochondrial alterations and the impairment in the activity of the cytoprotective enzymes NAD(P)H: quinone oxidoreductase 1 (NQO1) and γ glutamyl cysteine ligase (γGCL). Studies were performed in renal epithelial LLC-PK1 cells and in isolated renal mitochondria from CIS, SFN or CIS + SFN treated rats. SFN effectively prevented the CIS-induced increase in reactive oxygen species (ROS) production and the decrease in NQO1 and γGCL activities and in glutathione (GSH) content. The protective effect of SFN on ROS production and cell viability was prevented by buthionine sulfoximine (BSO), an inhibitor of γGCL, and by dicoumarol, an inhibitor of NQO1. SFN was also able to prevent the CIS-induced mitochondrial alterations both in LLC-PK1 cells (loss of membrane potential) and in isolated mitochondria (inhibition of mitochondrial calcium uptake, release of cytochrome c, and decrease in GSH content, aconitase activity, adenosine triphosphate (ATP) content and oxygen consumption). It is concluded that the protection exerted by SFN on mitochondrial alterations and NQO1 and γGCL enzymes may be involved in the renoprotection of SFN against CIS.  相似文献   

10.
We investigated an involvement of ROS, such as H2O2 and O2- and GSH in the As4.1 cell death by antimycin A and examined whether ROS scavengers rescue antimycin A-induced As4.1 cell death and its mechanism. Levels of intracellular H2O2 and O2- were markedly increased in antimycin A-treated cells. Antimycin A reduced the intracellular GSH content. A ROS scavenger, Tiron down-regulated the production of intracellular H2O2. However, the reduction of intracellular H2O2 level did not change the apoptosis parameters, such as sub-G1 DNA content and annexin V binding. Interestingly, treatment of Tiron could partially prevent the loss of mitochondrial transmembrane potential (DeltaPsi(m)). Treatment of SOD and catalase also reduced the intracellular H2O2 and loss of mitochondrial transmembrane potential (DeltaPsi(m)) without reducing O2- level and apoptosis in antimycin A-treated As4.1 cells. All the ROS scavengers, SOD and catalase did not inhibit GSH depletion induced by antimycin A, resulting in failure of preventing the apoptosis. In addition, all the reagents including antimycin A did not induce any specific phase arrest of cell cycle in As4.1 cells. In summary, these results demonstrate that antimycin A generates potently ROS, H2O2 and O2- and induces the depletion of GSH content in As4.1 JG cells, and that Tiron, SOD and catalase inhibited partially the loss of mitochondrial transmembrane potential (DeltaPsi(m)) via the reduction of intracellular H2O2 level.  相似文献   

11.
Cisplatin is one of the most potent chemotherapeutic anticancer drugs for the treatment of various cancers. The cytotoxic action of the drug is often thought to be associated with its ability to bind DNA to form cisplatin–DNA adducts. Impaired DNA repair processes including base excision repair (BER) play important roles on its cytotoxicity. XRCC1 is a key protein known to play a central role at an early stage in the BER pathway. However, whether XRCC1 contributes to decrease the cisplatin cytotoxicity and cisplatin-induced DNA damage in HepG2 still remains unknown. Hence, the purpose of this study was to explore whether abrogation of XRCC1 gene expression by short hairpin RNAs (shRNA) could reduce DNA repair and thus sensitize liver cancer cells to cisplatin. We abrogated the XRCC1 gene in HepG2 cell using shRNA transfection. Cell viability was measured by MTT assay and clonogenicity assay. Comet assay was used to detect the DNA damage induced by cisplatin. The host cell reactivation was employed to assess the DNA repair capacity of cisplatin-damaged luciferase reporter plasmid. Flow cytometry analysis was used to determine cisplatin-induced apoptosis, cell cycle and reactive oxygen species (ROS). The results showed that abrogation of XRCC1 could sensitize HepG2 cells to cisplatin. This enhanced cytotoxicity could be attributed to the increased DNA damage and reduced DNA repair capacity. Increasing cell cycle arrest and intracellular ROS production lead to more tumor cell apoptosis and then enhanced the cisplatin cytotoxicity. Our results suggested that the cisplatin cytotoxicity may increase by targeting inhibition of XRCC1.  相似文献   

12.
An excessive and sustained increase in reactive oxygen species (ROS) production and oxidative stress have been implicated in the pathogenesis of many diseases. In the present study, we have demonstrated that 4-hydroxynonenal (4-HNE), a product of lipid peroxidation, alters glutathione (GSH) pools and induces oxidative stress in PC12 cells in culture. This increase was accompanied by alterations in subcellular ROS and glutathione (GSH) metabolisms. The GSH homeostasis was affected as both mitochondrial and extramitochondrial GSH levels, GSH peroxidase and glutathione reductase activities were inhibited and glutathione S-transferase (GST) activity was increased after 4-HNE treatment. A concentration- and time-dependent increase in cytochrome P450 2E1 (CYP 2E1) activity in the mitochondria and postmitochondrial supernatant was also observed. 4-HNE-induced oxidative stress also caused an increase in the expression of GSTA4-4, CYP2E1 and Hsp70 proteins in the mitochondria. Increased oxidative stress in PC12 cells initiated apoptosis as indicated by the release of mitochondrial cytochrome c, activation of poly-(ADP-ribose) polymerase (PARP), DNA fragmentation and decreased expression of antiapoptotic Bcl-2 proteins. Mitochondrial respiratory and redox functions also appeared to be affected markedly by 4-HNE treatment. These results suggest that HNE-induced oxidative stress and apoptosis might be associated with altered mitochondrial functions and a compromised GSH metabolism and ROS clearance.  相似文献   

13.
The mechanisms of bromate (BrO3)-induced toxicity in Normal Rat Kidney (NRK) and human embryonic kidney 293 (HEK293) cells were investigated. BrO3 (added as KBrO3) induced concentration-dependent decreases in 3-(4, dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide (MTT) staining after 48 h. BrO3-induced necrosis based on tandem increases in annexin V and PI staining. Cell cycle analysis demonstrated that BrO3 also induced G2/M arrest and nuclear fragmentation, prior to alterations in MTT staining or annexin V and PI staining. Immunoblot analysis demonstrated that the G2/M arrest correlated to induction of phosphorylated (p)-p53, p21, cyclin B1 and p-cdc2. Further, BrO3 induced time-dependent increases in the activity of the mitogen activated protein kinases p38 and ERK1/2. Treatment of cells with the p38 inhibitor SB202190, but not the ERK1/2 inhibitor PD98059, partially reversed BrO3-induced G2/M arrest and decreased BrO3-induced p-p53, p21 and cyclin B1 expression. In addition, BrO3 treatment induced reactive oxygen species (ROS) based on increases in CM-H2DCFDA fluorescence. The antioxidant ascorbic acid inhibited BrO3-induced p38 activation, G2/M arrest, p-p53, p21 and cyclin B1 expression; however, ascorbic acid had no effect on BrO3-induced formation of 8-OHdG, a marker of DNA oxidative damage, whose increases preceded cell death by 24 h. These data suggest that ROS mediated MAPK activation is involved in the molecular mechanisms of BrO3-induced cell cycle arrest, which occurs independently of 8-OH-dG production. The similar mode of action in both NRK and HEK293 cells suggests that the mechanisms of BrO3-induced renal cell death are model-independent.  相似文献   

14.
15.
Acute exposure of acetaminophen (APAP), a widely used analgesic and antipyretic drug, causes severe renal damage and no specific agent has been reported so far that plays any beneficial role in this organ pathophysiology. In the present study, the protective role of taurine on APAP-induced nephrotoxicity was investigated in mice. In order to induce acute nephrotoxicity, APAP was administered at a single dose of 2 g/kg body weight orally to male adult albino mice of Swiss strain. APAP exposure for 24 h significantly increased plasma level of blood urea nitrogen (BUN), creatinine, uric acid, TNF-α, NO production, urinary γ-glutamyl transpeptidase (γ-GT) activity, total urinary protein and urinary glucose level accompanied by a decrease in Na+–K+–ATPase activity. Moreover, APAP administration significantly increased MDA, protein carbonylation, GSSG level, intracellular ROS production and cytochrome P450 enzyme (CYPP450) activity. The same exposure decreased GSH level, ferric reducing/antioxidant power (FRAP) as well as the activities of antioxidant enzymes indicating that APAP-induced renal damage was mediated through oxidative stress. Besides, APAP exposure significantly reduced mitochondrial membrane potential and induced up-regulation of CYP2E1 in renal tissues although JNK did not play any significant role in this APAP-induced renal pathophysiology. Caspase 9/3 immunoblot and DNA fragmentation analyses showed that APAP-induced renal cell damage was mostly necrotic in nature, although some apoptosis also occurred simultaneously. Taurine treatment both pre and post (150 mg/kg body weight for 3 days, orally) to APAP exposure, however, significantly reduced APAP-induced nephrotoxicity through its antioxidant properties, urinary excretion of APAP and suppression of CYP2E1. Results suggest that taurine might be a potential therapeutic candidate against APAP-induced acute nephrotoxicity.  相似文献   

16.
Aristolochic acid I (AAI) has been widely found in herbal remedies and linked to the development of nephropathy and urothelial carcinoma in humans. This study elucidated the mechanism of oxidative stress and DNA damage mediated by AAI in human cells. Treatment of human promyelocytic leukemia cells (HL-60) and human renal proximal tubular cells (HK-2) with AAI led to a dose-dependent increase of reactive oxygen species (ROS). AAI also elevated the levels of DNA strand breaks and 8-hydroxy guanosine in HL-60 and HK-2 cells. Antioxidants, including Tiron, N-acetyl-l-cysteine (NAC) and glutathione (GSH), effectively suppressed the AAI-induced ROS and AAI-elicited genotoxicity, indicating that AAI induced the DNA damage through oxidative stress. GSH depletion was also found in AAI-treated cultures and proceeded prior to ROS formation. Exposure of HL-60 cells with AAI activated both ERK1/2 and p38 kinase phosphorylation, while only MEK1/2 inhibitor, U0126, significantly decreased AAI-mediated ROS. Preincubation of cells with thiol-containing compounds (NAC and GSH) inhibited the caspase 3 activity triggered by AAI, but non-thiol Tiron did not show a similar effect. This study demonstrated that AAI treatment results in oxidative stress-related DNA damage through GSH depletion and ERK1/2 activation; AAI-induced apoptosis is associated with GSH loss, but is independent of ROS generation.  相似文献   

17.
Casiopeínas are a series of mixed chelate copper complexes that are being evaluated as anticancer agents. Their effects in the cell include oxidative damage and mitochondrial dysfunction, yet the molecular mechanisms leading to such effects remain unclear. We tested whether [Cu(4,7-dimethyl-phenanthroline)(glycinate)]NO3 (Casiopeína IIgly or Cas IIgly) could alter cellular glutathione (GSH) levels by redox cycling with GSH to generate ROS and cellular oxidative stress. Cas IIgly induced a dramatic drop in intracellular levels of GSH in human lung cancer H157 and A549 cells, and is able to use GSH as source of electrons to catalyze the Fenton reaction. In both cell lines, the toxicity of Cas IIgly (2.5–5 μM) was potentiated by the GSH synthesis inhibitor l-buthionine sulfoximine (BSO) and diminished by the catalytic antioxidant manganese(III) meso-tetrakis(N,N′-diethylimidazolium-2-yl)porphyrin (MnTDE-1,3-IP5+), thus supporting an important role for oxidative stress. Cas IIgly also caused an over-production of reactive oxygen species (ROS) in the mitochondria and a depolarization of the mitochondrial membrane. Moreover, Cas IIgly produced mitochondrial DNA damage that resulted in an imbalance of the expression of the apoproteins of the mitochondrial respiratory chain, which also can contribute to increased ROS production. These results suggest that Cas IIgly initiates multiple possible sources of ROS over-production leading to mitochondrial dysfunction and cell death.  相似文献   

18.
Titanium dioxide nanoparticles (TiO(2)-NPs) induced cytotoxicity and DNA damage have been investigated using human amnion epithelial (WISH) cells, as an in vitro model for nanotoxicity assessment. Crystalline, polyhedral rutile TiO(2)-NPs were synthesized and characterized using X-ray diffraction (XRD), UV-Visible spectroscopy, Fourier transform infra red (FTIR) spectroscopy, and transmission electron microscopic (TEM) analyses. The neutral red uptake (NRU) and [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] (MTT) assays revealed the concentration dependent cytotoxic effects of TiO(2)-NPs (30.6nm) in concentration range of 0.625-10μg/ml. Cells exposed to TiO(2)-NPs (10μg/ml) exhibited significant reduction (46.3% and 34.6%; p<0.05) in catalase activity and glutathione (GSH) level, respectively. Treated cells showed 1.87-fold increase in intracellular reactive oxygen species (ROS) generation and 7.3% (p<0.01) increase in G(2)/M cell cycle arrest, as compared to the untreated control. TiO(2)-NPs treated cells also demonstrated the formation of DNA double strand breaks with 14.6-fold (p<0.05) increase in Olive tail moment (OTM) value at 20μg/ml concentration, vis-à-vis untreated control, under neutral comet assay conditions. Thus, the reduction in cell viability, morphological alterations, compromised antioxidant system, intracellular ROS production, and significant DNA damage in TiO(2)-NPs exposed cells signify the potential of these NPs to induce cyto- and genotoxicity in cultured WISH cells.  相似文献   

19.
The effect of the depletion or oxidation of cellular GSH on cytotoxicity of MG132 was assessed. Viability loss and decrease in GSH contents in small cell lung cancer (SCLC) cells treated with MG132 was attenuated by caspase inhibitors (z-IETD.fmk, z-LEHD.fmk and z-DQMD.fmk). Thiol compounds (N-acetylcysteine and N-(2-mercaptopropionyl)glycine) and free radical scavengers reduced MG132-induced cell death. Antioxidants, including N-acetylcysteine, inhibited the MG132-induced nuclear damage, loss in mitochondrial transmembrane potential, cytosolic accumulation of cytochrome c and caspase-3 activation. Depletion of GSH due to buthionine sulfoxime did not affect the cell viability loss, ROS formation and GSH depletion due to MG132 in SCLC cells. A thiol oxidant monochloramine, p-chloromercuribenzoate and N-ethylmaleiamide also did not affect cytotoxicity of MG132. The results suggest that the toxicity of MG132 on SCLC cells is mediated by activation of caspase-8, -9 and -3. Removal of free radicals and recovery of GSH contents may attenuate MG132-induced apoptotic cell death. Nevertheless, depletion or oxidation of cellular GSH may not affect toxicity of MG132.  相似文献   

20.
The formation of reactive oxygen species (ROS) plays a critical role in 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-induced toxicities in mammalian cells since it promotes cell proliferation, growth arrest, and apoptosis. In this study, we investigated whether TCDD induces oxidative stress and DNA damage in human ERalpha(+)/MCF-7 and ERalpha(-)/MDA-MB-231 breast cancer cells and whether this is accompanied by the initiation of DNA repair events. Results indicated that viability of MCF-7 and MDA-MB-231 cells was concentration- and time-dependently reduced by TCDD. Further, we observed significant increases in ROS formation and decreases in intracellular glutathione (GSH) in these two cell lines after TCDD treatment. Overall, the extent of cell death was greater in MCF-7 cells than in MDA-MB-231 cells whereas the magnitude of ROS formation and GSH depletion was greater in MDA-MB-231 cells than in MCF-7 cells. In addition, we observed that at non-cytotoxic concentration (1nM for 5h), TCDD induced decreases in intracellular NAD(P)H and NAD(+) in MCF-7 and MDA-MB-231 cells. These decreases were completely blocked by three types of poly(ADP-ribose) polymerase-1 (PARP-1) inhibitors. The catalytic activation of PARP-1 in cells treated with TCDD was confirmed by detection of the presence of polymers of ADP-ribose-modified PARP-1 using Western blotting. Moreover, we demonstrated increases in the number of DNA strand breaks in MCF-7 and MDA-MB-231 cells exposed to TCDD as measured by the single-cell gel electrophoresis (Comet) assay. Overall, this evidence confirms that TCDD induces decreases in intracellular NAD(P)H and NAD(+) through PARP-1 activation mediated by formation of DNA strand breaks. In addition, we demonstrated that the extent of oxidative stress and DNA damage was greater in MDA-MB-231 cells than in MCF-7 cells, with a strong correlation to estrogen receptor (ER) status. In conclusions, our findings add further support to the theme that ROS formation is a significant determinant factor in mediating the induction of oxidative DNA damage and repair in human breast cancer cells exposed to TCDD and that the TCDD-induced oxidative stress and DNA damage may, in part, contribute to TCDD-induced carcinogenesis.  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