用户名: 密码: 验证码:
氧化应激介导的喹烯酮遗传毒性及普洱茶水提取液保护作用研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
兽药残留不仅可能影响到畜禽等动物的生长发育和正常生理机能的维持,而且可能通过食物链的生物富集作用影响人类的健康。相关研究证实人类肿瘤发病率的升高与许多环境污染有关,来自动物食品的某些残留兽药的致突变和致癌作用可能是重要的原因之一。喹恶啉类药物(Quinoxalines)是人工合成的一类具有喹恶啉-1,4-二氧化物(quinoxaline-1,4-dioxides, QdNOs)基本结构的化合物,可抑制多种细菌的生长和繁殖,包括金黄色葡萄球菌、大肠杆菌和沙门氏菌等。卡巴氧和喹乙醇作为两种传统的QdNOs,体内和体外的大量研究发现这两种药物存在着潜在的致突变性和致癌性。正因为如此,欧洲共同体委员会(Commission of the European Community, CEC)于1999年将这两种化学物列为禁止使用的动物促生长剂。然而,作为一种新型的替代产品,喹烯酮(Quinocetone, QCT)与其他QdNOs也存在着相同的主体结构,在大量关注其药效的同时,喹烯酮药物使用的安全性也日益受到人们的关注。有限的资料显示,体外研究报道,QCT可能导致细菌和哺乳动物细胞株的致突变作用,并能引起HepG2细胞株的DNA损伤。但是目前关于QCT在体内引起致突变作用的研究很有限,且存在较大争议,因此选用对致癌性化学物较敏感的实验动物种群,并结合短期和长期实验,对于准确评价喹烯酮对哺乳动物的遗传毒性评价有重要的科学价值,并为制定相应的使用规范和标准有参考价值。氧化应激是许多外源化合物导致机体毒性作用的主要早期分子机制之一,有研究证明QdNOs的一些成员如卡巴氧和喹乙醇诱导的DNA损伤与在细胞内产生的ROS堆积直接相关。由于QCT与其他QdNOs一样具有相同的主体结构,其代谢过程中是否也能引起机体氧化应激,进而导致氧化性DNA损伤,目前还未见报道。
     许多研究已经证实,应激性反应蛋白的激活是机体应对外源化合物引起毒性作用的早期保护性机制,其中热休克蛋白家族(heat shock protein, HSP)是当前研究的热点。在众多候选基因之中,血红素氧化酶-1(heme oxygenase-1, HO-1)因具有较强的抗炎、抗氧化和抗凋亡作用,因此与多种氧化应激相关性损伤密切相关。正是由于HO-1对氧化损伤的多重防御效应和良好的应激性、可诱导性,近年来它已经成为预防氧化损伤的重要靶基因,被广泛关注。大量研究发现,多种膳食抗氧化物能够诱导HO-1的表达并发挥抗氧化功能。普洱茶是生长在我国云贵高原特有的茶种,相关研究发现普洱茶提取液(BTE)具有较强的抗氧化作用,抗高脂血症和减肥功能,而发挥其功能作用的主要机制是能有有效清除体内过多的活性氧自由基和活性氮物质。提示普洱茶可能通过其强的抗氧化活性,降低外源性毒物引起机体的氧化性DNA损伤,进而发挥其抗突变作用,但是这一推断目前尚未在体内实验中证明,此外关于普洱茶对HO-1的调节作用目前还没有相关的研究。
     因此,为进一步评价喹烯酮在体内的遗传毒性及其分子毒作用机制,本研究通过急性和亚慢性暴露动物染毒,探讨氧化应激与遗传毒性之间的关系,并研究HO-1在外源性化合物作用下在遗传毒性中的分子靶点作用。同时,通过普洱茶提取液补充实验,探索研究BTE对QCT诱导的遗传毒性的保护作用及其相关分子机制。
     第一部分急性暴露下喹烯酮对Balb/c小鼠遗传毒性研究
     目的:探讨QCT对Balb/c小鼠的致突变作用,并初步探讨QCT致突变作用与氧化应激的相互关系。
     方法:雄性Balb/c小鼠(48只)及雌性小鼠(48只),同一性别小鼠随机分为以下8组:(1)阴性对照组(Control):0.5%羧甲基纤维素钠水溶液灌胃;(2)喹烯酮高剂量组(QCT-H):12000 mg/kg·bw;(3)喹烯酮中剂量组(QCT-M):mg/kg-bw;(4)喹烯酮低剂量组(QCT-L):3000 mg/kg·bw;(5)阳性对照组(CY):环磷酰胺40 mg/kg-bw腹腔注射。第二次灌胃4~6h颈椎脱臼处死动物,取胸骨骨髓涂片,Giemsa染色检查嗜多染红细胞(PCE)微核发生率。取新鲜肝、肾组织,制作单细胞悬液,碱性凝胶电泳实验评价肝肾分离细胞DNA损伤程度。制作肝、肾组织冰冻切片,DHE染色,荧光显微镜下观察荧光强度,并计算平均荧光密度。制备肝、肾组织线粒体匀浆,检测GSH水平和总抗氧化能力(T-AOC),以及GPx, SOD和CAT活力。
     结果:(1)高剂量QCT组雌、雄性Balb/c小鼠骨髓细胞微核发生率分别为9.2‰和9.4‰,均显著性高于阴性对照组。中、低剂量QCT组小鼠微核发生率与阴性对照组相比无统计学差异。
     (2)彗星实验结果显示急性QCT暴露均可引起Balb/c小鼠肝、肾分离细胞的DNA损伤。对于肝分离细胞,除了3000 mg/kg·bw QCT组,其余QCT剂量组OTM值显著性高于阴性对照组;对于肾分离细胞,所有QCT剂量组OTM均显著性高于阴性对照组。
     (3)肝、肾细胞中ROS水平呈剂量依赖性升高,同时GSH水平和T-AOC水平在高剂量QCT组线粒体匀浆中显著性低于对照组,GPx, SOD和CAT活性也受到明显抑制。
     结论:(1)喹烯酮急性暴露能够引起Balb/c小鼠骨髓PCE微核实验阳性和肝、肾分离细胞DNA损伤程度增加,提示喹烯酮对哺乳动物具有潜在的遗传毒性。
     (2)喹烯酮急性暴露引起的遗传毒性跟其代谢中产生过多的ROS以及对抗氧化系统的抑制有直接关系。
     第二部分:亚慢性暴露下喹烯酮对SD大鼠遗传毒性及分子机制研究
     目的:以QCT在SD大鼠中的亚慢性暴露造模,以肝脏为主要的靶器官,进一步探讨QCT较长时期的暴露对大鼠的遗传毒性及其分子机制。
     方法:成年雄性SD大鼠(160~180g)72只随机分为4组:喹烯酮高剂量组(QCT-H):2400 mg/kg/day;喹烯酮中剂量组(QCT-M):800 mg/kg/day;喹烯酮低剂量组(QCT-H):50 mg/kg/day;正常对照组(control):0 mg/kg/day。在喂养期间分别于第5 week(8只)和第14 week(10只)处死一批老鼠,分别检测血清ALT, AST, Cr和BUN水平和尿8-OHdG含量。制备13 week SD大鼠肝细胞悬液,彗星实验测定肝细胞DNA损伤程度。检测肝细胞ROS水平以及肝组织匀浆中GSH, MDA含量,GPx, GST, GR, SOD和CAT活性。RT-PCR法检测碱基切除修复相关基因OGG1, XRCC1, PARP1, PARP2和PARG mRNA水平。RT-PCR和western blot法分别检测炎症相关基因和凋亡相关基因mRNA和蛋白表达水平。同时,分别检测喹烯酮暴露4 week和13 week SD大鼠肝脏HO-1蛋白表达水平。
     结果:(1) 2400 mg/kg/day喹烯酮暴露组SD大鼠体重显著性低于对照组,肝脏相对脏器重量显著性高于正常对照组。血清生化结果显示:QCT染毒大鼠血清ALT, AST水平呈剂量依赖型和时间依赖型升高,且高剂量QCT组大鼠血清ALT和AST水平均显著性高于对照组(P<0.05)。此外肝脏病理结果显示肝细胞内呈现大量空泡样改变,汇管区出现单核细胞侵润和结缔组织增生性改变,可见少量肝细胞坏死和凋亡现象。
     (2)彗星结果显示高剂量QCT组大鼠肝脏分离细胞DNA损伤程度明显增加,其OTM和Tail DNA%值显著性高于对照组(P<0.01)。
     (3)肝细胞内ROS,尿8-OHdG和肝组织匀浆MDA水平呈剂量依赖性升高,而抗氧化系统包括GSH水平,GPx, GST, GR, SOD和CAT活力呈剂量依赖性降低。其中高剂量QCT组各个指标与对照组相比,差异均具有显著性(P<0.05或P<0.01)。
     (4)与对照组相比,QCT亚慢性暴露后OGG1 mRNA表达呈剂量依赖性升高,其中高剂量QCT组大鼠肝脏OGG1 mRNA升至对照组的5.15倍(P<0.01)。此外,在OGG1介导的碱基切除修复通路相关酶mRNA水平,如Xrcc1, PARP1和IPARP2 mRNA表达水平均显著性升高(P<0.05)。
     (5)高剂量QCT的亚慢性暴露,可直接造成NF-κB mRNA表达的上升,同时也增加iNOS, TNF-a和Cox-2多个基因InRNA的表达水平(P<0.05)。高剂量QCT可造成p53 mRNA水平的显著性增高,同时casepase-3 mRNA水平显著性升高(P<0.05),以及对凋亡反应有一定抑制作用的Bcl-xl mRNA水平一定程度的下降,但是与对照组相比其差异没有显著性(P>0.05)。而各个基因蛋白表达水平与mRNA水平相一致。
     (6)高剂量组HO-1蛋白水平呈剂量依赖性和时间依赖性降低,其中QCT染毒三个月SD大鼠肝脏中HO-1蛋白水平显著性低于对照组(P<0.05), Nrf-2mRNA水平和蛋白水平也显著性低于对照组(P<0.05),而Keap-1蛋白水平在各个组别之间没有显著性差异(P>0.05)。
     结论:(1)QCT亚慢性染毒可造成肝细胞DNA损伤程度增加和OGG1修复系统活化,并产生大量的DNA加合物,进一步提示其具有潜在的遗传毒性。
     (2)HO-1的表达抑制造成的细胞内高氧化应激可能是QCT诱导的遗传毒性的重要分子机制。
     第三部分:普洱茶水提取液对喹烯酮诱导的遗传毒性的保护作用及其机制研究
     目的:探讨普洱茶水提取液对喹烯酮亚慢性暴露所致遗传的保护作用及其机制
     方法:成年雄性SD大鼠(160~180g)40只随机分为5组:喹烯酮染毒组(QCT):2400mg/kg/day;普洱茶补充高剂量组(QCT+BTE-H):2400 mg/kg/day QCT+1000 mg/kg/dayBTE;普洱茶补充低剂量组(QCT+BTE-L):2400 mg/kg/day QCT+500 mg/kg/day BTE;正常对照组(control)正常进食;普洱茶对照组:1000 mg/kg/day BTE灌胃。所有SD大鼠喂养13周,每周记录体重和进食量,于13周末处死所有大鼠。分离血清检测ALT和AST水平,分离整个肝脏计算肝脏相对重量并做病理切片H&E染色。彗星实验检测肝细胞DNA损伤程度,高效液相色谱电化学法检测尿液8-OHdG水平,冰冻切片检测肝细胞ROS水平,制作组织匀浆检测GSH和MDA含量,GPx, GST, GR, SOD和CAT活性。RT-PCR法检测HO-1, iNOS, TNF-a, Cox-2, p53, Bcl-xl和casepase-3 mRNA水平,western blot法检测HO-1, Nrf-2, Keap-1, iNOS, NF-κB, Cox-2, Bcl-xl casepase-3, ERK 42/44和pERK 42/44 (Thr202/Tyr204)蛋白表达水平。
     结果:(1)喹烯酮染组SD大鼠体重,进食量显著性低于对照组,肝脏脏器系数,血清ALT和AST水平显著性高于对照组(P<0.05或P<0.01)。肝脏病理结果显示肝细胞内呈现大量空泡样改变,汇管区出现单核细胞侵润和结缔组织增生性改变,可见少量肝细胞坏死和凋亡现象。BTE补充组能在一定程度上改善QCT染毒造成的肝功能损伤,表现为降低血清ALT和AST水平,减轻QCT染毒诱导的肝脏病理损伤。
     (2)各个剂量的BTE补充均能够降低QCT染毒所致的DNA损伤,而BTE本身对肝细胞DNA没有DNA损伤作用。
     (3)各个剂量的BTE补充能提高肝脏中HO-1 mRNA和蛋白水平,并能提高Nrf-2蛋白水平。此外Keap-1蛋白在BTE对照组呈高表达状态,而其他各组间的差别不显著(P>0.05)。
     (4)各个组别间ERK42/44蛋白表达水平没有明显差别(P>0.05),但是BTE补充能提高ERK42/44的磷酸化水平。
     (5)BTE补充能有效降低QCT染毒引起的高ROS,8-OHdG和MDA状态,并能上调肝脏线粒体抗氧化系统,包括GSH含量,GPx, GST, GR, SOD和CAT活力。
     (6)与QCT染毒组相比,BTE补充能够改善SD大鼠肝脏炎症反应和细胞凋亡。表现为降低iNOS, NF-κB, Cox-2, p53和casepase-3 mRNA和蛋白表达水平,提高Bcl-xlmRNA和蛋白表达水平。
     结论:(1)BTE补充能改善QCT亚慢性染毒所致的DNA损伤。
     (2)BTE通过提高ERK1/2的磷酸化水平,活化Nrf-2/HO-1通路,并抑制氧化应激,继而发挥抗突变作用。
     本研究的创新之处
     (1)本研究第一次证实喹烯酮药物能够对实验动物造成遗传毒性。
     (2)本研究从DNA损伤和修复、氧化应激两方面探讨喹烯酮遗传毒性的分子毒理学机制。
     (3)喹烯酮可能通过抑制HO-1的表达,造成肝脏细胞内高氧化应激状态而导致DNA损伤。进而证明HO-1可能是QCT导致SD大鼠遗传毒性的分子靶点。
     (4)普洱茶作为一种抗氧化物质,可通过活化ERK1/2上调HO-1的表达,并抑制氧化应激,继而发挥抗DNA损伤作用。
Veterinary drugs residues may not only affect the growth and normal physiological function of livestock and other animals but also may threaten human health though the bioaccumulation of the food chain. Studies have reported that the increased incidence of human cancer could associate with some environmental pollution, Veterinary drugs residues in animal food induced mutagenic and carcinogenic may be important. Quinoxaline drugs are a class of compounds with the basic structure of synthetic Quinoxaline-1,4-dioxide (QdNOs), they can inhibit against kinds of bacteria such as Staphylococcus aureus, Escherichia coli and Salmonella, and accelerate the animal growth. Both carbadox and olaquindox, two traditional QdNOs drugs, have shown mutagenicity and potential carcinogenicity in various test systems, so the Commission of the European Community has forbidden the use of carbadox and olaquindox as anima growth promoters in 1999. Quinocetone (QCT) is a new QdNOs and has been approved as an animal growth promoter in China since 2003 because of its higher effective and less toxic. However, based on available evidences from the literature, QCT may lead to bacterial and mammalian cell lines mutagenesis, and can cause DNA damage in HepG2 cell lines. But the studies on the QCT caused mutagenesis in vivo were limited, and the results of related studies were considerable controversy. Therefore, much research, used of sensitive laboratory animal species with carcinogenic chemicals, is necessary to evaluate accurately the genetic toxicity induced by QCT in vivo, which is important to establish reference norms and standards. Oxidative stress caused by many exogenous compounds is one of early molecular mechanisms of toxicity. The related studies showed that some QdNOs members, such as carbadox and qlaquindox, can lead to DNA damage in vitro and this injure was directly related to accumulation of ROS. With the same structure to some QdNOs, whether the oxidative stress was also occurred during the metabolic process and associated with oxidative DNA damage is largely unknown.
     Many studies confirmed that activation of stress response proteins is the early protective mechanism of the toxic effects induced by exogenous compounds. Heat shock protein (heat shock protein, HSP) family has become the focus in this field. Among the panel of potential candidate genes, heme oxygenase-1 (HO-1) seems to draw much attention with its potent anti-inflammatory, anti-oxidant, and anti-proliferative effects, close correlation with oxidative stress-related injury. Due to the function of multiple defenses against oxidative damage, good irritability and inducibility, HO-1 has become an important target gene to prevent oxidative damage. Some studies reported that a variety of dietary antioxidants can induce HO-1 expression and play antioxidant function. Chinese black tea (Pu-erh fermented tea), originally produced in the Yunnan province of China, is obtained by first parching crude green tea leaves and then fermenting them with microorganisms such as Aspergillus sp. Favorable evidence support the benefits of a diet rich in Pu-erh black tea and its associated bioactive components, such as antioxidation, hypocholesterolemia, and anti-obesity effects, and the main mechanism with these benefits may associate with its strong function by scavenging excess reactive oxygen species and reactive nitrogen substances. Suggesting that Pu-erh black tea may develop protective effect against oxidative DNA damage induced exogenous carcinogens and this could play an important role in anti-mutation effect. However, this hypothesis has not been approved in vivo study, and there is no related research on the regulation mechanism of HO-1 by Pu-erh black tea.
     Therefore, to further evaluate QCT-induced genetic toxicity and molecular toxicology mechanism in vivo, the aim of this study is to discuss the relationship among oxidative stress, inflammatory reaction, apoptosis and oxidative damage induced by QCT though acute or subchronic exposure, and the target function of HO-1 in exogenous carcinogens induced DNA damage. Besides, the protective effect and relative molecular mechanism of Pu-erh black tea extract are also evaluated as one part of this study.
     Part 1:Evaluation of genotoxicity induced by quinocetone in Balb/c mice though acute exposure
     Objectives:The aim of this study is to evaluate the genotoxicity of quinocetone though acute exposure in vivo, and discuss the relationship between genotoxicity and oxidative stress.
     Methods:Mice were divided randomly into total 10 groups (5 groups with females and males, respectively) of six animals with the same sex each and treated for acute QCT exposure as follows:(QCT groups) mice received the treatment comprised two successive administrations of QCT which was given as a solution in 0.5% sodium carboxymethylcellulose (Sinopharm Chemical Reagent Co., Ltd. P. R. China) at 24-h intervals by gavage for 12000 mg/kg·bw (QCT-H group),6000 mg/kg·bw (QCT-M group) and 3000 mg/kg-bw (QCT-L group), respectively; (Negative control group) normal control group received only the vehicle (0.5% sodium carboxymethylcellulose); (Positive control group) the mice received 40 mg/kg bw cyclophosphamidum injection. At the sampling time (between 3 and 6 h after the second dosing), all the animals were sacrificed. Giemsa staining method was used for micronucleus test of bone marrow polychromatic erythrocyte (PCE). The in vivo comet assay was used to evaluate the DNA damage in hepatic and nephric isolated cells. Besides, the ROS level, as well as GSH and T-AOC, and the activity of glutathione peroxidase, superoxide dismutase and catalase in liver and kidney were determined.
     Results:(1) The micronucleus rates of bone marrow PCE in 12000 mg/kg-bw of females and males were 9.2%o and 9.4%o, respectively. The differences between QCT groups and negative groups were statistically significantly. The 6000 and 3000 mg/kg-bw QCT groups can not lead to significant increase of micronucleus rate as compared to negative groups.
     (2) The DNA damage was observed in all the groups treated with single QCT, the OTM value in QCT-treated group were significantly higher than controls except the liver with dose of 3000 mg/kg-bw (P<0.01).
     (3) The ROS level in liver and kidney appeared a dose dependent increase during all QCT group, it was accompanied by decreased GSH and T-AOC level and repression of antioxidative enzymes activity, including GPx, SOD and CAT.
     Conclusion:(1) These data demonstrate that acute QCT exposure can develop a potential genotoxicity in mammalian.
     (2) The genotoxicity induced by QCT was mediated by oxidative stress though generating excessive ROS and inhibiting antioxidative systerm.
     Part 2:Evaluation of genotoxicity induced by QCT though subchronic exposure in SD rats
     Objectives:The aim of this study was aimed to further determine the genotoxicity induced by QCT though subchronic exposure in SD rats.
     Methods:A total of 72 male SD rats were randomly divided into four groups as below:(1) high dose of QCT (QCT-H):2400 mg/kg/day; (2) middle dose of QCT (QCT-M):800 mg/kg/day; (2) low dose of QCT (QCT-L):50 mg/kg/day; (4) Control group:only received the vehicle. All the animals were sacrificed at end of one-month (n=8) and three-month (n=10), respectively. The serum ALT, AST, Cr and BUN levels, as well as the urinary 8-hydroxy 2-deoxyguanosine (8-OHdG) among all groups were determined and compared. Comet assay was used to evaluate the endocellular DNA damage in hepatic cell suspension. The ROS level, as well as GSH and MAD, and the activities of GPx, GST, GR, SOD and CAT were measured using commercial assay kits. The mRNA levels of related enzymes of OGG1-mediated base excision repair pathway, including Xrccl, PARP1 and PARP2, were analyzed by RT-PCR test. RT-PCR and western blot were used to detect inflammation and apoptosis related gene mRNA and protein expression levels, respectively. Besides, liver HO-1 protein expression was detected in 4 week and 13 week QCT exposure in SD rats.
     Results:(1) During the 13-week toxicological experiment, QCT-fed rats had significantly lower body weight and food consumption than control rats from the 1st week and 9th week of experiment, respectively (P<0.05). These trends were still evident throughout the rest of study period. Compared to the control group, QCT administration group in higher doses resulted in greater values for relative liver weight (P<0.01). Subchronic QCT administration resulted in a sustained elevation of ALT, AST, Cr and BUN levels. Hepatic histopathological examination showed that QCT caused some morphological abnormalcy in the liver characterized by physaliferous cells, inflammatory cell infiltration, slight fibration, cell necrosis and lysis.
     (2) The DNA damage was observed in all the higher doses groups treated with QCT, the OTM value in high dose of QCT group was significantly higher than controls (P<0.01), while the OTM value in QCT-L group was significantly lower than controls (P<0.01).
     (3) Increased hepatic cell ROS level, urinary 8-hydroxy 2-deoxyguanosine (8-OHdG) contents and lipid peroxidation level were observed in the 2400 mg/kg/day QCT groups. Inhibited antioxidant system, i.e. glutathione glutathione S-transferase, glutathione peroxidase and glutathione reductase was also observed in the liver homogenate of high dose of QCT groups (P<0.01 or P<0.05).
     (4) The OGG1 mRNA level in QCT-H group was almost 5.15 times higher than controls (P <0.01), this result accorded with the 8-OHdG results during the two groups. Besides, the mRNA levels of related enzymes of OGG1-mediated base excision repair pathway, including Xrcc1, PARP1 and PARP2, were also significantly increased in QCT-H group as compared to controls (P<0.01 or P<0.05).
     (5) High dose of QCT subchronic exposure can not only directly lead to increases NF-κB mRNA expression, but also enhance multiple inflammatory related genes expression, including iNOS, TNF-a and Cox-2 (P<0.05). One the other hand, the mRNA levels of some apoptosis related gene like p53 and casepase-3 were also increased significantly as compared to controls (P<0.05). The Bcl-xl mRNA level was decreased, but the difference was not statistically significantly (P>0.05). The changes of protein expression levels were parallel with the mRNA levels among the groups.
     (6) The dose dependent and time dependent decrease of HO-1 protein expression was observed in the QCT-treated groups. As compared to controls, the level of HO-1 protein was not significantly decreased in the liver of four week QCT exposure (P<0.05), while the difference was significant at the end of thirteen week (P>0.05).
     Conclusion:(1) QCT, which is a generator of ROS, induces DNA damage in hepatic cells tested by comet assay and activiates OGG1-mediated base excision repair pathway in SD rats, which lead to accumulation of DNA adduts. The results of this study demonstrate that QCT could induce potential genotoxicity after a 13-week subchronic exposure in vivo.
     (2) The endocellular high oxdatived stress accompanied by inhibition of Nrf-2/HO-1 pathway could be the main molecular mechanism of QCT-induced genotoxicity. HO-1 could be an important molecular target which mediated the genotoxicity induced by some exogenous toxicants.
     Part 3:Protective effects of Pu-erh black tea extract supplementation against quinocetone-induced genotoxicity through subchronic exposure in SD rats
     Objectives:The present study was designed to investigate the protective effects and molecular mechanisms of Pu-erh black tea extract (BTE) in a rat model of QCT-induced genotoxicity.
     Methods:A total of 40 male SD rats were divided into five groups (eight animals for each group) and treated for 13-week as follows:(Control group) normal control group received only the basal diet; (QCT groups) SD rats were feed the diet with 2400 mg/kg/day QCT adjusted by daily food consumption; (QCT+BTE groups) QCT plus BTE groups received BTE once daily with doses of 1000 mg/kg/day (QCT+BTE-H group) and 500 mg/kg/day (QCT+BTE-L group); (BTE control group) single BTE administration with dose of 1000 mg/kg/day was selected as the BTE control group. Body weights and food consumption were measured weekly. All the animals were sacrificed at the 92-day. Serum from blood samples was collected for biochemical examination. The fresh liver was removed and weighted, then parts of liver in every group were placed in 10% neutral buffered formalin, sectioned and stained with hematoxylin and eosin (H&E) for histopathological examination. The DNAdamage in liver cell was evaluated by comet assay. The ROS level, as well as urinary 8-OHdG contents and lipid peroxidation level, was measured as the describtion in the second part. The mRNA levels of HO-1, iNOS, TNF-a, Cox-2, p53, Bcl-xl and casepase-3 were determined by RT-PCT. The protein expression levels of HO-1, Nrf-2, Keap-1, iNOS, NF-κB, Cox-2, Bcl-xl, casepase-3, ERK 42/44 and pERK 42/44 (Thr202/Tyr204) were measured by western blot.
     Results:(1) QCT-fed rats had significantly lower body weight and food consumption than control rats. Compared to the control group, QCT administration group resulted in greater values for relative liver weight and higher levels of serum ALT and AST (P<0.01 or P 0.05). Hepatic histopathological examination showed that QCT caused some morphological abnormalcy in the liver characterized by physaliferous cells, inflammatory cell infiltration, slight fibration, cell necrosis and lysis. BTE supplementation could effectively ameliorate QCT-induced hepatosis, including descreasing the serum ALT and AST levels, alleviating the pathological lesions in the liver.
     (2) BTE supplementation groups of both doses could significantly increase the DNA damage induced by QCT (P<0.01), the BTE itself can not lead to DNA damage in the SD rats though subchronic exposure.
     (3) BTE supplementation groups of both doses could increase the HO-1 and Nrf-2 mRNA and protein level in the liver as compared to the QCT-fed group. The Keap-1 protein level in the BTE control group was significantly higher than any other groups (P<0.01), but there was no significant difference among the four other groups.
     (4) The difference of ERK 42/44 protein in every group was not statistically significantly (P>0.05), but the phosphorylation of ERK 42/44 in BTE plus groups was higher than QCT-treated group.
     (5) The ROS level in hepatic cell, as well as the urinary 8-OHdG contents and lipid peroxidation level, was significantly increased in the 1000 mg/kg/day BTE supplementation group (P<0.01 or P<0.05). These were accompanied by modulation of antioxidative system for increasing the GSH level and activities of GPx, GST, GR, SOD and CAT in the liver homogenate.
     (6) As compared to QCT-fed group, BTE supplementation could alleviate the inflammatory response and cell apoptosis throgh inhibiting multiple inflammatory and apoptosis related genes and protein expression, including iNOS, TNF-a, Cox-2, p53 and casepase-3 and upregulating the mRNA and protein levels of Bcl-xl.
     Conclusion:(1) BTE supplementation can effectively protect the liver against QCT-induced DNA damage in SD rats.
     (2) BTE could upregulate the Nrf-2/HO-1 pathway via activation of ERK1/2 in liver through inhibiting oxidative stress, which plays a positive function of antimutagenic.
引文
1. Paige, JC; Tollefson, L; Miller, MA. Health implications of residues of veterinary drugs and chemicals in animal tissues. Vet Clin North Am Food Anim Pract.1999,15, (1), 31-43, viii.
    2. Carta, A; Corona, P; Loriga, M. Quinoxaline 1,4-dioxide:a versatile scaffold endowed with manifold activities. Curr Med Chem.2005,12, (19),2259-72.
    3. Huang, XJ; Zhang, HH; Wang, X, et al. ROS mediated cytotoxicity of porcine adrenocortical cells induced by QdNOs derivatives in vitro. Chem Biol Interact.2010,185, (3),227-34.
    4. Chen, Q; Tang, S; Jin, X, et al. Investigation of the genotoxicity of quinocetone, carbadox and olaquindox in vitro using Vero cells. Food Chem Toxicol.2009,47, (2), 328-34.
    5. Situa, C; Elliott, CT. Simultaneous and rapid detection of five banned antibiotic growth promoters by immunoassay. Analytica Chimica Acta.2005,529, ((1-2)),89-96.
    6. Wu, Y; Y., W. Simultaneous determination of five quinoxaline-1,4-dioxides in animal feeds using ultrasonic solvent extraction and high-performance liquid chromatography. Analytica Chimica Acta.2006,569, (1-2),97-102.
    7. Wang, X; Zhang, W; Wang, Y, et al. Acute and sub-chronic oral toxicological evaluations of quinocetone in Wistar rats. Regul Toxicol Pharmacol.2010,58, (3),421-7.
    8. Ding, MX; Yuan, ZH; Wang, YL, et al. Olaquindox and cyadox stimulate growth and decrease intestinal mucosal immunity of piglets orally inoculated with Escherichia coli. J Anim Physiol Anim Nutr (Berl).2006,90, (5-6),238-43.
    9. Ding, MX; Wang, YL; Zhu, HL, et al. Effects of cyadox and olaquindox on intestinal mucosal immunity and on fecal shedding of Escherichia coli in piglets. J Anim Sci.2006, 84, (9),2367-73.
    10.张伟;彭大鹏;黄玲丽,et al喹烯酮遗传毒性的研究.毒理学杂志.2007,21,(4),335-335.
    11. Hao, L; Chen, Q; Xiao, X. Molecular mechanism of mutagenesis induced by olaquindox using a shuttle vector pSP189/mammalian cell system. Mutat Res.2006,599, (1-2),21-5.
    12. Bendaly, J; Zhao, S; Neale, JR, et al.2-Amino-3,8-dimethylimidazo-[4,5-f]quinoxaline-induced DNA adduct formation and mutagenesis in DNA repair-deficient Chinese hamster ovary cells expressing human cytochrome P4501A1 and rapid or slow acetylator N-acetyltransferase 2. Cancer Epidemiol Biomarkers Prev.2007,16, (7),1503-9.
    13.郝利华;肖希龙;周宗灿.穿梭质粒pSP189/哺乳动物Vero细胞检测系统在喹乙醇诱变分子机制研究中的应用癌变·畸变·突变.2006,18,(1),23-25.
    14. Jin, X; Chen, Q; Tang, SS, et al. Investigation of quinocetone-induced genotoxicity in HepG2 cells using the comet assay, cytokinesis-block micronucleus test and RAPD analysis. Toxicol In Vitro.2009,23, (7),1209-14.
    15. Azqueta, A; Pachon, G; Cascante, M, et al. DNA damage induced by a quinoxaline 1,4-di-N-oxide derivative (hypoxic selective agent) in Caco-2 cells evaluated by the comet assay. Mutagenesis.2005,20, (3),165-71.
    16. Bernstein, C; Bernstein, H; Payne, CM, et al. DNA repair/pro-apoptotic dual-role proteins in five major DNA repair pathways:fail-safe protection against carcinogenesis. Mutat Res.2002,511, (2),145-78.
    17. Christmann, M; Tomicic, MT; Roos, WP, et al. Mechanisms of human DNA repair:an update. Toxicology.2003,193, (1-2),3-34.
    18. Friedberg, EC. A brief history of the DNA repair field. Cell Res.2008,18, (1),3-7.
    19. Ichihara, T; Wanibuchi, H; Totsuka, Y, et al. Induction of DNA-adducts and increase of 8-hydroxy-2-deoxyguanosine, but no development of preneoplastic lesions in offspring liver with transplacental and trans-breast milk exposure to 2-amino-3,8-dimethylimidazo [4,5-f]quinoxaline (MelQx) in rats. Cancer Sci.2004,95, (12),943-8.
    20. Iwai, S; Karim, R; Kitano, M, et al. Role of oxidative DNA damage caused by carbon tetrachloride-induced liver injury--enhancement of MeIQ-induced glutathione S-transferase placental form-positive foci in rats. Cancer Lett.2002,179, (1),15-24.
    21. Kushida, M; Wanibuchi, H; Morimura, K, et al. Dose-dependence of promotion of 2-amino-3,8-dimethylimidazo[4,5-f]quinoxaline-induced rat hepatocarcinogenesis by ethanol:evidence for a threshold. Cancer Sci.2005,96, (11),747-57.
    22. Westerheide, SD; Morimoto, RI. Heat shock response modulators as therapeutic tools for diseases of protein conformation. J Biol Chem.2005,280, (39),33097-100.
    23. Takahashi, T; Morita, K; Akagi, R, et al. Heme oxygenase-1:a novel therapeutic target in oxidative tissue injuries. Curr Med Chem.2004,11,(12),1545-61.
    24. Morse, D; Choi, AM. Heme oxygenase-1:from bench to bedside. Am J Respir Crit Care Med.2005,172, (6),660-70.
    25. Kirkby, KA; Adin, CA. Products of heme oxygenase and their potential therapeutic applications. Am J Physiol Renal Physiol.2006,290, (3), F563-71.
    26. Ogborne, RM; Rushworth, SA; Charalambos, CA, et al. Haem oxygenase-1:a target for dietary antioxidants. Biochem Soc Trans.2004,32, (Pt 6),1003-5.
    27. Wang, D; Xiao, R; Hu, X, et al. Comparative safety evaluation of Chinese Pu-erh green tea extract and Pu-erh black tea extract in Wistar rats. J Agric Food Chem.2010,58, (2), 1350-8.
    28. Duh, PD; Yen, GC; Yen, WJ, et al. Effects of pu-erh tea on oxidative damage and nitric oxide scavenging. J Agric Food Chem.2004,52, (26),8169-76.
    29. Fujita, H; Yamagami, T. Efficacy and safety of Chinese black tea (Pu-Ehr) extract in healthy and hypercholesterolemic subjects. Ann Nutr Metab.2008,53, (1),33-42.
    30. Fujita, H; Yamagami, T. Extract of black tea (pu-ehr) inhibits postprandial rise in serum cholesterol in mice, and with long term use reduces serum cholesterol and low density lipoprotein levels and renal fat weight in rats. Phytother Res.2008,22, (10), 1275-81.
    31. Wang, Y; Zhao, R; Yan, X, et al. The effects of quinocetone on promoting broilers growth. Veterinary Science in China.1995,25, (7),36-38.
    32. Fischler, F, Commision regulation (EC) No.2788/98. In Communities, O. J. o. t. E., Ed. 1998; Vol.41, pp 31-32.
    33. Zhang, W. Preclinical toxicological study of quinoceton. Masters Degree Dissertations of Central China Agricultural University.2007, pp.27-31.
    34. Tu, H. The genotoxicity of quinoxalines to mammalian cell.. Masters Degree Dissertations of Central China Agricultural University.2007,22-24.
    35. Nesslany, F; Zennouche, N; Simar-Meintieres, S, et al. In vivo Comet assay on isolated kidney cells to distinguish genotoxic carcinogens from epigenetic carcinogens or cytotoxic compounds. Mutat Res.2007,630, (1-2),28-41.
    36. Sasaki, YF; Nishidate, E; Izumiyama, F, et al. Simple detection of chemical mutagens by the alkaline single-cell gel electrophoresis (Comet) assay in multiple mouse organs (liver, lung, spleen, kidney, and bone marrow). Mutat Res.1997,391, (3),215-31.
    37. Lowry, OH; Rosebrough, NJ; Farr, AL, et al. Protein measurement with the Folin phenol reagent. J Biol Chem.1951,193, (1),265-75.
    38. Kono, Y. Generation of superoxide radical during autoxidation of hydroxylamine and an assay for superoxide dismutase. Arch Biochem Biophys.1978,186, (1),189-95.
    39. Sazuka, Y; Tanizawa, H; Takino, Y. Effect of adriamycin on the activities of superoxide dismutase, glutathione peroxidase and catalase in tissues of mice. Jpn J Cancer Res.1989,80,(1),89-94.
    40. Goth, L. A simple method for determination of serum catalase activity and revision of reference range. Clin Chim Acta.1991,196, (2-3),143-51.
    41. Moron, MS; Depierre, JW; Mannervik, B. Levels of glutathione, glutathione reductase and glutathione S-transferase activities in rat lung and liver. Biochim Biophys Acta.1979, 582, (1),67-78.
    42. Shen, J; Yang, C; Wu, C, et al. Identification of the major metabolites of quinocetone in swine urine using ultra-performance liquid chromatography/electrospray ionization quadrupole time-of-flight tandem mass spectrometry. Rapid Commun Mass Spectrom.24, (3),375-83.
    43. Thukral, SK; Nordone, PJ; Hu, R, et al. Prediction of nephrotoxicant action and identification of candidate toxicity-related biomarkers. Toxicol Pathol.2005,33, (3), 343-55.
    44. Liu, Z; Huang, L; Dai, M, et al. Metabolism of cyadox in rat, chicken and pig liver microsomes and identification of metabolites by accurate mass measurements using electrospray ionization hybrid ion trap/time-of-flight mass spectrometry. Rapid Commun Mass Spectrom.2009,23, (13),2026-34.
    45. Liu, Z; Huang, L; Dai, M, et al. Metabolism of olaquindox in rat liver microsomes: structural elucidation of metabolites by high-performance liquid chromatography combined with ion trap/time-of-flight mass spectrometry. Rapid Commun Mass Spectrom.2008,22, (7),1009-16.
    46. Kabuto, H; Hasuike, S; Minagawa, N, et al. Effects of bisphenol A on the metabolisms of active oxygen species in mouse tissues. Environ Res.2003,93, (1),31-5.
    47. Ihsan, A; Wang, X; Liu, Z, et al. Long-term mequindox treatment induced endocrine and reproductive toxicity via oxidative stress in male Wistar rats. Toxicol Appl Pharmacol. 252, (3),281-8.
    1. Durackova, Z. Some current insights into oxidative stress. Physiol Res.59, (4),459-69.
    2. Jabs, T. Reactive oxygen intermediates as mediators of programmed cell death in plants and animals. Biochem Pharmacol.1999,57, (3),231-45.
    3. Fridovich, I. The biology of oxygen radicals. Science.1978,201, (4359),875-80.
    4. Hussain, SP; Hofseth, LJ; Harris, CC. Radical causes of cancer. Nat Rev Cancer.2003, 3, (4),276-85.
    5. Schraufstatter, I; Hyslop, PA; Jackson, JH, et al. Oxidant-induced DNA damage of target cells. J Clin Invest.1988,82, (3),1040-50.
    6. Visconti, R; Grieco, D. New insights on oxidative stress in cancer. Curr Opin Drug Discov Devel.2009,12, (2),240-5.
    7. Huang, XJ; Zhang, HH; Wang, X, et al. ROS mediated cytotoxicity of porcine adrenocortical cells induced by QdNOs derivatives in vitro. Chem Biol Interact.185, (3), 227-34.
    8. Wang, D; Zhong, Y; Luo, X, et al. Pu-erh black tea supplementation decreases quinocetone-induced ROS generation and oxidative DNA damage in Balb/c mice. Food Chem Toxicol.2011,49, (2),477-84.
    9. Wang, X; Zhang, W; Wang, Y, et al. Acute and sub-chronic oral toxicological evaluations of quinocetone in Wistar rats. Regul Toxicol Pharmacol.2010,58, (3),421-7.
    10. Hao, L; Chen, Q; Xiao, X. Molecular mechanism of mutagenesis induced by olaquindox using a shuttle vector pSP189/mammalian cell system. Mutat Res.2006,599, (1-2),21-5.
    11. Paehler, A; Richoz, J; Soglia, J, et al. Analysis and quantification of DNA adducts of 2-amino-3,8-dimethylimidazo[4,5-f]quinoxaline in liver of rats by liquid chromatography/electrospray tandem mass spectrometry. Chem Res Toxicol.2002,15, (4), 551-61.
    12. Azqueta, A; Arbillaga, L; Pachon, G, et al. A quinoxaline 1,4-di-N-oxide derivative induces DNA oxidative damage not attenuated by vitamin C and E treatment. Chem Biol Interact.2007,168, (2),95-105.
    13. Cooke, MS; Evans, MD; Herbert, KE, et al. Urinary 8-oxo-2'-deoxyguanosine--source, significance and supplements. Free Radic Res.2000,32, (5),381-97.
    14. Kasai, H. Analysis of a form of oxidative DNA damage,8-hydroxy-2'-deoxyguanosine, as a marker of cellular oxidative stress during carcinogenesis. Mutat Res.1997,387, (3), 147-63.
    15.郝利华;肖希龙;周宗灿.穿梭质粒pSP189/哺乳动物Vero细胞检测系统在喹乙醇诱变分子机制研究中的应用.癌变·畸变·突变.2006,18,(1),23-25.
    16. Valls-Belles, V; Torres Mdel, C; Boix, L, et al. alpha-Tocopherol, MDA-HNE and 8-OHdG levels in liver and heart mitochondria of adriamycin-treated rats fed with alcohol-free beer. Toxicology.2008,249, (2-3),97-101.
    17. Azqueta, A; Pachon, G; Cascante, M, et al. DNA damage induced by a quinoxaline 1,4-di-N-oxide derivative (hypoxic selective agent) in Caco-2 cells evaluated by the comet assay. Mutagenesis.2005,20, (3),165-71.
    18. Klungland, A; Bjelland, S. Oxidative damage to purines in DNA:role of mammalian Oggl. DNA Repair (Amst).2007,6, (4),481-8.
    19. Sunaga, N; Kohno, T; Shinmura, K, et al. OGG1 protein suppresses G:C-->T:A mutation in a shuttle vector containing 8-hydroxyguanine in human cells. Carcinogenesis. 2001,22, (9),1355-62.
    20. Hodges, NJ; Chipman, JK. Down-regulation of the DNA-repair endonuclease 8-oxo-guanine DNA glycosylase 1 (hOGGl) by sodium dichromate in cultured human A549 lung carcinoma cells. Carcinogenesis.2002,23, (1),55-60.
    21. Lee, MR; Kim, SH; Cho, HJ, et al. Transcription factors NF-YA regulate the induction of human OGG1 following DNA-alkylating agent methylmethane sulfonate (MMS) treatment. J Biol Chem.2004,279, (11),9857-66.
    22. Dolphin, D, Glutathione:chemical, biochemical and medical aspects.. Wiley:New York (NY),1989.
    23. Maellaro, E; Casini, AF; Del Bello, B, et al. Lipid peroxidation and antioxidant systems in the liver injury produced by glutathione depleting agents. Biochem Pharmacol. 1990,39, (10),1513-21.
    24. Grivennikov, SI; Karin, M. Inflammation and oncogenesis:a vicious connection. Curr Opin Genet Dev.20, (1),65-71.
    25. Gonda, TA; Tu, S; Wang, TC. Chronic inflammation, the tumor microenvironment and carcinogenesis. Cell Cycle.2009,8, (13),2005-13.
    26. Reuter, S; Gupta, SC; Chaturvedi, MM, et al. Oxidative stress, inflammation, and cancer:how are they linked? Free Radic Biol Med.49, (11),1603-16.
    27. Hussain, SP; Harris, CC. Inflammation and cancer:an ancient link with novel potentials. Int J Cancer.2007,121, (11),2373-80.
    28. Domitrovic, R; Jakovac, H; Blagojevic, G. Hepatoprotective activity of berberine is mediated by inhibition of TNF-alpha, COX-2, and iNOS expression in CCl(4)-intoxicated mice. Toxicology.280, (1-2),33-43.
    29. Zhou, BB; Elledge, SJ. The DNA damage response:putting checkpoints in perspective. Nature.2000,408, (6811),433-9.
    30. Szabadkai, G; Rizzuto, R. Participation of endoplasmic reticulum and mitochondrial calcium handling in apoptosis:more than just neighborhood? FEBS Lett.2004,567, (1), 111-5.
    31. Truong-Tran, AQ; Grosser, D; Ruffin, RE, et al. Apoptosis in the normal and inflamed airway epithelium:role of zinc in epithelial protection and procaspase-3 regulation. Biochem Pharmacol.2003,66, (8),1459-68.
    32. Wagener, FA; Volk, HD; Willis, D, et al. Different faces of the heme-heme oxygenase system in inflammation. Pharmacol Rev.2003,55, (3),551-71.
    33. Deshane, J; Wright, M; Agarwal, A. Heme oxygenase-1 expression in disease states. Acta Biochim Pol.2005,52, (2),273-84.
    34. Otterbein, LE; Choi, AM. Heme oxygenase:colors of defense against cellular stress. Am J Physiol Lung Cell Mol Physiol.2000,279, (6), L1029-37.
    35. Otterbein, LE; Soares, MP; Yamashita, K, et al. Heme oxygenase-1:unleashing the protective properties of heme. Trends Immunol.2003,24, (8),449-55.
    36. Farombi, EO; Surh, YJ. Heme oxygenase-1 as a potential therapeutic target for hepatoprotection. J Biochem Mol Biol.2006,39, (5),479-91.
    37. Balogun, E; Hoque, M; Gong, P, et al. Curcumin activates the haem oxygenase-1 gene via regulation of Nrf2 and the antioxidant-responsive element. Biochem J.2003,371, (Pt 3), 887-95.
    38. Nguyen, T; Yang, CS; Pickett, CB. The pathways and molecular mechanisms regulating Nrf2 activation in response to chemical stress. Free Radic Biol Med.2004,37, (4),433-41.
    39. Alam, J; Killeen, E; Gong, P, et al. Heme activates the heme oxygenase-1 gene in renal epithelial cells by stabilizing Nrf2. Am J Physiol Renal Physiol.2003,284, (4), F743-52.
    40. Itoh, K; Tong, KI; Yamamoto, M. Molecular mechanism activating Nrf2-Keap1 pathway in regulation of adaptive response to electrophiles. Free Radic Biol Med.2004,36, (10),1208-13.
    41. da Silva, JL; Morishita, T; Escalante, B, et al. Dual role of heme oxygenase in epithelial cell injury:contrasting effects of short-term and long-term exposure to oxidant stress. J Lab Clin Med.1996,128, (3),290-6.
    42. Galbraith, R. Heme oxygenase:who needs it? Proc Soc Exp Biol Med.1999,222, (3), 299-305.
    43. Yun, N; Eum, HA; Lee, SM. Protective role of heme oxygenase-1 against liver damage caused by hepatic ischemia and reperfusion in rats. Antioxid Redox Signal.13, (10), 1503-12.
    44. Song, F; Chen, W; Jia, W, et al. A natural sweetener, Momordica grosvenori, attenuates the imbalance of cellular immune functions in alloxan-induced diabetic mice. Phytother Res.2006,20, (7),552-60.
    45. Faraonio, R; Vergara, P; Di Marzo, D, et al. p53 suppresses the Nrf2-dependent transcription of antioxidant response genes. J Biol Chem.2006,281, (52),39776-84.
    1. Halliwell, B. Free radicals, proteins and DNA:oxidative damage versus redox regulation. Biochem Soc Trans.1996,24, (4),1023-7.
    2. Bu-Abbas, A; Nunez, X; Clifford, MN, et al. A comparison of the antimutagenic potential of green, black and decaffeinated teas:contribution of flavanols to the antimutagenic effect. Mutagenesis.1996,11, (6),597-603.
    3. Azqueta, A; Arbillaga, L; Pachon, G, et al. A quinoxaline 1,4-di-N-oxide derivative induces DNA oxidative damage not attenuated by vitamin C and E treatment. Chem Biol Interact.2007,168, (2),95-105.
    4. Khuri, FR; Shin, DM. Head and neck cancer chemoprevention gets a shot in the arm. J Clin Oncol.2008,26, (3),345-7.
    5. Liang, Y; Zhang, L; Lu, J. A study on chemical estimation of pu-erh tea quality. J Sci Food Agric.2005,85,381-390.
    6. Jeng, KC; Chen, CS; Fang, YP, et al. Effect of microbial fermentation on content of statin, GABA, and polyphenols in Pu-Erh tea. J Agric Food Chem.2007,55, (21),8787-92.
    7. Yang, TT; Koo, MW. Hypocholesterolemic effects of Chinese tea. Pharmacol Res. 1997,35, (6),505-12.
    8. Catterall, F; Copeland, E; Clifford, MN, et al. Contribution of theafulvins to the antimutagenicity of black tea:their mechanism of action. Mutagenesis.1998,13, (6), 631-6.
    9. Murugan, RS; Uchida, K; Hara, Y, et al. Black tea polyphenols modulate xenobiotic-metabolizing enzymes, oxidative stress and adduct formation in a rat hepatocarcinogenesis model. Free Radic Res.2008,42, (10),873-84.
    10. Duh, PD; Yen, GC; Yen, WJ, et al. Effects of pu-erh tea on oxidative damage and nitric oxide scavenging. J Agric Food Chem.2004,52, (26),8169-76.
    11. Wang, D; Xiao, R; Hu, X, et al. Comparative safety evaluation of Chinese Pu-erh green tea extract and Pu-erh black tea extract in Wistar rats. J Agric Food Chem.2010,58, (2), 1350-8.
    12. Fujita, H; Yamagami, T. Efficacy and safety of Chinese black tea (Pu-Ehr) extract in healthy and hypercholesterolemic subjects. Ann Nutr Metab.2008,53, (1),33-42.
    13. Fujita, H; Yamagami, T. Extract of black tea (pu-ehr) inhibits postprandial rise in serum cholesterol in mice, and with long term use reduces serum cholesterol and low density lipoprotein levels and renal fat weight in rats. Phytother Res.2008,22, (10),1275-81.
    14. Venugopal, R; Jaiswal, AK. Nrfl and Nrf2 positively and c-Fos and Fral negatively regulate the human antioxidant response element-mediated expression of NAD(P)H:quinone oxidoreductasel gene. Proc Natl Acad Sci U S A.1996,93, (25), 14960-5.
    15. Owuor, ED; Kong, AN. Antioxidants and oxidants regulated signal transduction pathways. Biochem Pharmacol.2002,64, (5-6),765-70.
    16. Roux, PP; Blenis, J. ERK and p38 MAPK-activated protein kinases:a family of protein kinases with diverse biological functions. Microbiol Mol Biol Rev.2004,68, (2),320-44.
    17. Na, HK; Kim, EH; Jung, JH, et al. (-)-Epigallocatechin gallate induces Nrf2-mediated antioxidant enzyme expression via activation of PI3K and ERK in human mammary epithelial cells. Arch Biochem Biophys.2008,476, (2),171-7.
    18. Wu, CC; Hsu, MC; Hsieh, CW, et al. Upregulation of heme oxygenase-1 by Epigallocatechin-3-gallate via the phosphatidylinositol 3-kinase/Akt and ERK pathways. Life Sci.2006,78, (25),2889-97.
    19. Lee-Hilz, YY; Boerboom, AM; Westphal, AH, et al. Pro-oxidant activity of flavonoids induces EpRE-mediated gene expression. Chem Res Toxicol.2006,19, (11),1499-505.
    20. Sang, S; Lambert, JD; Hong, J, et al. Synthesis and structure identification of thiol conjugates of (-)-epigallocatechin gallate and their urinary levels in mice. Chem Res Toxicol.2005,18, (11),1762-9.
    21. Jozkowicz, A; Was, H; Dulak, J. Heme oxygenase-1 in tumors:is it a false friend? Antioxid Redox Signal.2007,9, (12),2099-117.
    22. Deshane, J; Wright, M; Agarwal, A. Heme oxygenase-1 expression in disease states. Acta Biochim Pol.2005,52, (2),273-84.
    23. Otterbein, LE; Soares, MP; Yamashita, K, et al. Heme oxygenase-1:unleashing the protective properties of heme. Trends Immunol,2003,24, (8),449-55.
    24. Kirkby, KA; Adin, CA. Products of heme oxygenase and their potential therapeutic applications. Am J Physiol Renal Physiol.2006,290, (3), F563-71.
    25. Stocker, R; Yamamoto, Y; McDonagh, AF, et al. Bilirubin is an antioxidant of possible physiological importance. Science.1987,235, (4792),1043-6.
    26. Kaur, H; Hughes, MN; Green, CJ, et al. Interaction of bilirubin and biliverdin with reactive nitrogen species. FEBS Lett.2003,543, (1-3),113-9.
    27. Hayashi, S; Takamiya, R; Yamaguchi, T, et al. Induction of heme oxygenase-1 suppresses venular leukocyte adhesion elicited by oxidative stress:role of bilirubin generated by the enzyme. Circ Res.1999,85, (8),663-71.
    28. Brouard, S; Otterbein, LE; Anrather, J, et al. Carbon monoxide generated by heme oxygenase 1 suppresses endothelial cell apoptosis. J Exp Med.2000,192, (7),1015-26.
    29. Ogborne, RM; Rushworth, SA; Charalambos, CA, et al. Haem oxygenase-1:a target for dietary antioxidants. Biochem Soc Trans.2004,32, (Pt 6),1003-5.
    1. Maines, MD. The heme oxygenase system:a regulator of second messenger gases. Annu Rev Pharmacol Toxicol.1997,37,517-54.
    2. Farombi, EO; Surh, YJ. Heme oxygenase-1 as a potential therapeutic target for hepatoprotection. J Biochem Mol Biol.2006,39, (5),479-91.
    3. McCoubrey, WK, Jr.; Huang, TJ; Maines, MD. Isolation and characterization of a cDNA from the rat brain that encodes hemoprotein heme oxygenase-3. Eur J Biochem. 1997,247,(2),725-32.
    4. Deshane, J; Wright, M; Agarwal, A. Heme oxygenase-1 expression in disease states. Acta Biochim Pol.2005,52, (2),273-84.
    5. Otterbein, LE; Soares, MP; Yamashita, K, et al. Heme oxygenase-1:unleashing the protective properties of heme. Trends Immunol.2003,24, (8),449-55.
    6. Kirkby, KA; Adin, CA. Products of heme oxygenase and their potential therapeutic applications. Am J Physiol Renal Physiol.2006,290, (3), F563-71.
    7. Stocker, R; Yamamoto, Y; McDonagh, AF, et al. Bilirubin is an antioxidant of possible physiological importance. Science.1987,235, (4792),1043-6.
    8. Kaur, H; Hughes, MN; Green, CJ, et al. Interaction of bilirubin and biliverdin with reactive nitrogen species. FEBS Lett.2003,543, (1-3),113-9.
    9. Hayashi, S; Takamiya, R; Yamaguchi, T, et al. Induction of heme oxygenase-1 suppresses venular leukocyte adhesion elicited by oxidative stress:role of bilirubin generated by the enzyme. Circ Res.1999,85, (8),663-71.
    10. Brouard, S; Otterbein, LE; Anrather, J, et al. Carbon monoxide generated by heme oxygenase 1 suppresses endothelial cell apoptosis. J Exp Med.2000,192, (7),1015-26.
    11. Oates, PS; West, AR. Heme in intestinal epithelial cell turnover, differentiation, detoxification, inflammation, carcinogenesis, absorption and motility. World J Gastroenterol.2006,12, (27),4281-95.
    12. Zhu, X; Fan, WG; Li, DP, et al. Heme oxygenase-1 system and gastrointestinal tumors. World J Gastroenterol.16, (21),2633-7.
    13. Busserolles, J; Megias, J; Terencio, MC, et al. Heme oxygenase-1 inhibits apoptosis in Caco-2 cells via activation of Akt pathway. Int J Biochem Cell Biol.2006,38, (9),1510-7.
    14. Fang, J; Akaike, T; Maeda, H. Antiapoptotic role of heme oxygenase (HO) and the potential of HO as a target in anticancer treatment. Apoptosis.2004,9, (1),27-35.
    15.刘秉乾;武玉东;马腾骥,et al血红素氧合酶-1对大鼠肾缺血再灌注损伤的保护作用.中化实验外科杂志.2004,12,1531-1532.
    16. Liu, ZM; Chen, GG; Ng, EK, et al. Upregulation of heme oxygenase-1 and p21 confers resistance to apoptosis in human gastric cancer cells. Oncogene.2004,23, (2),503-13.
    17. Ohyama, K; Akaike, T; Hirobe, C, et al. Cytotoxicity and apoptotic inducibility of Vitex agnus-castus fruit extract in cultured human normal and cancer cells and effect on growth. Biol Pharm Bull.2003,26, (1),10-8.
    18. Hill, M; Pereira, V; Chauveau, C, et al. Heme oxygenase-1 inhibits rat and human breast cancer cell proliferation:mutual cross inhibition with indoleamine 2,3-dioxygenase. Faseb J.2005,19, (14),1957-68.
    19. Berberat, PO; Dambrauskas, Z; Gulbinas, A, et al. Inhibition of heme oxygenase-1 increases responsiveness of pancreatic cancer cells to anticancer treatment. Clin Cancer Res. 2005,11,(10),3790-8.
    20. Was, H; Cichon, T; Smolarczyk, R, et al. Overexpression of heme oxygenase-1 in murine melanoma:increased proliferation and viability of tumor cells, decreased survival of mice. Am J Pathol.2006,169, (6),2181-98.
    21. Jozkowicz, A; Was, H; Dulak, J. Heme oxygenase-1 in tumors:is it a false friend? Antioxid Redox Signal.2007,9, (12),2099-117.
    22. Iwasa, H; Han, J; Ishikawa, F. Mitogen-activated protein kinase p38 defines the common senescence-signalling pathway. Genes Cells.2003,8, (2),131-44.
    23. Kim, HP; Wang, X; Galbiati, F, et al. Caveolae compartmentalization of heme oxygenase-1 in endothelial cells. Faseb J.2004,18, (10),1080-9.
    24. Ollinger, R; Bilban, M; Erat, A, et al. Bilirubin:a natural inhibitor of vascular smooth muscle cell proliferation. Circulation.2005,112, (7),1030-9.
    25. Ball, KL. p21:structure and functions associated with cyclin-CDK binding. Prog Cell Cycle Res.1997,3,125-34.
    26. Straume, O; Akslen, LA. Importance of vascular phenotype by basic fibroblast growth factor, and influence of the angiogenic factors basic fibroblast growth factor/fibroblast growth factor receptor-1 and ephrin-Al/EphA2 on melanoma progression. Am J Pathol. 2002,160,(3),1009-19.
    27. Genter, MB; Burman, DM; Vijayakumar, S, et al. Genomic analysis of alachlor-induced oncogenesis in rat olfactory mucosa. Physiol Genomics.2002,12, (1), 35-45.
    28. Sacca, P; Caballero, F; Batlle, A, et al. Cell cycle arrest and modulation of HO-1 expression induced by acetyl salicylic acid in hepatocarcinogenesis. Int J Biochem Cell Biol.2004,36, (10),1945-53.
    29. Yan, Y; Higashi, K; Yamamura, K, et al. Different responses other than the formation of DNA-adducts between the livers of carcinogen-resistant rats (DRH) and carcinogen-sensitive rats (Donryu) to 3'-methyl-4-dimethylaminoazobenzene administration. Jpn J Cancer Res.1998,89, (8),806-13.
    30. Mayerhofer, M; Florian, S; Krauth, MT, et al. Identification of heme oxygenase-1 as a novel BCR/ABL-dependent survival factor in chronic myeloid leukemia. Cancer Res.2004, 64, (9),3148-54.
    31. Marinissen, MJ; Tanos, T; Bolos, M, et al. Inhibition of heme oxygenase-1 interferes with the transforming activity of the Kaposi sarcoma herpesvirus-encoded G protein-coupled receptor. J Biol Chem.2006,281, (16),11332-46.
    32. Torisu-Itakura, H; Furue, M; Kuwano, M, et al. Co-expression of thymidine phosphorylase and heme oxygenase-1 in macrophages in human malignant vertical growth melanomas. Jpn J Cancer Res.2000,91, (9),906-10.
    33. Deininger, MH; Meyermann, R; Trautmann, K, et al. Heme oxygenase (HO)-1 expressing macrophages/microglial cells accumulate during oligodendroglioma progression. Brain Res.2000,882, (1-2),1-8.
    34. Doi, K; Akaike, T; Fujii, S, et al. Induction of haem oxygenase-1 nitric oxide and ischaemia in experimental solid tumours and implications for tumour growth. Br J Cancer. 1999,80, (12),1945-54.
    35. Sunamura, M; Duda, DG; Ghattas, MH, et al. Heme oxygenase-1 accelerates tumor angiogenesis of human pancreatic cancer. Angiogenesis.2003,6, (1),15-24.
    36. Kimpara, T; Takeda, A; Watanabe, K, et al. Microsatellite polymorphism in the human heme oxygenase-1 gene promoter and its application in association studies with Alzheimer and Parkinson disease. Hum Genet.1997,100, (1),145-7.
    37. Okamoto, I; Krogler, J; Endler, G, et al. A microsatellite polymorphism in the heme oxygenase-1 gene promoter is associated with risk for melanoma. Int J Cancer.2006,119, (6),1312-5.
    38. Chen, YH; Lin, SJ; Lin, MW, et al. Microsatellite polymorphism in promoter of heme oxygenase-1 gene is associated with susceptibility to coronary artery disease in type 2 diabetic patients. Hum Genet.2002,111, (1),1-8.
    39. Hirai, H; Kubo, H; Yamaya, M, et al. Microsatellite polymorphism in heme oxygenase-1 gene promoter is associated with susceptibility to oxidant-induced apoptosis in lymphoblastoid cell lines. Blood.2003,102, (5),1619-21.
    40. Exner, M; Schillinger, M; Minar, E, et al. Heme oxygenase-1 gene promoter microsatellite polymorphism is associated with restenosis after percutaneous transluminal angioplasty. J Endovasc Ther.2001,8, (5),433-40.
    41. Schillinger, M; Exner, M; Mlekusch, W, et al. Heme oxygenase-1 gene promoter polymorphism is associated with abdominal aortic aneurysm. Thromb Res.2002,106, (2), 131-6.
    42. Kikuchi, A; Yamaya, M; Suzuki, S, et al. Association of susceptibility to the development of lung adenocarcinoma with the heme oxygenase-1 gene promoter polymorphism. Hum Genet.2005,116, (5),354-60.
    43. Chang, KW; Lee, TC; Yeh, WI, et al. Polymorphism in heme oxygenase-1 (HO-1) promoter is related to the risk of oral squamous cell carcinoma occurring on male areca chewers. Br J Cancer.2004,91, (8),1551-5.
    44. Becker, JC; Fukui, H; Imai, Y, et al. Colonic expression of heme oxygenase-1 is associated with a better long-term survival in patients with colorectal cancer. Scand J Gastroenterol.2007,42, (7),852-8.
    45. Nowis, D; Legat, M; Grzela, T, et al. Heme oxygenase-1 protects tumor cells against photodynamic therapy-mediated cytotoxicity. Oncogene.2006,25, (24),3365-74.
    46. Jalili, A; Makowski, M; Switaj, T, et al. Effective photoimmunotherapy of murine colon carcinoma induced by the combination of photodynamic therapy and dendritic cells. Clin Cancer Res.2004,10, (13),4498-508.
    47. Hellmuth, M; Wetzler, C; Nold, M, et al. Expression of interleukin-8, heme oxygenase-1 and vascular endothelial growth factor in DLD-1 colon carcinoma cells exposed to pyrrolidine dithiocarbamate. Carcinogenesis.2002,23, (8),1273-9.
    48. Fang, J; Sawa, T; Akaike, T, et al. Enhancement of chemotherapeutic response of tumor cells by a heme oxygenase inhibitor, pegylated zinc protoporphyrin. Int J Cancer.2004,109, (1),1-8.

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700