新型一氧化氮供体-β半乳糖基化偶氮烯翁二醇的抗菌、抗肿瘤作用途径研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
一氧化氮(NO)是一种在胞内由三种不同的一氧化氮合酶(iNOS、nNOS、eNOS)产生的气态小分子,它在松弛血管平滑肌、抑制血小板凝集、协助免疫系统破坏肿瘤细胞和胞内病原体(病毒和细菌)、参与神经突触传递等多种生理和病理的过程中起到了重要作用。一氧化氮具有极短的半衰期并且容易变成多种活性氮基团(RNS),如三氧化二氮(N_2O_3)和过氧化亚硝酸根离子(ONOO~-)等,因此在治疗和研究中利用这个活泼的小分子是很困难的。由于气态一氧化氮不稳定且不方便利用,人们对一氧化氮供体能在体内通过分解、氧化、还原的多种机制产生RNS产生了越来越浓的兴趣。NO供体是指能够储存并稳定释放活性氮基团(RNS)的一类有机化合物。以前应用常规NO供体作为药物前体遇到许多困难,例如药品的运输缺乏靶向性,难以达到胞内有效剂量以及有毒副作用等。迄今为止,许多NO供体的前体药物已经得到了发展,例如,亚硝基铁氰化钠(SNP),羟基脲以及合成的羟基脲衍生物,NONOates。但是至今大多数传统的NO供体的应用受到限制,主要因为缺乏特异性、缺乏适当的靶向传递途径,导致了许多副作用发生。因此,现在迫切需要设计出一种新型的能使NO靶向定点释放的NO供体,该供体能直接将可调节剂量的药物传递至位点作用。我们小组合成并报导的β-半乳糖基化偶氮烯翁二醇(β-Gal-NONOate)是一种新型定点释放NO的化合物,它一旦受到β-半乳糖苷酶的激活作用就能够释放RNS(NO)。
     1.在本研究中,我们将β-Gal=NONOate作为杀菌剂来研究其杀菌效果、作用机理。通过测定胞内的NO水平及比较用β-Gal-NONOate和NONOate分别处理的E.coli DH5α(pUC18)的存活率,我们可以很明显地观察到β-Gal-NONOate比传统的NONOate具有更高的杀菌活性。同时我们也比较了分别用β-Gal-NONOate和NONOate处理的E.coli DH5α的存活率,这二者都显示出较低的杀菌活性。这结果说明了β-Gal-NONOate是一个高效的并且很有前景的NO供体。β-Gal-NONOate被证实可借助于己糖载体很容易传送进入细菌胞内。因此,一旦内部细胞产生β-半乳糖苷酶就会引起β-Gal-NONOate的水解,NO也就能迅速地从该化合物上释放下来,在胞内达到有效浓度。证明了β-Gal-NONOate的释放方式是一种新颖、有效的在细菌胞内定点释放RNS(NO)的途径。
     2.基于以上研究,我们确认β-Gal-NONOate是一个有显著杀菌活性的定位胞内释放的NO供体,在临床应用和相关研究中都具有重要意义。第三章初步探索了β-Gal-NONOate在消除细菌普遍存在的抗生素耐药性方面的作用。首先,采用E-test法测定最低抑菌浓度(MIC)来确定原始菌株对抗生素的耐受度,经抗生素多代诱导后得到Amp耐药性菌株,再用β-Gal-NONOate与NONOate分别处理,在一系列连续的传代培养后使细菌恢复对抗生素的敏感性。研究结果显示,β-Gal-NONOate较之于NONOate,能更有效地降低Amp抗性菌株的MIC值,即消除了细菌的耐药性;当与Amp一起进行亚培养时,β-Gal-NONOate与NONOate相比,能在更短的代数内杀死细菌,也就是说大大的缩短了抗生素耐药性的持续时间。这些结果进一步证实了β-Gal-NONOate作为一种NO供体,不仅可被用作单纯的杀菌剂,还可用于消除目前日益严重的细菌的抗生素耐药性。基于前期的研究,我们断定β-Gal-NONOate通过向胞内运送更多的NO,依靠迅速达到有效浓度的NO消除了细菌对抗生素的耐药性。
     3.深入研究抗生素耐药性的分子机制有利于提高现有抗菌药物的作用效果。双向电泳(two dimensional electrophoresis,2-DE)是蛋白质组学研究的关键核心技术之一。第四章应用蛋白质组学方法比较分析了E.coli K-12中与Amp抗性产生和消除相关的蛋白质,以了解抗性产生的途径。利用2-DE技术比较了三种大肠杆菌菌株(E.coli K-12、E.coli K-12 Amp抗性菌、E.coli K-12 Amp抗性消除菌)相互间在蛋白质水平上的差异。分别提取三种菌的蛋白质,依次进行等电聚焦电泳(IFE)和十二烷基磺酸钠聚丙烯酰胺凝胶电泳(SDS-PAGE),使用考马斯亮兰进行染色,经PDQuest7.2软件对结果进行比对、解析,以此研究β-Gal-NONOate对菌株在蛋白质水平上的影响。最终确认了β-Gal-NONOate可以通过改变细菌中蛋白质的水平来发挥消除Amp耐药性的作用。
     4.迄今为止,NO供体已经被用于抗肿瘤治疗的各个方面。为了能选择性地杀伤肿瘤细胞、避免不必要的副作用,第五章中β-Gal-NONOate被用到了抗肿瘤研究上。首次在9L/LacZ细胞中通过测定胞内释放NO的水平和抗肿瘤活性,证明了它更优于其前体NONOate。β-Gal-NONOate只在β-半乳糖苷酶激活下才会在9L/LacZ细胞内水解释放NO,这决定了它比NONOate具有更强的细胞毒性。结果显示:在9L/LacZ细胞内等浓度的情况下,β-Gal-NONOate产生的NO水平要高于NONOate,从而导致更高的抗肿瘤活性。然而,β-Gal-NONOate对9L细胞的毒性却不及NONOate。因此,β-Gal-NONOate是一种具有靶点特异性的药物前体,它是依赖于β-半乳糖苷酶的存在而发挥作用的定位胞内释放NO的NO供体,并很有希望成为一种有前途的探针和有潜力的新型抗肿瘤治疗药物。本研究首次设计并证明了一个新颖的肿瘤细胞内定点释放NO的药物传递途径,更容易使药物在胞内达到有效浓度,减毒增效降低毒副作用。这为新型抗肿瘤药物的开发提供了新的思路。
     5.第六章进一步比较研究了偶氮烯翁二醇(diazeniumdiolate,NONOate)与β-Gal-NONOate(β-galactosyl-diazeniumdiolate)的抗肿瘤作用。利用阳离子脂质体介导法,将真核表达载体pcDNA3-LacZ转染到HeLa细胞中;经G418(Geneticin)筛选,获得了稳定表达β-半乳糖苷酶的HeLa/LacZ细胞株。本研究由大鼠胶质瘤C6和C6/LacZ细胞,拓展到了人宫颈癌细胞模型,更全面地证实了β-Gal-NONOate的抗肿瘤作用和给药途径。本章通过Griess法比较了β-Gal-NONOate与NONOate在四种细胞中NO的释放量,根据细胞克隆形成率、细胞计数和MTT法比较β-Gal-NONOate与NONOate对细胞的抑制作用。结果显示β-Gal-NONOate在含有LacZ基因的两个细胞系中的NO释放量与药物浓度呈剂量依赖关系,并在相同药物浓度条件下明显高于NONOate;而β-Gal-NONOate在不表达β-半乳糖苷酶的C6、HeLa细胞中未检测到NO释放;β-Gal-NONOate对带有LacZ的两个细胞系的抗肿瘤活性显著高于NONOate(P<0.05),但对C6、HeLa细胞则没有明显的抑制作用;NONOate在含或不含LacZ的四个细胞系中的NO释放量和抑制作用效果都没有显著差异。本章通过C6细胞及在人宫颈癌HeLa细胞中转入常见报告基因LacZ,再次证实了β-Gal-NONOate比NONOate更加稳定并具有更高的抗肿瘤活性,其作用的发挥依赖于对β-半乳糖苷酶的选择性,是一个有效的定位胞内释放的NO供体。作为抗肿瘤研究中的一种新型探针,本研究将β-Gal-NONOate的应用范围扩大到了对人肿瘤细胞的基因治疗层面上,预示了其未来在肿瘤的治疗和研究中的应用前景。
     6.第七章研究了新型NO供体β-Gal-NONOate对大鼠胶质瘤9L/LacZ、C6/LacZ细胞以及人子宫颈癌HeLa/LacZ细胞的诱导凋亡作用。将稳定表达β-半乳糖苷酶的三个细胞株经β-Gal-NONOate处理后,通过:光镜、荧光显微观察细胞形态学结构的改变和典型凋亡特征的出现、琼脂糖凝胶电泳观察凋亡典型的DNA ladder、原位末端标记法(TUNEL)检测凋亡细胞的DNA断裂、流式细胞仪(FCM)Annexin V-FITC/PI双染色测定细胞凋亡比例,证明了β-Gal-NONOate能够诱导三种细胞凋亡,药物处理组的细胞呈现出典型的凋亡特征,并且随着药物浓度和时间的增加,这种变化更加明显;细胞凋亡大于坏死,比例在一定范围内与药物浓度成正相关。应用激光共聚焦显微镜(LSCM)和特异荧光探针,对β-Gal-NONOate作用前后三种细胞内游离Ca~(2+)浓度([Ca~(2+)]_i)和线粒体跨膜电位(ΔΨm)的变化进行了精确测定。β-Gal-NONOate处理后,细胞内游离钙离子的荧光强度持续、较大幅度的升高,而线粒体跨膜电位迅速降低。由此可见,β-Gal-NONOate在胞内释放了NO后可能通过直接调节细胞膜上的Ca~(2+)通道或同时在胞内释放内质网、线粒体等细胞内钙库中与钙结合蛋白松弛结合的游离钙,引起胞内[Ca~(2+)]_i的变化,并将这种信号传递给线粒体。线粒体随即发生通透性变化,跨膜电位下降,引发线粒体功能改变,激活凋亡的关键酶Caspase级联放大,导致线粒体释放Cytc、ROS等一系列诱导凋亡因子。最后在Caspase、Bcl-2及核酸内切酶等的共同作用下,使细胞进入死亡程序。β-Gal-NONOate释放的NO或RNS可能进入细胞核,影响复制、转录、翻译等各个环节,进一步引起染色体的聚集、断裂、DNA ladder等凋亡特征性的形态和生化改变。本研究还利用MTT法证实了β-Gal-NONOate与常用抗肿瘤药cisplatin联用有增效作用(q>1),这对临床抗肿瘤药物的减毒增效研究有重要的指导意义。
NO, which is a paradoxical gaseous messenger enzymatically generated in vivo by three isoforms of nitric oxide synthase (iNOS, nNOS and eNOS), plays an important role in numerous physiological and pathophysiological processes including relaxing vascular smooth muscle, inhibiting platelet aggregation, assisting the immune system in destroying tumor cells and intracellular pathogens and participating in neuronal synaptic transmission. Unfortunately, NO has extremely short half-life and is easily converted into a variety of reactive nitrogen species (RNS), such as dinitrogen trioxide (N_2O_3), nitrogen dioxide (NO_2), and the peroxynitrite anion (ONOO~-), so it is difficult to utilize this active small molecule in therapy and research. Thus site-specific delivery of exogenous NO becomes an attractive therapeutic option in the treatment of disease. Due to the instability and inconvenient handling of aqueous NO solutions, there has been an increasing interest in using NO donors capable of generating RNS in vivo by a variety of mechanisms including decomposition,
    oxidation, or reduction.
    1. β-galactosyl-pyrrolidinyl diazeniumdiolates (β-Gal-NONOate) is a new
    site-specific NO-releasing compound reported by our group, which releases RNS(NO) once activated by β-galactosidase. In Chapter 2, β-Gal-NONOate was used as a bactericidal reagent to determine its effectiveness of NO releasing. Through the evaluation of intracellular NO level and the comparison of survival of E. coli transformed with lacZ gene but treated with β-Gal-NONOate and NONOate, respectively, it's evident that β-Gal-NONOate had a higher bactericidal activity than conventional NONOate. While either β-Gal-NONOate- or NONOate-treated-E. coli without transferred lacZ gene manifested low bactericidal activity. The results revealed that β-Gal-NONOate was a high effective and promising NO donor. It was presumed that β-Gal-NONOate could be easily transported into the cells via sugar transporters. Thus, once it was hydrolyzed by β-galactosidase produced inside cells, NO was readily released from the compound. Therefore it took on a novel and efficient approach to deliver RNS (NO) into cells.
    2. β-Gal-NONOate, which has been reported as a site-specific NO donor with high antibacterial activity(in Chapter 2) is an intriguing agent in therapy and related research. Through this study, β-Gal-NONOate was proved to be promising in overcoming prevalent antibiotic resistance of bacteria. It was applied to get rid of ampicillin resistance of E. coli K-12 with β-galactosidase activity. Bacterial susceptibility to antibiotics was evaluated with the minimum inhibitory concentration (MIC), which could be measured with E-test After successful inducement of ampicillin-resistant strains, β-Gal-NONOate and NONOate were separately applied to restore antibiotic susceptibility of bacteria through a series of sequential subcultures. The results demonstrated that β-Gal-NONOate possessed greater potential to decrease the MIC of ampicillin-resistant strains than NONOate; while combined with ampicillin for subculture, β-Gal-NONOate was able to kill off the resistant strains within fewer passages than NONOate, that is, to greatly shorten the duration of antibiotic resistance. These results further substantiated that β-Gal-NONOate was a very prospective NO donor, which could be explored and utilized not only as antimicrobial but also as a promising agent to remove increasingly serious antibiotic resistance of bacteria in current times. Based on our previous studies, it is concluded that elimination of antibiotic resistance may be due to more site-specific NO delivery into cells, accessibility of efficaciously intracellular NO level and high efficient antimicrobial roles of β-Gal-NONOate.
    3. The elucidation of the molecular details of antibiotic resistance will lead to improvements in extending the efficacy of current antimicrobials. Two dimensional electrophoresis(2-DE) is a key technique in studies of proteome. In Chapter 4, proteomic methodologies were applied for the comparative analysis of proteome of E. coli K-12 responded to ampicillin(Amp) resistance and Amp re-susceptivity for understanding of universal pathways that form barriers for antimicrobial agents. For this purpose, three kinds of E. coli K-12(E coli K-12 ancestor strain, E. coli K-12 Amp resistant strain, E. coli K-12 re-susceptible strain) proteome were characterized with the use of 2-DE methods. Then, differential proteins due to Amp resistance were determined by comparing with each other using PDQuest7.2 to study the influence on bacterium in proteome by β-Gal-NONOate. Our findings will be helpful for further understanding of antibiotic-resistant / overcoming resistance mechanism related to proteome. This study also confirmed that β-Gal-NONOate certainly contributes the elimination of Amp by changing the protein expression. 4. So far, nitric oxide (NO) donors have been applied to various aspects of antitumor therapy. To selectively sensitize tumor cells and avoid unwanted side effects, β-Gal-NONOate was firstly used in the cancer research. In this study, we first verified its superiority over its parent NONOate in terms of targeted intracellular NO-releasing and antitumor activity with 9L/LacZ cells in vitro. β-Gal-NONOate only released NO when hydrolyzed by β-galactosidase in 9L/LacZ cells, which led to its more powerful cytotoxicity than that of NONOate. The results showed that β-Gal-NONOate produced higher NO levels than NONOate in 9L/LacZ cells at equal concentration, and hence induced optimal NO levels for antitumor activity. However, in 9L cells, β-Gal-NONOate showed less toxicity than NONOate. Therefore, it is demonstrated that β-Gal-NONOate is a site-specific prodrug for targeting NO intracellularly as a β-galactosidase-sensitive NO donor, and it is also expected to be a promising probe in numerous experimental settings and a potential therapeutic drug for antitumor treatment.
    5. In order to evaluate the antitumor effects of β-Gal-NONOate in details and to clarify its mechanism to action as an antitumor prodrug, four cell lines were used in Chapter 6. Through cationic liposome-mediated transfection, the eukaryotic expression vector pcDNA3-LacZ was transferred into the HeLa cells; through G418 screening, the HeLa/LacZ cell line steadily expressing β-galactosidase was obtained. With C6/LacZ、 C6、 HeLa/LacZ and HeLa cells as in vitro model, β-galactosidase activities of tumor cells were determined through X-gal staining. NO levels in these four cell lines released from β-Gal-NONOate and NONOate were evaluated with Griess assay. Antitumor effects of NO donors on the four cell lines were estimated by cell counting, cell colony forming rate and MTT (Methylthiazoletetrazolium) assay, respectively. We found that NO levels released from β-Gal-NONOate in C6/LacZ and HeLa/LacZ cells were dependent on its concentration and evidently higher than that from NONOate. And similarly, its antitumor activity to C6/LacZ and HeLa/LacZ cells were obviously more powerful than NONOate(P < 0.05), but it did not exhibit conspicuous cytotoxicity to C6 cells. However, the NO released from NONOate inside the four cell lines and the effects on all kinds of cells didn't have great difference. We could draw a conclusion that β-Gal-NONOate was more stable and possessed higher antitumor activity than NONOate. In addition, its action greatly depended on β-galactosidase. It was an effective intracellularly NO release compound, which would be very promising in tumor therapy and relevant research.
    6. The apoptosis effects of 9L/LacZ, C6/LacZ and HeLa/LacZ cells induced by β-Gal-NONOate were investigated in Chapter 7. Typical apoptotic morphological features included cell shrinkage and condensation and margination of nuclear chromatin were showed by light microscopy. Condensed nuclear chromatin and the morphology of nuclei were demonstrated by fluorescent microscopy with AO and Hoechst 33342 staining. β-Gal-NONOate induced apoptotic DNA breaks were confirmed by a typical "DNA ladder" on agarose gel electrophoresis as well as evidenced by TUNEL assay. β-Gal-NONOate induced apoptosis in a concentration-dependent manner. The apoptotic rates of three cells were measured by flow cytometry with Annexin V-FITC and PI staining. The changes of intracellular [Ca~(2+)]_i and mitochondrial transmembrane potential (ΔΨm) were detected using fluorescence indicator Fluo-3/AM and Rhodamine 123 with laster scanning confocal microscopy. β-Gal-NONOate resulted in a rapid increase in [Ca~(2+)]_i and a great decrease in ΔΨm. The results of this study confirmed the ability of β-Gal-NONOate to suppress the proliferation of three kinds of cells with typical apoptosis feature in vitro. Disturbance of homeostasis in calcium signaling system might play pivotal roles in apoptosis of three cells. Effects of β-Gal-NONOate were assessed both alone and in paired combination with cisplatin. Q values were used to characterize the interactions as synergistic, additive, or antagonistic. Significant synergistic effects in growth inhinition of β-Gal-NONOate (0.25-10 mM) with cisplatin (2.5-10 μM) on three cell lines were observed (q>1). We conclude that β-Gal-NONOate may be worth of further studies assessing its value in cancer therapy in combination with the other chemotherapeutic agents.
引文
[1] Marietta, M. A., Hurshman, A. R., Rusche, K. M. Catalysis by nitric oxide synthase. Curr. Opin. Chem. Biol., 1998, 2(5): 656-663.
    [2] 许样裕,朱敏生,潘英,等诱生型一氧化氮合酶抗体的制备与初步应用.中国生化药物杂志,2001,22(4):169-171.
    [3] Ignarro, L., Murad, F. "Nitric Oxide: Biochemistry, Molecular Biology, and Therapeutic Implications," Academic, San Diego, CA, 1995,
    [4] 赵志光,谭玲玲,王锁民,等植物一氧化氮(NO)研究进展.植物学通报,2002,19(6):659-665.
    [5] Denninger, J. W., Marietta, M. A. Guanylate cyclase and the NO/cGMP signaling pathway. Biochim. Biophys. Acta, 1999, 1411(2-3): 334-350.
    [6] 马雪莲,李菁,李跃华,等Glucan对氰化钾致肝细胞损伤的保护作用.中国生化药物杂志,2003,24(1):20-22.
    [7] 吴俊芳,王平,张均田 神经生长因子对一氧化氮介导的大鼠脑皮质神经元毒性的影响.中国生化药物杂志,1999,20(4):170-173.
    [8] 嵇扬 胸腺肽对小鼠腹腔巨噬细胞释放一氧化氮的影响.中国生化药物杂志,2001,22(2):74-75.
    [9] 徐军发,侯敢,黄迪南,等酵母多糖对小鼠腹腔巨噬细胞产生一氧化氮和白细胞介素-1的影响.中国生化药物杂志,2002,23(2):67-68.
    [10] Jia, Q., Janezuk, A. J., Cai, T, Xian, M., Wen, Z. NO donors with anticancer activity. Expert Opin. Ther. Patents, 2002, 12(6): 819-826.
    [11] 陈莉,张奕华 前药原理在非甾体抗炎药中的应用.中国药师,2003,6(1):50-52.
    [12] Bandarage, U. K., Chen, L., Fang, X., Garvey, D. S, Glavin, A., Janero, D. R., Letts, L. G., Mercer, G. J., Saha, J. K., Schroeder, J. D., Shumway, Mo J., Tam, S. W. Nitrosothiol esters of diclofenac: synthesis and pharmacological characterization as gastrointestinal-sparing prodrugs. J Med. Chem., 2000, 43(21): 4005-4016.
    [13] de Tejada, I. S., Garvey, D. S, Schroeder, J. D., Shelekhin, T, Letts, L. G., Fernandez, A., Cuevas, B., Gabancho, S., Martinez, V., Angulo, J., Trocha, M., Marek, P., Cuevas, P., Tam, S. W. Design and evaluation of nitrosylated alpha-adrenergic receptor antagonists as potential agents for the treatment of impotence. J. Pharmacol. Exp. Ther., 1999, 290(1): 121-128.
    [14] Whittle, B. J. Temporal relationship between cyclooxygenase inhibition, as measured by prostacyclin biosynthesis, and the gastrointestinal damage induced by indomethacin in the rat. Gastroenterology, 1981, 80(1): 94-98.
    [15] Wallace, J. L. Nonsteroidal anti-inflammatory drugs and gastroenteropathy: the second hundred years. Gastroenterology, 1997, 112(3): 1000-1016.
    [16] Lechi, C., Andrioli, G., Gaino, S., Tommasoli, R., Zuliani, V., Ortolani, R., Degan, M., Benoni, G., Bellavite, P, Lechi, A., Minuz, P. The antiplatelet effects of a new nitroderivative of acetylsalicylic acid——an in vitro study of inhibition on the early phase of platelet activation and on TXA2 production. Thromb. Haemost., 1996, 76(5): 791-798.
    [17] Wallace, J. L., Museara, M. N., McKnight, W., Dicay, M., Del Soidato, P., Chino, G. In vivo antithrombotic effects of a nitric oxide-releasing aspirin derivative, NCX-4016. Thromb. Res., 1999, 93(1): 43-50.
    [18] Wallace, J. L., Vergnolle, N., Muscara, M. N., Asfaha, S., Chapman, K., McKnight, W., Del Soldato, P., Moreili, A., Fiorucci, S. Enhanced anti-inflammatory effects of a nitric oxide-releasing derivative of mesalamine in rats. Gastroenterology, 1999, 117(3): 557-566.
    [19] Paul-Clark, M., Del Soldato, P., Fiorucci, S., Flower, R. J., Perretti, M. 21-NO-prednisolone is a novel nitric oxide-releasing derivative of prednisolone with enhanced anti-inflammatory properties. Br. J. Pharmacol., 2000, 131(7): 1345-1354.
    [20] Rieardo, d. L. B., Roque, D. C. C., Marcelo, N. M. Nitric oxide: properties and therapeutic use. Quim. Nova, 2005, 28(6): 1046-1054.
    [21] Endres, S., Hacker, A., Noaek, E., Kojda, G., Lehmann, J. NO-Donors, part 3: nitrooxyacylated thiosalicylates and salicylates - synthesis and biological activities. Eur. J. Med Chem., 1999, 34(11): 895-901.
    [22] Lehmann, J., Kahlich, R., Meyer zum Gottesberge, C., Fricke, U. Organic nitrates. Ⅱ. Synthesis and biological activities of 4-nitrooxymethylphenyl-1,4-dihydropyridines. Arch. Pharm. (Weinheim), 1997, 330(8): 247-252.
    [23] Haj-Yehia, A., Nassar, T., Lotan, C. Development of 3-nitratomethyl-proxyl (NMP): a novel, bifunctional superoxide dismutase-mimic-nitricoxide-donor. Drug Dev. Res., 2000, 50(314): 528-536.
    [24] Fruttero, R., Beschi, D, Di Stile, A, Gasee, A. The furoxan system as a useful tool for balancing "hybrids" with mixed alpha 1-antagonist and NO-like vasodilator activities. J Med. Chem., 1995, 38(25): 4944-4949.
    [25] Di Stilo, A., Visentin, S., Cena, C, Gasco, A. M., Ermondi, G., Gasco, A. New 1,4-dihydropyridines conjugated to furoxanyl moieties, endowed with both nitric oxide-like and calcium channel antagonist vasodiator activities. J Med. Chem, 1998, 41(27): 5393-5401.
    [26] Visentin, S., Amid, P, Fruttero, R., Besehi, D, Roussel, C., Giusta, L., Carbone, E., Gasco, A. Synthesis and voltage-clamp studies of methyl 1,4-dihydro-2, 6-dimethyl-5-nitro-4-(benzofurazanyl)pyridine-3-carboxylate racemates and enantiomers and of their benzofuroxanyl analogues. J Med. Chem., 1999, 42(8): 1422-1427.
    [27] Mu, L., Feng, S. S., Go, M. L. Study of synthesis and cardiovascular activity of some furoxan derivatives as potential NO-donors. Chem. Pharm. Bull., 2000, 48(6): 808-816.
    [28] Keefer, L. K. Nitric oxide-releasing compounds: From basic research to promising drugs. Chemtech, 1998, 28(8): 30-35.
    [29] Saavedra, J. E., Shami, P. J., Wang, L. Y., Davies, K. M., Booth, M. N., Citro, M. L., Keefer, L. K. Esterase-sensitive nitric oxide donors of the diazeniumdiolate family: in vitro antileukemic activity. J. Med Chem., 2000, 43(2): 261-269.
    [30] Saavedra, J. E., Billiar, T. R., Williams, D. L., Kim, Y. M., Watkins. S. C., Keefer, L. K. Targeting nitric oxide (NO) delivery in vivo. Design of a liver-selective NO donor prodrug that blocks tumor necrosis factor-alpha-induced apoptosis and toxicity in the liver. J. Med. Chem., 1997, 40(13): 1947-1954.
    [31] Hou, Y., Xie, W., Janczuk, A. J., Wang, P. G. O-Alkylation of Cupferron: Aiming at the Design and Synthesis of Controlled Nitric Oxide Releasing Agents. Nature, 1994, 368: 62.
    [32] Tang, X., Xian, M., Trikha, M., Honn, K. V., Wang, P. G. Synthesis of peptide-diazeniumdiolate conjugates: towards enzyme activated antitumor agents. Tetrahedron Lett., 2001, 42(14): 2625-2629.
    [33] Wu, X., Tang, X., Xian, M., Wang, P. G. Glycosylated diazeniumdiolates: a novel class of enzyme-activated nitric oxide donors. Tetrahedron Lett., 2001, 42(23): 3779-3782.
    [34] 任昉,方治平 青霉素结合蛋白与金黄色葡萄球菌对β-内酰胺类抗生素的耐药机制.四川生理科学杂志,2001,23(2):52-54.
    [35] 刘永芳,吕晓菊 铜绿假单胞菌对碳青霉烯类抗生素的耐药性.中国抗生素杂志,2005,30(11):699-704.
    [36] McMurry, L., Petrucci, R. E., Jr., Levy, S. B. Active efflux of tetracycline encoded by four genetically different tetracycline resistance determinants in Escherichia coli. Proc. Natl. Acad Sci. U. S. A., 1980, 77(7): 3974-3977.
    [37] Ross, J. I., Eady, E. A., Cove, J. H., Cunliffe, W. J., Baumberg, S., Wootton, J. C. Inducible erythromycin resistance in staphylococci is encoded by a member of the ATP-binding transport super-gene family. Mol. Microbiol., 1990, 4(7): 1207-1214.
    [38] Li, X. Z., Ma, D., Livermore, D. M., Nikaido, H. Role of efflux pump(s) in intrinsic resistance of Pseudomonas aeruginosa: active efflux as a contributing factor to beta-lactam resistance. Antimicrob. Agents Chemother., 1994, 38(8): 1742-1752.
    [39] Aires, J. R., Kohler, T., Nikaido, H., Plesiat, P. Involvement of an active efflux system in the natural resistance of Pseudomonas aeruginosa to aminoglycosides. Antimicrob. Agents Chemother., 1999, 43(11): 2624-2628.
    [40] Nikaido, H. Prevention of drug access to bacterial targets: permeability barriers and active efflux. Science, 1994, 264(5157): 382-388.
    [41] Okusu, H., Ma, D., Nikaido, H. AcrAB efflux pump plays a major role in the antibiotic resistance phenotype of Escherichia coli multiple-antibiotic-resistance (Mar) mutants. J Bacteriol., 1996, 178(1): 306-308.
    [42] Wright, G. D. Bacterial resistance to antibiotics: enzymatic degradation and modification. Adv. Drug Delivery Rev., 2005, 57(10): 1451-1470.
    [43] 马涛,徐文方 β-内酰胺酶与细菌耐药性的研究进展.国外医药—合成药,生化药,制剂分册,2000,21(4):199-201.
    [44] Park, B. H., Levy, S. B. The cryptic tetracycline resistance determinant on Tn4400 mediates tetracycline degradation as well as tetracycline efflux. Antimicrob. Agents Chemother., 1988, 32(12): 1797-1800.
    [45] Whittle, G., Hund, B. D., Shoemaker, N. B., Salyers, A. A. Characterization of the 13-kilobase ermF region of the Bacteroides conjugative transposon CTnDOT. Appl. Environ. Microbiol., 2001, 67(8): 3488-3495.
    [46] Speer, B. S., Bedzyk, L., Salyers, A. A. Evidence that a novel tetracycline resistance gene found on two Bacteroides transposons encodes an NADP-requiring oxidoreductase. J. Bacteriol., 1991, 173(1): 176-183.
    [47] Maisnier-Patin, S., Anderson, D. I. Adaptation to the deleterious effects of antimicrobial drug resistance mutations by compensatory evolution. Res. Microbiol., 2004, 155(5): 360-369.
    [48] 李明,蔡锦兰,张瑞苓科学合理的使用抗生素.中华临床医学研究杂志,2006,12(5):633-634.
    [49] Bonfigfio, G., Russo, G., Nieoletti, G. Recent developments in carbapenems. Expert Opin. Invest. Drugs, 2002, 11(4): 529-544.
    [50] 陆玉峰 生物代谢基因调控在抗生素生物合成中的应用.中国生化药物杂志志,2003,24(2):91-94.
    [51] 诸林俏,黄晨 β-内酰胺酶抑制剂复合制剂的临床应用.海峡药学,2003,15(5):97-98.
    [52] 苏金石,谢峥,杨宗芳 抗生素耐药性的产生及预防.广西药学杂志,2003,18(4):279-280.
    [53] Qiu, X. Q., Wang, H., Lu, X. F., Zhang, J., Li, S. F., Cheng, G., Wan, L., Yang, L., Zuo, J. Y., Zhou, Y. Q., Wang, H. Y., Cheng, X., Zhang, S. H., On, Z. R., Zhong, Z. C., Cheng, J. Q., Li, Y. P., Wu, G. Y. An engineered multidomain bactericidal peptide as a model for targeted antibiotics against specific bacteria. Nat. Biotechnol., 2003, 21(12): 1480-1485.
    [54] Guerrier-Takada, C., Salavati, R., Altman, S. Phenotypic conversion of drug-resistant bacteria to drug sensitivity. Proc. Natl. Acad. Sci. U. S. A., 1997, 94(16): 8468-8472.
    [55] 万齐全,齐俊英,陈如外源指导序列逆转临床大肠埃希菌耐药性研究.中国传染病杂志,2004,22(1):27-29.
    [56] Khuller, G. K., Kapur, M., Sharma, S. Liposome technology for drug delivery against mycobacterial infections. Curr. Pharm. Des., 2004, 10(26): 3263-3274.
    [57] 罗云敬,刘海涛,李天剑,等 头孢菌素类抗生素脂质体的包封率测定与渗漏研究.中国生化药物杂志,1999,20(1):4-6.
    [58] 李松,陈畅,史艳秋,顾黎,王鹏 一氧化氮供体最新研究进展及应用前景.中国生化药物杂志,2006,27(6):374-378.
    [59] 胡章颜 一氧化氮与细胞凋亡的研究进展.江西医学院学报,2005,45(3):176-178.
    [60] Nicotera, P., Brune, B., Bagetta, G. Nitric oxide: inducer or suppressor of apoptosis? Trends Pharmacol. Sci., 1997, 18(6): 189-190.
    [61] Dimmeler, S., Zeiher, A. M. Nitric oxide and apoptosis: another paradigm for the double-edged role of nitric oxide. Nitric Oxide, 1997, 1(4): 275-281.
    [62] Boyd, C. S., Cadenas, E. Nitric oxide and cell signaling pathways in mitochondrial-dependent apoptosis. Biol. Chem., 2002, 383(3-4): 411-423.
    [63] Bosca, L., Hortelano, S. Mechanisms of nitric oxide-dependent apoptosis: involvement of mitochondrial mediators. Cell. Signal., 1999, 11(4): 239-244.
    [64] Daiber, A., Frein, D., Namgaladze, D., Ullrich, V. Oxidation and nitrosation in the nitrogen monoxide/superoxide system. J. Biol. Chem., 2002, 277(14): 11882-11888.
    [65] Moriya, R., Uehara, T., Nomura, Y. Mechanism of nitric oxide-induced apoptosis in human neuroblastoma SH-SYSY cells. FEBS Lett., 2000, 484(3): 253-260.
    [66] Kim, Y. M., Chung, H. T., Simmons, R. L., Billiar, T. R. Cellular non-heme iron content is a determinant of nitric oxide-mediated apoptosis, necrosis, and caspase inhibition. J. Biol. Chem., 2000, 275(15): 10954-10961.
    [67] Bal-Price, A., Brown, C,. C. Nitric-oxide-induced necrosis and apoptosis in PC12 cells mediated by mitochondria. J. Neurochem., 2000, 75(4): 1455-1464.
    [68] Kurose, I., Miura, S., Higuchi, H., Watanabe, N., Kamegaya, Y., Takaishi, M., Tomita, K., Fukumura, D., Kato, S., Ishii, H. Increased nitric oxide synthase activity as a cause of mitochondrial dysfunction in rat hepatocytes: roles for tumor necrosis factor alpha. Hepatology, 1996, 24(5): 1185-1192.
    [69] Martin, S., Diaz, C,., Caside, M. Bacterial lipoplysaccaride antagonizes Transforming growth factor-13: induced apoptosis in primary cultures of hepatocytes. Hepatology, 1996, 23(5): 1200.
    [70] Zeini, M., Hortelano, S., Traves, P. G., Gomez-Valades, A. G., Pujol, A., Perales, J. C., Bartrons, R., Bosca, L. Assessment of a dual regulatory role for NO in liver regeneration after partial hepatectomy: protection against apoptosis and retardation of hepatocyte proliferation. FASEB J., 2005, 19(8): 995-997.
    [71] Maulik, N., Kagan, V. E., Tyurin, V. A., Das, D. K. Redistribution of phosphatidylethanolamine and phosphatidylserine precedes reperfusion-induced apoptosis. Am. J. Physiol., 1998, 274(1 Pt 2): H242-248.
    [72] 陈宏,蔡德鸿,王梅,等氨基胍对白介素-1β损伤的离体大鼠胰岛细胞保护作用 中国病理生理杂志,1999,15(7):628-630.
    [73] Henningsson, R., Salehi, A., Lundquist, I. Role of nitric oxide synthase isoforms in glucose-stimulated insulin release. Am. H. Physiol., 2002, 283(1): C296-304.
    [74] 袁涛,王姮 Ⅰ型糖尿病和β细胞凋亡.国外医学内分泌学分册,2002,22(1):28-31.
    [75] Andren-Sandberg, A., Backman, P. L., Andersson, R. Results of adjuvant therapy in resected pancreatic cancer. Int. J. Pancreatol., 1997, 21(1): 31-38.
    [76] Matthews, N. E., Adams, M. A., Maxwell, L. R., Gofton, T. E., Graham, C. H. Nitric oxide-mediated regulation of chemosensitivity in cancer cells. J. Natl. Cancer Inst., 2001, 93(24): 1879-1885.
    [77] Louis, J. I. Nitric Oxide: A Unique Endogenous Signaling Molecule in Vascular Biology (Nobel Lecture). Angew. Chem. Int. Ed. Engl., 1999, 38(13-14): 1882-1892.
    [78] Robert, F. F. Endothelium-Derived Relaxing Factor: Discovery, Early Studies, and Identification as Nitric Oxide (Nobel Lecture). Angew. Chem. Int. Ed. Engl., 1999, 38(13-14): 1870-1880.
    [79] Pfeiffer, S., Mayer, B., Hemmens, B. Nitric Oxide: Chemical Puzzles Posed by a Biological Messenger. Angew. Chem. Int. Ed. Engl., 1999, 38(12): 1714-1731.
    [80] Feldman, P. L., Griffith, O. W., Stuehr, D. J. The surprising life of nitric oxide. Chem. Eng. News, 1993, 71(51): 26-38.
    [81] Bredt, D. S., Snyder, S. H. Nitric oxide: a physiologic messenger molecule. Annu. Rev. Biochem., 1994, 63: 175-195.
    [82] Liu, J., Waalkes, M. P. Nitric oxide and chemically induced hepatotoxicity: beneficial effects of the liver-selective nitric oxide donor, V-PYRRO/NO. Toxicology, 2005, 208(2): 289-297.
    [83] Weller, R. Nitric oxide donors and the skin: useful therapeutic agents? Clin. Sci., 2003, 105(5): 533-535.
    [84] Serra, P. A., Rocchitta, G., Delogu, M. R., Migheli, R., Taras, M. G., Mura, M. P., Esposito, G., Miele, E., Desole, M. S., Miele, M. Role of the nitric oxide/cyclic GMP pathway and extracellular environment in the nitric oxide donor-induced increase in dopamine secretion from PC12 cells: a microdialysis in vitro study. J. Neurochem., 2003, 86(6): 1403-1413.
    [85] Wang, P. G., Xian, M., Tang, X., Wu, X., Wen, Z., Cai, T., Janczuk, A. J. Nitric oxide donors: chemical activities and biological applications. Chem. Roy., 2002, 102(4): 1091-1134.
    [86] Sambrook, J., Fritsch, E. F., Maniatis, T. "Molecular Cloning," A laboratory manual 2ned., cold spring harbor laboratory press, New York, 1989, pp. 808-809.
    [87] Green, L. C., Wagner, D. A., Glognwski, J., Skipper, P. L., Wishnok, J. S., Tannenbaum, S. R. Analysis of nitrate, nitrite, and [15N]nitrate in biological fluids. Anal. Biochem., 1982, 126(1): 131-138.
    [88] Schmidt, F., Ungureanu, I., Duvai, R., Pompon, A., Monneret, C. Cancer Chemotherapy: A Paclitaxel Prodrug for ADEPT (Antibody-Directed Enzyme Prodrug Therapy). Eur. J. Org. Chem., 2001, 2001(11): 2129-2134.
    [89] Desbene, S., Van, H. D., Michel, S., Tillequin, F., Koch, M., Schmidt, F., Fiorent, J. C., Monneret, C., Straub, R., Czech, J., Gerken, M., Bosslet, K. Application of the ADEPT strategy to the MDR resistance in cancer chemotherapy. Anticancer. Drug Des., 1999, 14(2): 93-106.
    [90] Radomski, M. W., Moncada, S. "Clinical Relevance of Nitric Oxide in the Cardiovascular System," eds. by Moncada, S., Higgs, E. A., Berrazueta, E. R., Edicomplet, Madrid, Spain, 1991, pp. 45-56.
    [91] Battinelli, E. M., Loscalzo, J. "Nitric Oxide Cardiovascular System," eds. by Loscalzo, J., Vita, J. A., Humana Press, Totowa, 2000, pp. 123-138.
    [92] Brennan, P. A. The actions and interactions of nitric oxide in solid tumours. Eur. J. Surg. Oncol., 2000, 26(5): 434-437.
    [93] Brunelli, L., Crow, J. P., Beckman, J. S. The comparative toxicity of nitric oxide and peroxynitrite to Escherichia coli. Arch. Biochem. Biophys., 1995, 316(1): 327-334.
    [94] Saier, M. H., Jr. Families of transmembrane sugar transport proteins. Mol. Microbiol., 2000, 35(4): 699-710.
    [95] Binder, S., Levitt, A. M., Sacks, J. J., Hughes, J. M. Emerging infectious diseases: public health issues for the 21st century. Science, 1999, 284(5418): 1311-1313.
    [96] Shank, J. L., Silliker, J. H., Harper, R. H. The effect of nitric oxide on bacteria. Appl. Microbial., 1962, 10: 185-189.
    [97] Wolkow, P. P. Involvement and dual effects of nitric oxide in septic shock. Inflamm. Res., 1998, 47(4): 152-166.
    [98] Brune, B. Nitric oxide: NO apoptosis or turning it on? Cell Death Differ., 2003, 10(8): 864-869.
    [99] Johnson, M. L., Billiar, T. R. Roles of nitric oxide in surgical infection and sepsis. World J. Surg., 1998, 22(2): 187-196.
    [100] Wink, D. A., Cook, J. A., Christodoulou, D., Krishna, M. C., Pacelli, R., Kim, S., DeGraff, W., Gamson, J., Vodovotz, Y., Russo, A., Mitchell, J. B. Nitric oxide and some nitric oxide donor compounds enhance the cytotoxicity of cisplatin. Nitric Oxide, 1997, 1(1): 88-94.
    [101] Konovalova, N. P., Goncharova, S. A., Volkova, L. M., Rajewskaya, T. A., Eremenko, L. T., Korolev, A. M. Nitric oxide donor increases the efficiency of cytostatic therapy and retards the development of drug resistance. Nitric Oxide, 2003, 8(1): 59-64.
    [102] Chen, C., Shi, Y. Q., Song, J., Qi, Q. S., Gu, L., Wang, P. G. Delivery of nitric oxide released from beta-Gal-NONOate activation by beta-galactosidase and its activity against Escherichia coli. Biol. Pharm. Bull., 2006, 29(6): 1239-1241.
    [103] Davies, J. Origins and evolution of antibiotic resistance. Microbiologia, 1996, 12(1): 9-16.
    [104] Levy, S. B. Antibiotic and antiseptic resistance: impact on public health. Pediatr. Infect. Dis. J., 2000, 19(10 Suppl): S120-122.
    [105] Xu, C., Lin, X., Ren, H., Zhang, Y., Wang, S., Peng, X. Analysis of outer membrane proteome of Escherichia coil related to resistance to ampicillin and tetracycline. Proteomics, 2006, 6(2): 462-473.
    [106] Bren, L. Battle of the bugs: fighting antibiotic resistance. FDA Consum., 2002, 36(4): 28-34.
    [107] Wink, D. A., Hanbauer, I., Grisham, M. B., Laval, F., Nims, R. W., Lavai, J., Cook, J., Pacelli, R., Liebmann, J., Krishna, M., Ford, P. C., Mitchell, J. B. Chemical biology of nitric oxide: regulation and protective and toxic mechanisms. Curr. Top. Cell. Regul., 1996, 34: 159-187.
    [108] Wink, D. A., Darbyshire, J. F., Nims, R. W., Saavedra, J. E., Ford, P. C. Reactions of the bioregulatory agent nitric oxide in oxygenated aqueous media: determination of the kinetics for oxidation and nitrosation by intermediates generated in the NO/O_2 reaction. Chem. Res. Toxicol., 1993, 6(1): 23-27.
    [109] Fang, F. C. Perspectives series: host/pathogen interactions. Mechanisms of nitric oxide-related antimicrobial activity. J. Clin. Invest., 1997, 99(12): 2818-2825.
    [110] Girlich, D., Naas, T., Bellais, S., Poirel, L., Karim, A., Nordmann, P. Heterogeneity of AmpC cephalosporinases of Hafnia alvei clinical isolates expressing inducible or constitutive ceftazidime resistance phenotypes. Antimicrob. Agents Chemother., 2000, 44(11): 3220-3223.
    [111] Thomson, K. S., Smith Moland, E. Version 2000: the new [beta]-lactamases of Gram-negative bacteria at the dawn of the new millennium. Microbes Infect., 2000, 2(10): 1225-1235.
    [112] Tang, X., Cai, T., Wang, P. G. Synthesis of beta-lactarnase activated nitric oxide donors. Bioorg. Med. Chem. Lett., 2003, 13(10): 1687-1690.
    [113] Fluit, A. C., Visser, M. R., Schmitz, F. J. Molecular detection of antimicrobial resistance. Clin. Microbiol. Rev., 2001, 14(4): 836-871, table of contents.
    [114] Luckey, M., Nikaido, H. Specificity of diffusion channels produced by lambda phage receptor protein of Escherichia coli. Proc. Natl. Acad. Sci. U. S. A., 1980, 77(1): 167-171.
    [115] Nikaido, H., Rosenberg, E. Y. Effect on solute size on diffusion rates through the transmembrane pores of the outer membrane of Escherichia coli. J. Gen. Physiol., 1981, 77(2): 121-135.
    [116] Nikaido, H., Rosenberg, E. Y. Porin channels in Escherichia coli: studies with liposomes reconstituted from purified proteins, J. Bacteriol., 1983, 153(1): 241-252.
    [117] Nikaido, H., Rosenberg, E. Y., Foulds, J. Porin channels in Escherichia coli: studies with beta-lactams in intact cells. J. Bacteriol., 1983, 153(1): 232-240.
    [118] Nikaido, H. Molecular basis of bacterial outer membrane permeability revisited. Microbiol. Mol. Biol. Rev., 2003, 67(4): 593-656.
    [119] Nikaido, H., Vaara, M. Molecular basis of bacterial outer membrane permeability. Microbiol. Rev., 1985, 49(1): 1-32.
    [120] Rubbo, H., Radi, R., Trnjillo, M., Telleri, R., Kalyanaraman, B., Barnes, S., Kirk, M., Freeman, B. A. Nitric oxide regulation of superoxide and peroxynitrite-dependent lipid peroxidation. Formation of novel nitrogen-containing oxidized lipid derivatives. J. Biol. Chem., 1994, 269(42): 26066-26075.
    [121] Sehweizer, H. P. Efflux as a mechanism of resistance to antimicrobials in Pseudomonas aeruginosa and related bacteria: unanswered questions. Genet. Mol. Res., 2003, 2(1): 48-62.
    [122] Swinbanks, D. Government backs proteome proposal. Nature, 1995, 378(6558): 653.
    [123] Cash, P. Proteomics in medical microbiology. Electrophoresis, 2000, 21(6): 1187-1201.
    [124] 许克新,王云川 双向电泳及质谱分析与蛋白组研究.第四军医大学学报,2002,23(22):2017-2022.
    [125] O'Farrell, P. H. High resolution two-dimensional electrophoresis of proteins. J. Biol. Chem., 1975, 250(10): 4007-4021.
    [126] 郭尧君 SDS电泳技术的实验考虑及最新进展.生物化学与生物物理进展,1991,18(1):32-37.
    [127] Ignarro, L. J., Buga, G. M., Wood, K. S., Byrns, R. E., Chaudhuri, G. Endothelium-derived relaxing factor produced and released from artery and vein is nitric oxide. Proc. Natl. Acad. Sci. U. S. A., 1987, 84(24): 9265-9269.
    [128] Bredt, D. S., Snyder, S. H. Nitric oxide: a physiologic messenger molecule. Annu. Rev. Biochem., 1994, 63: 175-195.
    [129] Davis, K. L., Martin, E., Turko, I. V., Murad, F. Novel effects of nitric oxide. Annu. Rev. Pharmacol. Toxicol., 2001, 41: 203-236.
    [130] Wink, D. A., Osawa, Y., Darbyshire, J. F., Jones, C. R., Eshenaur, S. C., Nims, R. W. Inhibition of cytochromes P450 by nitric oxide and a nitric oxide-releasing agent. Arch. Biochem. Biophys., 1993, 300(1): 115-123.
    [131] Thatcher, G. R., Nicolescu, A. C., Bennett, B. M., Toader, V. Nitrates and NO release: contemporary aspects in biological and medicinal chemistry. Free Radical Biol. Meal, 2004, 37(8): 1122-1143.
    [132] Hou, Y. C., Janczuk, A., Wang, P. G. Current trends in the development of nitric oxide donors. Curn. Pharm. Des., 1999, 5(6): 417-441.
    [133] Wang, P. G., Xian, M., Tang, X., Wu, X., Wen, Z., Cai, T., Janczuk, A. J. Nitric oxide donors: chemical activities and biological applications. Chem. Rev., 2002, 102(4): 1091-1134.
    [134] Scatena, R., Bottoni, P., Martorana, G. E., Giardina, B. Nitric oxide donor drugs: an update on pathophysiology and therapeutic potential. Expert Opin. Invest. Drugs, 2005, 14(7): 835-846.
    [135] Keefer, L. K., Nims, R. W., Davies, K. M., Wink, D. A. "NONOates" (1-substituted diazen-1-ium-1,2-diolates) as nitric oxide donors: convenient nitric oxide dosage forms. Methods Enzymol., 1996, 268: 281-293.
    [136] 张奕华,田季德,彭司勋 靶向作用的一氧化氮供体及其相关药物.药学学报,2006,41(6):481-486.
    [137] 张继瑜 细菌抗药性的分子机制.中兽医医药杂志,2002,21(2):18-22.
    [138] 张国庆,廖杰,于力方双向电泳技术在蛋白质组研究中的应用.标记免疫分析与临床,2003,10(3):171-173.
    [139] 杨何义,应万涛,钱小红 蛋白质组技术的研究进展.自然科学进展,2002,12(1):13-17.
    [140] 翁瑜,曾群力,姜槐,许正平双向凝胶电泳比较三种常用蛋白质提取方法.中国生物化学与分子生物学报,2005,21(5):691-694.
    [141] 王新,王伯良,张宗友,等蛋白质组分析中双向电泳技术的改良和应用.细胞与分子免疫学杂志,2001,17(2):191-192.
    [142] 万晶宏,钱小红,郭尧君,等蛋白质组分析中双向聚丙烯酰胺凝胶电泳技术的初步建立与优化.军事医学科学院院刊,1998,22(4):297-300.
    [143] 王凤茹 双向电泳应注意的几个关键问题.生物技术通报,2005,5(6):62-64.
    [144] 秦慧,刘霆,柳斌,等几种双向凝胶电泳蛋白质检测方法的比较.中国实验血液学杂志,2006,14(1):168-172.
    [145] 刘健平,陈国华,陈本美,等蛋白质组双向电泳实验中一些常见失误的分析.生命科学研究,2003,7(2):177-180.
    [146] 贾宇峰,林秋霞,郭尧君,等蛋白质双向电泳图像分析.生物化学与生物物理进展,2001,28(2):246-250.
    [147] 熊兴东,许丽艳,沈忠英,等双向凝胶电泳图像分析方法的建立.癌变·畸变·突变,2002,14(1):139-143.
    [148] Moneada, S., Palmer, R. M., Higgs, E. A. Nitric oxide: physiology, pathophysiology, and pharmacology. Pharmacol. Rev., 1991, 43(2): 109-142.
    [149] Jenkins, D. C., Charles, I. G., Thomsen, L. L., Moss, D. W., Holmes, L. S., Baylis, S. A., Rhodes, P., Westmore, K., Emson, P. C., Moneada, S. Roles of nitric oxide in tumor growth. Proc. Natl. Acad. Sci. U. S. A., 1995, 92(10): 4392-4396.
    [150] 张剑,何生 一氧化氮在肿瘤生长转移中的作用.中国普外基础与临床杂志,2004,11(6):561-563.
    [151] Megson, I. L., Webb, D. J. Nitric oxide donor drugs: current status and future trends. Expert Opin. Invest. Drugs, 2002, 11(5): 587-601.
    [152] Saavedra, J. E., Billiar, T. R., Williams, D. L., Kim, Y. M., Watkins, S. C., Keefer, L. K. Targeting nitric oxide (NO) delivery in vivo. Design of a liver-selective NO donor prodrug that blocks tumor necrosis factor-alpha-induced apoptosis and toxicity in the liver. J. Med. Chem., 1997, 40(13): 1947-1954.
    [153] Zhang, R., Zhang, L., Zhang, Z., Wang, Y., Lu, M., LaPointe, M., Chopp, M. A nitric oxide donor induces neurogenesis and reduces functional deficits after stroke in rats. Ann. Neurol., 2001, 50(5): 602-611.
    [154] Hrabie, J. A., Keefer, L. K. Chemistry of the Nitric Oxide-Releasing Diazeniumdiolate ("Nitrosohydroxylamine") Functional Group and Its Oxygen-Substituted Derivatives. Chem. Rev., 2002, 102(4): 1135-1154.
    [155] Keefer, L. K. Nitric oxide (NO)- and nitroxyl (HNO)-generating diazeniumdiolates (NONOates): emerging commercial opportunities. Cure. Top. Med. Chem., 2005, 5(7): 625-636.
    [156] Cai, T. B., Lu, D., Landerholm, M., Wang, P. G. Sialated diazeniumdiolate: a new sialidase-activated nitric oxide donor. Org. Lett., 2004, 6(23): 4203-4205.
    [157] Ma, W., Rogers, K., Zbar, B., Schmidt, L. Effects of different fixatives on beta-galactosidase activity. J. Histochem. Cytochem., 2002, 50(10): 1421-1424.
    [158] Umansky, V., Sehirrmacher, V. Nitric oxide-induced apoptosis in tumor cells. Adv. Cancer Res., 2001, 82: 107-131.
    [159] Nam, M., Johnston, P., Lal, B., Indurti, R., Wilson, M. A., Laterra, J. Endothelial cell-based cytokine gene delivery inhibits 9L glioma growth in vivo. Brain Res., 1996, 731(1-2): 161-170.
    [160] Thorens, B. Facilitated glucose transporters in epithelial cells. Annu. Rev. Physiol., 1993, 55: 591-608.
    [161] Tamai, I., Tsuji, A. Cartier-mediated approaches for oral drug delivery. Adv. Drug Delivery Rev., 1996, 20(1): 5-32.
    [162] Wong, A., Toth, I. Lipid, sugar and liposaccharide based delivery systems. Curr. Med. Chem., 2001, 8(9): 1123-1136.
    [163] Cantuaria, G., Magalhaes, A., Angioli, R., Mendez, L., Mirhashemi, R., Wang, J., Wang, P., Penalver, M., Averette, H., Braunschweiger, P. Antitumor activity of a novel glyco-nitric oxide conjugate in ovarian carcinoma. Cancer, 2000, 88(2): 381-388.
    [164] Lin, W. C., Pretlow, T. P., Pretlow, T. G., 2nd, Culp, L. A. Bacterial lacZ gene as a highly sensitive marker to detect micrometastasis formation during tumor progression. Cancer Res., 1990, 50(9): 2808-2817.
    [165] Kobayashi, K., Nakanishi, H., Masuda, A., Tezuka, N., Mural, M., Tatematsu, M. Sequential observation of micrometastasis formation by bacterial lacZ gene-tagged Lewis lung carcinoma cells. Cancer Lett., 1997, 112(2): 191-198.
    [166] Kobayashi, K., Nakanishi, H., Inada, K., Fujimitsu, Y., Yamachika, T., Shirai, T., Tatematsu, M. Growth characteristics in the initial stage of micrometastasis formation by bacterial LacZ gene-tagged rat prostatic adenocarcinoma cells. Jpn. J. Cancer Res., 1996, 87(12): 1227-1234.
    [167] Visted, T., Thorsen, J., Thorsen, F., Read, T. -A., Ulvestad, E., Engebraaten, O., Sorensen, D., Yla-Herttuala, S., Tyynela, K., Rucldidge, G., Edvardsen, K., Bjerkvig, R., Lund-Johansen, M. LacZ-neoR transfected glioma cells in syngeneic rats: Growth pattern and characterization of the host immune response against cells transplanted inside and outside the CNS. Int. J. Cancer, 2000, 85(2): 228-235.
    [168] Nakanishi, H., Kobayashi, K., Nishimura, T., Inada, K., Tsukamoto, T., Tatematsu, M. Chemosensitivity of micrometastases and circulating tumor cells to uracil and tegafur as evaluated using LacZ gene-tagged Lewis lung carcinoma cell. Cancer Lett., 1999, 142(1): 31-41.
    [169] Steiner, M. S. World Pat. Appl. WO 200020608.2000.
    [170] Steiner, M. S. World Pat. Appl. WO 200020038. 2000.
    [171] Tietze, L. F., Herzig, T., Fecher, A., Haunert, F., Sehuberth, I. Highly Selective Glycosylated Prodrugs of Cytostatic CC-1065 Analogues for Antibody-Directed Enzyme Tumor Therapy. Chembiochem, 2001, 2(10): 758-765.
    [172] Bakina, E., Farquhar, D. Intensely cytotoxic anthracycline prodrugs: galactosides. Anticancer. Drug Des., 1999, 14(6): 507-515.
    [173] Bagshawe, K. D., Napier, M. Early clinical studies with ADEFF, in Enzyme-Prodrug Strategies for Cancer Therapy (Eds.: Knox, R. J., Melton, R. G.), Kluwer Academic/Plenum Publishers, New York, 1999, pp. 199-207.
    [174] Burke, P. J. in Enzyme-Prodrug Strategies for Cancer Therapy (Eds.: Knox, R. J., Melton, R. G.), Kluwer Academic/Plenum Publishers, New York, 1999, pp. 133-154.
    [175] Drapier, J. C., Hibbs, J. B., Jr. Murine cytotoxic activated macrophages inhibit aconitase in tumor cells. Inhibition involves the iron-sulfur prosthetic group and is reversible, d. Clin. Invest., 1986, 78(3): 790-797.
    [176] Lancaster Jr, J. R. Nitric Oxide in Cells. Am. Sci., 1992, 80(3): 248-259.
    [177] Stuehr, D. J., Nathan, C. F. Nitric oxide. A macrophage product responsible for cytostasis and respiratory inhibition in tumor target cells. J. Exp. Med., 1989, 169(5): 1543-1555.
    [178] Sokolowska, M., Rokita, H., Wlodek, L. Activation of DNA biosynthesis in human hepatoblastoma HEPG2 cells by the nitric oxide donor, sodium nitroprusside. Fundam. Clin. Pharmacol., 2003, 17(5): 599-607.
    [179] Chen, C., Shi, Y., Li, S., Qi, Q., Gu, L., Song, J., Wang, P. G. A Glycosylated Nitric Oxide Donor, beta-Gal-NONOate, and its Site-specific Antitumor Activity. Arch Pharm. (Weinheim), 2006, 339(7): 366-371.
    [180] 边城,徐永红,李定国 一氧化氮对大鼠实验性肝纤维化的影响.中国药理学通报,2002,18(3):312-314.
    [181] Bell, G. I., Kayano, T., Buse, J. B., Burant, C. F., Takeda, J., Lin, D., Fukumoto, H., Seino, S. Molecular biology of mammalian glucose transporters. Diabetes Care, 1990, 13(3): 198-208.
    [182] Gould, G. W., Bell, G. I. Facilitative glucose transporters: an expanding family. Trends Biochem. Sci., 1990, 15(1): 18-23.
    [183] Schiller, D., Cavalla, P., Migheli, A., Chio, A., Giordana, M. T., Marina, S., Attanasio, A. Apoptosis and cell proliferation in human neuroepithelial tumors. Neurosci. Lett., 1995, 195(2): 81-84.
    [184] Ormerod, M. G., Orr, R. M., Peacock, J. H. The role of apoptosis in cell killing by cisplatin: a flow cytometric study. Br. J. Cancer, 1994, 69(1): 93-100.
    [185] Lee, J. U., Hosotani, R., Wada, M., Doi, R., Kosiba, T., Fujimoto, K., Miyamoto, Y., Mori, C., Nakamura, N., Shiota, K., Imamura, M. Mechanism of apoptosis induced by cisplatin and VP-16 in PANC-1 cells. Anticancer Res., 1997, 17(5A): 3445-3450.
    [186] 马文斌,陈松森,任祖渊,等顺铂诱导大鼠C6胶质瘤细胞凋亡.基础医学与临床,2000,20(3):272-275.
    [187] Brown, D. G., Sun, X. M, Cohen, G, M. Dexamethasone-indueed apoptosis involves cleavage of DNA to large fragments prior to intemucleosomal fragmentation. J. Biol. Chem., 1993, 268(5): 3037-3039.
    [188] Orrenius, S., McCabe, M. J., Jr., Nicotera, P. Ca~(2+)-dependent mechanisms of cytotoxicity and programmed cell death. Toxicol. Lett., 1992, 64-65 Spec No: 357-364.
    [189] 郭建友,霍海如,姜廷良衡量联合用药作用研究方法评价.中药药理与临床,2005,21(3):60-64.
    [190] 姜泊“细胞凋亡基础与临床,”人民军医出版社,北京,1997,p.119.
    [191] Catchpoole, D. R., Stewart, B. W. Etoposide-induced cytotoxicity in two human T-cell leukemic lines: delayed loss of membrane permeability rather than DNA fragmentation as an indicator of programmed cell death. Cancer Res., 1993, 53(18): 4287-4296.
    [192] Ormerod, M. G., O'Neill, C. F., Robertson, D., Harrap, K. R. Cisplatin induces apoptosis in a human ovarian carcinoma cell line without concomitant intemueleosomal degradation of DNA. Exp. Cell Res., 1994, 211 (2): 231-237.
    [193] Mathiasen, I. S., Sergeev, I. N., Basthoim, L., Elling, F., Norman, A. W., Jaattela, M. Calcium and calpain as key mediators of apoptosis-like death induced by vitamin D compounds in breast cancer cells. J. Biol. Chem., 2002, 277(34): 30738-30745.
    [194] Zhivotovsky, B., Cedervall, B., Jiang, S., Nicotera, P, Orrenius, S. Involvement of Ca~(2+) in the formation of high molecular weight DNA fragments in thymoeyte apoptosis. Biochem. Biophys. Res. Commun., 1994, 202(1): 120-127.
    [195] Jiang, S., Chow, S. C., Nicotera, P., Orrenius, S. Intracellular Ca~(2+) signals activate apoptosis in thymocytes: studies using the Ca(~(2+))-ATPase inhibitor thapsigargin. Exp. Cell Res., 1994, 212(1): 84-92.
    [196] Jones, D. P., Meeonkey, D. J., Nieotera, P., Orrenius, S. Calcium-activated DNA fragmentation in rat liver nuclei, J. Biol. Chem., 1989, 264(11): 6398-6403.
    [197] Chen, L., Wang, Q., Gao, H. Involvement of Ca~(2+) and CaN singaling pathway in CORT-induced activation of fasl during apoptosis in rat leydig cell. Chinese J. Androl., 2006, 20(2): 4-8.
    [198] Yang, Q., Jiang, H., Lin, B., Xi, X., Liu, Y., Wang, L., Lu, X., Gu, J., Li, D. Inhibition of intracellular Ca~(2+)-chelating agent on HeLa cell apoptosis induced by HSV-1. Clin. J. Cell Mol. Immunol., 2003, 19(5): 421-424.
    [199] Huigsloot, M., Tijdens, I. B., Mulder, G. J., van de Water, B. Differential regulation of doxorubicin-induced mitochondrial dysfunction and apoptosis by Bcl-2 in mammary adenocarcinoma (MTLn3) cells, d. Biol. Chem., 2002, 27709): 35869-35879,
    [200] Carriedo, S. G., Yin, H. Z., Semi, S. L., Weiss, J. H. Rapid Ca~(2+) entry through Ca~(2+)-permeable AMPA/Kainate channels triggers marked intracellular Ca~(2+) rises and consequent oxygen radical production, J. Neurosci., 1998, 18(19): 7727-7738.
    [201] Herrmann, M., Lorenz, H. M., Voll, R., Grunke, M., Woith, W., Kalden, J. R. A rapid and simple method for the isolation of apoptotic DNA fragments. Nucleic Acids Res., 1994, 22(24): 5506-5507.
    [202] Maneiro, E., Lopez-Armada, M. J., de Andres, M. C., Carames, B., Martin, M. A., Bonilla, A., Del Hoyo, P., Galdo, F., Arenas, J., Blaneo, F. J. Effect of nitric oxide on mitochondrial respiratory activity of human articular chondrocytes. Ann. Rheum. Dis., 2005, 64(3): 388-395.

© 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