糖苷酶抑制剂的戈谢病分子伴侣机制和HDAC抑制剂的抗肿瘤机制研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
酶蛋白几乎参与机体所有的生命活动,其功能对于机体的正常运行至关重要,许多疾病的发生都与酶功能的失调有关。因此,以酶为靶点的药物开发一直都是研究的热点。其中,酶抑制剂类药物占据临床用药的三分之一,成为新药来源的主要途径。
     糖昔酶是一类将寡糖等糖基复合物的糖苷键水解的酶,主要参与食物的消化、多糖和糖脂复合物的降解。糖苷酶抑制剂可以抑制糖苷酶的活性,阻断碳水化合物的分解,对一些糖代谢紊乱疾病如溶酶体贮积症、糖尿病等有重要药用价值。同时,以糖苷酶抑制剂为工具,可以更好的了解糖苷酶的水解机制,分析糖基化的生物学作用,利于揭示阿尔茨海默病、肿瘤等重大疾病的发生机制。因此,围绕酶抑制剂药物的开发,本课题开展了糖苷酶抑制剂和组蛋白去乙酰化酶抑制剂的设计、筛选和机制分析,为相关疾病靶向药物的开发提供了依据和新思路。
     戈谢病是一种由于酸性p-葡萄糖脑苷酶(acid β-glucocerebrosidase, GCase)基因突变引起的发病率最高的溶酶体贮积病。该酶基因的突变将导致GCase的提前降解,最终不能定位于溶酶体中发挥活性,从而使底物葡萄糖脑苷脂(Glucosylcera-mide, GC)大量积累。基于酶的竞争性抑制剂机制,人体提出了分子伴侣设想:竞争性抑制剂与突变酶的活性中心结合,诱导酶形成正确折叠,并辅助酶定位于溶酶体,在高浓度底物的溶酶体中,竞争性抑制剂被底物竞争性解离,酶分子得以释放,从而发挥生物功能。依据该酶的催化机制,本课题组前期设计、合成系列葡萄糖咪唑类化合物,本课题中通过生物学活性测试,发现化合物具有强抑制活性和高选择性,且细胞膜透过性好,细胞毒性低。以发病率最高的两种成淋巴细胞(N370S, L444P)为模型,我们发现该类化合物均具有较好的伴侣活性,尤其是化合物11(3,3-二甲基-N-苯基-4-酰胺-1-丁基取代的葡萄糖咪唑),2.5μM的用药浓度下,可使N370S型GCase酶活提高2.1倍。利用计算机模拟、脉冲酶解稳定性试验、Western blot和细胞免疫荧光试验等方法分析,发现化合物11可以结合并稳定突变GCase酶的结构,提高其蛋白水平,促进其溶酶体定位,最终提高细胞中GCase的活性。
     N-乙酰葡萄糖胺(0-linked N-acetylglucosamine,O-GlcNAc)修饰是一种重要的蛋白翻译后修饰,参与生物体中的多种细胞活动,O-GlcNAcase (OGA)是体内唯一水解该修饰的糖苷酶。有人发现阿尔茨海默病人的大脑中Tau蛋白的O-GlcNAc修饰严重不足,因此,我们设想通过OGA抑制剂来提高患者体内的O-GlcNAc修饰水平。利用前期设计、合成的系列化合物,通过酶学筛选,获得了一个高效选择性的OGA抑制剂(Ki=5.9μM),与阳性化合物PUGNAc相比它能更多地提高细胞内的O-GlcNAc水平。同时,利用体外分段表达,我们获得了一个OGA变体(细胞凋亡过程中的OGA剪切体,nOGA)的专一性抑制剂。所筛选的这些OGA抑制剂将有助于进一步揭示细胞内O-GlcNAc修饰的机制,阐明与O-GlcNAc修饰相关疾病的病因,为相关药物的研发提供了基础。
     组蛋白去乙酰化酶(Histone deacetylases, HDACs)是一类调控染色体形成和基因表达的关键酶。当组蛋白处于低乙酰化状态时,核小体结构紧密,使各种促进细胞分化和凋亡的基因受到抑制,引起肿瘤的发生。HDACs抑制剂(HDACi)通过抑制HDAC去乙酰化活性,阻滞肿瘤细胞生长,诱导其分化和凋亡。因此,以HDACs为靶点,设计合成抗肿瘤药物具有广阔的前景。依据前期设计、合成的系列HDACi,通过体外酶活和细胞毒性筛选,我们获得了化合物Z-13,它对HDAC酶活性和肿瘤细胞增殖的抑制都要比阳性药SAHA (suberoylanilide hydroxamic acid)高一个数量级。通过对它抗肿瘤机制的研究,发现它主要通过阻遏细胞周期和诱导细胞凋亡,来抑制肿瘤细胞增殖,延缓体内乳腺癌肿瘤的生长。同时,我们做了SAHA和taxol联合用药对乳腺癌细胞抑制的影响,发现,SAHA能增强taxol对乳腺癌细胞的抑制,尤其是增强了taxol对其不敏感的乳腺癌细胞增殖的抑制。在对体内建立的移植瘤模型上,也表现出了SAHA能增强taxol对肿瘤抑制的作用。所以二者的联合用药为临床上的进一步研究提供了理论基础。
Enzymes involve in almost all of our life process, so their activities play a vital role in maintaining the normal operation of the body. Many diseases are caused by disorder of the activity of some enzymes, so it has been widely recognized in recently years that enzymes are promising targets for drug development. So far, enzyme inhibitor drugs supply a third of drugs clinical applied, and as a mainly source of new drugs.
     Glycosidases catalyze the hydrolysis of the glycosidic linkage to release smaller sugars. They widely involve in digesting of food, polysaccharides and glycoconjugates metabolism, glycosidase inhibitors can inhibit the activity of glycosidase, and block the decomposition of carbohydrates. they have important therapeutic value against some glycometabolism disorder diseases, such as lysosomal storage diseases, diabetes, etc. Meanwhile, The study of the glycosidase inhibitors contribute to better understanding of the the hydrolytic mechanism and biological functions of glycosylation, revealing the pathogenesises of Alzheimer's disease, cancer, and other major diseases. Surrounding the development of enzyme inhibitor drugs, we carried out the topic:design, screening and mechanism analysis of glycosidase inhibitors and HDAC inhibitors. These results provide a basis for the development of the targeted drugs.
     Gaucher disease, the highest incidence of lysosomal storage diseases, is caused by the mutations in acid β-glucocerebrosidase (GCase). The mutation of GCase leads to the earlier degradation of GCase in endoplasmic reticulum, so GCase cannot be located in Iysosome, eventually, cause the accumulation of glucosylceramide (GlcCer). Based on the mechanism of competitive enzymes inhibitors, people put forward the hypothesis of pharmacological chaperone therapy (PCT), which refers to the use of competitive inhibitors to specifically bind and potentially stabilize catalytically competent GCase variants at neutral pH in the ER, assisting with the folding of these mutant proteins and helping these proteins pass the quality control checks for trafficking to the lysosome. Due to the high substrate concentrations and low pH in the lysosome, the pharmacological chaperones (PCs) would be displaced from the active site of the enzyme, and the stored GlcCer would be degraded. According to the catalytic mechanism of GCase, we synthesized a series of glucoimidazoles. By biological activity screening, we found all of the compounds had strong inhibitory activity, high selectivity, good cell membrane permeability, and low cytotoxicity. A cell-based assay using patient-derived lymphoblasts (N370S or L444P mutation) demonstrated that the administration of these compounds can significantly increase GCase activity. Interestingly, the moderate inhibitor, compound11, which bears a3,3-dimethyl-N-phenyl-4-amide-l-butyl substituent, had the greatest effect on activity, yielding a2.1-fold increase in N370S lymphoblasts at2.5μM and a1.2-fold increase in L444P at0.5μM following a three-day incubation. Next, computer docking studies and a protease protection assay were used to elucidate the ligand-enzyme interactions responsible for the chaperone activity of11. Western blot and immuno-fluorescence assays verified the trafficking of the restored GCase to the lysosome.
     O-linked N-acetylglucosamine (O-GlcNAc) modification, which involes in a variety of cellular processes, is an essential posttranslational modification in metazoans. O-GlcNAcase (OGA) is the only glycosidase which is responsible for the removal of O-GlcNAc. The study found that O-GlcNAcylation of tau in the brains of alzheimer's patients is severe insufficient. So, We wanted to improve the level of O-GlcNAcby OGA inhibitors. We designed and synthesized series of compounds. By enzyme activity screening, we got a powerful selective OGA inhibitor (Kj=5.9μM) which effectively induced more cellular hyper-O-GlcNAcylation than PUGNAc. Using piecewise expression of OGA in vitro, we obtained a specific nOGA (N r terminal fragment of OGA during apoptosis) inhibitor (Kj=48μM). These OGA inhibitors contributed to revealing the mechanism of O-GlcNAc modifications, and pathogenesises of diseases for the abnormalities of O-GlcNAc modification, in order to provide a basis for the development of the relevant drugs.
     HDACs (Histone deacetylases) are a kind of key enzymes which play fundamental roles in the regulation of gene expression and maintenance of chromatin structures. When the histones are in low acetylation state, which can condense chromatin, promote cell growth, differentiation and inhibit the expression of apoptosis related genes, finally cause the occurrence of tumor. Histone deacetylase inhibitors (HDACi) can reverse the hypoacetylation status of histones, thereby inducing the tumor cell differentiation and apoptosis. It has been widely recognized in recently years that HDACs are promising targets for cancer therapy. We synthesized a novel calss of HDAC inhibitors. After in vitro enzyme activity and cell toxicity screening, we discovered a compound, Z-13, which showed about one order of magnitude more potent than SAHA (suberoylanilide hydroxamic acid) in both enzymatic and cellular assays. Based on the research of antitumor mechanism of Z-13, we found that it inhibited the proliferation of tumor cell in vitro and in vivo mainly by inducing cell apoptosis and cell cycle arrest. We also want to know whether SAHA can enhance the growth inhibitory effect induced by taxol against breast cancer cell. By experiment, we found that SAHA can enhance taxol-induced cell death against human breast cancer cells, especially in taxol-resistant and multi-resistant breast cancer cells. Based on BALB/c nude mice breast cancer xenograft model, the synergetic effect was also observed in the in vivo xenograft tumor model. The combination of SAHA and taxol may have therapeutic potential in the treatment of breast cancer.
引文
[1]黄熙泰,于自然,李翠凤.现代生物化学(第二版),化学工业出版社,2005.
    [2]王镜岩,朱圣庚,徐长发.生物化学第三版(上册),高等教育出版社,2002,327-332.
    [3]Moruno-Davila M A, Garrido-del Solo C, Garcia-Moreno M, et al. Kinetic analysis of enzyme systems with suicide substrate in the presence of a reversible competitive inhibitor, tested by simulated progress curves. The international journal of biochemistry & cell biology,2001,33, 181-191.
    [4]王树青.基于生物信息学的SARS冠状病毒主蛋白酶抑制剂的设计[硕士毕业论文],天津:天津师范大学,2005.
    [5]刘洋洋.酶抑制剂类药物筛选研究进展.海南省药学会○一○年学术年会会议论文集,2010,9-15.
    [6]唐章勇,唐灿.酶抑制剂筛选的研究进展.上海医药,2007,28,117-119.
    [7]李越中.药物微生物技术,化学工业出版社,2004,3-5.
    [8]朱明燕,柳景景,雷石.血管紧张素转换酶抑制剂研究进展与临床应用.中国医药导报,2011,89-10.
    [9]黄哲,王礼琛.血管肽酶抑制剂的研究与开发.药学进展,2004,214-217.
    [10]李能刚,徐进宜,吴晓明.血管紧张素转化酶和中性肽链内切酶的双重抑制剂.药学进展,2000,133-136.
    [11]张小平.血管紧张素转换酶抑制剂和血管肽酶抑制剂研究进展.食品与药品,2006,15-18.
    [12]梁艳,徐向伟,赵专友.血管肽酶抑制剂新药研发的进展.现代药物与临床,2012,493-497.
    [13]郑晓克.抗肿瘤药物的研究进展.中山大学研究生学刊(自然科学、医学版),2008,7-12.
    [14]车文军,李志裕,尤启冬.抗肿瘤药物新靶点与新药研究.药学进展,2007,247-253.
    [15]蒋鸿超,李鸿钧,孙茂盛.靶向Survivin肿瘤治疗研究进展.医学综述,2006,163-166.
    [16]李乾斌,徐文方,张震.抗癌作用新靶点及其抑制剂的研究进展.中国药物化学杂志,2004,65-70.
    [17]刘霞,冯长根.酶抑制剂在抗糖尿病药物中的应用研究.中国药学杂志,2003,11-13.
    [18]de Bruijne J, Bergmann J F, Reesink H W, et al. Antiviral activity of narlaprevir combined ith ritonavir and pegylated interferon in chronic hepatitis C patients. Hepatology,2010,52, 1590-1599.
    [19]Knechten H, Lutz T, Pulik P, et al. Safety and Efficacy in HIV-1-Infected Patients Treated with Ritonavir-Boosted Saquinavir Mesylate. Archives of drug information,2010,3,26-36.
    [20]Rago A, Lichtner M, Mecarocci S, et al. Antiviral treatment including entecavir plus tenofovir disoproxil fumarate for HBV reactivation following a rituximab-based regimen. Antiviral therapy,2010,15,929-932.
    [21]Margot N A, Enejosa J, Cheng A K, et al. Development of HIV-1 drug resistance through 144 weeks in antiretroviral-naive subjects on emtricitabine, tenofovir disoproxil fumarate, and efavirenz compared with lamivudine/zidovudine and efavirenz in study GS-01-934. J Acquir Immune Defic Syndr,2009,52,209-221.
    [22]Soriente A, De Rosa M M, Scettri A, et al. Manoalide. Current medicinal chemistry,1999,6, 415-431.
    [23]Rao N L, Dunford P J, Xue X, et al. Anti-inflammatory activity of a potent, selective leukotriene A4 hydrolase inhibitor in comparison with the 5-lipoxygenase inhibitor zileuton. The Journal of pharmacology and experimental therapeutics,2007,321,1154-1160.
    [24]Bernardi A, Frozza R L, Horn A P, et al. Protective effects of indomethacin-loaded nanocapsules against oxygen-glucose deprivation in organotypic hippocampal slice cultures: involvement of neuroinflammation. Neurochemistry international,2010,57,629-636.
    [25]Goldstein J L, Aisenberg J, Lanza F, et al. A multicenter, randomized, double-blind, active-comparator, placebo-controlled, parallel-group comparison of the incidence of endoscopic gastric and duodenal ulcer rates with valdecoxib or naproxen in healthy subjects aged 65 to 75 years. Clinical therapeutics,2006,28,340-351.
    [26]Cruz-Hernandez C, Oliveira M, Pescia G, et al. Lipase inhibitor orlistat decreases incorporation of eicosapentaenoic and docosahexaenoic acids in rat tissues. Nutr Res,2010, 30,134-140.
    [27]Canesi M, Zecchinelli A L, Pezzoli G, et al. Clinical experience of tolcapone in advanced Parkinson's disease. Neurological sciences:official journal of the Italian Neurological Society and of the Italian Society of Clinical Neurophysiology,2008,29 Suppl 5, S380-382.
    [28]林邦和.溶酶体.生物学通报,1989,3-6.
    [29]王洪允,江骥,胡蓓.串联质谱在新生儿遗传代谢性疾病筛查中的应用.质谱学报,2011,24-30.
    [30]陆志城.溶酶体贮积症治疗药物研究进展.中国处方药,2005,60-64.
    [31]何灵江,原丽红.人类溶酶体与溶酶体疾病.生物学教学,2010,2-5.
    [32]Methods for a prompt and reliable laboratory diagnosis of Pompe disease:Report from an international consensus meeting. Molecular Genetics and Metabolism 2008,93,275-281.
    [33]黄萍,俞玲娜,刘丽.黏多糖贮积症致心血管病变及其处理.中国实用儿科杂志,2013,28,62-64.
    [34]郭奕斌.黏多糖病的诊断及防治策略.中华临床医师杂志,2009,3,1620-1627.
    [35]BeutlerE G,G.A.,2001.
    [36]Futerman A H, van Meer G. The cell biology of lysosomal storage disorders: Nat Rev Mol Cell Biol,2004,5,554-565.
    [37]吴希如;包新华.溶酶体贮积症的诊断与治疗进展.北京大学学报(医学版),2005,27,440-444.
    [38]Hruska K S, LaMarca M E, Scott C R, et al. Gaucher disease:mutation and polymorphism spectrum in the glucocerebrosidase gene (GBA). Hum Mutat,2008,29,567-583.
    [39]Zeller J L, Burke A E, Glass R M. JAMA patient page. Gaucher disease. JAMA,2007,298, 1358.
    [40]Harmanci O, Bayraktar Y. Gaucher disease:new developments in treatment and etiology. World J Gastroenterol,2008,14,3968-3973.
    [41]Elstein D, Hollak C, Aerts J M, et al. Sustained therapeutic effects of oral miglustat (Zavesca, N-butyldeoxynojirimycin, OGT 918) in type I Gaucher disease. J Inherit Metab Dis,2004,27, 757-766.
    [42]Weinreb N J, Barranger J A, Charrow J, et al. Guidance on the use of miglustat for treating patients with type 1 Gaucher disease. Am J Hematol,2005,80,223-229.
    [43]Benito J M, Garcia Fernandez J M, Ortiz Mellet C. Pharmacological chaperone therapy for Gaucher disease:a patent review. Expert Opin Ther Pat,2011,21,885-903.
    [44]Dvir H, Harel M, McCarthy A A, et al. X-ray structure of human acid-beta-glucosidase, the defective enzyme in Gaucher disease. EMBO reports,2003,4,704-709.
    [45]Schueler U H, Kolter T, Kaneski C R, et al. Correlation between enzyme activity and substrate storage in a cell culture model system for Gaucher disease. J Inherit Metab Dis,2004, 27,649-658.
    [46]Suzuki Y, Ogawa S, Sakakibara Y. Chaperone therapy for neuronopathic lysosomal diseases: competitive inhibitors as chemical chaperones for enhancement of mutant enzyme activities. Perspectives in medicinal chemistry,2009,3,7-19.
    [47]Sawkar A R, Cheng W C, Beutler E, et al. Chemical chaperones increase the cellular activity of N370S beta-glucosidase:a therapeutic strategy for Gaucher disease. Proceedings of the National Academy of Sciences of the United States of America,2002,99,15428-15433.
    [48]Kelly J W, Sawkar A R, Beutler E, et al. Chemical chaperones and their effect upon the cellular activity of beta-glucosidase. PCT, US2003/033670,2004.
    [49]Compain P, Martin O R, Boucheron C, et al. Design and synthesis of highly potent and selective pharmacological chaperones for the treatment of Gaucher's disease. Chembiochem, 2006,7,1356-1359.
    [50]Steet R A, Chung S, Wustman B, et al. The iminosugar isofagomine increases the activity of N370S mutant acid beta-glucosidase in Gaucher fibroblasts by several mechanisms. Proceedings of the National Academy of Sciences of the United States of America,2006,103, 13813-13818.
    [51]Yu Z, Sawkar A R, Whalen L J, et al. Isofagomine and 2,5-anhydro-2,5-imino-D-glucitol-based glucocerebrosidase pharmacological chaperones for Gaucher disease intervention. Journal of medicinal chemistry,2007,50,94-100.
    [52]Khanna R, Benjamin E R, Pellegrino L, et al. The pharmacological chaperone isofagomine increases the activity of the Gaucher disease L444P mutant form of beta-glucosidase. FEBS J, 2010,277,1618-1638.
    [53]Wang G N, Reinkensmeier G, Zhang S W, et al. Rational design and synthesis of highly potent pharmacological chaperones for treatment of N370S mutant Gaucher disease. Journal of medicinal chemistry,2009,52,3146-3149.
    [54]Luan Z, Higaki K, Aguilar-Moncayo M, et al. Chaperone activity of bicyclic nojirimycin analogues for Gaucher mutations in comparison with N-(n-nonyl)deoxynojirimycin. Chembiochem,2009,10,2780-2792.
    [55]Wang G N, Twigg G, Butters T D, et al. Synthesis of N-substituted epsilon-hexonolactams as pharmacological chaperones for the treatment of N370S mutant Gaucher disease. Org Biomol Chem,2012,10,2923-2927.
    [56]Goddard-Borger E D, Tropak M B, Yonekawa S, et al. Rapid assembly of a library of lipophilic iminosugars via the thiol-ene reaction yields promising pharmacological chaperones for the treatment of Gaucher disease. Journal of medicinal chemistry,2012,55,2737-2745.
    [57]Fan J Q, Zhu X; Sheth K. New glucoimidazole and polyhydroxycyclohexenyl amine derivatives are glucocerebrosidase inhibitors useful to treat Gaucher's disease. PCT, US2004/037703,2005.
    [58]Hersher R. Small biotechs raring to cash in on the orphan disease market. Nat Med,2012,18, 330-331.
    [59]Clarke J T, Mahuran D J, Same S, et al. An open-label Phase I/II clinical trial of pyrimethamine for the treatment of patients affected with chronic GM2 gangliosidosis (Tay-Sachs or Sandhoff variants). Mol Genet Metab,2011,102,6-12.
    [60]Diaz L, Casas J, Bujons J, et al. New glucocerebrosidase inhibitors by exploration of chemical diversity of N-substituted aminocyclitols using click chemistry and in situ screening. Journal of medicinal chemistry,2011,54,2069-2079.
    [61]Park C, Marqusee S. Pulse proteolysis:a simple method for quantitative determination of protein stability and ligand binding. Nat Methods,2005,2,207-212.
    [62]Clark N E, Metcalf M C, Best D, et al. Pharmacological chaperones for human alpha-N-acetylgalactosaminidase. Proceedings of the National Academy of Sciences of the United States of America,2012,109,17400-17405.
    [63]Trapero A, Alfonso I, Butters T D, et al. Polyhydroxylated bicyclic isoureas and guanidines are potent glucocerebrosidase inhibitors and nanomolar enzyme activity enhancers in Gaucher cells. Journal of the American Chemical Society,2011,133,5474-5484.
    [64]Trapero A, Gonzalez-Bulnes P, Butters T D, et al. Potent aminocyclitol glucocerebrosidase inhibitors are subnanomolar pharmacological chaperones for treating gaucher disease. Journal of medicinal chemistry,2012,55,4479-4488.
    [65]Hill T, Tropak M B, Mahuran D, et al. Synthesis, kinetic evaluation and cell-based analysis of C-alkylated isofagomines as chaperones of beta-glucocerebrosidase. Chembiochem,2011, 12,2151-2154.
    [66]Norgan A P, Coffman P K, Kocher J P, et al. Multilevel Parallelization of AutoDock 4.2. J Cheminform,2011,3,12.
    [67]Goodsell D S. Computational docking of biomolecular complexes with AutoDock. Cold Spring Harb Protoc,2009,2009, pdb prot5200.
    [68]Wang Y, Bolton E, Dracheva S, et al. An overview of the PubChem BioAssay resource. Nucleic acids research,2010,38, D255-266.
    [69]Schrodinger, LLC,2010.
    [70]Laskowski R A, Swindells M B. LigPlotf+:multiple ligand-protein interaction diagrams for drug discovery. Journal of chemical information and modeling,2011,51,2778-2786.
    [71]Li T, Guo L, Zhang Y, et al. Structure-activity relationships in a series of C2-substituted gluco-configured tetrahydroimidazopyridines as beta-glucosidase inhibitors. Bioorganic & medicinal chemistry,2011,19,2136-2144.
    [72]Pasmanik-Chor M, Elroy-Stein O, Aerts H, et al. Overexpression of human glucocerebrosidase containing different-sized leaders. The Biochemical journal,1996,317 (Pt 1),81-88.
    [73]Brumshtein B, Greenblatt H M, Butters T D, et al. Crystal structures of complexes of N-butyl-and N-nonyl-deoxynojirimycin bound to acid beta-glucosidase:insights into the mechanism of chemical chaperone action in Gaucher disease. The Journal of biological chemistry,2007,282,29052-29058.
    [74]Lieberman R L, Wustman B A, Huertas P, et al. Structure of acid beta-glucosidase with pharmacological chaperone provides insight into Gaucher disease. Nature chemical biology, 2007,3,101-107.
    [75]Hanahan D, Weinberg R A. The hallmarks of cancer. Cell,2000,100,57-70.
    [76]Kinzler K W, Vogelstein B. Cancer-susceptibility genes. Gatekeepers and caretakers. Nature, 1997,386,761,763.
    [77]Baylin S B, Ohm J E. Epigenetic gene silencing in cancer-a mechanism for early oncogenic pathway addiction? Nature reviews. Cancer,2006,6,107-116.
    [78]Lund A H, van Lohuizen M. Epigenetics and cancer. Genes & development,2004,18, 2315-2335.
    [79]Choudhary C, Kumar C, Gnad F, et al. Lysine acetylation targets protein complexes and co-regulates major cellular functions. Science,2009,325,834-840.
    [80]Wolffe A P. Histone deacetylase:a regulator of transcription. Science,1996,272,371-372.
    [81]Roth S Y, Denu J M, Allis C D. Histone acetyltransferases. Annual review of biochemistry, 2001,70,81-120.
    [82]Marmorstein R, Roth S Y. Histone acetyltransferases:function, structure, and catalysis. Current opinion in genetics & development,2001,11,155-161.
    [83]Marmorstein R. Structure of histone deacetylases:insights into substrate recognition and catalysis. Structure,2001,9,1127-1133.
    [84]Hodawadekar S C, Marmorstein R. Chemistry of acetyl transfer by histone modifying enzymes:structure, mechanism and implications for effector design. Oncogene,2007,26, 5528-5540.
    [85]Yang X J, Seto E. HATs and HDACs:from structure, function and regulation to novel strategies for therapy and prevention. Oncogene,2007,26,5310-5318.
    [86]Schlissel M S. Perspectives:transcription. A tail of histone acetylation and DNA recombination. Science,2000,287,438-440.
    [87]Daly K, Shirazi-Beechey S P. Microarray analysis of butyrate regulated genes in colonic epithelial cells. DNA and cell biology,2006,25,49-62.
    [88]Marks P A. Discovery and development of SAHA as an anticancer agent. Oncogene,2007, 26,1351-1356.
    [89]Zhang Y, Fang H, Jiao J, et al. The structure and function of histone deacetylases:the target for anti-cancer therapy. Current medicinal chemistry,2008,15,2840-2849.
    [90]Corminboeuf C, Hu P, Tuckerman M E, et al. Unexpected deacetylation mechanism suggested by a density functional theory QM/MM study of histone-deacetylase-like protein. Journal of the American Chemical Society,2006,128,4530-4531.
    [91]Vannini A, Volpari C, Filocamo G, et al. Crystal structure of a eukaryotic zinc-dependent histone deacetylase, human HDAC8, complexed with a hydroxamic acid inhibitor. Proceedings of the National Academy of Sciences of the United States of America,2004,101, 15064-15069.
    [92]Somoza J R, Skene R J, Katz B A, et al. Structural snapshots of human HDAC8 provide insights into the class I histone deacetylases. Structure,2004,12,1325-1334.
    [93]Nielsen T K, Hildmann C, Riester D, et al. Complex structure of a bacterial class 2 histone deacetylase homologue with a trifluoromethylketone inhibitor. Acta crystallographica. Section F, Structural biology and crystallization communications,2007,63,270-273.
    [94]Kim H M, Ryu D K, Choi Y, et al. Structure-activity relationship studies of a series of novel delta-lactam-based histone deacetylase inhibitors. Journal of medicinal chemistry,2007,50, 2737-2741.
    [95]Fraga M F, Ballestar E, Villar-Garea A, et al. Loss of acetylation at Lys16 and trimethylation at Lys20 of histone H4 is a common hallmark of human cancer. Nature genetics,2005,37, 391-400.
    [96]Yasui W, Oue N, Ono S, et al. Histone acetylation and gastrointestinal carcinogenesis. Annals of the New York Academy of Sciences,2003,983,220-231.
    [97]Lin R J, Sternsdorf T, Tini M, et al. Transcriptional regulation in acute promyelocytic leukemia. Oncogene,2001,20,7204-7215.
    [98]Wilson A J, Byun D S, Popova N, et al. Histone deacetylase 3 (HDAC3) and other class I HDACs regulate colon cell maturation and p21 expression and are deregulated in human colon cancer. The Journal of biological chemistry,2006,281,13548-13558.
    [99]Choi J H, Kwon H J, Yoon B I, et al. Expression profile of histone deacetylase 1 in gastric cancer tissues. Japanese journal of cancer research:Gann,2001,92,1300-1304.
    [100]Halkidou K, Gaughan L, Cook S, et al. Upregulation and nuclear recruitment of HDAC1 in hormone refractory prostate cancer. The Prostate,2004,59,177-189.
    [101]Zhang Z, Yamashita H, Toyama T, et al. Quantitation of HDAC1 mRNA expression in invasive carcinoma of the breast*. Breast cancer research and treatment,2005,94,11-16.
    [102]Huang B H, Laban M, Leung C H, et al. Inhibition of histone deacetylase 2 increases apoptosis and p21Cipl/WAF1 expression, independent of histone deacetylase 1. Cell death and differentiation,2005,12,395-404.
    [103]Song J, Noh J H, Lee J H, et al. Increased expression of histone deacetylase 2 is found in human gastric cancer. APMIS:acta pathologica, microbiologica, et immunologica Scandinavica,2005,113,264-268.
    [104]Zhang Z, Yamashita H, Toyama T, et al. HDAC6 expression is correlated with better survival in breast cancer. Clinical cancer research:an official journal of the American Association for Cancer Research,2004,10,6962-6968.
    [105]Ropero S, Esteller M. The role of histone deacetylases (HDACs) in human cancer. Molecular oncology,2007,1,19-25.
    [106]Bi G, Jiang G. The molecular mechanism of HDAC inhibitors in anticancer effects. Cellular & molecular immunology,2006,3,285-290.
    [107]谢华峰,陆涛.抗肿瘤药物组蛋白去乙酰化酶抑制剂的研究进展(下).中国药师,2010,807-810.
    [108]Kozikowski A P, Chen Y, Gaysin A, et al. Functional differences in epigenetic modulators-superiority of mercaptoacetamide-based histone deacetylase inhibitors relative to hydroxamates in cortical neuron neuroprotection studies. Journal of medicinal chemistry,2007, 50,3054-3061.
    [109]Itoh Y, Suzuki T, Kouketsu A, et al. Design, synthesis, structure--selectivity relationship, and effect on human cancer cells of a novel series of histone deacetylase 6-selective inhibitors. Journal of medicinal chemistry,2007,50,5425-5438.
    [110]Lindemann R K, Gabrielli B, Johnstone R W. Histone-deacetylase inhibitors for the treatment of cancer. Cell Cycle,2004,3,779-788.
    [111]Drummond D C, Noble C O, Kirpotin D B, et al. Clinical development of histone deacetylase inhibitors as anticancer agents. Annual review of pharmacology and toxicology, 2005,45,495-528.
    [112]Bhalla K N. Epigenetic and chromatin modifiers as targeted therapy of hematologic malignancies. Journal of clinical oncology:official journal of the American Society of Clinical Oncology,2005,23,3971-3993.
    [113]Cote S, Rosenauer A, Bianchini A, et al. Response to histone deacetylase inhibition of novel PML/RARalpha mutants detected in retinoic acid-resistant APL cells. Blood,2002,100, 2586-2596.
    [114]He L Z, Tolentino T, Grayson P, et al. Histone deacetylase inhibitors induce remission in transgenic models of therapy-resistant acute promyelocytic leukemia. The Journal of clinical investigation,2001,108,1321-1330.
    [115]Warrell R P, Jr., He L Z, Richon V, et al. Therapeutic targeting of transcription in acute promyelocytic leukemia by use of an inhibitor of histone deacetylase. Journal of the National Cancer Institute,1998,90,1621-1625.
    [116]Reynolds C P, Lemons R S. Retinoid therapy of childhood cancer. Hematology/oncology clinics of North America,2001,15,867-910.
    [117]Coffey D C, Kutko M C, Glick R D, et al. The histone deacetylase inhibitor, CBHA, inhibits growth of human neuroblastoma xenografts in vivo, alone and synergistically with all-trans retinoic acid. Cancer research,2001,61,3591-3594.
    [118]Cameron E E, Bachman K E, Myohanen S, et al. Synergy of demethylation and histone deacetylase inhibition in the re-expression of genes silenced in cancer. Nature genetics,1999, 21,103-107.
    [119]Gore S D, Baylin S, Sugar E, et al. Combined DNA methyltransferase and histone deacetylase inhibition in the treatment of myeloid neoplasms. Cancer research,2006,66, 6361-6369.
    [120]Kim M S, Blake M, Baek J H, et al. Inhibition of histone deacetylase increases cytotoxicity to anticancer drugs targeting DNA. Cancer research,2003,63,7291-7300.
    [121]Fuino L, Bali P, Wittmann S, et al. Histone deacetylase inhibitor LAQ824 down-regulates Her-2 and sensitizes human breast cancer cells to trastuzumab, taxotere, gemcitabine, and epothilone B. Molecular cancer therapeutics,2003,2,971-984.
    [122]Catley L, Weisberg E, Tai Y T, et al. NVP-LAQ824 is a potent novel histone deacetylase inhibitor with significant activity against multiple myeloma. Blood,2003,102,2615-2622.
    [123]Liu H L, Chen Y, Cui G H, et al. Curcumin, a potent anti-tumor reagent, is a novel histone deacetylase inhibitor regulating B-NHL cell line Raji proliferation. Acta pharmacologica Sinica,2005,26,603-609.
    [124]Loprevite M, Tiseo M, Grossi F, et al. In vitro study of CI-994, a histone deacetylase inhibitor, in non-small cell lung cancer cell lines. Oncology research,2005,15,39-48.
    [125]Zhang X D, Gillespie S K, Borrow J M, et al. The histone deacetylase inhibitor suberic bishydroxamate regulates the expression of multiple apoptotic mediators and induces mitochondria-dependent apoptosis of melanoma cells. Molecular cancer therapeutics,2004,3, 425-435.
    [126]Nakata S, Yoshida T, Horinaka M, et al. Histone deacetylase inhibitors upregulate death receptor 5/TRAIL-R2 and sensitize apoptosis induced by TRAIL/APO2-L in human malignant tumor cells. Oncogene,2004,23,6261-6271.
    [127]Inoue S, MacFarlane M, Harper N, et al. Histone deacetylase inhibitors potentiate TNF-related apoptosis-inducing ligand (TRAIL)-induced apoptosis in lymphoid malignancies. Cell death and differentiation,2004,11 Suppl 2, SI93-206.
    [128]Guo F, Sigua C, Tao J, et al. Cotreatment with histone deacetylase inhibitor LAQ824 enhances Apo-2L/tumor necrosis factor-related apoptosis inducing ligand-induced death inducing signaling complex activity and apoptosis of human acute leukemia cells. Cancer research,2004,64,2580-2589.
    [129]Singh T R, Shankar S, Srivastava R K. HDAC inhibitors enhance the apoptosis-inducing potential of TRAIL in breast carcinoma. Oncogene,2005,24,4609-4623.
    [130]Kim-E H, Kim H S, Kim S U, et al. Sodium butyrate sensitizes human glioma cells to TRAIL-mediated apoptosis through inhibition of Cdc2 and the subsequent downregulation of survivin and XIAP. Oncogene,2005,24,6877-6889.
    [131]Earel J K, Jr., VanOosten R L, Griffith T S. Histone deacetylase inhibitors modulate the sensitivity of tumor necrosis factor-related apoptosis-inducing ligand-resistant bladder tumor cells. Cancer research,2006,66,499-507.
    [132]Rosato R R, Almenara J A, Dai Y, et al. Simultaneous activation of the intrinsic and extrinsic pathways by histone deacetylase (HDAC) inhibitors and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) synergistically induces mitochondrial damage and apoptosis in human leukemia cells. Molecular cancer therapeutics,2003,2, 1273-1284.
    [133]Rosato R R, Almenara J A, Yu C, et al. Evidence of a functional role for p21WAFl/CIP1 down-regulation in synergistic antileukemic interactions between the histone deacetylase inhibitor sodium butyrate and flavopiridol. Molecular pharmacology,2004,65,571-581.
    [134]Rahmani M, Yu C, Reese E, et al. Inhibition of PI-3 kinase sensitizes human leukemic cells to histone deacetylase inhibitor-mediated apoptosis through p44/42 MAP kinase inactivation and abrogation of p21(CIPlAVAF1) induction rather than AKT inhibition. Oncogene,2003, 22,6231-6242.
    [135]Bali P, George P, Cohen P, et al. Superior activity of the combination of histone deacetylase inhibitor LAQ824 and the FLT-3 kinase inhibitor PKC412 against human acute myelogenous leukemia cells with mutant FLT-3. Clinical cancer research:an official journal of the American Association for Cancer Research,2004,10,4991-4997.
    [136]Yu C, Dasmahapatra G, Dent P, et al. Synergistic interactions between MEK1/2 and histone deacetylase inhibitors in BCR/ABL+human leukemia cells. Leukemia,2005,19,1579-1589.
    [137]Deininger M, Buchdunger E, Druker B J. The development of imatinib as a therapeutic agent for chronic myeloid leukemia. Blood,2005,105,2640-2653.
    [138]Nimmanapalli R, Fuino L, Bali P, et al. Histone deacetylase inhibitor LAQ824 both lowers expression and promotes proteasomal degradation of Bcr-Abl and induces apoptosis of imatinib mesylate-sensitive or-refractory chronic myelogenous leukemia-blast crisis cells. Cancer research,2003,63,5126-5135.
    [139]Yu C, Rahmani M, Almenara J, et al. Histone deacetylase inhibitors promote STI571-mediated apoptosis in STI571-sensitive and-resistant Bcr/Abl+human myeloid leukemia cells. Cancer research,2003,63,2118-2126.
    [140]George P, Bali P, Annavarapu S, et al. Combination of the histone deacetylase inhibitor LBH589 and the hsp90 inhibitor 17-AAG is highly active against human CML-BC cells and AML cells with activating mutation of FLT-3. Blood,2005,105,1768-1776.
    [141]Kim J S, Jeung H K, Cheong J W, et al. Apicidin potentiates the imatinib-induced apoptosis of Bcr-Abl-positive human leukaemia cells by enhancing the activation of mitochondria-dependent caspase cascades. British journal of haematology,2004,124,166-178.
    [142]Kawano T, Horiguchi-Yamada J, Iwase S, et al. Depsipeptide enhances imatinib mesylate-induced apoptosis of Bcr-Abl-positive cells and ectopic expression of cyclin D1, c-Myc or active MEK abrogates this effect. Anticancer research,2004,24,2705-2712.
    [143]Nimmanapalli R, Fuino L, Stobaugh C, et al. Cotreatment with the histone deacetylase inhibitor suberoylanilide hydroxamic acid (SAHA) enhances imatinib-induced apoptosis of Bcr-Abl-positive human acute leukemia cells. Blood,2003,101,3236-3239.
    [144]Rahmani M, Reese E, Dai Y, et al. Cotreatment with suberanoylanilide hydroxamic acid and 17-allylamino 17-demethoxygeldanamycin synergistically induces apoptosis in Bcr-Abl+ Cells sensitive and resistant to STI571 (imatinib mesylate) in association with down-regulation of Bcr-AbI, abrogation of signal transducer and activator of transcription 5 activity, and Bax conformational change. Molecular pharmacology,2005,67,1166-1176.
    [145]Wang X, Ju W, Renouard J, et al.17-allylamino-17-demethoxygeldanamycin synergistically potentiates tumor necrosis factor-induced lung cancer cell death by blocking the nuclear factor-kappaB pathway. Cancer research,2006,66,1089-1095.
    [146]Bali P, Pranpat M, Bradner J, et al. Inhibition of histone deacetylase 6 acetylates and disrupts the chaperone function of heat shock protein 90:a novel basis for antileukemia activity of histone deacetylase inhibitors. The Journal of biological chemistry,2005,280, 26729-26734.
    [147]Pei X Y, Dai Y, Grant S. Synergistic induction of oxidative injury and apoptosis in human multiple myeloma cells by the proteasome inhibitor bortezomib and histone deacetylase inhibitors. Clinical cancer research:an official journal of the American Association for Cancer Research,2004,10,3839-3852.
    [148]Yu C, Rahmani M, Conrad D, et al. The proteasome inhibitor bortezomib interacts synergistically with histone deacetylase inhibitors to induce apoptosis in Bcr/Abl+cells sensitive and resistant to STI571. Blood,2003,102,3765-3774.
    [149]Kopito R R. Aggresomes, inclusion bodies and protein aggregation. Trends in cell biology, 2000,10,524-530.
    [150]Kawaguchi Y, Kovacs J J, McLaurin A, et al. The deacetylase HDAC6 regulates aggresome formation and cell viability in response to misfolded protein stress. Cell,2003,115,727-738.
    [151]Hideshima T, Bradner J E, Wong J, et al. Small-molecule inhibition of proteasome and aggresome function induces synergistic antitumor activity in multiple myeloma. Proceedings of the National Academy of Sciences of the United States of America,2005,102,8567-8572.
    [152]Nome R V, Bratland A, Harman G, et al. Cell cycle checkpoint signaling involved in histone deacetylase inhibition and radiation-induced cell death. Molecular cancer therapeutics,2005,4, 1231-1238.
    [153]Karagiannis T C, Harikrishnan K N, El-Osta A. The histone deacetylase inhibitor, Trichostatin A, enhances radiation sensitivity and accumulation of gammaH2A.X. Cancer biology& therapy,2005,4,787-793.
    [154]Camphausen K, Cerna D, Scott T, et al. Enhancement of in vitro and in vivo tumor cell radiosensitivity by valproic acid. International journal of cancer. Journal international du cancer,20055114,380-386.
    [155]Zhang Y, Jung M, Dritschilo A. Enhancement of radiation sensitivity of human squamous carcinoma cells by histone deacetylase inhibitors. Radiation research,2004,161,667-674.
    [156]Kim J H, Shin J H, Kim I H. Susceptibility and radiosensitization of human glioblastoma cells to trichostatin A, a histone deacetylase inhibitor. International journal of radiation oncology, biology, physics,2004,59,1174-1180.
    [157]Camphausen K, Scott T, Sproull M, et al. Enhancement of xenograft tumor radiosensitivity by the histone deacetylase inhibitor MS-275 and correlation with histone hyperacetylation. Clinical cancer research:an official journal of the American Association for Cancer Research, 2004,10,6066-6071.
    [158]Shiloh Y. ATM and related protein kinases:safeguarding genome integrity. Nature reviews. Cancer,2003,3,155-168.
    [159]Bakkenist C J, Kastan M B. DNA damage activates ATM through intermolecular autophosphorylation and dimer dissociation. Nature,2003,421,499-506.
    [160]Kim G D, Choi Y H, Dimtchev A, et al. Sensing of ionizing radiation-induced DNA damage by ATM through interaction with histone deacetylase. The Journal of biological chemistry, 1999,274,31127-31130.
    [161]Munshi A, Kurland J F, Nishikawa T, et al. Histone deacetylase inhibitors radiosensitize human melanoma cells by suppressing DNA repair activity. Clinical cancer research:an official journal of the American Association for Cancer Research,2005,11,4912-4922.
    [162]Yaneva M, Li H, Marple T, et al. Non-homologous end joining, but not homologous recombination, enables survival for cells exposed to a histone deacetylase inhibitor. Nucleic acids research,2005,33,5320-5330.
    [163]Chung Y L, Wang A J, Yao L F. Antitumor histone deacetylase inhibitors suppress cutaneous radiation syndrome:Implications for increasing therapeutic gain in cancer radiotherapy. Molecular cancer therapeutics,2004,3,317-325.
    [164]Torres C R, Hart G W. Topography and polypeptide distribution of terminal N-acetylglucosamine residues on the surfaces of intact lymphocytes. Evidence for O-linked GlcNAc. The Journal of biological chemistry,1984,259,3308-3317.
    [165]Holt G D, Hart G W. The subcellular distribution of terminal N-acetylglucosamine moieties. localization of a novel protein-saccharide linkage, O-linked GlcNAc. The Journal of biological chemistry,1986,261,8049-8057.
    [166]Davis L I, Blobel G. Identification and characterization of a nuclear pore complex protein. Cell,1986,45,699-709.
    [167]Snow C M, Senior A, Gerace L. Monoclonal antibodies identify a group of nuclear pore complex glycoproteins. The Journal of cell biology,1987,104,1143-1156.
    [168]Hanover J A, Cohen C K, Willingham M C, et al. O-linked N-acetylglucosamine is attached to proteins of the nuclear pore. Evidence for cytoplasmic and nucleoplasmic glycoproteins. The Journal of biological chemistry,1987,262,9887-9894.
    [169]Turner J R, Tartakoff A M, Greenspan N S. Cytologic assessment of nuclear and cytoplasmic O-linked N-acetylglucosamine distribution by using anti-streptococcal monoclonal antibodies. Proceedings of the National Academy of Sciences of the United States of America,1990,87,5608-5612.
    [170]Comer F I, Vosseller K, Wells L, et al. Characterization of a mouse monoclonal antibody specific for O-linked N-acetylglucosamine. Analytical biochemistry,2001,293,169-177.
    [171]Holt G D, Snow C M, Senior A, et al. Nuclear pore complex glycoproteins contain cytoplasmically disposed O-linked N-acetylglucosamine. The Journal of cell biology,1987, 104,1157-1164.
    [172]Hart G W, Housley M P, Slawson C. Cycling of O-linked beta-N-acetylglucosamine on nucleocytoplasmic proteins. Nature,2007,446,1017-1022.
    [173]Chou C F, Smith A J, Omary M B. Characterization and dynamics of O-linked glycosylation of human cytokeratin 8 and 18. The Journal of biological chemistry,1992,267,3901-3906.
    [174]Roquemore E P, Chevrier M R, Cotter R J, et al. Dynamic O-GlcNAcylation of the small heat shock protein alpha B-crystallin. Biochemistry,1996,35,3578-3586.
    [175]O'Donnell N, Zachara N E, Hart G W, et al. Ogt-dependent X-chromosome-linked protein glycosylation is a requisite modification in somatic cell function and embryo viability. Molecular and cellular biology,2004,24,1680-1690.
    [176]Haltiwanger R S, Grove K, Philipsberg G A. Modulation of O-linked N-acetylglucosamine levels on nuclear and cytoplasmic proteins in vivo using the peptide O-GlcNAc-beta-N-acetylglucosaminidase inhibitor O-(2-acetamido-2-deoxy-D-glucopyranosylidene) amino-N-phenylcarbamate. The Journal of biological chemistry,1998,273,3611-3617.
    [177]Sakabe K, Wang Z, Hart G W. Beta-N-acetylglucosamine (O-GlcNAc) is part of the histone code. Proceedings of the National Academy of Sciences of the United States of America,2010, 107,19915-19920.
    [178]Strahl B D, Allis C D. The language of covalent histone modifications. Nature,2000,403, 41-45.
    [179]Zhang Y, Reinberg D. Transcription regulation by histone methylation:interplay between different covalent modifications of the core histone tails. Genes & development,2001,15, 2343-2360.
    [180]Liu F, Iqbal K, Grundke-Iqbal I, et al. O-GlcNAcylation regulates phosphorylation of tau:a mechanism involved in Alzheimer's disease. Proceedings of the National Academy of Sciences of the United States of America,2004,101,10804-10809.
    [181]Tallent M K, Varghis N, Skorobogatko Y, et al. In vivo modulation of O-GlcNAc levels regulates hippocampal synaptic plasticity through interplay with phosphorylation. The Journal of biological chemistry,2009,284,174-181.
    [182]Wang Z, Gucek M, Hart G W. Cross-talk between GlcNAcylation and phosphorylation: site-specific phosphorylation dynamics in response to globally elevated O-GlcNAc. Proceedings of the National Academy of Sciences of the United States of America,2008,105, 13793-13798.
    [183]Wang Z, Pandey A, Hart G W. Dynamic interplay between O-linked N-acetylglucosaminylation and glycogen synthase kinase-3-dependent phosphorylation. Molecular & cellular proteomics:MCP,2007,6,1365-1379.
    [184]Cheng X, Cole R N, Zaia J, et al. Alternative O-glycosylation/O-phosphorylation of the murine estrogen receptor beta. Biochemistry,2000,39,11609-11620.
    [185]Khidekel N, Ficarro S B, Clark P M, et al. Probing the dynamics of O-GlcNAc glycosylation in the brain using quantitative proteomics. Nature chemical biology,2007,3, 339-348.
    [186]Manning G, Whyte D B, Martinez R, et al. The protein kinase complement of the human genome. Science,2002,298,1912-1934.
    [187]van Putten J P, Krans H M. Glucose as a regulator of insulin-sensitive hexose uptake in 3T3 adipocytes. The Journal of biological chemistry,1985,260,7996-8001.
    [188]Nawano M, Oku A, Ueta K, et al. Hyperglycemia contributes insulin resistance in hepatic and adipose tissue but not skeletal muscle of ZDF rats. American journal of physiology. Endocrinology and metabolism,2000,278, E535-543.
    [189]Kim J K, Zisman A, Fillmore J J, et al. Glucose toxicity and the development of diabetes in mice with muscle-specific inactivation of GLUT4. The Journal of clinical investigation,2001, 108,153-160.
    [190]Nelson B A, Robinson K A, Buse M G Defective Akt activation is associated with glucose-but not glucosamine-induced insulin resistance. American journal of physiology. Endocrinology and metabolism,2002,282, E497-506.
    [191]Erol A. Insulin resistance is an evolutionarily conserved physiological mechanism at the cellular level for protection against increased oxidative stress. BioEssays:news and reviews in molecular, cellular and developmental biology,2007,29,811-818.
    [192]Hoehn K L, Salmon A B, Hohnen-Behrens C, et al. Insulin resistance is a cellular antioxidant defense mechanism. Proceedings of the National Academy of Sciences of the United States of America,2009,106,17787-17792.
    [193]Porte D, Jr., Schwartz M W. Diabetes complications:why is glucose potentially toxic? Science,1996,272,699-700.
    [194]Walgren J L, Vincent T S, Schey K L, et al. High glucose and insulin promote O-GlcNAc modification of proteins, including alpha-tubulin. American journal of physiology. Endocrinology and metabolism,2003,284, E424-434.
    [195]Yki-Jarvinen H, Virkamaki A, Daniels M C, et al. Insulin and glucosamine infusions increase O-linked N-acetyl-glucosamine in skeletal muscle proteins in vivo. Metabolism: clinical and experimental,1998,47,449-455.
    [196]Hazel M, Cooksey R C, Jones D, et al. Activation of the hexosamine signaling pathway in adipose tissue results in decreased serum adiponectin and skeletal muscle insulin resistance. Endocrinology,2004,145,2118-2128.
    [197]Marshall S, Bacote V, Traxinger R R. Discovery of a metabolic pathway mediating glucose-induced desensitization of the glucose transport system. Role of hexosamine biosynthesis in the induction of insulin resistance. The Journal of biological chemistry,1991, 266,4706-4712.
    [198]Crook E D, Daniels M C, Smith T M, et al. Regulation of insulin-stimulated glycogen synthase activity by overexpression of glutamine:fructose-6-phosphate amidotransferase in rat-1 fibroblasts. Diabetes,1993,42,1289-1296.
    [199]Akimoto Y, Hart G W, Wells L, et al. Elevation of the post-translational modification of proteins by O-linked N-acetylglucosamine leads to deterioration of the glucose-stimulated insulin secretion in the pancreas of diabetic Goto-Kakizaki rats. Glycobiology,2007,17, 127-140.
    [200]Federici M, Menghini R, Mauriello A, et al. Insulin-dependent activation of endothelial nitric oxide synthase is impaired by O-linked glycosylation modification of signaling proteins in human coronary endothelial cells. Circulation,2002,106,466-472.
    [201]Hanover J A. Glycan-dependent signaling:O-linked N-acetylglucosamine. The FASEB journal:official publication of the Federation of American Societies for Experimental Biology, 2001,15,1865-1876.
    [202]Baron A D, Zhu J S, Zhu J H, et al. Glucosamine induces insulin resistance in vivo by affecting GLUT 4 translocation in skeletal muscle. Implications for glucose toxicity. The Journal of clinical investigation,1995,96,2792-2801.
    [203]Zachara N E, O'Donnell N, Cheung W D, et al. Dynamic O-GlcNAc modification of nucleocytoplasmic proteins in response to stress. A survival response of mammalian cells. The Journal of biological chemistry,2004,279,30133-30142.
    [204]Taylor R P, Parker G J, Hazel M W, et al. Glucose deprivation stimulates O-GlcNAc modification of proteins through up-regulation of O-linked N-acetylglucosaminyltransferase. The Journal of biological chemistry,2008,283,6050-6057.
    [205]Cheung W D, Hart G W. AMP-activated protein kinase and p38 MAPK activate O-GlcNAcylation of neuronal proteins during glucose deprivation. The Journal of biological chemistry,2008,283,13009-13020.
    [206]Jones S P, Zachara N E, Ngoh G A, et al. Cardioprotection by N-acetylglucosamine linkage to cellular proteins. Circulation,2008,117,1172-1182.
    [207]Garvey W T, Olefsky J M, Griffin J, et al. The effect of insulin treatment on insulin secretion and insulin action in type II diabetes mellitus. Diabetes,1985,34,222-234.
    [208]McCIain D A. Hexosamines as mediators of nutrient sensing and regulation in diabetes. Journal of diabetes and its complications,2002,16,72-80.
    [209]McCIain D A, Lubas W A, Cooksey R C, et al. Altered glycan-dependent signaling induces insulin resistance and hyperleptinemia. Proceedings of the National Academy of Sciences of the United States of America,2002,99,10695-10699.
    [210]Love D C, Hanover J A. The hexosamine signaling pathway:deciphering the "O-GlcNAc code". Science's STKE:signal transduction knowledge environment,2005,2005, re13.
    [211]Issad T, Combettes M, Ferre P. Isoproterenol inhibits insulin-stimulated tyrosine phosphorylation of the insulin receptor without increasing its serine/threonine phosphorylation. European journal of biochemistry/FEBS,1995,234,108-115.
    [212]Combettes-Souverain M, Issad T. Molecular basis of insulin action. Diabetes & metabolism, 1998,24,477-489.
    [213]Lefebvre T, Dehennaut V, Guinez C, et al. Dysregulation of the nutrient/stress sensor O-GlcNAcylation is involved in the etiology of cardiovascular disorders, type-2 diabetes and Alzheimer's disease. Biochimica et biophysica acta,2010,1800,67-79.
    [214]Yang X, Ongusaha P P, Miles P D, et al. Phosphoinositide signalling links O-GlcNAc transferase to insulin resistance. Nature,2008,451,964-969.
    [215]Issad T, Kuo M. O-GlcNAc modification of transcription factors, glucose sensing and glucotoxicity. Trends in endocrinology and metabolism:TEM,2008,19,380-389.
    [216]Kuo M, Zilberfarb V, Gangneux N, et al. O-GlcNAc modification of FoxOl increases its transcriptional activity:a role in the glucotoxicity phenomenon? Biochimie,2008,90, 679-685.
    [217]Laczy B, Hill B G, Wang K, et al. Protein O-GlcNAcylation:a new signaling paradigm for the cardiovascular system. American journal of physiology. Heart and circulatory physiology, 2009,296, H13-28.
    [218]Chatham J C, Not L G, Fulop N, et al. Hexosamine biosynthesis and protein O-glycosylation:the first line of defense against stress, ischemia, and trauma. Shock,2008,29, 431-440.
    [219]Ngoh G A, Facundo H T, Zafir A, et al. O-GlcNAc signaling in the cardiovascular system. Circulation research,2010,107,171-185.
    [220]Yang S, Zou L Y, Bounelis P, et al. Glucosamine administration during resuscitation improves organ function after trauma hemorrhage. Shock,2006,25,600-607.
    [221]Zachara N E, Hart G W. O-GlcNAc a sensor of cellular state:the role of nucleocytoplasmic glycosylation in modulating cellular function in response to nutrition and stress. Biochimica et biophysica acta,2004,1673,13-28.
    [222]Zou L, Yang S, Champattanachai V, et al. Glucosamine improves cardiac function following trauma-hemorrhage by increased protein O-GlcNAcylation and attenuation of NF-{kappa}B signaling. American journal of physiology. Heart and circulatory physiology,2009,296, H515-523.
    [223]Ngoh G A, Facundo H T, Hamid T, et al. Unique hexosaminidase reduces metabolic survival signal and sensitizes cardiac myocytes to hypoxia/reoxygenation injury. Circulation research,2009,104,41-49.
    [224]Ngoh G A, Watson L J, Facundo H T, et al. Augmented O-GlcNAc signaling attenuates oxidative stress and calcium overload in cardiomyocytes. Amino acids,2011,40,895-911.
    [225]Slawson C, Pidala J, Potter R. Increased N-acetyl-beta-glucosaminidase activity in primary breast carcinomas corresponds to a decrease in N-acetylglucosamine containing proteins. Biochimica et biophysica acta,2001,1537,147-157.
    [226]Krzeslak A, Pomorski L, Lipinska A. Elevation of nucleocytoplasmic beta-N-acetylglucosaminidase (O-GlcNAcase) activity in thyroid cancers. International journal of molecular medicine,2010,25,643-648.
    [227]Gu Y, Mi W, Ge Y, et al. GlcNAcylation plays an essential role in breast cancer metastasis. Cancer research,2010,70,6344-6351.
    [228]Mi W, Gu Y, Han C, et al. O-GlcNAcylation is a novel regulator of lung and colon cancer malignancy. Biochimica et biophysica acta,2011,1812,514-519.
    [229]Hirata-Fukae C, Li H F, Ma L, et al. Levels of soluble and insoluble tau reflect overall status of tau phosphorylation in vivo. Neuroscience letters,2009,450,51-55.
    [230]Duara R, Barker W, Loewenstein D, et al. The basis for disease-modifying treatments for Alzheimer's disease:the Sixth Annual Mild Cognitive Impairment Symposium. Alzheimer's & dementia:the journal of the Alzheimer's Association,2009,5,66-74.
    [231]Sigurdsson E M. Immunotherapy targeting pathological tau protein in Alzheimer's disease and related tauopathies. Journal of Alzheimer's disease:JAD,2008,15,157-168.
    [232]Iqbal K, Chohan M O, Grundke-Iqbal I. Stratification of patients is the way to go to develop neuroprotective/disease-modifying drugs for Alzheimer's disease. Journal of Alzheimer's disease:JAD,2008,15,339-345.
    [233]李铁海,郭利娜,李中华.O-GlcNAcase抑制剂.化学进展(PROGRESS IN CHEMISTRY),2011,23,1657-1664.
    [234]Fischer P M. Turning down tau phosphorylation. Nature chemical biology,2008,4, 448-449.
    [235]Gao Y, Wells L, Comer F I, et al. Dynamic O-glycosylation of nuclear and cytosolic proteins:cloning and characterization of a neutral, cytosolic beta-N-acetylglucosaminidase from human brain. The Journal of biological chemistry,2001,276,9838-9845.
    [236]Dong D L, Hart G W. Purification and characterization of an O-GlcNAc selective N-acetyl-beta-D-glucosaminidase from rat spleen cytosol. The Journal of biological chemistry, 1994,269,19321-19330.
    [237]Li J, Li Z, Li T, et al. Identification of a specific inhibitor of nOGA-a caspase-3 cleaved O-GlcNAcase variant during apoptosis. Biochemistry. Biokhimiia,2012,77,194-200.

© 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