用户名: 密码: 验证码:
TRAP1基因沉默和基因过表达对A549细胞和MDA-MB-231细胞信号传导通路影响的研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
研究背景和目的
     肿瘤坏死因子α(Tumor necrosis factorα, TNF-α)是一种多功能细胞因子,由单核细胞和巨噬细胞等分泌产生,在生物机体的各个反应中具有重要作用,包括炎症反应、免疫调节、抗肿瘤与微生物感染等方面。TNF的生物学功能通过细胞表面相应的受体来实现。TNF受体家族主要是Ⅰ型膜蛋白受体,TNF-α有两类受体,分子量分别是55kD(TNFR-I)和75kD(TNFR-Ⅱ)。TNFR-Ⅰ广泛分布于正常细胞膜以及多种肿瘤细胞膜表面,TNFR-Ⅱ主要分布于造血细胞和内皮细胞。两类受体的胞外区有28%的同源性,而胞内区没有同源性,提示两者介导了不同的信号传递途径。TNFR-Ⅰ参与介导细胞活化信号和增殖信号,诱导一氧化氮合成酶和白介素8的活性,活化NF-κB,亦可诱导凋亡、介导炎症反应、抗病毒、促进成纤维细胞增殖等;而TNFR-Ⅱ主要调控胸腺细胞的增殖,抑制造血和调节内皮细胞或中性粒细胞活性等。TNF/TNFR系统不仅是重要的炎症因子,而且对细菌感染,某些自身免疫疾病和退化性疾病有保护功能。TNF与TNFR结合以后,一方面介导凋亡信号,在抗肿瘤和抗病毒感染中发挥重要作用,另一方面也参与自身免疫性疾病和败血症中对自身组织细胞的损伤。TNFR的生物学活性受诸多因素影响,包括遗传和环境因素,细胞分化以及细胞因子网络间的信息传递等。
     TNF受体相关蛋白1(TNFR associated protein 1, TRAP 1)是在1995年鉴定出来的HSP90家族的一个成员,与肿瘤坏死因子1类受体的细胞内结构域结合。其结构与热休克蛋白75相似,主要定位于线粒体,在心脏肌原纤维、心脏细胞核、胰腺以及血管内皮细胞表面也有定位。近年来研究发现,TRAP1不但具有HSP90家族的共同特点,如在氧化应激时保护细胞,维持线粒体膜稳定、抑制活性氧的产生,避免凋亡;TRAP1还有不同于HSP90家族其他成员的独特特点,在于其特有的LxCxE模序,该模序可与Rb蛋白的SV40 T抗原结合结构域结合,在有丝分裂和热休克应激后,TRAP1可从线粒体转移至细胞核内,并借此结构域与Rb结合,参与辅助变性的Rb重新折叠,恢复其天然构象及功能,因此,TRAP1可做为Rb的分子伴侣,协助其调控细胞周期的进程,而HSPs其他成员则没有此模序及此功能。
     热休克蛋白(Heat Shock Proteins, HSPs)又被称作“分子伴侣”(molecular chaperone),当细胞受到应激因素作用或处于病理生理状态下(如低氧、感染性炎症、缺血等)均可诱发热休克蛋白合成和表达增加。研究发现,许多肿瘤细胞可高度表达或特异表达不同亚族的HSP,认为HSP的表达参与了基因调控及细胞周期调控,与肿瘤细胞的增殖、分化、转移及凋亡有关,并与癌基因和抑癌基因有着密切关系,可能与肿瘤的发生、发展、治疗及预后及机体对抗肿瘤药物的耐药性等有关。我们实验室前期研究发现,TRAP1虽然在多数肿瘤中高表达,在某些肿瘤如非小细胞肺癌、非霍奇金淋巴瘤、胰腺癌中反而低表达。后继研究发现TRAP1在调节Rb/E2F1途径中发挥重要作用,影响细胞周期进程中G1/S期的转换。这一结果促使我们继续研究TRAP1在肿瘤细胞中的相关基因和信号通路,以期发现TRAP1不同于其他分子伴侣的独特特点,为治疗提供新的靶点。
     本研究运用人类寡核苷酸芯片技术筛选TRAP1在肿瘤细胞中的相关基因,选用高表达TRAP1的A549肺腺癌细胞系和低表达TRAP1的MDA-MB-231乳腺癌细胞系,该两细胞系均为高度侵袭性,均表达Rb及E2F1。利用小干扰RNA技术和逆转录病毒载体在两个细胞系中分别敲低和过表达TRAP1,进行16小时的常氧或缺氧处理,在两个细胞系中分别比较在常氧和急性缺氧的初期,TRAP1表达高低不同所引起的相关基因的变化,将得到的差异表达基因列表上传至在线生物信息学分析软件进行基因功能注释、归纳聚类并建立基因相互作用网络图,研究TRAP1所影响的信号途径,探索其在肿瘤发病机制中的作用。
     材料和方法
     1.细胞培养:用siRNA技术转染A549细胞,同时以scramble RNA做为阴性对照;而用逆转录病毒载体在MDA-MB-231中构建长效稳定过表达TRAP1细胞株,同时以空载体转染该细胞做为阴性对照。即得到4组细胞株:A549/siRNA和A549/Scramble, MDA231/TRAP1+和MDA231/Mock。分别分组进行常氧或缺氧(0.1%02浓度)处理16小时。
     2.收获细胞,提取总RNA,用Taqman实时荧光定量RT-PCR检测TRAP1表达水平,验证转染的效率。
     3.体外扩增RNA并纯化,与荧光染料耦合,每个样本对应一个对照RNA,即人类通用参照总RNA。耦合后再次纯化,检测RNA浓度。
     4.对芯片进行紫外偶联并预杂交、洗片、离心干燥备用。
     5.将每个样本和一个对照RNA以相同质量混合加样于同一张芯片上,置于42℃杂交20小时。次日洗片、离心干燥。
     6.在暗室用GenePix(?) 4000B芯片扫描仪扫描芯片,每个样本获得一个图像,用GenePix Pro 6.0软件进行图像分析,将图像转换为数据保存。用GeneSpringTM GX10软件对获得的图像数据进行分析。采用LOWESS方法对数据进行标准化、标记过滤、倍值过滤和表达水平过滤、采用对数值转换,去除每张芯片上变异系数(CV)大于25%的数据,设定比值改变(Fold change)>2.0为筛选标准,获得4个差异表达基因列表,即:常氧16小时后,A549/siRNA Vs A549/Scr, MDA231/TRAP 1+Vs MDA231/Mock;缺氧16小时后,A549/siRNA Vs A549/Scr, MDA231/TRAP1+Vs MDA231/Mock。
     7.使用DAVID (Database for Annotation, Visualization and Integrated Discovery)在线数据库对差异表达基因的注解、功能聚类、通路分析,再用pSTIING在线数据库建立基因相互作用网络图。
     8.挑选6个差异表达基因,用Taqman实时荧光定量RT-PCR验证芯片结果的可靠性。
     结果
     1. Taqman qRT-PCR显示,常氧和缺氧下,A549/siRNA组TRAP1的表达水平均明显低于A549/Scr组;而MDA231/TRAP1+组TRAP1的表达水平明显高于MDA231/Mock组,说明在两个细胞系中转染均成功。
     2.常氧处理16小时后,A549细胞的两个比较组中有373个差异表达基因,其中A549/Scr组表达上调的基因198个,下调的175个;而MDA231的两个比较组中有163个差异表达基因,在MDA231/TRAP1+组上调的基因91个,下调的72个,A549细胞的差异表达基因数目大于MDA231细胞;而在缺氧处理16小时后,A549细胞系差异表达基因数目为377个,其中在A549/Scr组226个上调,151个下调;MDA231细胞系差异表达基因为421个,比在常氧时明显增多。
     3.经DAVID在线功能聚类分析,敲低TRAP1基因时差异表达基因所参与的生物过程以及信号通路,与过表达TRAP1基因时差异表达基因所参与的生物过程以及信号通路基本相似,包括细胞周期调控、转录调节、免疫/炎症反应、血管生成、G蛋白偶联受体蛋白信号转导、细胞粘附、凋亡调节、磷酸化、离子转运和蛋白水解等方面。其中转录调节和细胞周期相关基因占的比例较大。
     4.挑选的6个差异表达基因,用Taqman qRT-PCR实验验证了其表达水平与芯片结果一致,说明芯片结果基本可靠。
     5.经pSTIING在线进行基因相互作用网络可视化,TNF途径、NF-κB途径、Rho激酶途径、JNK途径和MAPK级联反应途径受TRAP1的调节比较明显。
     结论
     1. TRAP1在两个细胞系中表达水平不同,其生理作用的重要性可能不同。在常氧下,TRAP1可能在正常表达有该蛋白的肿瘤细胞中发挥着更重要的作用,而低表达该蛋白的肿瘤细胞可能存在其他不依赖TRAP1的机制而促进肿瘤生长。
     2.敲低TRAP1基因时差异表达基因所参与的生物过程以及信号通路,与过表达TRAP1基因时差异表达基因所参与的生物过程以及信号通路基本相似,证实了TRAP1所影响的生物通路。
     3. TRAP1可能促进肿瘤细胞的细胞周期进程和增殖,与TNF途径、Rho激酶途径和G-蛋白偶联信号转导途径有关。
     4. TRAP1可激活天然免疫反应,激活补体途径、调节巨噬细胞活性;也参与适应性免疫反应,参与T/B细胞活化、调节T细胞分化,影响Th1或Th2细胞亚群分泌细胞因子。
     5. TRAP1促进细胞的磷酸化过程,其中MAPK级联反应途径发挥了重要作用。
     6. TRAP1可能抑制肿瘤细胞转移,与细胞粘附相关基因的差异表达上调有关。
     7. TRAP1可能促进肿瘤细胞血管生成,以满足或维持局部肿瘤组织的血供,以尽快使肿瘤细胞摆脱缺氧状态。
     8. TRAP1对肿瘤细胞的凋亡无明显促进或抑制的趋势,有待进一步在具体肿瘤细胞中研究。
Backgrounds and objective
     Tumor necrosis factor-α(TNF-α) is a multifunctional cytokine produced by monocytes/macrophages and lymphocytes, it is important in inflammatory and immune response, and for resistance to infection and cancers. TNF-αexerts many of its effects by binding to the cell membrane receptor of either 55 kDa termed TNFR-I or 75 kDa termed TNFR-Ⅱ. The two receptors share limited homology in the extracellular region, no homology is seen in the intracellular regions of the proteins, suggests the two receptors activate distinct signalling pathways. TNFR-I is involved in the activation of NF-κB, the induction of apoptosis, antivirus response, and activation of fibroblasts proliferation, etc, while TNFR-Ⅱis mainly involved in the proliferation of the thymus and regulation of the activity of endothelial cells and neutrophils. But the biological functions of the two receptors are not independent as many activities are completed by both of them, such as cytotoxicity and hepatotoxicity. The TNF/TNFR signaling pathway is not only important in inflammatory response, but also protective for bacterial infection and some autoimmune diseases. The binding of TNF to TNFR will initiate two opposite signaling pathways, which to some extent explain why TNFR can have contradictory biological functions. Therefore, the activity of TNFR depends on many factors that affect the balance of TNF/TNFR, it is also influenced by heredity, environment, stage of cell differentiation and many other signaling pathways among cytokines.
     TNF receptor associated protein 1 (TRAP1) was first identified in 1995 as a member of HSP90 family, it binds to the intracellular domain of TNFR-I. TRAPl is identical to Hsp75 in its structure and locates mainly at the mitochondria but are also observed in certain tissues including cardiac sarcomeres, nuclei of pancreatic and heart cells, and on the cell surface of blood vessel endothelial cells. Recent studies reveal that TRAR1 has the general functions of HSPs, such as protecting cells from oxidative stress and apoptosis via maintaining the homeostasis of mitochondria membrane and inhibition of reactive oxygen species (ROS) generation. However, TRAP1 also has distinct functions from other HSP90 family members in that it has a unique LxCxE motif that allows it to bind to retinoblastoma protein (Rb) during mitosis and after heat shock when TRAP1 moves from its predominantly mitochondria location to the nucleus, to assist in the refolding of denatured Rb.
     HSPs are also known as molecular chaperones, they can induce the change of the expression model of HSPs genes leading to the increasing generation of a series of HSPs when cells encounter stresses or are under pathophysiological conditions (i.e. hypoxia, inflammation and ischemia). Studies have shown that many tumor cells can express high level of different subgroups of HSPs, it is deemed that HSPs are implicated in the cell cycle regulation, the proliferation, differentiation, metastasis and apoptosis of tumor cells, also HSPs have close relationship with oncogenes and tumor suppressor genes and might be associated with the tumorigenesis and the tolerance of anticancer drugs. The preliminary study in our lab revealed that although TRAP1 is over-expressed in many tumors, its expression level is low in some tumors such as non-small cell lung cancer, non-Hodgkin lymphoma and pancreatic cancer. The preceding experiment discovered TRAPl plays key role in regulating Rb/E2F1 pathway and influences the G1/S progression of the cell cycle. This finding promoted us to further study the related genes and pathways of TRAP1 in tumor cells in order to find the distinct functions of TRAP1 and to find new therapeutic targets.
     This study will use oligonucleotide microarray platform to detect the associated genes of TRAP1 in tumor cells, A549 lung adenocarcinoma cell line which expresses high level of TRAP 1 and MDA-MB-231 breast cancer cell line which expresses low level of TRAP 1 are used, both of them are highly invasive and express Rb and E2F1. Small interfering RNA (siRNA) and retroviral vector are used to construct the knock down model and the over-expression model of TRAP1, cells are treated under 16 hours of either normoxia or hypoxia. The gene expression profiles are compared between TRAP1 transfected group and control group, and the biological signaling pathways that TRAP1 affected are explored.
     Materials and methods 1. Cell culture:Transient transfection of TRAP1 by siRNA in A549 cell line, scramble RNA transfected cells were negative control. Meanwhile using retroviral vector to construct stable over-expression model of TRAP 1 in MDA-MB-231 cell line, empty vector transfected cells were negative control. Thus four groups were constructed as following:A549/siRNA and A549/Scramble, MDA231/TRAP1+ and MDA231/Mock. Splited them into subgroups followed by 16 hours of either normoxic or hypoxic (0.1%O2) treatment.
     2. Cells were harvested, total RNA was extracted and TRAP1 expression was verified by Taqman real time PCR.
     3. Total RNA was amplified in vitro and purified followed by dye coupling of each sample. Universal Human Reference total RNA was used as reference for each sample. The coupled aRNA was again purified and the concentration was checked.
     4. Microarray slides were UV-cross linked and pre-hybridized before use.
     5. Each sample was mixed with same amount of reference RNA and then added onto one slide, the slides were placed into 42℃oven for 20 hours of hybridization. Slides were then washed and spinned to dry.
     6. Slides were scanned using GenePix 4000B microarray scanner, the picture images were analyzed using GenePix 6.0 software, then the data were analyzed by GeneSpringTM GX10 software. Data were background subtracted and normalized by LOWESS and retrieved as the log transformed values. The filtering criteria were set as the coefficient of variation (CV)<25%and fold change>2.0 as cut-off. Thus 4 lists of differentially expressed genes were obtained, as:in normoxia, A549/siRNA Vs A549/Scr, MDA231/TRAP1+Vs MDA231/Mock; inhypoxia, A549/siRNA Vs A549/Scr, MDA231/TRAP1+Vs MDA231/Mock.
     7. The gene lists were then analyzed using DAVID bioinformatics resources (Database for Annotation, Visualization and Integrated Discovery), finally genes of interest and their interaction networks were visualized by using pSTIING (Protein, Signalling, Transcriptional Interactions & inflammation Networks Gateway).
     8. Taqman real time PCR was carried out for the expression of 6 genes chosen from the gene lists to verify the results of the microarray experiment.
     Results
     1. In either normoxia or hypoxia, the expression level of TRAP1 was obviously lower in A549/siRNA group than in A549/Scr group, and was obviously higher in MDA231/TRAP1+group than in MDA231/Mock group, indicating the high efficiency of TRAP 1 transfection.
     2. After 16 hours of normoxic treatment, there were 373 differentially expressed genes in the A549 comparison groups, while in MDA231 comparison groups there were only 163 differentially expressed genes. After 16 hours of hypoxic treatment, the numbers of differentially expressed genes were 377 and 421 in the A549 comparison groups and MDA231 comparison groups respectively.
     3. The analysis by using DAVID showed the affected biological pathways when TRAP1 was knocked down in A549 cells were similar to those when TRAP1 was restored in MDA231 cells, including the regulation of cell cycle, transcription, immune/inflammatory response, angiogenesis, G-protein coupled receptor protein pathway, cell adhesion, regulation of apoptosis, phosphorylation, etc. The percentage of genes involved in the regulation of cell cycle and transcription is relatively large.
     4. The expression model of the 6 picked genes by Taqman real time PCR was the same as that of the microarray experiment, indicating the results of the microarray platform was trustable.
     5. The analysis by pSTIING to visualize the interaction network of those differentially expressed genes showed that, TNF pathway, NF-κB pathway, Rho kinase pathway, JNK pathway and MAPK cascade pathway were influenced.
     Conclusions
     1. In normoxia, TRAP1 may be more important for tumor cells that normally express this protein rather than those that have lost it. Whereas at the beginning of a short period of hypoxia, tumor cells tend to start the mechanism of expressing this heat shock protein to survive through some common pathways.
     2. The affected biological pathways when TRAP1 was knocked down in A549 cells were similar to those when TRAP1 was restored in MDA231 cells, which proved the role of TRAP 1 in these pathways.
     3. TRAP1 may promote the cell cycle progression and proliferation of tumor cells, which is associated with TNF pathway and Rho kinase pathway and G-protein coupled receptor protein pathway.
     4. TRAP1 may activate innate immune response, activate complement pathways and regulate microphage activity; it may also be involved in adaptive immunity, regulate T/B activation and T cell differentiation, and influence the cytokine production by Thl or Th2 subgroups.
     5. TRAP1 promotes cell phosphorylation, in which MAPK cascade pathway plays a vital role.
     6. TRAP1 might inhibit the metastasis of tumor cells, which is associated with the up-regulation of cell adhesion related genes.
     7. TRAP1 may promote the angiogenesis of tumor cells, in order to maintain local blood supply and to avoid a hypoxic condition.
     8. There was no obvious trend was found as for the promotion or inbition of apoptosis, which needs further study in specific tumors.
引文
1. Aggarwal BB, Aiyer RA, Pennica D, et al:Human tumour necrosis factors: structure and receptor interactions. Ciba Found Symp.1987; 131:39-51.
    2. Klostergaard J:Role of tumor necrosis factor in monocyte/macrophage tumor cytotoxicity in vitro. Nat Immun Cell Growth Regul.1987; 6:161-166.
    3. Warren JS, Ward PA, Johnson KJ:Tumor necrosis factor:a plurifunctional mediator of acute inflammation. Mod Pathol.1988; 1:242-247.
    4. Idriss HT, Naismith JH:TNF alpha and the TNF receptor superfamily: structure-function relationship(s). Microsc Res Tech.2000; 50:184-195.
    5. Brockhaus M, Schoenfeld HJ, Schlaeger EJ, et al:Identification of two types of tumor necrosis factor receptors on human cell lines by monoclonal antibodies. Proc Natl Acad Sci U S A.1990; 87:3127-3131.
    6. Tartaglia LA, Weber RF, Figari IS, et al:The two different receptors for tumor necrosis factor mediate distinct cellular responses. Proc Natl Acad Sci U S A.1991; 88:9292-9296.
    7. Wong GH, Tartaglia LA, Lee MS, et al:Antiviral activity of tumor necrosis factor is signaled through the 55-kDa type I TNF receptor [corrected]. J Immunol. 1992; 149:3350-3353.
    8. Jacobsen FW, Rothe M, Rusten L, et al:Role of the 75-kDa tumor necrosis factor receptor:inhibition of early hematopoiesis. Proc Natl Acad Sci U S A.1994; 91:10695-10699.
    9. Tartaglia LA, Goeddel DV, Reynolds C, et al:Stimulation of human T-cell proliferation by specific activation of the 75-kDa tumor necrosis factor receptor. J Immunol.1993; 151:4637-4641.
    10. Song HY, Dunbar JD, Zhang YX, et al:Identification of a protein with homology to hsp90 that binds the type 1 tumor necrosis factor receptor. J Biol Chem. 1995; 270:3574-3581.
    11. Loetscher H, Pan YC, Lahm HW, et al:Molecular cloning and expression of the human 55 kd tumor necrosis factor receptor. Cell.1990; 61:351-359.
    12. Schall TJ, Lewis M, Koller KJ, et al:Molecular cloning and expression of a receptor for human tumor necrosis factor. Cell.1990; 61:361-370.
    13. Csermely P, Schnaider T, Soti C, et al:The 90-kDa molecular chaperone family: structure, function, and clinical applications. A comprehensive review. Pharmacol Ther.1998; 79:129-168.
    14. Felts SJ, Owen BA, Nguyen P, et al:The hsp90-related protein TRAP1 is a mitochondrial protein with distinct functional properties. J Biol Chem.2000; 275:3305-3312.
    15. Cechetto JD, Gupta RS:Immunoelectron microscopy provides evidence that tumor necrosis factor receptor-associated protein 1 (TRAP-1) is a mitochondrial protein which also localizes at specific extramitochondrial sites. Exp Cell Res.2000; 260:30-39.
    16. Argon Y, Simen BB:GRP94, an ER chaperone with protein and peptide binding properties. Semin Cell Dev Biol.1999; 10:495-505.
    17. Fu Y, Lee AS:Glucose regulated proteins in cancer progression, drug resistance and immunotherapy. Cancer Biol Ther.2006; 5:741-744.
    18. Masuda Y, Shima G, Aiuchi T, et al:Involvement of tumor necrosis factor receptor-associated protein 1 (TRAP1) in apoptosis induced by beta-hydroxyisovalerylshikonin. J Biol Chem.2004; 279:42503-42515.
    19. Hua G, Zhang Q, Fan Z:Heat shock protein 75 (TRAP1) antagonizes reactive oxygen species generation and protects cells from granzyme M-mediated apoptosis. J Biol Chem.2007;282:20553-20560.
    20. Costantino E, Maddalena F, Calise S, et al:TRAP1,a novel mitochondrial chaperone responsible for multi-drug resistance and protection from apoptotis in human colorectal carcinoma cells. Cancer Lett.2009; 279:39-46.
    21. Montesano Gesualdi N, Chirico G, Pirozzi G, et al:Tumor necrosis factor-associated protein 1 (TRAP-1) protects cells from oxidative stress and apoptosis. Stress.2007;10:342-350.
    22. Im CN, Lee JS, Zheng Y, et al:Iron chelation study in a normal human hepatocyte cell line suggests that tumor necrosis factor receptor-associated protein 1 (TRAP1) regulates production of reactive oxygen species. J Cell Biochem.2007; 100:474-486.
    23. Chen CF, Chen Y, Dai K, et al:A new member of the hsp90 family of molecular chaperones interacts with the retinoblastoma protein during mitosis and after heat shock. Mol Cell Biol.1996; 16:4691-4699.
    24. Lindquist S, Craig EA:The heat-shock proteins. Annu Rev Genet.1988; 22:631-677.
    25. Hartl FU:Molecular chaperones in cellular protein folding. Nature.1996; 381:571-579.
    26. Kazmierczuk A, Kilianska ZM:[The pleiotropic activity of heat-shock proteins]. Postepy Hig Med Dosw (Online).2009; 63:502-521.
    27. Gething MJ, Sambrook J:Protein folding in the cell. Nature.1992; 355:33-45.
    28. Sarto C, Binz PA, Mocarelli P:Heat shock proteins in human cancer. Electrophoresis.2000; 21:1218-1226.
    29. Creagh EM, Sheehan D, Cotter TG:Heat shock proteins-modulators of apoptosis in tumour cells. Leukemia.2000; 14:1161-1173.
    30. Taldone T, Sun W, Chiosis G:Discovery and development of heat shock protein 90 inhibitors. Bioorg Med Chem.2009;17:2225-2235.
    31. DeBoer C, Meulman PA, Wnuk RJ, et al:Geldanamycin, a new antibiotic. J Antibiot (Tokyo).1970; 23:442-447.
    32. Schulte TW, An WG, Neckers LM:Geldanamycin-induced destabilization of Raf-1 involves the proteasome. Biochem Biophys Res Commun.1997;239:655-659.
    33. Chiosis G, Huezo H, Rosen N, et al:17AAG:low target binding affinity and potent cell activity-finding an explanation. Mol Cancer Ther.2003;2:123-129.
    34. Porter JR, Ge J, Lee J, et al:Ansamycin inhibitors of Hsp90:nature's prototype for anti-chaperone therapy. Curr Top Med Chem.2009;9:1386-1418.
    35. Hanson BE, Vesole DH:Retaspimycin hydrochloride (IPI-504):a novel heat shock protein inhibitor as an anticancer agent. Expert Opin Investig Drugs.2009; 18:1375-1383.
    36. Ramanathan RK, Egorin MJ, Erlichman C, et al:Phase I pharmacokinetic and pharmacodynamic study of
    17-dimethylaminoethylamino-17-demethoxygeldanamycin, an inhibitor of heat-shock protein 90, in patients with advanced solid tumors. J Clin Oncol. 28:1520-1526.
    37. Chiosis Q Lucas B, Shtil A, et al:Development of a purine-scaffold novel class of Hsp90 binders that inhibit the proliferation of cancer cells and induce the degradation of Her2 tyrosine kinase. Bioorg Med Chem.2002; 10:3555-3564.
    38. Kang BH, Plescia J, Dohi T, et al:Regulation of tumor cell mitochondrial homeostasis by an organelle-specific Hsp90 chaperone network. Cell.2007; 131:257-270.
    39. Fang W, Li X, Jiang Q, et al:Transcriptional patterns, biomarkers and pathways characterizing nasopharyngeal carcinoma of Southern China. J Transl Med.2008; 6:32.
    40. Hu J:A genomic based approach to the understanding of the molecular biology of non-small cell carcinoma. Oxford:University of Oxford; 2006.
    41. Ramasamy A:Increasing statistical power and generalizability in genomics microarray research. DPhil. Oxford:University of Oxford; 2009.
    42. Flemington EK, Speck SH, Kaelin WG, Jr.:E2F-1-mediated transactivation is inhibited by complex formation with the retinoblastoma susceptibility gene product. Proc Natl Acad Sci U S A.1993; 90:6914-6918.
    43. Rubin SM, Gall AL, Zheng N, et al:Structure of the Rb C-terminal domain bound to E2F1-DP1:a mechanism for phosphorylation-induced E2F release. Cell. 2005; 123:1093-1106.
    44. Ahlander J, Bosco G:The RB/E2F pathway and regulation of RNA processing. Biochem Biophys Res Commun.2009;384:280-283.
    45. Lee JO, Russo AA, Pavletich NP:Structure of the retinoblastoma tumour-suppressor pocket domain bound to a peptide from HPV E7. Nature.1998; 391:859-865.
    46. Tan EY:Loss of protein folding gene expression in human tumours. DPhil. Oxford:University of Oxford; 2007.
    47. Ramsay G:DNA chips:state-of-the art. Nat Biotechnol.1998; 16:40-44.
    48. Gerhold D, Rushmore T, Caskey CT:DNA chips:promising toys have become powerful tools. Trends Biochem Sci.1999; 24:168-173.
    49. Thompson M, Furtado LM:High density oligonucleotide and DNA probe arrays for the analysis of target DNA. Analyst.1999; 124:1133-1136.
    50. McGall GH, Fidanza JA:Photolithographic synthesis of high-density oligonucleotide arrays. Methods Mol Biol.2001; 170:71-101.
    51. Hughes TR, Mao M, Jones AR, et al:Expression profiling using microarrays fabricated by an ink-jet oligonucleotide synthesizer. Nat Biotechnol.2001; 19:342-347.
    52. Heller MJ:DNA microarray technology:devices, systems, and applications. Annu Rev Biomed Eng.2002; 4:129-153.
    53. Lobenhofer EK, Bushel PR, Afshari CA, et al:Progress in the application of DNA microarrays. Environ Health Perspect.2001; 109:881-891.
    54. Ashburner M, Ball CA, Blake JA, et al:Gene ontology:tool for the unification of biology. The Gene Ontology Consortium. Nat Genet.2000; 25:25-29.
    55. Huang da W, Sherman BT, Lempicki RA:Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc.2009; 4:44-57.
    56. Dennis G, Jr., Sherman BT, Hosack DA, et al:DAVID:Database for Annotation, Visualization, and Integrated Discovery. Genome Biol.2003; 4:P3.
    57. Ng A, Bursteinas B, Gao Q, et al:pSTIING:a'systems' approach towards integrating signalling pathways, interaction and transcriptional regulatory networks in inflammation and cancer. Nucleic Acids Res.2006; 34:D527-534.
    58. Yang YH, Dudoit S, Luu P, et al:Normalization for cDNA microarray data:a robust composite method addressing single and multiple slide systematic variation. Nucleic Acids Res.2002; 30:e15.
    59. Livak KJ, Schmittgen TD:Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods.2001; 25:402-408.
    60. Mishra S, Mishra JP, Gee K, et al:Distinct role of calmodulin and calmodulin-dependent protein kinase-II in lipopolysaccharide and tumor necrosis factor-alpha-mediated suppression of apoptosis and antiapoptotic c-IAP2 gene expression in human monocytic cells. J Biol Chem.2005; 280:37536-37546.
    61. Zwicker J, Gross C, Lucibello FC, et al:Cell cycle regulation of cdc25C transcription is mediated by the periodic repression of the glutamine-rich activators NF-Y and Sp1. Nucleic Acids Res.1995; 23:3822-3830.
    62. Caretti G, Salsi V, Vecchi C, et al:Dynamic recruitment of NF-Y and histone acetyltransferases on cell-cycle promoters. J Biol Chem.2003; 278:30435-30440.
    63. Yun J, Chae HD, Choy HE, et al:p53 negatively regulates cdc2 transcription via the CCAAT-binding NF-Y transcription factor. J Biol Chem.1999; 274:29677-29682.
    64. Garkavtsev I, Grigorian IA, Ossovskaya VS, et al:The candidate tumour suppressor p33INGl cooperates with p53 in cell growth control. Nature.1998; 391:295-298.
    65. Leung KM, Po LS, Tsang FC, et al:The candidate tumor suppressor INGlb can stabilize p53 by disrupting the regulation of p53 by MDM2. Cancer Res.2002; 62:4890-4893.
    66. Liu F, Lee WH:CtIP activates its own and cyclin D1 promoters via the E2F/RB pathway during G1/S progression. Mol Cell Biol.2006; 26:3124-3134.
    67. Hershko T, Chaussepied M, Oren M, et al:Novel link between E2F and p53: proapoptotic cofactors of p53 are transcriptionally upregulated by E2F. Cell Death Differ.2005; 12:377-383.
    68. Zuchero JB, Coutts AS, Quinlan ME, et al:p53-cofactor JMY is a multifunctional actin nucleation factor. Nat Cell Biol.2009; 11:451-459.
    69. Coller HA, Grandori C, Tamayo P, et al:Expression analysis with oligonucleotide microarrays reveals that MYC regulates genes involved in growth, cell cycle, signaling, and adhesion. Proc Natl Acad Sci U S A.2000; 97:3260-3265.
    70. Paulsen MT, Starks AM, Derheimer FA, et al:The p53-targeting human phosphatase hCdcl4A interacts with the Cdkl/cyclin B complex and is differentially expressed in human cancers. Mol Cancer.2006; 5:25.
    71. Monte M, Benetti R, Buscemi G, et al:The cell cycle-regulated protein human GTSE-1 controls DNA damage-induced apoptosis by affecting p53 function. J Biol Chem.2003; 278:30356-30364.
    72. Ghosh AK, Steele R, Ryerse J, et al:Tumor-suppressive effects of MBP-1 in non-small cell lung cancer cells. Cancer Res.2006; 66:11907-11912.
    73. Janssen E, Zhu M, Zhang W, et al:LAB:a new membrane-associated adaptor molecule in B cell activation. Nat Immunol.2003; 4:117-123.
    74. Mosser DM, Zhang X:Interleukin-10:new perspectives on an old cytokine. Immunol Rev.2008;226:205-218.
    75. Willemsen RA, Sebestyen Z, Ronteltap C, et al:CD8 alpha coreceptor to improve TCR gene transfer to treat melanoma:down-regulation of tumor-specific production of IL-4, IL-5, and IL-10. J Immunol.2006; 177:991-998.
    76. Huang M, Wang J, Lee P, et al:Human non-small cell lung cancer cells express a type 2 cytokine pattern. Cancer Res.1995; 55:3847-3853.
    77. Javid B, MacAry PA, Lehner PJ:Structure and function:heat shock proteins and adaptive immunity. J Immunol.2007; 179:2035-2040.
    78. Schmitt E, Gehrmann M, Brunet M, et al:Intracellular and extracellular functions of heat shock proteins:repercussions in cancer therapy. J Leukoc Biol. 2007; 81:15-27.
    79. Bae J, Mitsiades C, Tai YT, et al:Phenotypic and functional effects of heat shock protein 90 inhibition on dendritic cell. J Immunol.2007;178:7730-7737.
    80. Tsan MF, Gao B:Heat shock protein and innate immunity. Cell Mol Immunol. 2004; 1:274-279.
    81. Srivastava P:Roles of heat-shock proteins in innate and adaptive immunity. Nat Rev Immunol.2002; 2:185-194.
    82. Takeda K, Shimozono R, Noguchi T, et al:Apoptosis signal-regulating kinase (ASK) 2 functions as a mitogen-activated protein kinase kinase kinase in a heteromeric complex with ASK1. J Biol Chem.2007; 282:7522-7531.
    83. Nadeau SI, Landry J:Mechanisms of activation and regulation of the heat shock-sensitive signaling pathways. Adv Exp Med Biol.2007; 594:100-113.
    84. Hanahan D, Weinberg RA:The hallmarks of cancer. Cell.2000; 100:57-70.
    85. Kubota K, Inoue K, Hashimoto R, et al:Tumor necrosis factor receptor-associated protein 1 regulates cell adhesion and synaptic morphology via modulation of N-cadherin expression. J Neurochem.2009; 110:496-508.
    86. Bierings R, Beato M, Edel MJ:An endothelial cell genetic screen identifies the GTPase Rem2 as a suppressor of p19ARF expression that promotes endothelial cell proliferation and angiogenesis. J Biol Chem.2008; 283:4408-4416.
    87. Jeimy SB, Fuller N, Tasneem S, et al:Multimerin 1 binds factor V and activated factor V with high affinity and inhibits thrombin generation. Thromb Haemost.2008; 100:1058-1067.
    88. Liu X, Zhao D, Qin L, et al:Transcription enhancer factor 3 (TEF3) mediates the expression of Down syndrome candidate region 1 isoform 1 (DSCR1-1L) in endothelial cells. J Biol Chem.2008; 283:34159-34167.
    89. Eto N, Miyagishi M, Inagi R, et al:Mitogen-activated protein 3 kinase 6 mediates angiogenic and tumongenic effects via vascular endothelial growth factor expression. Am J Pathol.2009;174:1553-1563.
    90. Marsters SA, Ayres TM, Skubatch M, et al:Herpesvirus entry mediator, a member of the tumor necrosis factor receptor (TNFR) family, interacts with members of the TNFR-associated factor family and activates the transcription factors NF-kappaB and AP-1.J Biol Chem.1997; 272:14029-14032.
    91. He L, Wu X, Siegel R, et al:TRAF6 regulates cell fate decisions by inducing caspase 8-dependent apoptosis and the activation of NF-kappaB. J Biol Chem.2006; 281:11235-11249.
    92. Chan FK, Shisler J, Bixby JG, et al:A role for tumor necrosis factor receptor-2 and receptor-interacting protein in programmed necrosis and antiviral responses. J Biol Chem.2003; 278:51613-51621.
    93. Kamradt MC, Lu M, Werner ME, et al:The small heat shock protein alpha B-crystallin is a novel inhibitor of TRAIL-induced apoptosis that suppresses the activation of caspase-3. J Biol Chem.2005; 280:11059-11066.
    94. Paulsen M, Ussat S, Jakob M, et al:Interaction with XIAP prevents full caspase-3/-7 activation in proliferating human T lymphocytes. Eur J Immunol.2008; 38:1979-1987.
    95. Bruno T, Iezzi S, De Nicola F, et al:Che-1 activates XIAP expression in response to DNA damage. Cell Death Differ.2008; 15:515-520.
    96. Jin H, Pan Y, He L, et al:p75 neurotrophin receptor inhibits invasion and metastasis of gastric cancer. Mol Cancer Res.2007;5:423-433.

© 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