猪瘟病毒感染蛋白质组学研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
本研究首次用2-DE、2D DIGE和质谱鉴定等高通量蛋白质组学技术对感染CSFV的体外PK-15细胞、体内靶细胞——外周血单核细胞以及感染猪血清进行了比较蛋白质组学分析,获得了迄今最为完善的CSFV感染宿主细胞、宿主血清差异表达蛋白质组数据。
     CSFV感染的PK-15细胞蛋白质组分析鉴定了21个差异表达蛋白,这些蛋白中膜联蛋白(annexin 2)、不均一核内核糖核酸蛋白(hnRNPs)、翻译延长因子1δ(EF-1δ)等蛋白在感染细胞中上调表达,根据文献报道推测这些蛋白参与了CSFV的感染、基因组复制、病毒粒子的包装、出芽等生命循环过程;参与细胞凋亡激活途径的糖酵解酶甘油醛-3-磷酸脱氢酶(GAPDH)下调表达、具有抗氧化应激和抑制细胞凋亡功能的糖酵解酶磷酸甘油酸变位酶1(PGAM1)和抗氧化应激蛋白过氧化物酶-6 (PRDX6)、硫氧还蛋白(thioredoxin- like)上调表达,这些变化抑制了PK-15细胞的凋亡,可能是PK-15细胞感染CSFV不产生细胞病变的机制之一。
     CSFV感染的体内靶细胞——外周血单核细胞的差异蛋白质组分析鉴定了34种差异表达蛋白。7个不同的蛋白点均鉴定为细胞骨架蛋白——肌动蛋白(actin),4个不同的蛋白点鉴定为膜联蛋白annexinA1,另有膜突蛋白(moesin)、粘着斑蛋白(Vinculin)、踝蛋白(talin)等细胞骨架蛋白发生上调或下调的表达变化,这表明CSFV感染诱导了细胞骨架的解聚、降解和紊乱失调。据文献报道,细胞骨架的紊乱可诱导细胞凋亡。此外,具有激活细胞凋亡途径功能的GAPDH上调表达,抗氧化应激反应蛋白过氧化物酶(Prx-1)下调表达。这些蛋白的调节变化显示CSFV感染可能通过多种途径诱导了外周血淋巴细胞的凋亡;与细胞免疫相关的血小板反应素1(Thrombospondin 1,TSP-1)下调表达,TSP-1参与免疫功能的调节,比如,TSP-1可调节T细胞行为并参与炎性T细胞的激活和克隆增殖。TSP-1的下调表达可能是CSFV诱导免疫抑制的机制之一。该研究还检测了CSFV石门株感染诱导外周血CD4+和CD8+ T淋巴细胞亚群变化动力学。
     此外,用2D DIGE对感染CSFV猪血清进行了差异蛋白质组分析,鉴定了14种差异表达蛋白,其中的急性期蛋白接联珠蛋白(haptoglobin)、补体成分C4a、载脂蛋白A-1(apolipoprotein A-1)已被鉴定为其它疾病如乙型肝炎病毒(HBV)或丙型肝炎病毒(HCV)感染或相关癌症的潜在生物标记。该研究鉴定的这些CSFV感染血清差异表达蛋白具有作为CSFV感染生物标记的潜在可能性,当然,这种可能性仍需和其它动物病毒感染性疾病进行比较和评估。
     本研究鉴定的大量差异表达蛋白体现着宿主细胞对CSFV感染的总体反应机制,为发现新的诊断、预防和治疗靶标以及揭示CSFV致病机制奠定了基础。
Classical swine fever virus (CSFV), is a small, enveloped virus with a non-segmented positive-stranded RNA genome, belonging to the genus Pestivirus within the family Flaviviridae. The viral genome is approximately 12.5 kb in size and contains a single large open reading frame (ORF). This ORF is translated into a polyprotein which is further cleaved into 11-12 mature proteins : NH2–Npro–C–Erns–E1–E2–p7–NS2–NS3–NS4A–NS4B–NS5A–NS5B–COOH. CSFV is the pathogen of classical swine fever (CSF), a highly contagious swine disease with high morbility and mortality, featuring symptoms of hemorrhagic fever and immune suppression. The disease is a notifiable one of the World organization for animal health (OIE), usually leading to substantial economic losses to the pig industry worldwide. In recent years, CSF with lower morbility and mortality and mild, chronic, atypical symptoms was often observed, even in immune pigs herd.
     At present, nothing is known about the interaction between CSFV and host cells. To uncover cellular protein responses in CSFV-infected PK-15 cells, a differentially proteomic analysis was conducted using 2D electrophoresis followed by MALDI-TOF-MS/MS identification. Altered expression of 35 protein spots with at least 1.5-fold quantitative differences in expression in infected cells at 48 p.i. were identified in 2D gels, with 21 of these being characterized by MALDI-TOF-MS/MS, including 16 upregulated proteins and 5 downregulated proteins. Western-bloting analysis confirmed the upregulation of annexin 2 and downregulation of GAPDH. The altered proteins could be sorted into 7 groups according to their cellular function: cytoskeleton, energy metabolism, replication/transcription and translation processes, protein processing, anti-oxidant stress, heat shock protein and signal transduction. The altered expression of these proteins provides a response profile of PK-15 host cells to CSFV infection. Further study on these altered proteins may facilitate uncovering the mechanisms of CSFV infection and pathogenesis.
     According cellular function of differentially expressed proteins, it was speculated that annexin 2, hnRNPs and EF-1δmight be play critical roles in CSFV infection, genome replication, virion assembly and egress. In CSFV infected samples, GAPDH with function of apoptosis activation were down-regulated, and PGAM1 and anti-oxidative proteins (PRDX6 and thioredoxin- like) with function of apoptosis inhibition were up-regulated, which may be one mechanism of CSFV inhibiting CPE of infected PK-15 and establishing persistent infection.
     CSFV has particular affinity to tissue macrophage and endothelial cell. Studies indicats that CSFV infection lead to leucopenia and immune suppression. To uncover the effects on immune cells following experimental infection with CSFV Shimen strain and the mechanism of response to CSFV infection , changes of kinetics of CD4+ and CD8+ T cell subpopulation and leukocytes apoptosis in peripheral blood was analyzed using flow cytometry. Results showed that CD4+ and CD8+ T cell subpopulations were significantly reduced, the percentage of CD4+ T cell subpopulation decreased from 26.87% prior to infection to 8.83% 4 days post-infection (dpi) and 13.86% 7 dpi, respectively, and CD8+ T cell subpopulation from 28.17% prior to infection to 14.19% 4 dpi and 17.55% 7 dpi, respectively. Numbers of apoptotic leukocytes and early apoptotic leukocytes in peripheral blood of the infected pigs were significantly more than those of the control pigs, which indicated that cells apoptosis induced by CSFV infection is the major cause of leucopenia and immune suppression. Proteomic analysis indicated that 73 differentially expressed protein spots were found in CSFV-infected PBMC samples, with 55 of these(37 upregulated proteins, 18 downregulated proteins) being characterized by MALDI-TOF-MS/MS, reprensentive 34 unique proteins. Western-blot analysis indicated that Annexin A1 was remarkbaly upregulated and cofilin was downregulated, which confirmed the data of 2DE. These altered proteins are involved in cellular function of cytoskeleton, energy metabolism, nucleic acid/protein processing and anti-stress. These data provides the changes in profile of protein expression and physiological functions in PBMC from CSFV-infected porcine, which provides basis for further uncovering mechanism of CSFV pathogenesis.
     According cellular function of differentially expressed proteins, it was speculated that moesin, EF-1α, Bip protein might be involved in the mechanism of CSFV infection and replication. Derangement and disruption of cytoskeleton of PBMC induced by CSFV infection and upregulation of GAPDH may active apoptosis pathway of PBMC. Apoptosis of PBMC and down-regulation of T lymphocyte immune regulation factor of TSP-1 may be tightly relate to mechanism of immune suppression induced by CSFV infection.
     To found relevant biomarker and proteins of CSFV infection in serum from infected porcine, differentially proteomic analysis was performed using 2D-DIGE followed by MALDI-TOF-MS and RP-HPLC-MS/MS identification. Results showed that 17 differentially expressed proteins were found in CSFV-infected serum, including upregulated 9 and downregulated 8 spots. 14 of them were identified by MALDI-TOF-MS and RP-HPLC MS/MS. These proteins embodies pathological response of CSFV-infected porcine, further study is needed for potentiality of these proteins as biomarker of CSFV infection.
     In summary, the present study provided the first proteomic analysis of CSFV infection, and established the most comprehensive differential proteomic index of CSFV-infected cells in vitro and vivo, and CSFV-infected porcine serum. Further functional investigation of these altered proteins may facilitate understanding the pathogenic mechanisms and molecular responses of host cell to CSFV infection. This may also permit identification of biomarkers of CSFV infection and therapeutic targets or development of new diagnostic methods.
引文
[1] Hanash S. Disease proteomics[J]. Nature, 2003;422:226–32.
    [2] Maxwell KL, Frappier L. Viral proteomics[J]. Microbiol Mol Biol Rev.2007, 71(2):398-411.
    [3] Unlu M, Morgan ME, Minden JS. Difference gel electrophoresis:a single gel method for detecting changes in protein extracts[J]. Electrophoresis, 1997, 18:2071–2077.
    [4] Gygi SP, Rist B, Gerber SA, et al. Quantitative analysis of complex protein mixtures using isotopecoded affinity tags[J]. Nat Biotechnol., 1999, 17:994–999.
    [5] Patton WF. Detection technologies in proteome analysis[J]. J Chromatogr B Analyt Technol Biomed Life Sci., 2002, 771:3–31.
    [6] Ong SE, Blagoev B, Kratchmarova I, et al. Stable isotope labeling by amino acids in cell culture, SILAC, as a simple and accurate approach to expression proteomics[J]. Mol Cell Proteomics, 2002, 1:376–386.
    [7] Ong SE, Foster LJ, Mann M. Mass spectrometric-based approaches in quantitative proteomics[J]. Methods, 2003, 29:124–130.
    [8] Ono M, Shitashige M, Honda K, et al. Label-free quantitative proteomics using large peptide data sets generated by nanoflow liquid chromatography and mass spectrometry[J]. Mol Cell Proteomics, 2006,5(7):1338-47.
    [9] Wang G, Wu WW, Zeng W, et al. Label-free protein quantification using LC-coupled ion trap or FT mass spectrometry: Reproducibility, linearity, and application with complex proteomes[J]. J Proteome Res., 2006,5(5):1214-23.
    [10] Cutillas PR, Vanhaesebroeck B. Quantitative profile of five murine core proteomes using label-free functional proteomics[J]. Mol Cell Proteomics, 2007, 6(9):1560-73.
    [11] Yu H, Wakim B, Li M, et al. Quantifying raft proteins in neonatal mouse brain by 'tube-gel' protein digestion label-free shotgun proteomics[J]. Proteome Sci., 2007, 5:17.
    [12] Issaq HJ, Veenstra TD, Conrads TP, et al. The SELDI-TOF MS approach toproteomics: protein profiling and biomarker identification[J]. Biochem Biophys Res Commun., 2002,292(3): 587-92.
    [13] Jensen ON, Houthaeve T, Shevchenko A, et al. Identification of the major membrane and core proteins of vaccinia virus by two-dimensional electrophoresis[J]. J Virol., 1996, 70: 7485–7497.
    [14]Chelius D, Hühmer AF, Shieh CH, et al. Analysis of the adenovirus type 5 proteome by liquid chromatography and tandem mass spectrometry methods[J]. J Proteome Res., 2002, 1(6): 501-13.
    [15] Chung CS, Chen CH, Ho MY, et al. Vaccinia Virus Proteome: Identification of Proteins in Vaccinia Virus Intracellular Mature Virion Particles[J]. J Virol. 2006,80(5):2127-2140.
    [16] Yoder JD, Chen TS, Gagnier CR, et al. Pox proteomics: mass spectrometry analysis and identification of Vaccinia virion proteins[J]. Virol J., 2006, 3:10.
    [17] Varnum SM, Streblow DN, Monroe ME, et al. Identification of proteins in human cytomegalovirus(HCMV) particle:the HCMV proteome[J]. J Virol., 2004,78(20):10960-6.
    [18] Kattenhorn LM, Mills R, Wagner M, et al. Identification of proteins associated with murine cytomegalovirus virions[J]. J Virol., 2004, 78:11187–11197.
    [19] Bortz E, Whitelegge JP, Jia Q, et al. Identification of proteins associated with murine gammaherpesvirus 68 virions[J]. J Virol., 2003, 77(24):13425-32.
    [20] Bechtel JT, Winant RC, Ganem D. Host and viral proteins in the virion of Kaposi's sarcoma-associated herpesvirus[J]. J Virol. 2005, 79(8):4952-64.
    [21] Sarracino E, Luftiq M, Chase MR, et al. Proteins of purified Epstein-Barr virus[J]. Proc Natl Acad Sci USA, 2004, 101:16286–16291.
    [22] Liu HC, Soderblom EJ, Goshe MB. A mass spectrometry-based proteomic approach to study Marek,s Disease Virus gene expression[J]. J Virol Methods, 2006, 135: 66-75.
    [23] Saphire AC, Gallay PA, Bark SJ. Proteomic analysis of human immunodeficiency virus using liquid chromatography/tandem mass spectrometry effectively distinguishes specific incorporated host proteins[J]. J Proteome Res. 2006, 5:530–538.
    [24] Chertova E, Chertov O, Coren LV, et al. Proteomic and biochemical analysis of purified human immunodeficiency virus type 1 produced from infected monocyte-derived macrophages[J]. J Virol. 2006, 80:9039–9052.
    [25] Ying W, Hao Y, Zhang Y, et al. Proteomic analysis on structural proteins of Severe Acute Respiratory Syndrome coronavirus[J]. Proteomics, 2004, 4: 492–504.
    [26] Zeng R, Ruan HQ, Jiang XS, et al. Proteomic analysis of SARS associated coronavirus using two-dimensional liquid chromatography mass spectrometry and one-dimensional sodium dodecyl sulfate-polyacrylamide gel electrophoresis followed by mass spectrometric analysis[J]. J Proteome Res. 2004, 3:549–555.
    [27] Tsai JM, Wang HC, Leu JH, et al.Genomic and proteomic analysis of thirty-nine structural proteins of shrimp white spot syndrome virus[J]. J Virol. 2004, 78(20):11360-70.
    [28] Xie X, Xu L, Yang F. Proteomic analysis of the major envelope and nucleocapsid proteins of white spot syndrome virus[J]. J Virol. 2006, 80(21):10615-23.
    [29] Huang C, Zhang X, Lin Q, et al. Proteomic analysis of shrimp white spot syndrome viral proteins and characterization of a novel envelope protein VP466[J]. Mol Cell Proteomics, 2002, 1:223–231.
    [30] Tsai JM, Wang HC, Leu JH, et al. Identification of the nucleocapsid, tegument, and envelope proteins of the shrimp white spot syndrome virus virion[J]. J Virol. 2006, 80:3021–3029.
    [31] Thali M, Bukovsky A, Kondo E, et al. Functional association of cyclophilinA with HIV-1 virions[J]. Nature, 1994, 372 (6504), 363-365.
    [32] McKeating JA, Griffiths PD, Grundy JE. Cytomegalovirus in urine specimens has host beta 2 microglobulin bound to the viral envelope: a mechanism of evading the host immune response[J]? J Gen Virol. 1987, 68 (Pt 3): 785-792.
    [33] Braaten D, Franke EK, Luban J. Cyclophilin A is required for the replication of group M human immunodeficiency virus type1 (HIV-1) and simian immunodeficiency virus SIV(CPZ)GAB but not group O HIV-1 or other primate immunodeficiency viruses[J]. J Virol. 1996, 70 (7): 4220-4227.
    [34] Braaten D, Franke EK, Luban J. Cyclophilin A is required for an early step in thelife cycle of human immunodeficiency virus type 1 before the initiation of reverse transcription[J]. J Virol. 1996, 70 (6): 3551-3560.
    [35] Braaten D, Luban J. Cyclophilin A regulates HIV-1 infectivity,as demonstrated by gene targeting in human T cells[J]. Embo J. 2001, 20 (6): 1300-1309.
    [36] MannováP, Fang R, Wang H, et al. Modification of host lipid raft proteome upon hepatitis C virus replication[J]. Mol Cell Proteomics, 2006, 5(12): 2319-25.
    [37] Jiang XS, Tang LY, Dai J, et al. Quantitative analysis of severe acute respiratory syndrome (SARS)-associated coronavirus-infected cells using proteomic approaches[J]. Mol Cell Proteomics, 2005, 4:902-913.
    [38] Dhingra V, Li Q, Allison AB, et al. Proteomic profiling and neurodegeneration in west-nile-virus-infected neurons[J]. J Biomed Biotechnol. 2005, (3): 271-9.
    [39] Chan EY, Qian WJ, Diamond DL, et al. Quantitative Analysis of Human Immunodeficiency Virus Type 1-Infected CD4+ Cell Proteome: Dysregulated Cell Cycle Progression and Nuclear Transport Coincide with Robust Virus Production[J]. J Virol. 2007, 81(14):7571-83.
    [40] Mayte C, Emilio C, Tomás U, et al. Modifications in the human T cell proteome induced by intracellular HIV-1 Tat protein expression[J].proteomics, 2006,6: 63-73.
    [41] Dhingra V, Li X, Liu Y, et al. Proteomic profiling reveals that rabies virus infection results in differential expression of host proteins involved in ion homeostasis and synaptic physiology in the central nervous system[J]. J Neurovirol. 2007, 13(2):107-17.
    [42] Bartee E, McCormack A, Früh K. Quantitative membrane proteomics reveals new cellular targets of viral immune modulators[J]. PLoS Pathog. 2006, 2(10):e107.
    [43] Greco A, Bienvenut W, Sanchez JC, et al. Identification of ribosome-associated viral and cellular basic proteins during the course of infection with herpes simplex virus type 1[J]. Proteomics, 2001, 1: 545–549.
    [44] Schlee M, Krug T, Gires O, et al. Identification of Epstein-Barr virus (EBV) nuclear antigen 2 (EBNA2) target proteins by proteome analysis: activation ofEBNA2 in conditionally immortalized B cells reflects early events after infection of primary B cells by EBV[J]. J Virol. 2004, 78(8):3941-52.
    [45] Brasier AR, Spratt HZ, Wu I, et al. Nuclear heat shock response and novel nuclear domain 10 reorganization in respiratory syncytial virus-infected identified by high-resolution two-dimensional gel electrophoresis[J] J Virol., 2004, 78:11461–11476.
    [46] Chang LY, Mohd Ali AR, Sharifah SH, et al. Human neuronal cell protein responses to Nipah virus infection [J]. Virology Journal, 2007, 4:54.
    [47] Rassmann A,HenkeA,Carlsohn M,et al. Proteome alterations in human host cells infected with coxsackievirus B3[J]. J Gen Virol. 2006 Sep;87(Pt 9):2631-8.
    [48] Alfonso P, Rivera J, Hernáez B, et al. Identification of cellular protens modified in response to African swine fever virus infection by proteomics[J]. proteomics, 2004, 4: 2037-2046.
    [49] Chongsatja PO, Bourchookarn A, Lo CF, et al. Proteomic analysis of differentially expressed proteins in Penaeus vannamei hemocytes upon Taura syndrome virus infection[J]. proteomics, 2007,7 (19): 3592-601.
    [50] Zheng X, Hong L, Shi L, et al. Proteomic analysis of host cells infected with infectious bursal disease virus[J]. MolCell proteomics. 2007,[Epub ahead of pront]
    [51] Ventelon-Debout M, Delalande F, Brizard JP, et al. Proteome analysis of cultivar-specific deregulations of Oryza sativa indica and O. sativa japonica cellular suspensions undergoing rice yellow mottle virus infection[J]. Proteomics, 2004 ,4(1):216-25.
    [52] Casado-Vela J, Casado-Vela J, Sellés S, et al. Proteomic analysis of tobacco mosaic virus-infected tomato (Lycopersicon esculentum M.) fruits and detection of viral coat protein[J]. Proteomics, 2006 , 1:S196-206.
    [53] Gygi SP, Rochon Y, Franza BR, et al. Correlation between protein and mRNA abundance in yeast[J]. Mol Cell Biol. 1999, 19:1720–1730.
    [54] Tian Q, Stepaniants SB, Mao M, et al. Integrated genomic and proteomic analyses of gene expression in mammalian cells[J]. Mol Cell Proteomics, 2004, 3:960–969.
    [55] Banks L, Pim D, Thomas M. viruses and the 26S proteasome: hacking into destruction [J]. Trends biochem sci. 2003, 28(8):452-459.
    [56] Issaq HJ, Xiao Z, Veenstra TD. Serum and plasma proteomics[J]. Chem Rev. 2007, 107(8): 3601-20.
    [57] He QY, George KKL, Zhou Y, et al. Serum biomarkers of hepatitis B virus infected liver inflammation:A protiomics study[J].Proteomics, 2003,3: 666-674.
    [58] Lee IN, Chen CH, Sheu JC, et al. Identification of complement C3a as a candidate biomarker in human chronic hepatitis C and HCV-related hepatocellular carcinoma using a proteomics approach[J].Proteomics, 2006, 6: 2865-2873.
    [59] Steel LF, Shumpert D, Michael T, et al. A strategy for the comparative analysis of serum proteomes for the discovery of biomarkers for hepatocellular carcinoma[J]. Proteomics, 2003, 3, 601–609.
    [60] Pang RT, Poon TC, Chan KC, et al. Serum proteomic fingerprints of adult patients with severe acute respiratory syndrome[J]. Clin Chem. 2006, 52(3): 421-9.
    [61] Kang X, Xu Y, Wu X, et al. Proteomic fingerprint for potential application to early diagnosis of severe acute respiratory syndrome[J]. Clin Chem., 2005, 51: 56-64.
    [62] Han J, Chang YW, Yao CW, et al. Plasma proteome of severe acute respiratory syndrome analyzed by two-dimensional gel electrophoresis and mass spectrometry[J]. PNAS., 2004, 101: 17039-17044.
    [63] Wan J, Sun W, Li X, et al. Inflammation inhibitors were remarkably up-regulated in plasma of severe acute respiratory syndrome patients at progressive phase[J]. Proteomics, 2006, 6(9): 2886-94.
    [64] Paradis V, Asselah T, Dargere D, et al. Serum proteome to predict virologic response in patients with hepatitis C treated by pegylated interferon plus ribavirin[J]. Gastroenterology, 2006, 130(7): 2189-97.
    [65]殷震,刘景华.动物病毒学[M].北京,科学出版社,1997.
    [66] Mennig V. Pestivirus: a review[J]. Vet Microbiol. 1990, 23:35-54
    [67] Meyers G, RumenAmpf T, Thiel HJ. Molecular cloning and nucleotide sequenceof the genome of hog cholera virus[J]. Virology, 1989,171:555-567.
    [68] Rümenapf T, Unger G, Strauss JH, et al. Processing of the envelope glycol- proteins of pestiviruses[J]. J Virol. 1993, 67 (6): 3288~3294.
    [69] Handschun G, Caselmann WH. Bacteerial expression and purification of hepatitis C virus capsid proteins of different size[J]. J Hepatol, 1995, 22: 2865-2876.
    [70] Liu JJ, Wong ML , Chen PF, et al. The recombinant nucleocapsid protein of classic -al swine fever virus can act as a transcriptional regulator[J]. Virus Res. 1998, 53(1):75-80.
    [71] Hulst MM, Moormann RJ. Erns protein of pestiviruses[J]. Methods Enzymol. 2001. 342:431-40.
    [72] Hausmann Y, Roman-Sosa G, Thiel HJ, et al. Classical swine fever virus glycoprotein Erns is an endoribonuclease with an unusual base specificity[J]. J Virol. 2004 May; 78(10): 5507-12.
    [73] Stark R, Meyers G, Rumenapf T, et al. Processing of pestivirus polyprotein: clesvage site between autoprotease and nucleocaspsid protein of classical swine fever[J]. J Virol.1996, 141:771-777.
    [74] Elbers K, Tautz N, Becher P, et al. Processing in the pestivirus E2-NS2 region: identification of proteins p7 and E2p7[J]. J Virol.1996,70 (6): 4131-4135.
    [75] Brownlie J. Pathogenesis of mucosal disease and molecular aspects of bovine viral diarrhea virus[J]. Vet Microbiol, 1990, 23:371-382.
    [76] Moser C, Stettler P, Tratschin JD, et al. Cytopathogenic and noncytopathogenic RNA replicons of classical swine fever virus[J]. J Virol. 1999, 73(9): 7787-94.
    [77] Kummerer BM, Tautz N, Becher P, et al. The genetic basis for cytopathogenicity of pestiviruses[J]. Vet Microbiol. 2000,,77(1-2):117-28.
    [78] Gu B, Liu C, Lin-Goerke J, et al. The RNA helicase and nucleotide triphosphatase activities of the bovine viral diarrhea virus NS3 protein are essential for viral replication[J]. J Virol, 2000,74 (4): 1794-1800.
    [79] Kou YH, Chou SM, Wang YM, et al. Hepatitis C virus NS4A inhibits cap-dependent and the viral IRES-mediated translation through interacting with eukaryotic elongation factor 1A[J]. J Biomed Sci. 2006, 13 (6), 861-874.
    [80] Johnson CM, Perez DR, French R, et al. The NS5A protein of bovine viral diarrhoea virus interacts with the alpha subunit of translation elongation factor-1[J]. J Gen Virol. 2001, 82: 2935-2943.
    [81] Yi GH, Zhang CY, Cao S, et al. De novo RNA synthesis by a recombinant classical swine fever virus RNA-dependent RNA polymerase[J]. Eur J Biochem. 2003 , 270 (24): 4952-61.
    [82] Wang Y, Wang Q, Lu X, et al. 12-nt insertion in 3' untranslated region leads to attenuation of classic swine fever virus and protects host against lethal challenge[J]. Virology. 2008, 10: 374(2):390-398.
    [83] Risatti GR, Borca MV, Kutish GF,et.al The E2 glycoprotein of classical swine fever virus is a virulence determinant in swine[J]. J Virol. 2005 Mar.79 (6):3787-96.
    [84] Risatti GR, Holinka LG, Fernandez Sainz I, et al. Mutations in the carboxyl terminal region of E2 glycoprotein of classical swine fever virus are responsible for viral attenuation in swine[J]. Virology 2007, 364: 371–382.
    [85] Risatti GR, Holinka LG, Carrillo C, et al. Identification of a novel virulence determinant within the E2 structural glycoprotein of classical swine fever virus[J]. Virology. 2006, 355(1): 94-101.
    [86] Risatti GR, Holinka LG, Fernandez Sainz I, et al. N-linked glycosylation status of classical swine fever virus strain Brescia E2 glycoprotein influences virulence in swine[J]. J Virol. 2007, 81(2): 924-933.
    [87] Risatti GR, Holinka LG, LuZ, et al. Mutation of E1 glycoprotein of classical swine fever virus affects viral virulence in swine[J]. Virology. 2005, 343(1):116-127.
    [88] Meyers G, Saalmüller A, Büttner M. Mutations abrogating the RNase activity in glycoprotein E(rns) of the pestivirus classical swine fever virus lead to virus attenuation[J]. J Virol. 1999,73(12): 10224-35.
    [89] Sainz IF, Holinka LG, Lu Z, et al. Removal of a N-linked glycosylation site of classical swine fever virus strain Brescia Erns glycoprotein affects virulence in swine[J]. Virology. 2008, 370(1):122-129.
    [90] Mayer D, Hofmann MA, Tratschin JD. Attenuation of classical swine fever virusby deletion of the viral Npro gene[J]. Vaccine 2004, 22(3-4): 317-328.
    [91] Doceul V, Charleston B, Crooke H, et al. The Npro product of classical swine fever virus interacts with IkappaBalpha, the NF-kappaB inhibitor[J]. J Gen Virol. 2008, 89(Pt 8): 1881-1889.
    [92] Pauly T, K?nig M, Thiel HJ, et al. Infection with classical swine fever virus: effects on phenotype and immune responsiveness of porcine T lymphocytes[J]. J Gen Virol. 1998, 79(Pt 1): 31-40.
    [93] Summerfield A, Mcneilly F,Walker I, et al. Depletion of CD4(+) and CD8(high+) T-cells before the onset of viraemia during classical swine fever [J]. Vet Immunol Immunopathol , 2001, 78(1):3-19.
    [94] Sánchez-Cordón PJ, Romanini S, Salguero FJ, et al. Apoptosis of thymocytes related to cytokine expression in experimental classical swine fever[J]. J Comp Pathol. 2002, 127(4): 239-248
    [95] Summerfield A, Knoetig SM, Tschudin R, et al. Pathogenesis of granulocytopenia and bone marrow atrophy during classical swine fever involves apoptosis and necrosis of uninfected cells[J]. Virology. 2000, 272(1): 50-60.
    [96] Choi C, Hwang KK, Chae C. Classical swine fever virus induces tumor necrosis factor-αand lymphocyte apoptosis[J]. Arch Virol. 2004, 149(5): 875-889.
    [97] Bruschke CJ, Hulst MM, Moormann RJ, et al. Glycoprotein Erns of pestiviruses induces apoptosis in lymphocytes of several species[J]. J Virol. 1997 , 71(9): 6692-6696.
    [98] Carrasco CP, Rigden RC, Vincent IE, et al. Interaction of classical swine fever virus with dendritic cells[J]. J Gen Virol. 2004, 85(pt6): 1633-41.
    [99] Glew EJ, Carr BV, Brackenbury LS, et al. Differential effects of bovine viral diarrhoea virus on monocytes and dendritic cells[J]. J Gen Virol. 2003, 84:1771–1780.
    [100] Suradhat S, Sada W, Buranapraditkun S, et al. The kinetics of cytokine production and CD25 expression by porcine lymphocyte subpopulations following exposure to classical swine fever virus (CSFV) [J]. Vet Immunol Immunopathol. 2005, 106(3-4): 197-208.
    [101] Jamin A, Gorin S, Cariolet R, et al. Classical swine fever virus induces activation of plasmacytoid and conventional dendritic cells in tonsil, blood, and spleen of infected pigs[J]. Vet Res. 2008, 39(1):7
    [102] Moore KW, de Waal Malefyt R, Coffman RL, et al. Interleukin-10 and the interleukin-10 receptor[J]. Annu Rev Immunol. 2001, 19: 683–765.
    [103] Redpath S, Ghazal P, Gascoigne NR. Hijacking and exploitation of IL-10 by intracellular pathogens[J]. Trends Microbiol. 2001, 9: 86–92.
    [104] Suradhat S, Thanawonguwech R, Poovorawan Y. Upregulation of IL-10 gene expression in porcine peripheral blood mononuclear cells by porcine reproductive and respiratory syndrome virus[J]. J Gen Virol. 2003, 84(pt2): 453-459.
    [105] Dolganiuc A, Kodys K, Kopasz A, et al. Hepatitis C virus core and non-structural protein 3 proteins induce pro- and anti-inflammatory cytokines and inhibit dendritic cell differentiation[J]. J Immunol. 2003, 170: 5615–5624.
    [106] Almonti JB, Ball TB, Fowke KR. Mechanisms of CD4+T lymphocyte cell death in human immunodeficiency virus infection and AIDS[J]. J Gen Virol. 2003, 84: 1649–1661.
    [107] Ruggli N, Tratschin JD, Schweizer M, et al. Classical Swine Fever Virus Interferes with Cellular Antiviral Defense: Evidence for a Novel Function of Npro[J] . J Virol. 2003, 77(13): 7645– 7654.
    [108] Ruggli N, Bird BH, Liu L, et al. Npro of classical swine fever virus is an antagonist of double-stranded RNA-mediated apoptosis and IFN-alpha/beta induction[J]. Virology, 2005, 340(2): 265-276.
    [109] La Rocca SA, Herbert RJ, Crooke H, et al. Loss of interferon regulatory factor 3 in cells infected with classical swine fever virus involves the N-terminal protease, Npro[J]. J Virol. 2005, 79(11): 7239-7247.
    [110] Bauhofer O, Summerfield A, Sakoda Y, et al. Classical swine fever virus Npro interacts with interferon regulatory factor 3 and induces its proteasomal degradation[J]. J Virol. 2007, 81(7):3087-3096.
    [111] Bensaude E, Turner JL, Wakeley PR, et al. Classical swine fever virus induces proinflammatory cytokines and tissue factor expression and inhibits apoptosisand interferon synthesis during the establishment of long-term infection of porcine vascular endothelial cells[J]. J Gen Virol. 2004, 85: 1029–1037.
    [112] Srikiatkhachorn A, Ajariyakhajorn C, Endy TP, et al. Virus-induced decline in soluble vascular endothelial growth receptor 2 is associated with plasma leakage in dengue hemorrhagic Fever[J]. J Virol. 2007, 81(4): 1592-1600.
    [113] Knoetig SM, Summerfield A, Spagnuolo-Weaver M, et al. Immunopathogenesis of classical swine fever: role of monocytic cells[J]. Immunology, 1999, 97(2): 359-366.
    [114] Heinz FX, Collett MS, Purcekk RH, et al. Family Flaviridae. In: Fauquet CM, Mayo M, Maniloff J, et al. (Eds.), Virus Taxonomy. Eighth Report of the International Committee on Taxonomy of Virus[M]. Academic Press, San Diego, 2004, pp.981-998.
    [115] van Gennip HGP, Vlot AC, Hulst MM, de Smit AJ, et al. Determination of virulence of classical swine fever virus strain Brecia[J]. J Virol. 2004, 78(16):8812-23.
    [116] Gr?nborg M, Kristiansen TZ, Iwahori A, et al. Biomarker discovery from pancreatic cancer secretome using a differential proteomic approach[J]. Mol. Cell Proteomics, 2006, 5, 157-171.
    [117] Drust DS, Creutz CE. Aggregation of chromaffin granules by calpactin at micromolar levels of calcium[J]. Nature, 1988, 331, 88–91.
    [118] Mayran N, Parton RG, Gruenberg J. Annexin II regulates multivesicular endosome biogenesis in the degradation pathway of animal cells[J]. EMBO J. 2003, 22, 3242–3253.
    [119] Zobiack N, Rescher U, Ludwig C, et al. The annexin 2/S100A10 complex controls the distribution of transferrin receptor containing recycling endosomes[J]. Mol Biol Cell, 2003, 14, 4896–4908.
    [120] Zeuschner D, Stoorvogel W, Gerke V. Association of annexin 2 with recycling endosomes requires either calcium- or cholesterol-stabilized membrane domains[J]. Eur J Cell Biol. 2001, 80, 499–507.
    [121] Donnelly SR, Moss SE. Annexins in the secretory pathway[J]. Cell Mol Life Sci. 1997, 53, 533–538.
    [122] Ma G, Greenwell-Wild T, Lei K, et al. Secretory leukocyte protease inhibitor binds to Annexin II, a cofactor for macrophage HIV-1 infection[J]. J Exp Med. 2004, 200, 1337–1346.
    [123] Ryzhova EV, Vos RM, Albright AV, et al. Annexin 2: a novel human immunodeficiency virus type 1 Gag binding protein involved in replication in monocyte-derived macrophages[J]. J Virol. 2006, 80(6), 2694-704.
    [124] Malhotra R, Ward M, Bright H, et al. Isolation and characterisation of potential respiratory syncytial virus receptor(s) on epithelial cells[J]. Microbes Infect. 2003, 5(2),123-33.
    [125] Wright JF, Kurosky A, Wasi S. An endothelial cell-surface form of annexin II binds human cytomegalovirus[J]. Biochem Biophys Res Commun. 1994, 198(3), 983-9.
    [126] Wright JF, Kurosky A, Pryzdial EL, et al. Host cellular annexin II is associated with cytomegalovirus particles isolated from cultured human fibroblasts[J]. J Virol. 1995, 69(8), 4784-91.
    [127] Pietropaolo RL, Compton T. Direct interaction between human cytomegalovirus glycoprotein B and cellular annexin II[J]. J Virol. 1997, 71(12), 9803-7.
    [128] Raynor CM, Wright JF, Waisman DM, et al. Annexin II enhances cytomegalovirus binding and fusion to phospholipid membranes[J]. Biochemistry. 1999, 38(16), 5089-95.
    [129] Bustin M, Reeves R. High-mobility-group chromosomal proteins: architectural components that facilitate chromatin function[J]. Prog. Nucleic Acid Res. Mol. Biol. 1996, 54, 35–100.
    [130] Grosschedl R, Giese K, Pagel J. HMG domain proteins: architectural elements in the assembly of nucleoprotein structures[J]. Trends Genet. 1994, 10, 94–100.
    [131] Krecic AM, Swanson MS. HnRNP complexes: composition, structure, and function[J]. Curr Opin Cell Biol. 1999, 11, 363–371.
    [132] Kim CS, Seol SK, Song OK, et al. An RNA-binding protein, hnRNP A1, and a scaffold protein, septin 6, facilitate hepatitis C virus replication[J]. J Virol. 2007, 81(8):3852-65
    [133] Li HP, Zhang X, Duncan R, et al. Heterogeneous nuclear ribonucleoprotein A1 binds to the transcription-regulatory region of mouse hepatitis virus RNA[J]. Proc Natl Acad Sci USA. 1997, 94: 9544-9549
    [134] Zhang X, Li HP, Xue W, et al. Formation of a ribonucleoprotein complex of mouse hepatitis virus involving heterogeneous nuclear ribonucleoprotein A1 and transcription-regulatory elements of viral RNA[J]. Virology 1999, 264: 115-124
    [135] Wang Y, Zhang X. The nucleocapsid protein of coronavirus mouse hepatitis virus interacts with the cellular heterogeneous nuclear ribonucleoprotein A1 in vitro and in vivo[J]. Virology, 1999, 265: 96–109
    [136] Shi ST, Yu GY, Lai MM. Multiple type A/B heterogeneous nuclear ribonucleoproteins (hnRNPs) can replace hnRNP A1 in mouse hepatitis virus RNA synthesis[J]. J Virol. 2003, 77: 10584-10593.
    [137] Woolaway K, Asai K, Emili A, et al. hnRNP E1 and E2 have distinct roles in modulating HIV-1 gene expression[J]. Retrovirology, 2007, 4: 28. (doi: 10.1186/1742-4690-4-28)
    [138] Asai K, Platt C, Cochrane A. Control of HIV-1 env RNA splicing and transport: investigating the role of hnRNP A1 in exon splicing silencer (ESS3a) function[J]. Virology, 2003, 314, 229-242.
    [139] Schaub MC, Lopez SR, Caputi M. Members of the heterogeneous nuclear ribonucleoprotein H family activate splicing of an HIV-1 splicing substrate by promoting formation of ATP-dependent spliceosomal complexes[J]. J Biol Chem. 2007, 282: 13617-13626.
    [140] Tange TO, Damgaard CK, Guth S, et al. The hnRNP A1 protein regulates HIV-1 tat splicing via a novel intron silencer element[J]. EMBO Journal,2001, 20: 5748-5758.
    [141] Marchand V, Mereau A, Jacquenet S, et al. A Janus splicing regulatory element modulates HIV-1 tat and rev mRNA production by coordination of hnRNP A1 cooperative binding[J]. J Mol Biol. 2002, 323: 629-652.
    [142] Bilodeau PS, Domsic JK, Mayeda A, et al. RNA splicing at human immunodeficiency virus type 1 3' splice site A2 is regulated by binding ofhnRNP A/B proteins to an exonic splicing silencer element[J]. J Virol. 2001, 75: 8487-8497.
    [143] Caputi M, Mayeda A, Krainer A, et al. hnRNP A/B proteins are required for inhibition of HIV-1 pre-mRNA splicing[J]. EMBO Journal.1999, 18: 4060- 4067.
    [144] Davis WG, Blackwell JL, Shi PY, et al. Interaction between the cellular protein eEF1A and the 3'-terminal stem-loop of West Nile virus genomic RNA facilitates viral minus-strand RNA synthesis[J]. J Virol. 2007, 81 (18):10172-10187.
    [145] De Nova-Ocampo M, Villegas-Sepúlveda N, del Angel RM. Translation elongation factor-1α, La, and PTB interact with the 3' untranslated region of dengue 4 virus RNA[J]. Virology, 2002, 295 (2): 337-347.
    [146] Xiao H, Neuveut C, Benkirane M, et al. Interaction of the second coding exon of Tat with human EF-1δdelineates a mechanism for HIV-1-mediated shut-off of host mRNA translation[J]. Biochem Biophy Res Commu. 1998, 244 (2): 384-389.
    [147] Ralser M, Wamelink MM, Kowald A, et al. Dynamic rerouting of the carbohydrate flux is key to counteracting oxidative stress[J]. J Biol. 2007, 6 (4):10 (doi:10.1186/jbiol61)
    [148] Kondoh H, Lleonart ME, Bernard D, et al. Protection from oxidative stress by enhanced glycolysis; a possible mechanism of cellular immortalization[J]. Histol Histopatho. 2007, 22 (1):85-90.
    [149] Kondoh H, Lleonart MF, Gil J, et al. Glycolytic enzymes can modulate cellular life span[J]. Cancer Res. 2005, 65 (1):177-185.
    [150] Manevich Y, Fisher AB. Peroxiredoxin 6, a 1-Cys peroxiredoxin, functions in antioxidant defense and lung phospholipids metabolism[J]. Free Radi Biol Med. 2005, 38: 1422-1432.
    [151] Chang XZ, Li DQ, Hou YF, et al. Identification of the functional role of peroxiredoxin 6 in the progression of breast cancer[J]. Breast Cancer Res. 2007, 9 (6): R76 (doi:10.1186/bcr1789).
    [152] Lincoln DT, Ali Emadi EM, Tonissen KF, et al. The thioredoxin–thioredoxinreductase system: over-expression in human cancer[J]. Anticancer Res. 2003, 23, 2425–2433.
    [153] Raffel J, Bhattacharyya AK, Gallegos A, et al. Increased expression of thioredoxin-1 in human colorectal cancer is associated with decreased patient survival[J]. J Lab Clin Med. 2003, 142: 46–51.
    [154] Madersbacher S, Gr?bl M, Kramer G, et al. Regulation of heat shock protein 27 expression of prostatic cells in response to heat treatment[J]. Prostate 1998, 37 (3): 174-181.
    [155] Patel YJ, Payne Smith MD, de Belleroche J, et al. Hsp27 and Hsp70 administered in combination have a potent protective effect against FALS-associated SOD1-mutant-induced cell death in mammalian neuronal cells[J]. Brain Res Mol Brain Res. 2005, 134 (2): 256-274.
    [156] Georgopoulos C, Welch WJ. Role of the major heat shock proteins as molecular chaperones[J]. Annu Rev Cell Biol. 1993, 9: 601-634.
    [157] Leong WF, Chow VT. Transcriptomic and proteomic analyses of rhabdomyosarcoma cells reveal differential cellular gene expression in response to enterovirus 71 infection[J]. Cell Microbiol, 2006, 8: 565-580.
    [158] Go EP, Wikoff WR, Shen Z, et al. Mass spectrometry reveals specific and global molecular transformations during viral infection[J]. J Proteome Res. 2006, 5: 2405-2416.
    [159] Edwards S, Fukusho A, Lefevre PC, et al. Classical swine fever: the global situation [J]. Vet Microbiol, 2000, 73:103-119.
    [160] Moennig V, Plagenann PG. The pestiviruses [J]. Adv Virus Res, 1992, 41: 53–98.
    [161] Summerfield A, Hofmann MA, Mccullough KC. Low density blood granulocytic cells induced during classical swine fever are targets for virus infection [J]. Vet Immunol Immunopathol, 1998, 63: 289–301.
    [162] Summerfield A, Knotig SM, Mccullough KC. Lymphocyte apoptosis during classical swine fever: implication of activation-induced cell death [J]. J Virol, 1998, 72:1853–1861.
    [163] Li Y, Zhao JJ, Shi Z, et al. A multiplex nested RT-PCR for the detection anddifferentiation of wild-type viruses from C-strain vaccine of classical swine fever virus [J]. J Virol Methods, 2007, 143(1):16-22.
    [164] Summerfield A, Mccullough KC. Porcine bone marrow myeloid cells: phenotype and adhesion molecule expression [J]. J Leukoc Biol, 1997, 62: 176-185.
    [165] Fischer VU, Kaden V, Beyer J. Detection of classical swine fever antigen and differentiation of field and vaccine virus antigen using immunofluorescent test on pigs tissues [J]. Mh Vet Med, 1991, 46:133–136.
    [166] Hoffmann R, Hoffmann-Fezer G, Weiss E. Bone marrow lesions in acute hog cholera with special reference to thrombopoietic cells [J]. Berl Munch Tierarztl Wochenschr, 1971, 84:301–305.
    [167] Summerfield A, Zingle K, Inumaru S, et al. Induction of apoptosis in bone marrow neutrophil-lineage cells by classical swine fever virus [J]. J Gen Virol, 2001, 82: 1309–1318.
    [168] Gomez-Villamandos JC, Salguero FJ, Ruiz-Villamor, et al. Classical Swine Fever: pathology of bone marrow [J]. Vet Pathol, 2003, 40(2):157-63.
    [169] Naniche D, Oldstone MB. Generalized immunosuppression: how viruses undermine the immune response [J]. Cell Mol Life Sci, 2000, 57:1399–1407.
    [170] Rumenapf T, Stark R, Meyers G, et al. Structural proteins of hog cholera virus expressed by vaccinia virus: further characterization and induction of protectiveimmunity [J]. J Virol, 1991, 65: 589–597.
    [171] Schneider R, Unger G, Stark R, et al. Identification of a structural glycoprotein of an RNA virus as a ribonuclease [J]. Science, 1993, 261:1169–1171.
    [172] White SR, Williams P, Wojcik KR, et al. Initiation of apoptosis by actin cytoskeletal derangement in human airway epithelial cells[J]. Am J Respir Cell Mol Biol 2001; 24: 282–294.
    [173] van de Water B, Nagelkerke JF, Stevens JL. Dephosphorylation of focal adhesion kinase (FAK) and loss of focal contacts precede caspase-mediated cleavage of FAK during apoptosis in renal epithelial cells[J]. J Biol Chem 1999; 274: 13328–13337.
    [174] Kruidering M, van deWater B, Zhan Y, et al. Cisplatin effects on F-actin andmatrix proteins precede renal tubular cell detachment and apoptosis in vitro[J]. Cell Death Differ 1998; 5: 601–614.
    [175] GenescàM, Sola A, Hotter G. Actin cytoskeleton derangement induces apoptosis in renal ischemia/reperfusion[J]. Apotosis, 2006,11(4):563-71.
    [176] Hecker C, Weise C, Schneider-Schaulies J, Holmes HC, ter Meulen V. Specific binding of HIV-1 envelope protein gp120 to the structural membrane proteins ezrin and moesin[J]. Virus Res. 1997, 49(2):215-23.
    [177] Schneider-Schaulies J, Dunster LM, Schwartz-Albiez R, Krohne G, ter Meulen V. Physical association of moesin and CD46 as a receptor complex for measles virus[J]. J Virol. 1995, 69(4): 2248-56.
    [178] Scheuring UJ, Corbeil J, Mosier DF, Theofilopoulos AN. Early modification of host cell gene expression induced by HIV-1[J]. AIDS.1998, 12(6):563-70.
    [179] Naghavi MH, Valente S, Hatziioannou T, de Los Santos K, Wen Y, Mott C, Gundersen GG, Goff SP. Moesin regulates stable microtuble formation and limits retroviral infection in cultured cells[J]. EMBO J. 2007, 26(1):41-52.
    [180] Ziegler WH, Liddington RC, Critchley DR. The structure and regulation of vinculin[J]. Trends Cell Biol. 2006, 16 (9 ): 453–460.
    [181] Subauste MC, Pertz O, Adamson ED, et al. Vinculin modulation of paxillin-FAK interactions regulates ERK to control survival and motility[J]. J Cell Biol. 2004, 165(3): 371-381.
    [182] Propato A, Cutrona G, Francavilla V, et al. Apoptotic cells overexpress vinculin and induce vinculin-specific cytotoxic T-cell cross-priming[J].Nat Med.2001, 7(7): 807- 813.
    [183] Rodríguez Fernández JL, Geiger B, Salomon D, et al. Suppression of tumorigenicity in transformed cells after transfection with vinculin cDNA[J]. J Cell Biol. 1992, 119(2): 427-438.
    [184] Johnson CM, Perez DR, French R, et al. The NS5A protein of bovine viral diarrhoea virus interacts with the alpha subunit of translation elongation factor-1[J]. J Gen Virol. 2001, 82(Pt12): 2935-2943.
    [185] Condeelis J. Elongation factor 1a, translation and the cytoskeleton[J]. Trends in Biochemical Sciences, 1995, 20: 169-170.
    [186] Murray JW, Edmonds BT, Liu G, et al. Bundling of actin filaments by elongation factor 1a inhibits polymerization at filament ends[J]. Journal of Cell Biology 135: 1309-1321.
    [187] Yang F, Demma M, Warren V, et al. Identification of an actin-binding protein from Dictyostelium as elongation factor 1a[J]. Nature, 1990, 347: 494-496.
    [188] Shiina N, Gotoh Y, Kubomura N, et al. Microtubule severing by elongation factor 1a[J]. Science, 1994, 266, 282-285.
    [189] Gonen H, Smith CE, Siegel NR, et al. Protein synthesis elongation factor EF-1a is essential for ubiquitin-dependent degradation of certain N a-acetylated proteins and may be substituted for by the bacterial elongation factor EF-Tu[J]. Proceedings of the National Academy of Sciences, 1994, 91: 7648-7652.
    [190] Kruse C, Grunweller A, Willkomm DK, et al. tRNA is entrapped in similar, but distinct, nuclear and cytoplasmic ribonucleoprotein complexes, both of which contain vigilin and elongation factor 1a[J]. Biochemical Journal, 1998, 329: 615-621.
    [191] Sirover, M. A. Role of the glycolytic protein, glyceraldehyde-3-phosphate dehydrogenase, in normal cell function and in cell pathology[J]. J Cell Biochem. 1997, 66:133–140.
    [192] Ishitani R, Chuang DM. Glyceraldehyde-3-phosphate dehydrogenase antisense oligodeoxynucleotides protect against cytosine arabinonucleoside-induced apoptosis in cultured cerebellar neurons[J]. Neurobiology. 1996, 93: 9937–41.
    [193] Sawa A, Khan A, Hester L, Snyder S. Glyceraldehyde-3-phosphate dehydrogenase: Nuclear translocation participates in neuronal and nonneuronal cell death[J]. Proc Natl Acad Sci U S A. 1997, 94: 11669–74.
    [194] Saunders PA, Chen RW, Chuang DM. Nuclear translocation of glyceraldehydes -3 - phosphate dehydrogenase isoforms during neuronal apoptosis[J]. J Neurochem.1999, 72: 925–32.
    [195] Dastoor Z, Dreyer JL. Potential role of nuclear translocation of glyceraldehydes -3- phosphate dehydrogenase in apoptosis and oxidative stress[J]. J Cell Sci. 2001, 114: 1643–53.
    [196] Munro S, Pelham HR. An Hsp70-like protein in the ER: identity with the 78 kdglucose-regulated protein and immunoglobulin heavy chain binding protein[J]. Cell, 1986, 46: 291–300.
    [197] Lee AS, Bell J, Ting J. Biochemical characterization of the 94-and 78- kilodalton glucose-regulated proteins in hamster fibroblasts[J]. J Biol Chem. 1984, 259: 4616–4621.
    [198] Welch WJ, Garrels JI, Thomas GP, et al. Biochemical characterization of the mammalian stress proteins and identification of two stress proteins as glucose- and Ca2+-ionophore-regulated proteins[J]. J Biol Chem. 1983, 258:7102–7111.
    [199] Wu FS, Park YC, Roufa D, et al. Selective stimulation of the synthesis of an 80,000-dalton protein by calcium ionophores[J]. J Biol Chem. 1981, 256: 5309–5312.
    [200] Sarnow P. Translation of glucose-regulated protein 78/immunoglobulin heavy-chain binding protein mRNA is increased in poliovirus-infected cells at a time when cap-dependent translation of cellular mRNAs is inhibited[J]. Proc Natl Acad Sci USA. 1989, 86: 5795–5799.
    [201] Bole DG, Hendershot LM, Kearney JF. Posttranslational association of immuno- globulin heavy chain binding protein with nascent heavy chains in nonsecreting and secreting hybridomas[J]. J Cell Biol. 1986, 102:1558–1566.
    [202] Pelham HR. Speculations on the functions of the major heat shock and glucose- regulated proteins[J]. Cell, 1986, 46: 959–961.
    [203] Berggren MI, Husbeck B, Samulitis B, et al. Thioredoxin peroxidase-1 (peroxiredoxin-1) is increased in thioredoxin-1 transfected cells and results in enhanced protection against apoptosis caused by hydrogen peroxide but not by other agents including dexamethasone, etoposide, and doxorubicin[J]. Arch Biochem Biophys. 2001, 392(1): 103-109.
    [204] Kim SY, Kim TJ, Lee KY. A novel function of peroxiredoxin 1 (Prx-1) in apoptosis signal-regulating kinase 1 (ASK1)-mediated signaling pathway[J]. FEBS Lett. 2008, 582(13): 1913-1918.
    [205] Qi Y, Chiu JF, Wang L, et al. Comparative proteomic analysis of esophageal squamous cell carcinoma[J]. Proteomics, 2005, 5(11): 2960-71.
    [206] Kim JH, Bogner PN, Baek SH, et al. Up-regulation of peroxiredoxin 1 in lungcancer and its implication as a prognostic and therapeutic target[J]. Clin Cancer Res. 2008,14(8): 2326-2333.
    [207] Geibed-Lynn R, Kursar M, Brown NV, et al. HIV-1 antiviral activity of recombinant natural killer cell enhancing factors, NKEF-A and NKEF-B, members of the peroxiredoxin family[J]. J Biol Chem. 2003, 278(3): 1569-74.
    [208] Li SS, Ivanoff A, Bergstr?m SE, et al. T lymphocyte expression of thrombospondin-1 and adhesion to extracellular matrix components[J]. Eur J Immunol. 2002, 32(4): 1069-1079.
    [209] Li Z, Calzada MJ, Sipes JM, et al. Interactions of thrombospondins with alpha4beta1 integrin and CD47 differentially modulate T cell behavior[J]. J Cell Biol. 2002, 157(3): 509-519.
    [210] Vallejo AN, Mugge LO, Klimiuk PA, et al. Central role of thrombospondin-1 in the activation and clonal expansion of inflammatory T cells[J]. J Immunol. 2000, 164:2947–2954.
    [211] Lawler J, Sunday M, Thibert V, et al. Thrombospondin-1 is required for normal murine pulmonary homeostasis and its absence causes pneumonia[J]. J. Clin. Invest. 1998, 101: 982–992.
    [212] Grossklaus DJ, Smith JA, Jr Shappell SB, et al. The free/total prostate-specific antigen ratio (%fPSA) is the best predictor of tumor involvement in the radical prostatectomy specimen among men with an elevated PSA[J]. Urol Oncol. 2002, 7:195–198.
    [213] Whitehouse C, Solomon E. Current status of the molecular characterization of the ovarian cancer antigen CA125 and implications for its use in clinical screening[J]. Gynecol Oncol. 2003, 88(1Pt): S152-7.
    [214] Zhang Z, Bast RC Jr, Yu Y, et al. Three biomarkers identified from serum proteomic analysis for the detection of early stage ovarian cancer[J]. Cancer Res. 2004,64(16), 5882–5890.
    [215] Sasaki K, Sato K, Akiyama Y, et al. Peptidomics-based approach reveals the secretion of the 29-residue COOH-terminal fragment of the putative tumor suppressor protein DMBT1 from pancreatic adenocarcinoma cell lines[J]. Cancer Res. 2002, 62: 4894–4898.
    [216] Kennedy S. The role of proteomics in toxicology: identification of biomarkers of toxicity by protein expression analysis[J]. Biomarkers 2002, 7(4): 269–290.
    [217] Anderson NL, Anderson NG. The human plasma proteome: history, character, and diagnostic prospects[J]. .Mol Cell Proteomics 2002,1(11): 845–867.
    [218] Lathrop JT, Anderson NL, Anderson NG, et al. Therapeutic potential of the plasma proteome[J]. Curr Opin Mol. Ther. 2003, 5(3):250–257.
    [219] Rosenblatt KP, Bryant-Greenwood P, Killian JK, et al. Serum proteomics in cancer diagnosis and management[J]. Annu Rev Med. 2004, 55: 97–112.
    [220] Jacobs IJ, Menon U. Progress and challenges in screening for early detection of ovarian cancer[J]. Mol Cell Proteomics 2004, 3: 355–366.
    [221] Ardekani AM, Liotta LA, Petricoin EF 3rd. Clinical potential of proteomics in the diagnosis of ovarian cancer[J]. Expert Rev Mol Diagn. 2002,2(4):312-20.
    [222] Feng JT, Liu YK, Song HY, et al. Heat-shock protein 27: a potential biomarker for hepatocellular carcinoma identified by serum proteome analysis[J]. Proteomics, 2005, 5 (17): 4581-4588.
    [223] Baumann H, Gauldie J. The acute phase response[J]. Immunol Today. 1994, 15(2): 74-80.
    [224] Kino K, Tsunoo H, Higa Y, et al. Kinetic aspects of hemoglobin- haptoglobin- receptor interaction in rat liver plasma membranes, isolated liver cells, and liver cells in primary culture[J]. J Biol Chem. 1982, 257: 4828-4833.
    [225] Kino K, Tsunoo H, Higa Y, et al. Hemoglobin-haptoglobin receptor in rat liver plasma membrane[J]. J Biol Chem. 1980, 255:9618-9620.
    [226] Kushner I. The phenomenon ofthe acute phase response[J]. Ann NY Acad Sci. 1982, 389:39-48.
    [227] Hansen JES, Iversen J, Lihme A, et al. Acute phase reaction, heterogeneity and microheterogeneity of serum proteins as nonspecific tumor markers in lung cancer[J]. Cancer. 1987, 60:1630-1635.
    [228] Giordano A, Spagnolo V, Colombo A, et al. Changes in some acute phase protein and immunoglobulin concentrations in cats affected by feline infectious peritonitis or exposed to feline coronavirus infection[J].Vet J. 2004,167(1): 38-44.
    [229] Shim BS. Increase in serum haptoglobin stimulated by prostaglandins[J].Nature, 1976, 259(5541): 326-327.
    [230] Kalsheker NA, Bradwell AR, Burnett D. The inhibition of cathepsin B by plasma haptoglobin. Experientia, 1981, 37: 447-448.
    [231] Cid MC, Grant DS, Hoffman GS, et al. Identification of Haptoglobin as an Angiogenic Factor in Sera from Patients with Systemic Vasculitis[J]. J Clin Invest. 1993,91(3): 977-85.
    [232] Fenton JW 2nd. Regulation of thrombin generation and functions[J]. Semin Thromb Hemost.1988, 14(3):234-240.
    [233] Dixon JL, Ginsberg HN. Hepatic synthesis of lipoproteins and apolipo- proteins [J]. Semin Liver Dis. 1992, 12: 364–72.
    [234] Danielsen EM, Hansen GH, Poulsen MD. Apical secretion of apolipoproteins from enterocytes[J]. J Cell Biol .1993, 120: 1347–56.
    [235] Barter PJ, Rye KA. The rationale for using apoA-I as a clinical marker of cardiovascular risk[J]. J Intern Med. 2006,259(5): 447-454.
    [236] Cockerill GW, Rye KA, Gamble JR, et al. High-density lipoproteins inhibit cytokine-induced expression of endothelial cell adhesion molecules[J]. Arterioscler Thromb Vasc Biol. 1995, 15: 1987–94.
    [237] Ashby DT, Rye KA, Clay MA, et al. Factors influencing the ability of HDL to inhibit expression of vascular cell adhesion molecule-1 in endothelial cells[J]. Arterioscler Thromb Vasc Biol, 1998, 18: 1450–5.
    [238] Bisoendial RJ, Hovingh GK, Levels JHM, et al. Restoration of endothelial function by increasing high-density lipoprotein in subjects with isolated low high-density lipoprotein[J]. Circulation, 2003, 107: 2944–8.
    [239] Law SKA, Dodds AW, Porter RR. The comparison of the properties of two classes, C4A and C4B, of the human complement component C4[J]. EMBO J. 1984, 3:1819-1823.
    [240] Carroll MD, Fathalla L, Bergamaschini EA, et al. Substitution of a single amino acid (aspartic acid for histidine) converts the functional activity of human C4B to C4A[J]. Proc Natl Acad Sci USA. 1990, 87:6868-6872.
    [241] Hauptmann G, Tappeiner G, Schifferli JA. Inherited deficiency of the fourthcomponent of human complement[J]. Immunodefic Rev. 1988, 1:3-22.
    [242] Devine DV, Siegel RS, Rosse WF. Interactions of the platelets in paroxysmal nocturnal hemoglobinuria with complement. Relationship to defects in the regulation of complement and to platalet survival in vivo[J]. J Clin Invest. 1987, 79: 131–137
    [243] DiScipio RG, Daffern PJ, Schraufstatter IU, et al. Human polymorphonuclear leukocytes adhere to complement factor H through an interaction that involves alphaMbeta2 (CD11b/CD18) [J]. J Immunol. 1998, 160: 4057–4066
    [244] Manuelian T, Hellwage J, Meri S, et al. Mutations in factor H reduce binding affinity to C3b and heparin and surface attachment to endothelial cells in hemolytic uremic syndrome[J]. J Clin Invest. 2003, 111: 1181–1190.

© 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