重组蓖麻毒素B链调节免疫细胞活化及其对信号传导通路的影响
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
蓖麻毒素(RT)由RTA、RTB两个肽链以二硫键共价相连接。RTB是能与半乳糖/N-乙酰基半乳糖胺特定结构结合的外源凝集素蛋白,具有凝集素活性。RTB本身无毒性,但可以帮助RTA转运到细胞溶质中,抑制蛋白质的合成。文献报道,RTB可以用作异源性疫苗抗原的载体,可融合不同的轮状病毒抗原VP7、P24、NSP4等,发挥粘膜佐剂作用。课题组前期研究亦发现RTB作为佐剂能诱导产生狂犬病毒中和抗体,抗体水平显著高于普通氢氧化铝佐剂疫苗,并能加速狂犬病毒疫苗诱导的体液免疫应答。然而对于RTB更多的生物学活性及机制研究国内外报道甚少。RTB对免疫细胞活化及细胞内的信号传导通路的影响国内外未见报道。本研究采用分子生物学与免疫学技术探讨了RTB对免疫细胞活化及细胞内的信号传导通路的影响。首先探讨了RTB对小鼠免疫细胞活化功能的影响。研究发现,RTB可明显促进小鼠脾脏淋巴细胞增殖;促进淋巴细胞培养上清中Th1型细胞因子的分泌;并发现RTB可促进CD4+T细胞的增殖和活化。RTB对小鼠RAW264.7巨噬细胞的活化功能与机制研究表明:RTB可促进RAW264.7细胞NO的分泌;促进iNOS基因与蛋白的表达;增强巨噬细胞的吞噬能力;促进IL-6和TNF-α基因与蛋白的表达。采用小鼠蛋白芯片抗体阵列对巨噬细胞与炎症相关的40种蛋白的变化进行检测,结果发现MCP-1、TNF-α、MIP-1α,MIP-1β,MIP-2,RANTES,TIMP-1,CD54的表达增高,G-CSF、KC的表达下调。RTB对RAW264.7巨噬细胞活化过程中的信号转导通路的影响研究表明:酪氨酸蛋白激酶介导的信号通路抑制剂、蛋白激酶C信号通路抑制剂和核因子κB信号通路抑制剂,可以抑制RTB诱导RAW264.7细胞NO分泌和iNOSmRNA表达以及TNF-α和IL-6mRNA和蛋白水平表达,提示酪氨酸蛋白激酶、蛋白激酶C和核因子κB信号通路可能参与RTB诱导巨噬细胞活化。采用RTB对酪氨酸蛋白激酶信号通路相关磷酸化位点分子进行蛋白组学分析,结果提示RTB通过影响RAW264.7细胞酪氨酸蛋白激酶信号途径SRC、JAK-STAT和核因子κB信号途径IκB-α蛋白磷酸化,激活酪氨酸蛋白激酶和NF-κB信号转导途径。
     Western blot和免疫荧光实验结果进一步证实RTB通过诱导巨噬细胞IκB-α磷酸化进而促进NF-κB核转位作用。
     上述结果提示RTB可以调节免疫细胞活化,对巨噬细胞的活化可通过细胞内的多条信号转导途径参与。提示RTB诱导巨噬细胞活化可能通过多条信号转导途径互相调节、共同作用。本研究为RTB免疫生物学活性研究及其免疫佐剂等方面的应用提供了新的科研资料,并奠定了实验依据与理论基础。
Ricin toxicity (RT) is covalently linked by the two peptide chains RTA and RTB indisulfide bonds.RTB consists of260amino acids is a galactose/N-acetylgalactosaminelectin protein and has a molecular weight about34000. The individualism of RTB has notoxicity and is believed to have two main functions at present: One is that it can helpRTA transfer to cytosol and inhibit the synthesis of protein. Another is that RTBligand has the capacity of specific binding with the receptor of different carbohydratestructure, to mediate various kinds of bioprocess, including interactions between cell,cell host and pathogen, and the innate immunity response. Also it can be used as thecarrier of heterologous antigens to the immune system. For instance, RTB can fusedifferent Rotavims antigens such as VP7, P24, NSP4, etc, and perform as mucosaladjuvants to enhance the immune reaction of organism. Our study also found that theRTB as adjuvant can induce the production of rabies neutralizing antibody, theantibody levels were significantly higher than ordinary aluminum hydroxide adjuvantvaccine and can accelerate the immune response of the body fluid induced by rabiesvaccine.With RTB as the object, this project investigates RTB's influence on mice's Tcell and macrophage activating function as well as on the signal transductionpathways inside the macrophage, in the hope of furnishing the experimental evidenceand theoretic foundation for the research on RTB immunobiological activation.
     The aspects of this research as follow:
     1. Preparation of RTB
     PET28a-RTB was transformed into BL21(DE3) cells to stimulate recombinantprotein synthesis and soluble protein was renaturated.The protein was purified usingnickel-NTA column chromatography. LPS was removed by Detoxi-Gel.
     2. RTB protein induced mouse T cell activation
     RTB was found was found to enhance the splenocytes proliferation comparedwith control. In the presence of ConA or LPS, the treatment with the RTBsignificantly enhanced the spleen index.The effect of RTB on the splenocytes proliferation was in a dose-dependent manner. The secretion of the IL-2, IL-4, IFN-γ,IL-5, and TNF-α cytokines was measured using a cytometric bead array immunoassay(CBA). After culturing spleen cells in vitro, the highest concentrations of TNF-α, IL-2,and IFN-γ that were induced were significant compared to the concentrations induced bythe control group. No significant differences were observed in the IL-4and IL-5cytokinelevels produced by experimental and control groups.CD4+T cell immunomagnetic negativeseparation and purification beads were used to study the CD4+T cell proliferation andcytokines secretion induced by recombinant RTB protein. The results showed that the RTBprotein can promote CD4+T cell proliferation and Th1-type cytokine secretion. Flowcytometry is adopted to test the number of cell subsets of CD4+and CD8+T, which showsthat RTB can promote the number of CD4+T, but have no significant effect on CD8+T.Flow cytometry is adopted to test the expression of CD25molecule, the mid-term activationmarker of mice's CD3+T cell, as a result of which RTB acting alone or cooperating withConA can both stimulate the expression of CD25molecule (P<0.01). Alarm blue approachis used to test and purify the proliferation activity of CD4+T cell, wherein the resultindicates that RTB can improve the purification of the proliferation activity CD4+T cellsubsets no matter it acts alone or cooperates with CD3monoclonal antibodies or CD28monoclonal antibodies (P<0.05).
     3. RTB protein induced mouse macrophages activation
     We researched RTB-induced effect on production of NO in mice macrophagic cell lineRAW264.7cells. It turns out that RTB can induce the release of NO in RAW264.7cells. Andit was dose-dependent and time-dependent. The expression of iNOS mRNA was rised with theincreasing density of RTB. Western blot confirm the expression of iNOS protein was rised asthe same reason. It suggested that RTB can induce the expression of iNOS, which can inducethe increasing immunoloregulation. RTB can also induce TNF-α and IL-6production, and thereis a time and dose-dependent relationship.We used flow cytometry to detect fluorescentmicrospheres engulfed by RAW264.7cells after stimulated with RTB, founding thatcomparing with the control, the percentage of fluorescent microspheres engulfed by RAW264.7cells had improved significantly.We used the RD mouse protein chip40proteins associated with inflammation to observed cytokine and chemokines changes. The resultsshowed that the expression of cytokine MCP-1、TNF-α、MIP-1α、 MIP-1β、 MIP-2、RANTES、 TIMP-1and CD54were increased and the expression of chemokines G-CSF andKC were decreased.
     4. Effect of signal transduction pathway on mouse macrophages activationinduced by RTB protein
     RAW264.7cells treated with in the presence of genistein, SB203580, LY294002,or staurosporine exhibited reduced NO production.The MAP kinase inhibitorsPD98059and SP600125did not affect the observed RTB-induced NO production.RAW264.7cells treated with RTB in the presence of the JAK2inhibitor tyrphostin(AG490) and NF-κB inhibitor BAY showed inhibited NO production. Similar resultswere obtained for iNOS mRNA expression by RT-PCR. The production of TNF-α andIL-6induced by RTB was inhibited by the co-treatment with genistein, SB203580,LY294002, BAY, staurosporine, and AG490. Similar effects were found with regard toTNF-α and IL-6gene transcription. These data suggested the probable role of tyrosinekinases, PI3K, PKC, and JAK2in RTB-mediated macrophage activation.weperformed a phospho-proteomics-based study using a phospho-antibody microarray.Without the inhibitor,20of the228sites were positive for phosphorylation, and thehighest value was observed for JAK2Tyr221. Two tyrosines (Tyr418and529) inSRC were phosphorylated. The JAK1Tyr1022, JAK2Tyr221, STAT1Tyr701, STAT3Tyr705, STAT4Tyr693, STAT5A Tyr694, and IkB-α Tyr42phosphorylation levelswere increased. Genistein reduced the phosphorylation of16/288tyrosine sites.Phosphoproteins expression spectrum indicates that by affecting RAW264.7celltyrosine protein kinase signaling pathway SRC, JAK-STAT and NF-κB signalingpathway IκB-α protein phosphorylation, RTB activates tyrosine protein kinase andNF-κB signal transduction pathway.
     5. Recombinant RTB protein induces NF-κB activation in RAW264.7cells
     We further confirmed that RTB was able to promote the IκB-α phosphorylationand NF-κB nuclear translocation by using Western blot and immunofluorescence.We considered that NF-κB activation was involved in the RAW264.7cells activationinduced by recombinant RTB.
     The above results demonstrate that RTB has regulating and activating functionon mice's immune cells and RAW264.7cell. The RTB's activating mechanism on RAW264.7cell may be closely related to RTB affecting tyrosine protein kinase and related proteinphosphorylation in NF-κB signal transduction pathways, so as to activate SRC, JAK-STATand NF-κB signal transduction pathways. RTB may induce macrophage activation by mutualadjustment and interaction of signal transduction pathways. Whether there are other signalpathways and related proteins participating in remains to be further studied.
引文
[1] Sharon N, Lis H. Lectins: cell-agglutinating and sugar-specific proteins[J].Science,1972,177(4053):949-959.
    [2] Bies C, Lehr C M, Woodley J F. Lectin-mediated drug targeting: history andapplications [J].Adv Drug Deliv Rev,2004,56(4):425-435.
    [3] Risberg T, Lund E, Wist E, et al. The use of non-proven therapy among patientstreated in Norwegian oncogical departments. A cross-sectionalnationalmulticentre study [J]. Eur J Cancer,1995,31A (11):1785-1789.
    [4] Fran D R, Aax N K. Medical aspects of chemical and biological warfare,chapter32, ricin toxin, fall church[C]VA: Office of the SurgeonGeneral,1997,631.
    [5] Olsnes S, Refsnes K, Pihl A. Mechanism of action of the toxic lectins abrin andRicin [J].Nature,1974,249(458):627-631.
    [6] Frankel A E, Fu T, Burbage, C, et al. Lectin-deficient ricin toxin intoxicatescells bearing the D-mannose receptor [J].Carbohydrate Research,1997,300(3):251-258.
    [7] Stirpe, F. Ribosome-inactivating proteins: From toxins to useful proteins [J].Toxicon,2013,67C:12-16.
    [8] Montfort W, Villafranca J E, Monzingo A F, et al. The three-dimensionalstructure of ricin at2.8A [J].J Biol Chem,1987,262(11):5398-5403.
    [9] Olsnes S, Pihl A. Construction and properties of chimeric toxins target specificcytotoxic agents. In: Dorner F, Drews J, eds. Pharmacology of Bacterial Toxins[J]. New York, NY: Pergamon Press,1986,709-739.
    [10] utenber E, Katzin B J, Ernst S, et al. Crystallographic refinement of ricin to2.5A [J].Proteins,1991,10(3):240-250.
    [11] Katzin B J, Collins E J, Robertus J D. Structure of ricin A-chain at2.5A [J].Proteins,1991,10(3):251-259.
    [12] Lebeda F J, Olson M A. Prediction of a conserved, neutralizing epitope inribosomeinactivating proteins [J].Int J Biol Macromol,1999,24(1):19-26.
    [13] Ready M P, Kim Y, Robertus J D. Site-directed mutagenesis of ricin A-chain andimplications for the mechanism of action [J].Proteins,1991,10(3):270-278.
    [14] Li X P, Chiou J C, Remacha M,etal. A two-step binding model proposed for theelectrostatic interactions of ricin a chain with ribosomes [J].Biochemistry,2009,48(18):3853-3863.
    [15] Mayerhofer P U, Cook J P, Wahlman J,et al. Ricin A chain insertion intoendoplasmic reticulum membranes is triggered by a temperature increase to37{degrees} C [J]. J Biol Chem,2009,284(15):10232-10242.
    [16] Sokolowska I, Walchli S, Wegrzyn G, et al. A single point mutation in ricinA-chain increases toxin degradation and inhibits EDEM1-dependent ERretrotranslocation [J]. Biochem J,2011,436(2):371-385.
    [17] Dietrich J B, Ribereau G G.Identity of the N-terminal sequences of the three Achains of mistletoe lectins: homology with ricin-like plant toxin andsingle-chain ribosome inhibiting proteins [J].Anticancer Drugs,1992,3(5):507-511.
    [18] Munishkin A, Wool IG. Systematic deletion analysis of ricin A-chain function.Single amino acid deletions [J].J Biol Chem,1995,270(51):30581-30587.
    [19] Wales R, Richardson P, Roberts L, et al. Mutational analysis of the galactosebinding activity of recombinant ricin B chain [J].J Biol Chem,1991,266(29):19172-19179.
    [20] Vijayan M, Chandra N. Lectins [J].Curr Opin Struct Biol,1999,9(6):707-714.
    [21] Sphyris N, Lord J M, Wales R, et al. Mutational analysis of the Ricinuscommunislectin B-chains [J].J Biol Chem,1995,270(35):20292-20297.
    [22] T onevitsky A, Toptygin A, Agapov I, et al. Renatured ricin toxin Bchain madein E. coli is soluble, stable, and biologically active [J].Biochem Mol Biol Int,1994,32(6):1139-1146.
    [23] Vitetta E and Yen N. Expression and functional properties of geneticallyengineered ricin B chain lacking galactose-binding activity. Biochim [J].Biophys Acta,1990,1049(2):151-157.
    [24] Olsnes S&Pihl A. Different biological properties of the two constituent peptidechains of ricin, a toxic protein inhibiting protein synthesis [J].Biochemistry,1973,12(16):3121-3126.
    [25] Sandvig K, Torgersen M L, Raa H A,et al. Clathrinindependent endocytosis:from nonexisting to an extreme degree of complexity [J].Histochem Cell Biol,2008,129(3):267-276.
    [26] Wieffer M, Maritzen T and Haucke V. SnapShot: endocytic trafficking [J].Cell,2009,137(2):382-383.
    [27] Garred O, van Deurs B and Sandvig K. Furin-induced cleavage and activation ofShiga toxin [J].J Biol Chem,1995,270(18):10817-10821.
    [28] Gordon V M and Leppla S H. Proteolytic activation of bacterial toxins: role ofbacterial and host cell proteases.Infect Immun,1994,62(2):333-340.
    [29] Mayor S and Pagano R E. Pathways of clathrin-independent endocytosis [J].NatRev Mol Cell Biol,2007,8(8):603-612.
    [30] Moya M, Dautry-Varsat A, Goud B, et al. Inhibition of coated pit formation inHep2cells blocks the cytotoxicity of diphtheria toxin but not that of ricin[J].Cell Biol,1985,101(2):548-559.
    [31] Sandvig K, Olsnes S, Petersen O W, et al. Acidification of the cytosol inhibitsendocytosis from coated pits [J].Cell Biol,1987,105(2):679-689.
    [32] Iversen T G, Frerker N, Sandvig K. Uptake of ricinB-quantum dot nanoparticlesby a macropinocytosis-like mechanism[J].J Nanobiotechnology,2012,10:33.
    [33] Garred O, Rodal S K, van Deurs B, et al. Reconstitution of clathrin-independentendocytosis at the apical domain of permeabilized MDCK II cells: Requirementfor a Rho-family GTPase [J].Traffic,2001,2(1):26-36.
    [34] Mallard F, Antony C, Tenza D, et al. Direct pathway from early/recyclingendosomes to the Golgi apparatus revealed through the study of shiga toxinB-fragment transport [J].Cell Biol,1998,143(4):973-990.
    [35] Iversen T G, Skretting G, Llorente A, et al. Endosome to Golgi transport ofricin is independent of clathrin and of the Rab9-and Rab11-GTPases [J].Mol.Biol. Cell,2001,12(7):2099-2107.
    [36] Johannes L and Popoff V. Tracing the retrograde route in protein trafficking [J].Cell,2008,135(7):1175-1187.
    [37] Sandvig K, Bergan J, Dyve A B, et al.Endocytosis and retrograde transport ofShiga toxin.[J].Toxicon,2010,56(7):1181-1185
    [38] Utskarpen A, Slagsvold H H, Iversen T G, et al. Retrograde transport of ricin isregulated by Rab6A/A’in a sequential manner [J].Traffic,2006,7:663–672.
    [39] Skanland S S, Walchli S, Brech A,et al. SNX4in complex with clathrin anddynein: implications for endosome movement [J].PLoS ONE,2009,4(6):e5935.
    [40] Sk nland S S, W lchli S, Wandinger-Ness A, et al. Phosphoinositide-regulatedretrograde transport of ricin: crosstalk between hVps34and sorting nexins [J].Traffic,2007,8(3):297–309.
    [41] Van Weering J R, Verkade P, Cullen P J.SNX-BAR proteins in phosphoinositide-mediated, tubular-based endosomal sorting. Semin [J].Cell Dev.Biol,2010,21(4):371-380.
    [42] Dyve A B, Bergan J, Utskarpen A, et al. Sorting nexin8regulates endosome-to-Golgi transport.[J].Biochem Biophys Res Commun,2009,390(1):109-114.
    [43] Skanland S S, Walchli S, Brech A,et al. SNX4in complex with clathrin anddynein: implications for endosome movement [J]. PLoS ONE,2009,4(6):e5935.
    [44] Bujny M V, Popoff V, Johannes L et al. The retromer component sorting nexin-1is required for efficient retrograde transport of Shiga toxin from early endosometo the trans Golgi network [J].Cell Sci,2007,120(7):2010-2021.
    [45] Popoff V, Mardones G A, Tenza D, et al. The retromer complex and clathrindefine an early endosomal retrograde exit site [J].Cell Sci,2007,120(12):2022-2031.
    [46] Utskarpen A,Slagsvold H H, Dyve A B, et al. SNX1and SNX2mediateretrograde transport of Shiga toxin [J].Biochem Biophys Res Commun,2007,358(2):566-570.
    [47] Hehnly H, Sheff D, Stamnes M. Shiga toxin facilitates its retrograde transport bymodifying microtubule dynamics [J].Mol Biol Cell,2006,17(10):4379-4389.
    [48] Hehnly H A, Longhini K M, Chen J L, et al. Retrograde Shiga toxin traffickingis regulated by ARHGAP21and Cdc42[J].Mol Biol Cell,2009,20(20):4303-4312.
    [49] Simpson J C, Lord J M, Roberts L M. Point mutations in the hydrophobicC-terminal region of ricin A chain indicate that Pro250plays a role inmembrane translocation [J].Eur Biochem,1995,232(2):458-463.
    [50] Deeks E, Cook J P, Day P J, et al. The low lysine content of ricin A chainreduces the risk of proteolytic degradation after translocation from theendoplasmic reticulum to the cytosol [J].Biochemistry,2002,41(10):3405-3413.
    [51] Neal L M, McCarthy E A, Morris C R,et al.Vaccine-induced intestinal immunityto ricin toxin in the absence of secretory IgA[J]. Vaccine.2011,29(4):681-689.
    [52] Simeral L S, Kapmeyer W, MacConnell W P, et al. On the role of the covalentcarbohydrate in the action of ricin [J].Biol Chem,1980,255(23):11098–11101.
    [53] Yermakova A, Vance D J, Mantis N J. Sub-Domains of Ricin’s B Subunit asTargets of Toxin Neutralizing and Non-Neutralizing Monoclonal Antibodies [J].PLoS One,2012,7(9): e44317.
    [54] Thorpe P E, Detre S I, Foxwell B M, et al. Modification of the carbohydrate inricin with metaperiodate-cyanoborohydride mixtures. Effects on toxicity and invivo distribution [J].Eur,Biochem,1985,147(1):197-206.
    [55] Vigerust DJ, Vick S, Shepherd VL. Characterization of functional mannosereceptor in a continuous hybridoma cell line[J]. BMC Immunol,2012,13:51.
    [56] Taylor P R, Gordon S, Martinez-Pomares L. The mannose receptor: linkinghomeostasis and immunity through sugar recognition [J].Trends Immunol,2005,26(2):104–110.
    [57] Kerrigan A M, Brown G D. C-type lectins and phagocytosis [J].Immunobiology,2009,214(7):562-75.
    [58] Upham J P, Pickett D, Irimura T, et al. Macrophage receptors for influenza Avirus: role of the macrophage galactose-type lectin and mannose receptor inviral entry [J]. J Virol,2010,84(8):3730-3737.
    [59] Magnusson S, Berg T. Endocytosis of ricin by rat liver cells in vivo and in vitrois mainly mediated by mannose receptors on sinusoidal endothelial cells [J].Biochem,1993,291(Pt3):749-755.
    [60] Shapira A, Benhar I. Toxin-based therapeutic approaches.[J]. Toxins(Basel),2010,2(11):2519-2583
    [61] Magnusson S, Kjeken R, Berg T. Characterization of two distinct pathways ofendocytosis of ricin by rat liver endothelial cells [J].Exp Cell Res,1993,205(1):118-125.
    [62] Skilleter D N, Paine A J, Stirpe F. A comparison of the accumulation of ricin byhepatic parenchymal and non-parenchymal cells and its inhibition of proteinsynthesis [J].Biochim Biophys Acta,1981,677(3-4):495-500.
    [63] Gage E, Hernandez M O, O’Hara J M, et al. Role of the mannose receptor(CD206) in immunity to ricin [J].Toxins (Basel),2011,3(9):1131-1145.
    [64] Leek M D, Griffiths G D, Green M A. Intestinal pathology followingintramuscular ricin poisoning [J].Pathol,1989,159(4):329-334.
    [65] Mantis N J, Morici L A, Roy C J. Mucosal vaccines for biodefense [J].Curr TopMicrobiol Immunol,2012,354:181-195.
    [66] Neal L M, McCarthy E A, Morris C R, et al. Vaccine-induced intestinalimmunity to ricin toxin in the absence of secretory IgA[J].Vaccine,2011,29(4):681-689.
    [67] Sekine I, Kawase Y, Nishimori I, et al. Pathological study on mucosal changes insmall intestine of rat by oral administration of ricin.I Microscopicalobservation[J]. Acta Pathologica Japonica,1986,36(8):1205-1212.
    [68] Smallshaw J E, Richardson J A, Vitetta E S. RiVax, a recombinant ricin subunitvaccine,protects mice against ricin delivered by gavage or aerosol [J].Vaccine,2007,25(42):7459-7469.
    [69] Yoder J M, Aslam R U, Mantis N J. Evidence for widespread epithelial damageand coincident production of monocyte chemotactic protein1in a murinemodel of intestinal ricin intoxication [J].Infect Immun,2007,75(4):1745-1750.
    [70] Mantis N J, McGuinness C R, Sonuyi O, et al. Immunoglobulin Aantibodiesagainst ricin A and B subunits protect epithelial cells from ricin intoxication[J].Infect Immun,2006,74(6):3455-3462.
    [71] Jandhyala D M, Ahluwalia A, Obrig T, et al. ZAK: a MAP3Kinase thattransduces Shiga toxin-and ricin-induced proinflammatory cytokine expression[J].Cell Microbiol,2008,10(7):1468-1477.
    [72] Kioka N, Minami K, Tamura A, Yoshikawa N. Chemokine expression in humanastrocytes in response to shiga toxin2[J]. Int J Inflam,2012,135803
    [73] Mallick EM, McBee ME, Vanguri VK, et al.A novel murine infection model forShiga toxin-producing Escherichia coli[J]. J Clin Invest,2012,122(11):4012-4024.
    [74] Yamasaki C, Nishikawa K, Zeng X T, et al. Induction of cytokines by toxinsthat have an identical RNA N-glycosidase activity: Shiga toxin, ricin, andmodeccin [J].Biochim Biophys Acta,2004,1671(1-3):44-50.
    [75] Friswell M, Campbell B, Rhodes J.The role of bacteria in the pathogenesis ofinflammatory bowel disease [J]. Gut Liver,2010,4(3):295-306
    [76] Smith D C, Lord J M, Roberts L M, et al.1st class ticket to class I: proteinstoxins as pathfinders for antigen presentation [J].Traffic,2002,3(10):697-704.
    [77] Day P J, Ernst S R, Frankel A E, et al. Structure and activity of an active sitesubstitution of ricin A chain [J].Biochemistry,1996,35(34):11098-11103.
    [78] Smith D C, Gallimore A, Jones E, et al. Exogenous peptides delivered by ricinrequire processing by signal peptidase for transporter associated with antigenprocessing-independent MHC class I-restricted presentation [J].The Journal ofImmunology,2002,169(1):99-107.
    [79] Grimaldi E, Claassen E A W, Lord J M, et al. Stimulation ofpneumonvirus-specifc CD8+T-cells using a non-toxic recombinant ricindelivery system [J].Mol. Immunol,2007,44(5):993-998.
    [80] Smith D C, Marsden C J, Lord J M, et al. Expression, purification andcharacterization of ricin vectors used for exogenous antigen delivery into theMHC class I presentation pathway [J].Biol,2003,5:13-19.
    [81] Medina-Bolivar F, Wright R, Funk V, et al. A non-toxic lectin for antigendelivery of plant-based mucosal vaccines [J].Vaccine,2003,21(9-10):997-1005.
    [82] Choi N W, Estes M K, Langridge W H R. Synthesis of a ricin toxin Bsubunit-rotavirus VP7fusion protein in potato[J].Mol. Biotechnol,2006,32(2):117-128.
    [83] Choi N W, Estes M K, Langridge W H R. Ricin toxin B subunit enhancement ofrotavirus NSP4immunogenicity in mice [J].Viral Immunol,2006,19(1):54-63.
    [84] Choi N, Estes M K, Langridge W H R. Mucosal immunization with a ricin toxinB-subunit-rotavirus NSP4fusion protein stimulates a Th1lymphocyte response[J]. Biotechnol,2006,121(2):272-283.
    [85] Zeng Q, Chen S, You Z, et al. Hepatocy te growth factor inhibit s anoikis in headand neck squamo us cell car cinoma cells by activation of ERK and Aktsignaling independent of NF-kappa B [J].J Bio l Chem,2002,277(28):25203-25208.
    [86] Summy JM, Ga llick GE. Treatm ent fo r advanced tumors: SRC reclaims centerstage [J]. C lin Cancer Res,2006,12(5):1398-1401.
    [87] Jiang Y, Ma Z, Xin G, et al. Th1and Th2immune response in chronic hepatitisB patients during a long-term treatment with adefovir dipivoxil [J].MediatorsInflamm,2010,143026.
    [88] Chakera A, Bennett SC, Cornall RJ. A whole blood monokine-based reporterassay provides a sensitive and robust measurement of the antigen-specific T cellresponse[J]. J Transl Med,2011,9:143.
    [89] Bruck R, Aeed H, Brazovsky E, et al. Allicin, the active component of garlic,prevents immune-mediated, concanavalin A-induced hepatic injury in mice[J].Liver Int,2005,25(3):613-621.
    [90] Wang Y, Cui X, Tai G, et al.A critical role of activin A in maturation of mouse peritonealmacrophages in vitro and in vivo[J]. Cell Mol Immunol,2009,6(5),387-6392.
    [91] Gordon S.Pattern recognition receptors: doubling up for the innate immuneresponse[J]. Cell,2002,111(7),927-930.
    [92] Bolander Jr FF.The role of nitric oxide in the biological activity of prolactin inthe mouse mammary gland[J]. Mol Cellular Endocrinol,2001,174(1-2),91-98.
    [93] Kozlowska K, Cichorek M, Wachulska M, et al.Role of interleukins and nitricoxide secretion by peritoneal macrophages in differential tumoricidal effect totransplantable melanomas as regarding their biological properties.Immunopharmacol [J]. Immunotoxicol,2006,28(2),305-317.
    [94] Renna NF, Diez ER, Lembo C.Role of cox-2in vascular inflammation: anexperimental model of metabolic syndrome[J]. Miatello RM MediatorsInflamm.,2013,513251.
    [95] Huang Z, Hoffmann F W, Fay J D, et al. Stimulation of unprimed macrophageswith immune complexes triggers a low output of nitric oxide by calcium-dependent neuronal nitric-oxide synthase[J].J Biol Chem,2012,287(7):4492-502.
    [96] Zoccal K F, Bitencourt Cda S, et al. Tityus serrulatus venom and toxins Ts1, Ts2and Ts6induce macrophage activation and production of immune mediators[J].Toxicon,2011,57(7-8),1101-1108.
    [97] Baugh J A, Bucala R. Mechanisms for modulating TNFa in immune andinflammatory disease[J]. Curr Opin Drug Discov Devel,2001,4(5),635-650.
    [98] Ghezzi P,Cerami A.Tumor necrosis factor as a pharmacological target[J]. Mol.Biotechnol.2005,31(3),239-244.
    [99] Zlotnik A, Yoshie O. Chemokines: a new classification system and their role inimmunity[J].Immunity,2000,12(2):121-127.
    [100] Yoshie O, Imai T, Nomiyama H. Chemokines in immunity [J].Adv Immunol,2001,78:57-110.
    [101] Gerard C, Rollins BJ. Chemokines and disease [J].Nat Immunol,2001,2(2):108-115.
    [102] Franco LH, Beverley SM, Zamboni DS.Innate immune activation andsubversion of Mammalian functions by leishmania lipophosphoglycan [J]. JParasitol Res,2012,165126.
    [103] Shishodia S, Shrivastava A, Sodhi A. Protein kinase C: a potential pathway ofmacrophage activation with cisplatin[J]. Immunol Lett.1998,61(2-3),179-186.
    [104] Robinson M J, Cobb M H. Mitogen-activated protein kinase pathways [J]. CurrOpin Cell Biol,1997,9(2),180–186.
    [105] Lewis TS, Shapiro P S, Ahn N G. Signal transduction through MAP kinasecascades [J]. Adv Cancer Res,1998,74,49–139.
    [106] Angel P, Karin M. The role of Jun, Fos and AP-1complex in thecell-proliferation and transformation[J]. Biochem Biophys Acta,1991(2-3),1072,129–157.
    [107] Chen C C, Wang J K. P38but not p44/42mitogen-activated protein kinase isrequired for nitric oxide synthase induction mediated by lipopolysaccharide inRAW264.7macrophages. Mol Pharmacol[J],1999,55(3),481-488.
    [108] Yoon Y D, Kang J S, Han S B, et al. Activation of mitogen-activated proteinkinases and AP-1by polysaccharide isolated from the radix of Platycodongrandiflorum in RAW264.7cells[J]. Int Immunopharmacol,2004,4(12),1477-1487.
    [109] Mendes A F, Carvalho A P, Caramona M M, et al. Role of nitric oxide in theactivation of NF-kappaB, AP-1and NOS II expression in articularchondrocytes[J]. Inflamm Res,2002,51(7),369-375.
    [110] Connelly L, Palacios-Callender M, Ameixa C, et al. Biphasic regulation ofNF-kappa B activity underlies the pro-and anti-inflammatory actions of nitricoxide[J]. J Immunol,2001,166(6),3873-3881.
    [111] Liu S F,Malik A B.NF-κB activation as a pathological mechanism of septicshock and inflammation [J].Am J Physiol Lung Cell Mol Physiol,2006,290(4):622-645
    [112] Eason SW, Martin W.Involvement of tyrosine kinase and protein kinase C inthe induction of nitric oxide synthase by LPS and IFN-γ in J774macrophages[J]. Arch Int Pharmacodyn,1995,330(2),225–236.
    [113] Liao X, Su J, Mrksich M. An adaptor domain-mediated autocatalytic interfacialkinase reaction [J]. Chemistry,2009,15(45):12303-12309

© 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