膜连蛋白Ⅰ参与地塞米松上调A549细胞吞噬凋亡的嗜酸性粒细胞
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
     近年来,嗜酸性粒细胞被认为是哮喘发病机制中的主要效应细胞。由于嗜酸性粒细胞侵入到支气管粘膜以及随后所释放的大量的毒性物质,因此常常引起支气管上皮细胞的损害,从而引起支气管的高反应性。另一方面,支气管上皮细胞也有吞噬凋亡的嗜酸性粒细胞的能力,这对炎症的消散有着重要的意义。这个过程受着许多因素的调节,这些调节因素可能有着重要的临床治疗意义。
     膜连蛋白Ⅰ是13个结构相关的钙依赖的膜连接蛋白Annexin超家族成员之一。Fan等研究发现膜联蛋白Ⅰ作为配体和受体促进巨噬细胞吞噬人外周血的淋巴细胞。另外,膜联蛋白Ⅰ存在于II型肺泡细胞系A549细胞的表面,地塞米松增加A549细胞表面膜联蛋白Ⅰ的表达,由此我们推测膜联蛋白Ⅰ可能参与地塞米松上调A549细胞吞噬凋亡的嗜酸性粒细胞的过程。
     目的
     在这次研究中,我们观察了A549细胞吞噬凋亡的嗜酸性粒细胞的动态过程,并且探讨了膜连蛋白I是否参与地塞米松上调A549细胞吞噬凋亡的嗜酸性粒细胞的过程,以及凋亡的嗜酸性粒细胞的吞噬对A549细胞分泌IL-6和IL-8的影响。
     方法
     1.嗜酸性粒细胞的分离和凋亡
     用红细胞裂解液去除红细胞后,利用磁性活化的细胞分离柱和CD15、CD16的磁珠抗体进行分离人外周血的嗜酸性粒细胞。纯化的嗜酸性粒细胞在培养箱中培养48h进行老化凋亡,然后用膜连蛋白V-FITC凋亡试剂盒进行凋亡评估。
     2. A549细胞吞噬凋亡的嗜酸性粒细胞的观察
     将A549细胞放入到热台系统中,显微镜下调整好界面。此时将凋亡的嗜酸性粒细胞从小孔中加入到35mm培养皿中,每隔一分钟自动拍摄吞噬过程。对吞噬的A549细胞在MetaMorph图像分析软件中进行周长、面积和密度的测量。GFP转染的A549吞噬7-AAD标记的凋亡的嗜酸性粒细胞用激光共聚焦拍摄。
     3.A549细胞吞噬凋亡的嗜酸性粒细胞的定量观察
     吞噬试验前,把长满A549细胞的玻片转移到用Parafilm膜包被好的12孔板中。把凋亡的嗜酸性粒细胞悬浮到DMEM中,加入到玻片上(1×104细胞/板),在37°C中孵育60分钟。吞噬结束后,用EDTA-PBS清洗以除去未吞噬的凋亡的嗜酸性粒细胞后,接着用4%的多聚甲醛固定,o-dianisidine进行染色,进行计数。
     3.膜连蛋白I的Western blot分析
     为了提供足够的A549细胞以测量A549细胞表面的膜连蛋白I,A549细胞接种于100mm的培养皿中培养96h,弃去培养基,用EDTA(1mmol/ml)洗A549细胞,收集上清蛋白,接着进行Western Blot分析。
     4.细胞因子的测定
     用地塞米松处理或未处理的A549细胞在48孔板中单独孵育或与凋亡的嗜酸性粒细胞共同孵育,收集上清。用IL-6和IL-8放射免疫试剂盒分析上清中的IL-6和IL-8水平。TGF-β1由ELISA试剂盒测定。
     5.统计方法
     所有试验分5次独立完成,数据用均数±标准误表示。组间的差异用单因素的方差分析,而两组之间的差异用t检验,统计在SPSS10.0软件统计包中进行,P<0.05表示差异有显著性。
     结果
     1.动态观察A549细胞吞噬凋亡的嗜酸性粒细胞
     自动地拍摄A549细胞吞噬凋亡的嗜酸性粒细胞的动态的过程。对吞噬前30分钟、吞噬时、吞噬后60分钟的A549细胞的周长、面积和密度进行测量发现,三个观察的指标在吞噬前30分钟、吞噬时、吞噬后60分钟差异有显著性,而在吞噬时和吞噬后60分钟三个观察指标并无明显的改变。激光共聚焦也证实了吞噬现象。
     2.地塞米松促进A549细胞吞噬凋亡的嗜酸性粒细胞在不同的时间内预先加入地塞米松后,A549细胞吞噬能力的发生了变化。在地塞米松孵育1小时开始吞噬功能增加,4小时达到最高峰为55%±2.2% ,而到24小时又降到38%±1.4%,但都显著高于对照组。
     3.地塞米松增加A549细胞表面膜连蛋白I的表达
     蛋白质印迹试验分析了地塞米松增加A549细胞表面膜连蛋白I的表达的作用。其密度值在2-4小时为最强,24小时降至正常范围,显示出地塞米松以时间依赖的方式增加A549细胞表面膜连蛋白I的表达。
     4.膜连蛋白I的单克隆抗体抑制地塞米松对A549细胞吞噬凋亡的嗜酸性粒细胞的增加作用
     为了探讨A549细胞表面膜连蛋白I对吞噬功能的影响,我们把膜连蛋白I单克隆抗体(10μg/ml)加入到经地塞米松在不同时间处理的A549细胞中进行孵育(30 min at 37°C),结果显示经地塞米松和单克隆抗体同时处理的A549细胞的吞噬能力明显地低于单用地塞米松处理的A549细胞的吞噬能力,差异有显著性。
     5.凋亡的嗜酸性粒细胞的表面膜连蛋白I参与了A549细胞的吞噬过程
     凋亡的嗜酸性粒细胞用EDTA洗以除去膜表面的膜连蛋白I,然后加入到A549细胞中,结果表明抑制了A549细胞的吞噬能力,与不处理组比较差异有显著性。
     6.地塞米松增加的A549细胞吞噬并不激发A549细胞分泌炎症因子的作用
     凋亡的嗜酸性粒细胞的吞噬和地塞米松对吞噬能力的增加并不能激发IL-6 IL-8和TGF-β1的释放。
     结论1.A549细胞在吞噬凋亡的嗜酸性粒细胞后其周长、面积和密度发生了变化,
     周长和面积缩小,而密度增加。
     2.地塞米松通过膜连蛋白I来增加A549细胞吞噬凋亡的嗜酸性粒细胞的能力。
     3.地塞米松增加的A549细胞吞噬并不激发A549细胞分泌炎症因子的作用。
Background
     Eosinophils are recently confirmed as one of the major effector cells in the immuno-inflammation changes in the airway in asthma. By infiltrating into the bronchial mucosa and subsequent release of histotoxic substances, eosinophils may cause epithelial damage, an event thought to be important in the development of airway hyperresponsiveness. On the other hand, the bronchial epithelial cells have the ability to engulf the apoptotic eosinophils, which may be crucial to the solution of inflammation. The process is regulated by many factors and the regulators of this process may have therapeutically potential.
     Annexin I is a member of annexin superfamily of 13 structurally related proteins that are able to bind to cell membrane in a calcium-dependent manner. Interestingly Annexin I may also be an endogenous ligand mediating the engulfment of apoptotic cells. Fan et al found that annexin I served as both ligand and receptor in promoting the phagocytosis of Jurkat T lymphocytes and human peripheral T lymphocytes by macrophages. In addition, annexin I is expressed on the membrane surface of A549 cell and is up-regulated by dexamethasone, indicating that annexin I may be involved in the dexamethasone’s upregulation of the engulfment of apoptotic eosinophils in A549 cell.
     Objective
     In the present study, we observed the real time process of phagocytosis of apoptotic eosinophils by A549 cells and investigated whether Annexin I was involved in the dexamethasone-mediated upregulation of phagocytosis of apoptotic eosinophils by A549 cells. In the meanwhile, we investigated the effect of phagocytosis of apoptotic eosinophils on IL-6 and IL-8 release in A549 cells.
     Method
     1. Eosinophil isolation and apoptosis induction
     After removal of red blood cells using red blood cell lysis buffer, the eosinophils were isolated by micromagnetic beads coated with anti-CD16, anti-CD15 and the magneticactivated separation column. Flow cytometric analysis assessed the apoptosis eosinophils rate after culturing for 48 hours.
     2. Real time process of phagocytosis of apoptotic eosionphils by A549 cells.
     A549 Cells were put into the heated-plate system. After this, apoptotic eosinophils were put into the dishes through the hole. The whole process of phagocytosis was automatically recorded every one minute. The curve length, area and density of A549 cells before and after phagocytosis were measured by MetaMorph software.A549 cells transfected by GFP engulf apoptotic eosinophils labled with 7-AAD, which was scanned by laser confocal scanning microscope.
     3. Quantitative observation of phagocytosis of apoptotic eosionphils by A549 cells.
     The coverslip with A549 cells were transferred to new 12-well plates coated with Parafilm membrane one by one. Washed apoptotic eosinophils were resuspended in DMEM and added to the coverslip (1×104/slide) with A549 cells for 60 minutes at 37°C under 5%CO2. After interaction, the noningested eosinophils were washed using oscillator with Ca2+ and Mg2+ free PBS solution supplemented with EDTA before fixation. The ingested eosinophils were visualized by o-dianisidine staining. At least 200 A549 cells which were conter-stained with haematoxylin in duplicate wells in randomly selected fields were counted by a blinded investigator, and the proportion of ingested eosinophils was expressed as a percentage.
     4. Western blotting analysis of annexin I
     In order to provide a sufficient number of cells for measurement of cell surface annexin I induction, subconfluent A549 cells were seeded in DMEM medium in 100mm dishes. The supernatants washed with EDTA were collected and followed by western blot analysis.
     5. Cytokine production
     A549 cells unstimulated or stimulated with dexamethasone (10-6 M) were incubated in DMEM in 48 wells plates alone or togather with apoptotic eosinophils. IL-6 and IL-8 were assayed in supernatants from cocultures of A549 cells and apoptotic eosinophils by RIA kit according to the manufacturer’s instruction. Concentration of TGF-β1 was measured by ELISA.
     6. Statistical method
     All experiments were performed for five independent times. Data were expressed as the means±standard error and were analyzed for significant differences by unpaired Student’s t test and one-way ANOVA with SPSS 10.0. Differences were considered statistically significant if P value <0.05.
     Result
     1. The curve length, area, and density of A549 cells were changed after phagocytosis
     The whole process of phagocytosis of apoptotic eosinophils by A549 cells was automatically recorded every one minute. The curve length, area and density of A549 cells before and after phagocytosis were measured by MetaMorph software. The results showed that the curve length and area of A549 cells were reduced after phagocytosis (P<0.001), while the density of A549 cells were increased (P<0.001).Phagocytosis was further confirmed by laser confocal scanning microscope.
     2. Dexamethasone promotes engulfment of apoptotic eosinophils by A549
     A549 epithelial cells recognized and ingested apoptotic eosinophils that were visualized as dark-staining peroxidase-positive cells. Effects of dexamethasone stimulation with various duration of preincubation on the engulfment of apoptotic eosinophils by A549 cells showed that the effect was not observed at shorter incubation time (< 1 h), but was evident at longer incubation time. The capacity of engulfment reached the highest at the 4h of preincubation (55%±2.2%) and reduced to the 38%±1.4% at the 24h of preincubation which was still significantly higher than that in control (0h).
     3. Dexamethasone treatment increased annexin I on cell surface of A549
     Western blot analysis showed the effect of dexamethasone treatment on the expression of annexin I on cell surface of A549. The result showed that the intensity was maximal at 4h after dexamethasone treatment and returned to the basal levels at 24h. Correlation analysis shown that the amount of cell surface annexin I was positively correlated with the capacity of engulfment induced by dexamethasone (r = 0.779, p<0.001).
     4. Increased phagocytosis of apoptotic eosinophils by A549 cells stimulated by treatment with dexamethasone was inhibited by annexin I mAb
     To investigate the functional contributions of A549 cell surface annexin I, we tried to use the annexin I mAb to inhibit the phagocytosis of apoptotic eosinophils by A549 cells. The capacity of phagocytosis of A549 cells treated with dexamethasone and annexin I mAb (10μg/ml; 30 min at 37°C) was compared with that of A549 cells treated with dexamethasone only, previously shown to significantly enhance phagocytosis. The results showed that pretreatment with dexamethasone and annexin I mAb downregulated the phagocytosis of apoptotic eosinophils.
     5. Annexin I expressed on apoptotic eosinophil surface was involved in its phagocytosis by A549
     Apoptotic eosinophils were washed with EDTA to remove annexin I before presented to A549 cells. The result showed removal of annexin I with EDTA and preincubation with annexin I mAb inhibited phagocytosis (p<0.001).
     6. Dexamethasone-promoted uptake of apoptotic eosinophils did not change the proinflammatory factor secretion in A549 cells
     There was no difference in the release of proinflammatory factors and TGF-β1 between the A549 cells stimulated with apoptotic eosinophils and non-stimulated A549 cells.
     Conclusion
     1. The curve length, area, and density of A549 cells were changed after phagocytosis. The curve length and area of A549 cells were reduced after phagocytosis, while the densities of A549 cells were increased.
     2. Dexamethasone promotes engulfment of apoptotic eosinophils by A549 through annexin I.
     3. Dexamethasone-promoted uptake of apoptotic eosinophils did not change the proinflammatory factor secretion in A549 cells.
引文
1. Cookson B. The alliance of genes and environment in asthma and allergy. Nature 1999; 402:BB5-11
    2. Holt PG, Macaubas C, Stumbles PA, et al. The role of allergy in the development of asthma. Nature 1999; 402: B12-7.
    3. Oettgen HC, Geha RS. IgE in asthma and atopy: cellular and molecular connections. J Clin Invest 1999; 104:829-35.
    4. Humbert M, Menz G, Ying S., et al. The immunopathology of extrinsic (atopic) and intrinsic (no-atopic) asthma: more similarities than differences. Immunol today 1999; 20:528-33
    5. Lee, J J, Dimina D, Macias MP, et al. Defining a link with asthma in mice congenitally deficient in eosinophils. Science 2004; 305: 1773
    6. Rothenberg ME. Eosinophilia. N Engl J Med 1998; 338:1592–1600.
    7. Strek M E and Leff AR. Eosinophils. In Barnes P J, Grunstein M M, Leff AR, and Woolcock AJ, editors. Asthma, Vol. 1. Lippincott Raven, New York. 1997; 399–417.
    8. Weller P F. Human eosinophils. J Allergy Clin Immunol 1997; 100:283–287.
    9. Giembycz MA and Lindsay MA. Pharmacology of the eosinophil. Pharmacol Rev 1999; 51:213–340.
    10. Kroegel C, Virchow JC,. Luttmann JW et al. Pulmonary immune cells in health and disease: the eosinophil leucocyte (Part I). Eur Respir J 1994; 7:519–543.
    11. Martin LB, Kita H, Leiferman KM, et al. Eosinophils in allergy: role in disease, degranulation, and cytokines. Int Arch Allergy Immunol 1996;109:207–215.
    12. Weller PF, Lim K, Wan HC, et al. Role of the eosinophil in allergic reactions. Eur Respir J Suppl. 1996; 22:109s–115s.
    13. Erjef?lt JS, Erjef?lt I, Sundler F,et al. Microcirculation-derived factors in airway epithelial repair in vivo. Microvasc Res 1994; 48:161–178.
    14. Wong DTW, Donoff RB, Yang J, et al. Sequential expression of TGF-α and TGF-β
    1 by eosinophils during cutaneous wound healing in the hamster. Am J Pathol 1993; 143:130–142
    15. Erjef?lt JS, Sundler F, and Persson CGA. Eosinophils, neutrophils, and venular gaps in the airway mucosa at epithelial removal–restitution. Am J Respir Crit Care Med 1996; 153:1666–1674.
    16. Gleich GJ,Motojima S, Frigas E, et al. The eosinophilic leukocyte and the pathology of fatal bronchial asthma: evidence for pathologic heterogeneity. J Allergy Clin Immunol 1987; 80:412–415.
    17. Venge P, and H?kansson L. Eosinophils. In Kaliner MA, Barnes PJ, and Persson CGA, editors. Asthma: Its Pathology and Treatment. Marcel Dekker, New York. 1991; 477–502.
    18. Fahy JV, Liu J, Wong H, et al. Cellular and biochemical analysis of induced sputum from asthmatic and from healthy subjects. Am Rev Respir Dis 1993; 147:1126–1131.
    19. Jeffery PK, Godfrey RW, Adelroth E, et al. Effects of treatment on airway inflammation and thickening of basement membrane reticular collagen in asthma: a quantitative light and electron microscopic study. Am Rev Respir Dis 1992; 145:890–899.
    20. Erjef?lt JS, Andersson M, Greiff L, et al. Cytolysis and piecemeal degranulation as distinct modes of activation of airway mucosal eosinophils. J Allergy Clin Immunol 1998; 102:286–294.
    21. Kato M, Kephart GM, Talley NJ, et al. Eosinophil infiltration and degranulation in normal human tissue. Anat. Rec. 1998; 252:418–425.
    22. Korsgren M, Erjef?lt JS, Korsgren O, et al. Allergic eosinophil-rich inflammation develops in lungs and airways of B cell-deficient mice. J Exp Med 1997; 185:885–892.
    23. Erjef?lt JS, Korsgren M, Nilsson MC, et al. Association between inflammation and epithelial damage-restitution processes in allergic airways in vivo. Clin Exp Allergy 1997; 27:1345–1357.
    24. Yamaguchi Y, Suda T, Ohta S, et al. Analysis of the survival of mature human eosinophils: interleukin-5 prevents apoptosis in mature human eosinophils. Blood 1991; 78:2542–2547.
    25. Stern M, Meagher L, Savill J, et al. Apoptosis in human eosinophils: programmed cell death in the eosinophil leads to phagocytosis by macrophages and is modulated by IL-5. J Immunol 1992; 148:3543–3549.
    26. Persson CGA, Erjef?lt JS. Eosinophil lysis and free granules: an in vivo paradigm for cell activation and drug development. Trends Pharmacol Sci 1997. 18:117–123.
    27. Stern M, Meagher L, Savill J, et al. Apoptosis in human eosinophils: Programmed cell death in the eosinophil leads to phagocytosis by macrophages and is modulated by IL-5. J Immunol 1992;148:3543
    28. Stern M, Savill J, Haslett C. Human monocyte-derived macrophage phagocytosis of senescent eosinophils undergoing apoptosis. Am J Pathol 149:911, 1996
    29. Walsh GM, Sexton DW, Blaylock MG, Convery CM. Resting and cytokine-stimulated human small airway epithelial cells recognize and ingest apoptotic eosinophils. Blood 1999; 94:2827-35.
    30. Chalfie M Y, Euskirchen Tu G, Ward W W, Prasher D C. Green fluorescent protein as a marker for gene expression.Science1994; 263:802–805.
    31. Adie EJ, Kalinka S, Smith L, Francis MJ, Marenghi A, Cooper ME , Briggs M, Michael NP, Milligan G, Game S. A pH-sensitive fluor, CypHer 5, used to monitor agonist-induced G protein-coupled receptor internalization in live cells.Biotechniques 2002; 33:1152–1154, 1156–1157.
    32. Eils R, Athale C. Computational imaging in cell biology. The Journal of Cell Biology.2003;05:477-481
    33. Yamamoto N, Yang M, Jiang P, Xu M, Tsuchiya H, Tomita K, Moossa AR, Robert Hoffman M. Real-time imaging of individual fluorescent-protein color-coded metastatic colonies in vivo. Clinical & Experimental Metastasis 2003; 20: 633–638
    34. Stence N, Waite M, Dailey ME. Dynamics of microglial activation: a confocal time-lapse analysis in hippocampal slices. Glia 2001; 33:256-266
    35. Maderna P, Yona S, Perertti M, et al, Modulation of Phagocytosis of Apoptotic Neutrophils by Supernatant from Dexamethasone-Treated Macrophages and Annexin-Derived Peptide Ac2-26. The Journal of Immunology 2005; 174(6): 3727-3733
    1. Wills-Karp M, Karp CL. Eosinophils in Asthma: Remodeling a Tangled Tale. Science 2004; 305(17): 1726-1728.
    2. Lee JJ, Dimina D, Macias MP, et al., Defining a Link with Asthma in Mice Congenitally Deficient in Eosinophils. Science 2004; 305(5691): 1773-1776.
    3. Humbles, A.A., Lloyd CM, McMillan SJ et al., A Critical Role for Eosinophils in Allergic Airways Remodeling. Science 2004; 305(5691): 1776-1779.
    4. Walsh G.M, Eosinophil apoptosis: mechanisms and clinical relevance in asthmatic and allergic inflammation. Br J Haematol 2000; 111: 61-67.
    5. Walsh, G.M., Sexton DW, Blaylock MG et al, Resting and Cytokine-Stimulated Human Small Airway Epithelial Cells Recognize and Engulf Apoptotic Eosinophils. Blood 1999; 94(8): 2827-2835.
    6. Wardlaw AJ. Molecular basis for selective eosinophil trafficking in asthma: a multistep paradigm. Journal of Allergy and Clinical Immunology 1999;104: 917-926.
    7. Duncan CJA, Lawrie A, Blaylock MG, et al, Reduced eosinophil apoptosis ininduced sputum correlates with asthma severity. European Respiratory Journal 2003; 22(3): 484-490
    8. Erjef?lt JS, Andersson M, Greiff L, et al. Cytolysis and piecemeal degranulation as distinct modes of activation of airway mucosal eosinophils. J Allergy Clin Immunol 1998; 102: 286-294
    9. Woolley KL, Gibson PG., Carty K, et al. Eosinophil apoptosis and the resolution of airway inflammation in asthma. Am J Respir Crit Care Med 1996; 154: 237–243.
    10. Erjef?lt JS, Greiff L, Matsson E, et al. Lack of apoptotic airway tissue eosinophils in vivo, even in steroid-treated allergic disease. Eur Respir J 1998; 12: 57-67
    11. Kodama T, Matsuyama T , Miyata S, et al. Kinetics of apoptotis in the lung of mice with allergic airway inflammation. Clin Exp Allergy 1998; 28: 1435-1443.
    12. Schneider TvV, Moqbel D, Issekutz R. Kinetics and quantitation of eosinophil and neutrophil recruitment to allergic lung inflammation in aBrown Norway rat model. Am J Respir Cell Mol. Biol, 1997. 17: 702-712.
    13. Erjef?lt JS, Korsgren M, Nilsson MC, et al. Association between inflammation and epithelial damage-restitution processes in allergic airways in vivo. Clin Exp Allergy 1997; 27: 1345-1357
    14. Erjefa¨lt JS, Malm-Erjefa¨lt M, Persson CG.. Rapid and efficient clearance of airway tissue granulocytes through transepithelial migration. Thorax 2004; 59: 136-143
    15. Miller LA, Butcher EC. Human airway epithelial monolayers promote selective transmigration of memory T cells: a transepithelial model of lymphocyte migration into the airways. Am J Respir Cell Mol. Biol 1998; 19: 892–900
    16. Kato M, Kephart G.M, Talley NJ, et al. Eosinophil infiltration and degranulation in normal human tissue. Anat Rec 1998; 252: 418-425
    17. Persson CGA, Erjef?lt JS. “Ultimate activation” of eosinophils in vivo: lysis andrelease of clusters of free eosinophil granules (Cfegs). . Thorax 1997; 52: 569-574
    18. Savill J, Fadok V. Corpse clearance defines the meaning of cell death. Nature 2000; 407: 784-788
    19. Stern M, Savill J, Haslett C. Human monocyte-derived macrophage phagocytosis of senescent eosinophils undergoing apoptosis. Mediation by alpha v beta 3/CD36/thrombospondin recognition mechanism and lack of phlogistic response. Am J Pathol 1996; 149: 911-921
    20. Sexton D, Blaylock MG, Walsh GM. Human alveolar epithelial cells engulf apoptotic eosinophils by means of integrin- and osphatidylserinereceptor-dependent mechanisms: a process upregulated by dexamethasone. J Allergy Clin Immunol 2001; 108: 962-969
    21. Walsh GM, Sexton DW, Blaylock MG. Corticosteroids, eosinophils and bronchial epithelial cells: new insights into the resolution of inflammation in asthma. Journal of Endocrinology, 2003; 178(1): 37-43
    22. Frey BM, Reber BFX, Vishwanath BS, et al., Annexin I modulates cell functions by controlling intracellular calcium release. The FASEB Journal 1999; 13(15): 2235-2245
    23. Traverso V, Morris JF, Flower RJ, et al. Lipocortin 1 (annexin 1) in patches associated with the membrane of a lung adenocarcinoma cell line and in the cell cytoplasm. Journal of Cell Science, 1998; 111(10): 1405-1418
    24. Arur S, Uche UE, Rezaul K , et al. Annexin I is an endogenous ligand that mediates apoptotic cell engulfment. Dev Cell 2003; 4: 587-598
    25. Fan XX, Krahling S, Smith D, et al. Macrophage Surface Expression of Annexins I and II in the Phagocytosis of Apoptotic Lymphocytes. Molecular Biology of the Cell 2004; 15(6): 2863-2872.
    26. Croxtall JD, Flower RJ. Lipocortin 1 Mediates Dexamethasone-Induced Growth Arrest of the A549 Lung Adenocarcinoma Cell Line. Proceedings of the NationalAcademy of Sciences 1992; 89(8):3571-3575
    27. Scannell M, Maderna P. Lipoxins and annexin-1: resolution of inflammation and regulation of phagoctosis of apoptotic cells. ScientificWorldJournal 2006; 6: 1555-1573
    28. Oliani SM, Damazo AS, Perretti M. Annexin 1 localisation in tissue eosinophils as detected by electron microscopy. Mediators Inflamm 2002; 11: 287-292
    29. Platt N. Gordon S. Is the class A macrophage scavenger receptor (SR-A) multifunctional? — The mouse’s tale. J Clin Invest 2001; 108: 649–654
    30. Krieger M. Scavenger receptor class B type I is a multiligand HDL receptor that influences diverse physiologic systems. J Clin Invest 2001; 108: 793–797
    31. Savill J, Fadok V. Corpse clearance defines the meaning of cell death. Nature 2000; 407: 784–788
    32. Fadok, V.A., Bratton DL, Henson PM. Bratton, and P.M. Henson, Phagocyte receptors for apoptotic cells: recognition, uptake, and consequences. Journal of Clinical Investigation 2001; 108(7): 957-962
    33. Fadok VA, Bratton DL, Frasch SC, et al. The role of phosphatidylserine in recognition of apoptotic cells by phagocytes see comments. Cell Death Differ 1998; 5: 551–562
    34. Fadok VA, de Cathelineau A, Daleke DL, et al. Loss of phospholipid asymmetry and surface exposure of phosphatidylserine is required for phagocytosis of apoptotic cells by macrophages and fibroblasts. J Biol Chem 2001; 276: 1071–1077
    35. Daleke DL, Lyles JV. Identification and purification of aminophospholipid flippases. Biochim Biophys Acta 2000; 1486: 108–127
    36. Bevers EM, Comfurius P, Dekkers DW, et al. Lipid translocation across the plasma membrane of mammalian cells. Biochim Biophys Acta 1999; 1439: 317–330
    37. Sims PJ, Wiedmer T. Unraveling the mysteries of phospholipid scrambling. Thromb Haemost 2001; 86: 266–275
    38. Hamon Y. ABC1 promotes engulfment of apoptotic cells and transbilayer redistribution of phosphatidylserine. Nat Cell Biol 2000; 2: 399-406
    39. Marguet D, Luciani MF, Moynault A, et al. Engulfment of apoptotic cells involves the redistribution of membrane phosphatidylserine on phagocyte and prey. Nat Cell Biol, 1999; 1: 454–456
    40. Callahan MK, Williamson P, Schegel, RA. Surface expression of phosphatidylserine on macrophages is required for phagocytosis of apoptotic thymocytes. Cell Death Differ 2000; 7: 645–653
    41. Taylor PR, Carugati A, Valerie A et al. A hierarchical role for classical pathway complement proteins in the clearance of apoptotic cells in vivo. J Exp Med 2000; 192: 359–366
    42. Ogden CA, deCathelineau A, Hoffmann PR, et al. C1q and mannose binding lectin (MBL) engagement of cell surface calreticulin and CD91 to initiates macropinocytosis and uptake of apoptotic cells. J Exp Med 2001; 194: 781–795
    43. Schagat TL, Wofford JA, Wright JR. Surfactant protein A enhances alveolar macrophage phagocytosis of apoptotic neutrophils. J Immunol 2001; 166: 2727–2733
    44. Franc NC, Heitzler P, Ezekowitz RA, et al. Requirement for croquemort in phagocytosis of apoptotic cells in Drosophila. Science 1999; 284: 1991–1994
    45. Zhou Z, Hartwieg E, Hrovitz HR. CED-1 is a transmembrane receptor that mediates cell corpse engulfment in C. elegans. Cell 2001; 104: 43–56
    46. Pearson AM. Scavenger receptors in innate immunity. Curr Opin Immunol 1996; 8: 20–28
    47. Hall SE, Lim S, Witherden IR, et al. Lung type II cell and macrophage annexin Ⅰrelease: differential effects of two glucocorticoids. The American Journal of Physiology 1999; 276(1): 114-121
    48. Croxtall JD, Choudhury Q, Newman S, et al. Lipocortin 1 and the control ofcPLA2 activity in A549 cells. Biochemistry Pharmacology 1996; 52:351-356
    49. Morand EF, Hall P, Hutchinson P, et al. Regulation of annexin I in rheumatoid synovial cells by glucocorticoids and interleukin-1. Mediators Inflamm 2006; 2006: 73835
    50. Parente L, Solito E. Annexin I: more than an anti-phospholipase protein. Inflamm Res 2004; 53: 125-132
    51. Podgorski MR, Goulding NJ, Hall ND, et al. Autoantibodies to lipocortin-1 are associated with impaired glucocorticoid responsiveness in rheumatoid arthritis. J Rheumatol 1992; 19: 1668-1671
    52. Maderna P, Yona S, Perretti M, et al, Modulation of Phagocytosis of Apoptotic Neutrophils by Supernatant from Dexamethasone-Treated Macrophages and Annexin-Derived Peptide Ac2-26. The Journal of Immunology, 2005; 174(6): 3727-3733
    53. Hannon RC, Getting JD, Roviezzo SJ, et al. Aberrant inflammation and resistance to glucocorticoids in Annexin 1-/- Mouse. The FASEB Journal 2003: p. 02-0239fje.
    54. Bandeira-Melo C, Bonavita AG, Diaz BL, et al. A novel effect for annexin 1-derived peptide ac2-26: reduction of allergic inflammation in the rat. J Pharmacol Exp Ther 2005; 313: 1416-22
    55. Gavins FN, Yona S, Kamal AM, et al. Leukocyte antiadhesive actions of annexin 1: ALXR- and FPR-related antiinflammatory mechanisms. Blood 2003; 101: 4140
    1. Cox G., Crossley J, Xing Z. Macrophage engulfment of apoptotic neutrophils contributes to the resolution of acute pulmonary inflammation in vivo. Am J Respir Cell Mol Biol 1995; 12: 232–237
    2. Haslett C, Savill JS, Whyte MKB, et al. Granulocyte apoptosis and the control of inflammation. Phil Trans R Soc Lond B 1994; 345: 327–333
    3. Pradhan D, Krahling S, Williamson P, et al. Multiple systems for recognition of apoptotic lymphocytes by macrophages. Mol Biol Cell 1997; 8: 767–778
    4. Sambrano G., Steinberg D. Recognition of oxidatively damaged and apoptotic cells by an oxidized low density lipoprotein receptor on mouse peritoneal macrophages: role of membrane phosphatidylserine. Proc Natl Acad Sci USA 1995; 92:1396–1400
    5. Platt N, Suzuki H, Kurihara Y, et al. Role for the class A macrophage scavenger receptor in the phagocytosis of apoptotic thymocytes in vitro. Proc Natl Acad Sci USA 1996; 93: 12456–12460
    6. Fukasawa M, Adachi H, Hirota K, et al. SRB1, a class B scavenger receptor, recognizes both negatively charged liposomes and apoptotic cells. Exp Cell Res 1996; 222: 246–250
    7. Luciani MF, Chimini G. Luciani MF, et al. The ATP binding cassette transporter ABC1, is required for the engulfment of corpses generated by apoptotic cell death.Eur Mol Biol Organ J 1996; 15: 226–235
    8. Homburg CHE, de Haas M, von dem Borne AEG..et al. Human neutrophils lose their surface FcgRIII and acquire annexin V binding sites during apoptosis in vitro. Blood 1995; 85: 532–540
    9. Martin SJ, Reutelingsperger CPM, McGahon AJ, et al. Early redistribution of plasma membrane phosphatidylserine is a general feature of apoptosis regardless of the initiating stimulus: inhibition by overexpression of bcl-2 and abl. J Exp Med 1995; 182: 1545–1556.
    10. Verhoven B, Schlegel RA, Williamson P. Mechanisms of phosphatidylserine exposure, a phagocyte recognition signal, on apoptotic T lymphocytes. J Exp Med 1995; 182: 1597–1601
    11. Gregory CD, Flora PK, Bhogal HS, et al. Phagocytic clearance of apoptotic cells: novel pathways identified by monoclonal antibodies to cell surface components. J Cell Biochem Suppl 1995; 19B: 312
    12. Platt N, Gordon S. Is the class A macrophage scavenger receptor (SR-A) multifunctional? — The mouse’s tale. J Clin Invest 2001; 108: 649–654
    13. Krieger M. Scavenger receptor class B type I is a multiligand HDL receptor that influences diverse physiologic systems. J Clin Invest 2001; 108: 793–797
    14. Savill J, Fadok V. Corpse clearance defines the meaning of cell death. Nature 2000; 407: 784–788
    15. Stern M, Savill J, Haslett C. Human monocyte-derived macrophage phagocytosis of senescent eosinophils undergoing apoptosis. Mediation by avb3/CD36/thrombospondin recognition mechanism and lack of phlogistic response. Am J Pathol 1996; 149: 911–921
    16. Persson CGA, Erjef?lt JS. “Ultimate activation” of eosinophils in vivo: lysis and release of clusters of free eosinophil granules (Cfegs). Thorax 1997; 52: 569-574
    17. Savill J, Fadok V. Corpse clearance defines the meaning of cell death. Nature 2000;407: 784-788
    18. Stern M, Savill J, Haslett C. Human monocyte-derived macrophage phagocytosis of senescent eosinophils undergoing apoptosis. Mediation by alpha v beta 3/CD36/thrombospondin recognition mechanism and lack of phlogistic response. Am J Pathol 1996; 149: 911-921
    19. Walsh, G.M., Sexton DW, Blaylock MG et al, Resting and Cytokine-Stimulated Human Small Airway Epithelial Cells Recognize and Engulf Apoptotic Eosinophils. Blood 1999; 94(8): 2827-2835.
    20. Sexton D, Blaylock MG, Walsh GM. Human alveolar epithelial cells engulf apoptotic eosinophils by means of integrin- and osphatidylserinereceptor-dependent mechanisms: a process upregulated by dexamethasone. J Allergy Clin Immunol 2001; 108: 962-969.
    21. Zhang ZH, Liu RY, Noordhoek JA, et al. Interaction of airway epithelial cells (A549) with spores and mycelium of Aspergillus fumigatus. J Infect, 2005; 51: 375-382
    22. Takizawa H. Bronchial epithelial cells in allergic reactions. Curr Drug Targets Inflamm Allergy 2005; 4: 305-311
    23. van den Berg A, Kuiper M., Snoek M, et al. Interleukin-17 induces hyperresponsive interleukin-8 and interleukin-6 production to tumor necrosis factor-alpha in structural lung cells. Am J Respir Cell Mol Boil 2005; 33: 97-104
    24. Kauffman HF, Tamm M, Timmerman JA et al. House dust mite major allergens Der p 1 and Der p 5 activate human airway-derived epithelial cells by protease-dependent and protease-independent mechanisms. Clin Mol Allergy 2006; 4: 5-12.
    25. Renald JC. New insights into the role of cytokines in asthma. J Clin Pathol 2001; 54: 577-589
    26. de Jager W, te Velthuis H, Prakken BJ, et al. Simultaneous detection of 15 humancytokines in a single sample of stimulated peripheral blood mononuclear cells. Clin Diagn Lab Immunol, 2003; 10: 133-139
    27. Message SD, Johnston SL. Host defense function of the airway epithelium in health and disease: clinical background. J Leukoc Biol 2004; 75: 5-17
    28. Bettiol J, Sele J, Henket M, et al. Cytokine production from sputum cells after allergenic challenge in IgE-mediated asthma. Allergy 2002; 57: 1145-1150
    29. Wang CB, Wong CK, Ip WK, et al. Induction of IL-6 in co-culture of bronchial epithelial cells and eosinophils is regulated by p38 MAPK and NF-kappaB. Allergy 2005; 60: 1378-1385
    30. Ordonez CL, Shaughenssy TE, Matthay MA, et al. Increased neutrophil numbers and IL-8 levels in airway secretions in acute severe asthma: Clinical and biologic significance. Am J Respir Crit Care Med 2000; 161: 1185-1190
    31. Dente FL, Carnevali S, Bartoli ML, et al. Profiles of proinflammatory cytokines in sputum from different groups of severe asthmatic patients. Ann Allergy Asthma Immunol 2006; 97: 312-320
    32. Sun YC, Zou Q, Yao WZ. Sputum interleukin-17 is increased and associated with airway neutrophilia in patients with severe asthma. Chin Med J (Engl), 2005; 118: 953-956
    33. Liu, Y, Cousin JM, Hughes J, et al. Glucocorticoids Promote Nonphlogistic Phagocytosis of Apoptotic Leukocytes. The Journal of Immunology 1999; 162(6): 3639-3646
    34. Maderna P, Yona S, Perretti M, et al. Modulation of phagocytosis of apoptotic neutrophils by supernatant from dexamethasone-treated macrophages and annexin-derived peptide Ac2–26. J Immunol 2005; 174: 3727–3733
    35. Huynh MLN, Fadok VA, Henson PM. Phosphatidylserine-dependent ingestion of apoptotic cells promotes TGF-{beta}1 secretion and the resolution of inflammation. Journal of Clinical Investigation 2002; 109(1): 41-50
    1. [1] Rosengarth A , Gerke V , Lueche H. X-ray structure of full-lengthe annexin 1 and implications for membrane aggregation. J Mol Biol 2001; 306:489
    2. [2] Frey BM, Reber BFX , Vishwanath BS, et al. Annexin I modulates cell functions by controlling intracellular calcium release. FASEB J 1999; 13:2235
    3. [3] Amandi-Burgermeister E, Tibes U, Kaiser BM, et al. Suppression of cytokine synthesis, integrin expression and chronic inflammation by inhibitors of cytosolic phospholipase A2. Eur J Pharmacol 1997; 326: 237
    4. [4] Kim S-W, Rhee HJ, Ko J, et al. Inhibition of cytosolic phospholipase A2 by annexin I. Specific interaction model and mapping of the interaction site. J Biol Chem 2001; 276: 15712.
    5. [5] Kim S-W, Ko J, Kim JH, et al. Differential effects of annexins I, II, III and V on cytosolic phospholipase A2 activity: specific interaction model. FEBS Lett 2001; 489: 243
    6. [6] Solito E, Raguenes-Nicol C, de Coupade C, et al. U937 Cells deprived of endogenous annexin 1 demonstrate an increased PLA2 activity. Br J Pharmacol 1998; 124:1675
    7. [7] Hannon R, Croxtall JD, Getting SJ, et al. Aberrant inflammation and resistance to glucocorticoids in annexin 1–/– mouse. FASEB J 2003; 17: 253
    8. [8] Croxtall JD, Choudhury Q, Flower RJ. Inhibitory effect of peptides derived from the N-terminus of lipocortin 1 on arachidonic acid release and proliferation in the A549 cell line: identification of E-QE-Y-V as a crucial component. Br J Pharmacol 1998; 123: 975
    9. [9] Smyth T, Harris HJ, Brown A, et al. Differential modulatory effects of annexin 1 on nitric oxide synthase induction by lipopolysaccharide in macrophages.Immunology 2006; 117: 340
    10. [10] Yona S, Ward B, Buckingham JC, et al. Macrophage biology in the Anx-A1-/- mouse. Prostaglandins Leukot Essent Fatty Acids 2005; 72: 95
    11. [11] Minghetti L, Nicolini A, Polazzi E, et al. Down-regulation of microglial cyclo-oxygenase-2 and inducible nitric oxide synthase expression by lipocortin 1. Br J Pharmacol 1999; 126: 1307.
    12. [12] Young KA, Hirst WD, Solito E, et al. De novo expression of lipocortin-1 in reactive microglia and astrocytes in kainic acid lesioned rat cerebellum. GLIA 1999; 26: 333.
    13. [13] Sampey AV, Hutchinson P, Morand EF. Annexin I and dexamethasone effects on phospholipase and cyclooxygenase activity in human synoviocytes. Med Inflamm 2000; 9: 125
    14. [14] Ferlazzo V, D’Agostino P, Milano S, et al. Anti-inflammatory effects of annexin-1: stimulation of IL-10 release and inhibition of nitric oxide synthesis. Int Immunopharmacol 2003; 3: 1363
    15. [15] Yi AK, Yoon JG, Yeo SJ, et al. Role of mitogen-activated protein kinases in CpG DNA-mediated IL-10 and IL-12 production: central role of extracellular signalregulated kinase in the negative feedback loop of the CpG DNAmediated Th1 response. J Immunol 2002; 168: 4711
    16. [16] Alldridge LC, Harris HJ, Plevin R, et al. The annexin protein lipocortin 1 regulates the MAPK/ERK pathway. J Biol Chem 1999; 274: 37620
    17. [17] Yang YH, Toh ML, Clyne CD, et al. Annexin 1 negatively regulates IL-6 expression via effects on p38 MAPK and MAPK phosphatase-1. J Immunol 2006; 177:8148
    18. [18] Getting SJ, Flower RJ, Perretti M. Inhibition of neutrophil and monocyte recruitment by endogenous and exogenous lipocortin 1. Br J Pharmacol 1997; 120: 1075
    19. [19] Hayhoe RP, Kamal AM, Solito E, et al. Annexin 1 and its bioactive peptide inhibit neutrophil-endothelium interactions under flow: indication of distinct receptor involvement. Blood 2006; 107:2123
    20. [20] Chatterjee BE, Yona S, Rosignoli G, et al. Annexin 1-deficient neutrophils exhibit enhanced transmigration in vivo and increased responsiveness in vitro. J Leukoc Biol 2005; 78:639
    21. [21] Damazo AS, Yona S, D'Acquisto F, et al. Critical protective role for annexin 1 gene expression in the endotoxemic murine microcirculation. Am J Pathol 2005; 166:1581
    22. [22] Lim LHK, Solito E, Russo-Marie F, et al. Promoting detachment of neutrophils adherent to murine postcapillary venules to control inflammation: effect of lipocortin 1. Proc Natl Acad Sci U S A 1998; 95: 14535
    23. [23] Oliani SM, Paul-Clark MJ, Christian HC, et al. Neutrophils interaction with inflamed postcapillary venule endothelium alters annexin 1 expression. Am J Pathol 2001; 158: 603
    24. [24] Bandeira-Melo C, Bonavita AG, Diaz BL, et al. A novel effect for annexin 1-derived peptide ac2-26: reduction of allergic inflammation in the rat. J Pharmacol Exp Ther 2005; 313: 1416
    25. [25] Solito E, Romero IA, Marullo S, et al. Annexin 1 binds to U937 monocytic cells and inhibits their adhesion to microvascular endothelium: involvement of the α4β1 integrin. J Immunol 2000; 165: 1573
    26. [26] Perretti M, Ingegnoli F, Wheller SK, et al. Annexin 1 modulates monocyte-endothelial cell interaction in vitro and cell migration in vivo in the human SCID mouse transplantation model. J Immunol 2002; 169: 2085
    27. [27] Walter A, Riehemann K, Gerke V. A novel ligand of the formyl peptide receptor: annexin I regulates neutrophil extravasation by interacting with the FPR. Molecular Cell 2000; 5: 831
    28. [28] Sawmynaden P, Perretti M. Glucocorticoid upregulation of the annexin-A1 receptor in leukocytes. Biochem Biophys Res Commun 2006; 349: 1351
    29. [29] Strausbaugh HJ, Rosen SD. A potential role for annexin 1 as a physiologic mediator of glucocorticoid-induced L-selectin shedding from myeloid cells. J Immunol 2001; 166: 6294
    30. [30] Perretti M, Getting SJ, Solito E,et al. Involvement of the receptor for formylated peptides in the in vivo antimigratory actions of annexin-1 and its mimetics. Am J Pathol 2001; 158: 1969
    31. [31] Chiang N, Fierro IM, Gronert K, et al. Activation of lipoxin A4 receptors by aspirin-triggered lipoxins ans select peptides evokes ligand-specific responses in inflammation. J Exp Med 2000; 191: 1197
    32. [32] Perretti M, Chiang N, La M, et al. Endogenous lipid- and peptide-derived anti-inflammatory pathways generated with glucocorticoids and aspirin treatment activate the lipoxin A4 receptor. Nature Med 2002; 8: 1296
    33. [33] Srikrishna G, Toomre DK, Manzi A, et al. A novel anionic modification of N-glycans on mammalian endothelial cells is recognised by activated neutrophils and modulate acute inflammatory response. J Immunol 2001; 166: 624
    34. [34] Srikrishna G, Panneerselvam K, Westphal V, et al. Two proteins modulating transendothelial migration of leukocytes recognize novel carboxylated glycans on endothelial cells. J Immunol 2001; 166: 4678
    35. [35] de Coupade C, Solito E, Levine JD. Dexamethasone enhances interaction of endogenous annexin I with L-selectin and triggers shedding of L-selectin in the monocytic cell line U-937. Br J Pharmacol 2003; 140: 133
    36. [36] Adams JM, Cory S. The Bcl-2 protein family: arbiters of cell survival. Science 1998; 281: 1322
    37. [37] Hengartner MO. The biochemistry of apoptosis. Nature 2000; 407: 770
    38. [38] Kothakota S, Azuma T, Reinhard C, et al. Caspase-3-generated fragment ofgelsolin: effector of morphological change in apoptosis. Science 1997; 278: 294
    39. [39] Enari M, Sakahira H, Yokoyama H,et al. A caspase-activated DNase that degrades DNA during apoptosis and its inhibitor ICAD. Nature 1998; 391: 43
    40. [40] Savill J. Apoptosis in resolution of inflammation. J Leukoc Biol 1997; 61: 375
    41. [41] Savill J, Dransfield I, Gregory C, et al. A blast from the past: clearance of apoptotic cells regulates immune responses. Nat Rev Immunol 2002; 2: 965
    42. [42] Schmidt M, Pauels H-G, Lügering A, et al. Glucocorticoids induce apoptosis in human monocytes: potential role of IL-1b. J Immunol 1999; 163: 3484
    43. [43]Distelhorst CW. Recent insights into the mechanism of glucocorticosteroid-induced apoptosis. Cell Death Differ 2002; 9: 6
    44. [44] Ramdas J, Harmon JM. Glucocorticoid-induced apoptosis and regulation of NF-kappaB activity in human leukemic T cells.Endocrinology 1998; 139: 3813
    45. [45] Dallaporta B, Pablo M, Maisse C, et al. Proteasome activation as a critical event of thymocyte apoptosis. Cell Death Differ 2000; 7: 368
    46. [46] Ivanov VN, Nikolic-Zugic J. Biochemical and kinetic characterization of the glucocorticoid-induced apoptosis of immature CD4+CD8+ thymocytes. Int Immunol 1998; 10: 1807
    47. [47] Yang Y, Fang S, Jensen JP,et al. Ubiquitin protein ligase activity of IAPs and their degradation in proteasomes in response to apoptotic stimuli. Science 2000; 288: 874
    48. [48] Gil-Gomez G, Berns A, Brady HJM. A link between cell cycle and cell death: Bax and Bcl-2 modulate Cdk2 activation during thymocyte apoptosis. EMBO J 1998; 17: 7209
    49. [49] Alam A, Braun MY, Hartgers F, et al. Specific activation of the cysteine protease CPP32 during the negative selection of T cells in the thymus. J Exp Med 1997; 186: 1503
    50. [50] Miyashita T, Nagao K, Krajewski S, et al. Investigation ofglucocorticoid-induced apoptotic pathway: processing of caspase-6 but not caspase-3. Cell Death Differ 1998; 5: 1034
    51. [51] Komoriya A, Packard BZ, Brown MJ, et al. Assessment of caspase activities in intact apoptotic thymocytes using cell-permeable fluorogrnic caspase substrates. J Exp Med 2000; 191: 1819
    52. [52] Solito E, de Coupade C, Canaider S, et al. Transfection of annexin 1 inmonocytic cells produces a high degree of spontaneous and stimulated apoptosis associated with caspase-3 activation. Br J Pharmacol 2001; 133: 217
    53. [53] Debret R, El Btaouri H, Duca L, et al. Annexin A1 processing is associated with caspase-dependent apoptosis in BZR cells. FEBS Lett 2003; 546: 195
    54. [54] Hsiang CH, Tunoda T, Whang YE, et al. The impact of altered annexin I protein levels on apoptosis and signal transduction pathways in prostate cancer cells. Prostate 2006; 66: 1413
    55. [55] Solito E, Kamal A, Russo-Marie F, et al. A novel calcium dependent pro-apoptotic effect of annexin I on human neutrophils. FASEB J 2003; 17: 1544
    56. [56] Wang HG, Pathan N, Ethell IM, et al. Ca2+-induced apoptosis through calcineurin dephosphorylation of BAD. Science 1999; 284: 339
    57. [57] Ravichandran KS. ‘Recruitment signals’ from apoptotic cells: invitation to a quiet meal. Cell 2003; 113: 817
    58. [58] Schlegel RA, Williamson P. Phosphatidylserine, a death knell. Cell Death Differ 2001; 8: 551
    59. [59] Fadok VA, Bratton DL, Konowal A, et al. Macrophages that have ingested apoptotic cells in vitroinhibit proinflammatory cytokine production throughautocrine/paracrine mechanisms involving TGF-β, PGE2, and PAF. J Clin Invest 1998; 101: 890
    60. [60] Huynh M-LN, Fadok VA, Henson PM. Phosphatidylserine-dependent ingestion of apoptotic cells promotes TGF-b1 secretion and the resolution ofinflammation. J Clin Invest 2003; 109: 41
    61. [61] Byrne A, Reen DJ. Lipopolysaccharide induces rapid production of IL-10 by monocytes in the presence of apoptotic neutrophils. J Immunol 2002; 168: 1968
    62. [62] Arur S, Uche UE, Rezaul K, et al. Annexin I is an endogenous ligand that mediates apoptotic cell engulfment.Dev Cell 2003; 4: 587
    63. [63] Fan XX, Krohling S, Smith D, et al. Macrophage surface expression of annexins I and II in the phagocytosis of apoptotic lymphocytes. Mol Biol Cell 2004; 15:2863
    64. [64] Maderna P, Yona S, Perretti M, et al. Modulation of phagocytosis of apoptotic neutrophils by supernatant from dexamethasone-treated macrophages and annexin-derived peptide Ac2–26. J Immunol 2005; 174: 3727
    65. [65] Scannell M, Maderna P. Lipoxins and annexin-1: resolution of inflammation and regulation of phagocytosis of apoptotic cells. ScientificWorldJournal 2006. 6: 1555
    66. [66] Muimo R , Hornickova Z , Riemen C E , et al. Histidine phosphorylation of annexin Ⅰ in airway epithelia. J Biol Chem 2000, 275: 36632
    67. [67] Jiyounga O , Rhee J , Kim S W, et al. Annexin2 Ⅰinhibits PMA2 induced c2fos SRE activation by suppressing cytosolic phospholipase A2 signal. FEBS Letters 2000; 476: 296
    68. [68] Miyachi T , Asai K, Tsuiki H , et al. Interleukin21 beta induces the expression of lipocortin 1 mRNA in cultured rat cortical astricytes. . Neuro Sci Res 2001; 40: 53
    69. [69] Buckingham JC, Flower RJ. Lipocortin Ⅰ: a second messenger of glucocorticoid action in the hypothalamo2pituitary2adrenocortical axis. Mol Med Today 1997; 3: 296
    70. [70] Morand EF, Hall P, Hutchinson P, et al. Regulation of annexin I in rheumatoid synovial cells by glucocorticoids and interleukin-1. Mediators Inflamm 2006; 2006:73835.
    71. [71] Brichory FM, Misek DE, Yim AM, et al. An immune response manifested by the common occurrence of annexin Ⅰ and Ⅱautoantibodies and high circulating levels of IL26 in lung cancer. Proc Natl Acad Sci USA 2001; 98: 9824
    72. [72] Wu YL, Jiang XK, Lillington DM, et al. Upregulation of lipocortin Ⅰ inhibits tumor necrosis factor2induced apoptosis in human leukaemic cells: a possible mechanism of resistance to immune surveillance. Br J Haematol 2000; 111: 807
    73. [73] Solito E , de Coupade C , Parente L , et al. Human annexin Ⅰis highly expressed during the differentiation of the epithelial cell line A549: involvement of nuclear factor interleukin 6 in phorbol ester induction of annexin Ⅰ. Cell Growth Differ1998; 9:327

© 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