用户名: 密码: 验证码:
促炎症消退介质脂氧素对内毒素性肺损伤的保护作用及机制
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
第一部分脂氧素对脂多糖诱导小鼠巨噬细胞钙信号转导及活性氧产生的影响及机制
     目的研究脂氧素对脂多糖诱导的小鼠RAW264.7巨噬细胞钙信号转导及活性氧产生的影响并探讨其可能机制。方法小鼠RAW264.7巨噬细胞,随机分为6组:①对照组;②单纯lipopolysaccharide(LPS)组:以终浓度2-10μg/ml LPS刺激巨噬细胞;③单纯Thapsigargin(钙池操纵的钙通道激活剂)组:以终浓度2μM Thapsigargin刺激巨噬细胞;④脂氧素+ LPS组:分别用终浓度为10-7、10-8或10-9M的脂氧素预处理,再以LPS(2-10μg/ml)刺激;⑤脂氧素+ Thapsigargin组:分别用终浓度为10-7、10-8或10-9M的脂氧素预处理,再以2μM Thapsigargin刺激;⑥2-Aminoethoxydiphenylborate(2-APB,钙池操纵的钙通道阻滞剂)+ Thapsigargin组:20μM 2-APB预处理,再以2μM Thapsigargin刺激。采用细胞内钙离子特异性荧光探针Fluo3-AM和活性氧特异性荧光探针DCFH-DA标记小鼠巨噬细胞。激光共聚焦显微镜动态观察脂多糖引起巨噬细胞胞内游离钙浓度变化,流式细胞仪测定巨噬细胞活性氧产生变化。结果脂多糖呈剂量依赖升高细胞内游离钙浓度和活性氧的产生,钙池操纵的钙通道激活剂Thapsigargin可完全抑制脂多糖引起的胞内钙超载。脂氧素呈剂量依赖抑制脂多糖引起的胞内游离钙浓度升高,脂氧素同时可抑制Thapsigargin激活钙池操纵的钙通道引起的钙内流。细胞外无钙液及脂氧素均可抑制脂多糖和Thapsigargin诱导的活性氧产生。结论脂多糖通过促进内钙释放而开放钙池操纵的钙通道,大量细胞外钙离子进入细胞使钙超载,活性氧大量产生,引起组织细胞损伤。脂氧素可能通过抑制钙池操纵的钙通道,使外钙内流减少,保持细胞钙稳态,减少活性氧的生成而起到抗炎促炎症消退作用。
     第二部分脂氧素对脂多糖诱导小鼠巨噬细胞TNF-α转化酶的影响
     目的研究脂氧素对脂多糖诱导的小鼠RAW264.7巨噬细胞TNF-α转化酶表达的影响。方法小鼠RAW264.7巨噬细胞,随机分为3组:①对照组;②单纯LPS组:以终浓度1- 2μg/ml LPS刺激巨噬细胞;③脂氧素+ LPS组:分别用终浓度为10-7、10-8或10-9 M的脂氧素预处理,再以LPS(1- 2μg/ml)刺激。孵育6小时和24小时后,收集细胞上清液,酶联免疫法测定上清液TNF-α浓度。提取细胞总mRNA, RT-PCR测定TNF-α转化酶和TNF-α基因表达,提取细胞总蛋白测定TNF-α转化酶蛋白表达,流式细胞仪检测膜型TNF-α蛋白表达。结果脂氧素抑制脂多糖诱导的RAW264.7小鼠巨噬细胞TNF-α基因和膜型TNF-α蛋白表达。脂氧素抑制脂多糖诱导的TNF-α转化酶蛋白表达但不影响其mRNA表达,并呈剂量依赖抑制脂多糖诱导的分泌型TNF-α产生。结论脂氧素通过干扰TNF-α转化酶蛋白质翻译过程,进而减少脂多糖诱导的RAW264.7巨噬细胞分泌型TNF-α的产生,这可能是脂氧素抗炎促炎症消退的重要作用机制之一。
     第三部分脂氧素对内毒素性肺损伤小鼠的保护作用及机制
     目的研究脂氧素对脂多糖诱导小鼠急性肺损伤的影响并探讨其可能机制。方法36只雄性C57BL/6小鼠,随机分为6组(n = 6):①对照组:小鼠雾化吸入0.9%生理盐水20 min,60 min后尾静脉注射10%乙醇5 ml/kg;②脂氧素组:小鼠雾化吸入0.9%生理盐水20 min,60 min后尾静脉注射脂氧素(0.1 mg/kg);③ZnPP组:小鼠雾化吸入0.9%生理盐水20 min,60 min后尾静脉注射Zinc protoporphyrin IX(血红素氧合酶-1抑制剂)25 mg/kg;④LPS组:小鼠雾化吸入LPS 20 min,60 min后尾静脉注射10%乙醇5 ml/kg;⑤脂氧素治疗组:以脂氧素0.1 mg/kg代替乙醇,其余处理同LPS组;⑥ZnPP+脂氧素治疗组:处理同脂氧素治疗组,但静脉注射脂氧素30 min前注射ZnPP 25 mg/kg。雾化吸入LPS或生理盐水后8 h处死动物。光镜下观察肺组织病理学变化,测定肺泡灌洗液白细胞、中性粒细胞计数和TNF-α含量及总蛋白浓度;测定肺组织髓过氧化物酶(MPO)活性、干湿重比值、丙二醛(MDA)活性和一氧化氮(NO)浓度;测定肺组织血红素氧合酶-1(HO-1)的蛋白表达和活性。结果与LPS组比较,脂氧素治疗组肺组织中性粒细胞浸润减少、肺水肿减轻,肺组织MPO活性、MDA活性、NO含量、TNF-α浓度均降低(P < 0.01),肺泡灌洗液总蛋白浓度降低(P < 0.01)。而HO-1的蛋白表达和活性明显升高,HO-1抑制剂ZnPP减弱脂氧素保护作用。结论脂氧素通过上调HO-1减轻内毒素诱导的小鼠急性肺损伤。
PartⅠThe effects of lipoxin on calcium signal transduction and production of reactive oxygen species in macrophages induced by LPS
     Objective To investigate the effects of lipoxin on calcium signal transduction and production of reactive oxygen species in macrophages induced by lipopolysaccharide (LPS). Methods Macrophages were randomly assigned to one of the following six groups:①control group;②LPS group;③Thapsigargin group;④Lipoxin + LPS group;⑤Lipoxin + Thapsigargin group;⑥2-Aminoethoxydiphenylborate + Thapsigargin group. The intracellular [Ca~(2+)]_i was analyzed by confocal laser microscopy. The production of ROS was assayed by flow cytometry. Results Both LPS and Thapsigargin induced an increase in intracellular [Ca~(2+)]_i, either in the presence or absence of extracellular Ca~(2+) in murine macrophages. The Ca~(2+) signal was sustained in the presence of external Ca~(2+) and only initiated a mild and transient rise in the absence of external Ca~(2+). SOC channel inhibitor 2-APB completely suppressed the Ca~(2+) entry signal evoked by thapsigargin and suppressed approximately 93% of the Ca~(2+) entry signal evoked by LPS. The increase in intracellular [Ca~(2+)]_i was associated with increased ROS production which was completely abolished in the absence of extracellular Ca~(2+) or in the presence of SOC channel inhibitors 2-APB or lipoxin. Conclusions These findings indicate that the LPS-induced intracellular [Ca~(2+)]_i increase depends on the Ca~(2+) entry through SOC channels, and lipoxin inhibits Ca~(2+) influx and ROS production through SOC channels in murine macrophages induced by LPS
     PartⅡThe effects of lipoxin on expression of TNF-αconverting enzyme in RAW264.7 macrophages induced by LPS
     Objective To observe the effects of lipoxin on expression of TNF-αconverting enzyme in RAW264.7 macrophages induced by LPS. Methods RAW264.7 macrophages were cultured in vitro with 1-2ug/ml lipopolysaccharide in the absence or presence of lipoxin at different concentrations for 6 and 24 hours, then the supernatant was collected for measuring TNF-αby ELISA kit and the expressions of TNF-αand TNF-αconverting enzyme were measured by semi-quantitative RT-PCR. Western blotting was applied to detect the expression of TNF-αconverting enzyme. The expressions of membrane-bound TNF-αprotein were assessed by flow cytrometry. Results Lipoxin inhibited the gene and protein expression of TNF-α, lipoxin inhibited up-regulation of TACE protein expression but did not inhibit TNF-αconverting enzyme mRNA expression. Conclusions Lipoxin inhibits the release of TNF-αthrough inhibition the protein expression of TNF-αconverting enzyme in RAW264.7 macrophages.
     PartⅢProtective effect of lipoxin on LPS-induced acute lung injury in mice
     Objective To investigate whether lipoxin could attenuate LPS induced acute lung injury in mice. Methods All of the animals were randomly assigned to one of six groups (n = 6 per group). In the sham-vehicle group, mice were treated with 0.9% saline 60 mins after they were challenged with saline. The sham-ATL group was identical to the sham-vehicle group except that ATL (0.7 mg/kg, intravenously) was administered instead of vehicle. The sham-ZnPP group was identical to the sham-vehicle group except that Zinc protoporphyrin IX (ZnPP, 25 mg/kg intravenously) was administered instead of vehicle. In the LPS-vehicle group, mice were treated with vehicle 60 mins after they were challenged with LPS. The LPS-ATL group was identical to the LPS-vehicle group but received ATL (0.7 mg/kg, intravenously) instead of vehicle. The ZnPP-ATL-LPS group was identical to the LPS-ATL group, but ZnPP (25 mg/kg intravenously) was administered 30 mins before ATL. Results Inhalation of LPS increased inflammatory cell counts, TNF (Tumor Necrosis Factor)-αand protein concentration in BALF and also induced lung histological injury and edema. Post-treatment with ATL inhibited TNF-α, nitric oxide and malondialdehyde production, with the outcome of decreased pulmonary edema, lipid peroxidation and the infiltration of neutrophils in lung tissues. In addition, Western blot and immunohistochemical analysis revealed that ATL promoted the formation of HO-1 in the lung tissues. Heme oxygenase-1(HO-1) activity was also increased in the lung tissues after ATL stimulation. The beneficial effects of ATL were abolished by ZnPP. Conclusions This study demonstrates that post-treatment with ATL significantly reduces LPS-induced acute lung injury in mice. HO-1 plays an essential role in the anti-inflammatory and pro-resolution bioactions of lipoxin.
引文
1. Bernard GR, Artigas A, Brigham KL, et al. The American-European Consensus Conference on ARDS. Definitions, mechanisms, relevant outcomes, and clinical trial coordination. Am J Respir Crit Care Med, 1994, 149: 818–824.
    2. Ware LB, Matthay MA. The acute respiratory distress syndrome. N Engl J Med, 2000, 342: 1334–1349.
    3. Balibrea JL, Arias-D?az J. Acute respiratory distress syndrome in the septic surgical patient. World J Surg, 2003, 27: 1275–1284.
    4. Crimi E, Slutsky AS. Inflammation and the acute respiratory distress syndrome. Best Practice Res Clin Anaesth, 2004, 18: 477–492.
    5. Bhatia M, Moochala S. Role of inflammatory mediators in the pathophysiology of acute respiratory distress syndrome. J Pathol, 2004, 202: 145–156.
    6. Lee WL, Downey GP. Neutrophil activation and acute lung injury. Curr Opin Crit Care, 2001, 7: 1–7.
    7. Abraham E. Neutrophils and acute lung injury. Crit Care Med, 2003, 31: S195–S199.
    8. Matthay MA, Zimmerman GA. Acute lung injury and the acute respiratory distress syndrome: four decades of inquiry into pathogenesis and rational management. Am J Respir Cell Mol Biol, 2005, 33:319-327.
    9. Opal SM, Fisher CJ Jr, Dhainau J-FA, et al. Confirmatory interleukin-1 receptor antagonist trial in severe sepsis: a phaseШ, randomized, double-blind, placebo-controlled, multicenter trial. The interleukin-1 Receptor Antagonist Sepsis Investgator Group. Crit Care Med, 1997, 25: 1115–1124.
    10. Abraham E, Laterre PF, Garbino J, et al. Lenercept (p55 tumor necrosis factor receptor fusion protein) in severe sepsis and early septic shock: a randomized, double-blind, placebo-controlled, multicenter phaseШtrial with 1342 patients. Crit Care Med, 2001, 29: 503–510.
    11. Bernard GR, Wheeler AP, Russell JA, et al. The effects of ibuprofen on the physiology and survival of patients with sepsis. The Ibuprofen in Sepsis Study Group. N Engl J Med,1997,336:912–918.
    12. The ARDS Network. Ketoconazole for early treatment of acute lung injury and acute respiratory distress syndrome: a randomized controlled trial. JAMA, 2000, 283:1995–2002.
    13. Lawrence T, Gilroy DW, Colville-Nash PR, et al. Possible new role for NF-kappaB in the resolution of inflammation. Nat Med, 2001, 7:1291–1297.
    14. Serhan CN, Savill J. Resolution of inflammation: the beginning programs the end. Nat Immunol, 2005, 6:1191–1197.
    15. Gilroy DW, Lawrence T, Perretti M, et al. Inflammatory resolution: new opportunities for drug discovery. Nat Rev Drug Discov, 2004, 3:401–416.
    16. Serhan CN. Lipoxins and novel aspirin-triggered 15-epi-lipoxin biosynthesis: an update and role in anti-inflammation and proresolution. Prostaglandins Other Lipid Mediators, 2002, 68-69:433–455.
    17. Chiang N, Arita M, Serhan CN. Anti-inflammatory circuitry: lipoxin, aspirin-triggered lipoxins and their receptor ALX. Prostaglandins Leukot Essent Fatty Acids, 2005, 73:163–177.
    18. Claria J, Serhan CN. Aspirin triggers previously undescribed bioactive eicosanoids by human endothelial cell-leukocyte interactions. Proc Natl Acad Sci U S A, 1995, 92:9475–9479.
    19. Godson C, Mitchell S, Harvey K, et al. Cutting edge: lipoxins rapidly stimulate nonphlogistic phagocytosis of apoptotic neutrophils by monocyte-derived macrophages. J Immunol, 2000, 164:1663–1667.
    20. Reville K, Crean JK, Vivers S, et al. Lipoxin A4 redistributes myosin IIA and Cdc42 in macrophages: implications for phagocytosis of apoptotic leukocytes. J Immunol, 2006, 176:1878–1888.
    21. Mitchell S, Thomas G, Harvey K, et al. Lipoxins, aspirin triggered epi-lipoxins, lipoxin stable analogues, and the resolution of inflammation: stimulation of macrophage phagocytosis of apoptotic neutrophils in vivo. J Am Soc Nephrol, 2002, 13:2497–2507.
    22. Leonard MO, Hannan K, Burne MJ, et al. 15-Epi-16-para- fluorophenoxy lipoxin A4 methyl ester, a synthetic analogue of 15-epi-lipoxin A(4), is protective in experimental ischemic acute renal failure. J Am Soc Nephrol, 2002, 13:1657–1662.
    23. Bannenberg G, Moussignac RL, Gronert K, et al. Lipoxins and novel 15-epi-lipoxin analogs display potent anti-inflammatory actions after oral administration. Br J Pharmacol, 2004, 143:43–52.
    24. Levy BD, De Sanctis GT, Devchand PR, et al. Lipoxins and aspirin-triggered lipoxins in airway responses. Adv Exp Med Biol, 2003, 525:19–23.
    25. Gewirtz AT, Collier-Hyams LS, Young AN, et al. Lipoxin A4 analogs attenuate induction of intestinal epithelial proinflammatory gene expression and reduce the severity of dextran sodium sulfate-induced colitis. J Immunol, 2002, 168:5260–5267.
    1. Parekh AB, Putney JW. Store-operated calcium channels. Physiol Rev,2005,85: 757-810.
    2. Parekh AB, Penner R . Store depletion and calcium influx. Physiol Rev, 1997, 77: 901- 930.
    3. Semenova SB, Kiselev KI, Mozhaeva GN. Low-conductivity calcium channels in the macrophage plasma membrane: activation by inositol-1,4,5-triphosphate. Neurosci Behav Physiol, 1999, 29: 339-345.
    4. Hoth M, Button DC, Lewis RS. Mitochondrial control of calcium-channel gating: a mechanism for sustained signaling and transcriptional activation in T lymphocytes. Proc Natl Acad Sci U S A, 2000, 97: 10607-10612.
    5. Rottingen J, Iversen JG. Ruled by waves? Intracellular and intercellular calcium signalling. Acta Physiol Scand, 2000, 169: 203-219.
    6. Chang WC, Parekh AB. Close functional coupling between Ca2+ release-activated Ca2+ channels, arachidonic acid release, and leukotriene C4 secretion. J Biol Chem, 2004, 279: 29994-29999.
    7. Feske S, Giltnane J, Dolmetsch R, et al. Gene regulation mediated by calcium signals in T lymphocytes. Nat Immunol, 2001, 2: 316-324.
    8. Parekh AB. Calcium signaling and acute pancreatitis: specific response to a promiscuous messenger. Proc Natl Acad Sci U S A, 2000, 97: 12933-124934.
    9. Putney JW. Presenilins, Alzheimer's disease, and capacitative calcium entry. Neuron, 2000, 27: 411-412.
    10. Lee C, Xu DZ, Feketeova E, et al. Store-operated calcium channel inhibition attenuates neutrophil function and postshock acute lung injury. J Trauma, 2005, 59: 56-63.
    11. Bonnans C, Mainprice B, Chanez P, et al. Lipoxin A4 stimulates a cytosolic Ca2+ increase in human bronchial epithelium. J Biol Chem, 2003 , 278:10879-10884.
    12. Lee TH, Horton CE, Kyan-Aung U, et al. Lipoxin A4 and lipoxin B4 inhibit chemotactic responses of human neutrophils stimulated by leukotriene B4 and formyl-L-methionyl-L-leucyl-L-phenylalanine. Clin Sci (Lond), 1989, 77:195-203.
    13. Gewirtz AT, McCormick B, Neish AS, et al. Pathogen-induced chemokine secretion from model intestinal epithelium is inhibited by lipoxin A4 analogs. J Clin Invest, 1998,101:1860-1869.
    14. Lee WL, Downey GP. Neutrophil activation and acute lung injury. Curr Opin Crit Care, 2001, 7: 1-7.
    15. Abraham E. Neutrophils and acute lung injury. Crit Care Med, 2003, 31: S195 - S199.
    16. Myers JT, Swanson JA. Calcium spikes in activated macrophages during Fcgamma receptor-mediated phagocytosis. J Leukoc Biol, 2002, 72: 677-684.
    17. Hsuan SL, Kannan MS, Jeyaseelan S, et al. Pasteurella haemolytica A1-derived leukotoxin and endotoxin induce intracellular calcium elevation in bovine alveolar macrophages by different signaling pathways. Infect Immun, 1998, 66: 2836-2844.
    18. Mustafa SB, Olson MS. Effects of calcium channel antagonists on LPS-induced hepatic iNOS expression. Am J Physiol, 1999, 277: G351- 360.
    19. Molinuevo MS, Etcheverry SB, Cortizo AM. Macrophage activation by a vanadyl-aspirin complex is dependent on L-type calcium channel and the eneration of nitric oxide. Toxicology, 2005, 210: 205-212.
    20. Lambeth JD. NOX enzymes and the biology of reactive oxygen. Nat Rev Immunol, 2004, 4: 181-189.
    21. Hensley K, Robinson KA, Gabbita SP, et al. Reactive oxygen species, cell signaling, and cell injury. Free Radic Biol Med, 2000, 28: 1456 -1462.
    22. Jacobson J, Duchen MR. Mitochondrial oxidative stress and cell death in astrocytes-requirement for stored Ca2+ and sustained opening of the permeabilitytransition pore. J Cell Sci, 2002, 115: 1175-1188.
    23. Granados MP, Salido GM, Pariente JA, et al. Generation of ROS in response to CCK-8 stimulation in mouse pancreatic acinar cells. Mitochondrion, 2004, 3: 285 - 296.
    24. Zahradnikova A Jr, Polakova E, Zahradnik I, et al. Kinetics of calcium spikes in rat cardiac myocytes. J Physiol, 2007, 578:677- 691.
    25. Thomas G, Gurung IS, Killeen MJ, et al. Effects of L-type Ca2+ channel antagonism on ventricular arrhythmogenesis in murine hearts containing a modification in the Scn5a gene modelling human long QT syndrome. J Physiol, 2007, 578:85-97.
    26. Ogata S, Kubota Y, Satoh S, et al. Ca2+ stimulates COX-2 expression through calcium-sensing receptor in fibroblasts. Biochem Biophys Res Commun, 2006, 351:808-814.
    27. Prakriya M, Lewis RS. Potentiation and inhibition of Ca2+ release-activated Ca2+ channels by 2-aminoethyldiphenyl borate (2-APB) occurs independently of IP3 receptors. J Physiol, 2001, 536: 3-19.
    28. Broad LM, Braun FJ, Lievremont JP, et al. Role of the phospholipase C-inositol 1,4,5-trisphosphate pathway in calcium release-activated calcium current and capacitative calcium entry. J Biol Chem, 2001, 276: 15945-15952.
    29. Hoth M, Penner R. Calcium release-activated calcium current in rat mast cells. J Physiol, 1993, 465: 359-386.
    30. Trebak M, Bird GS, McKay RR, et al. Comparison of human TRPC3 channels in receptor-activated and store-operated modes. Differential sensitivity to channel blockers suggests fundamental differences in channel composition. J Biol Chem, 2002, 277: 21617-21623.
    1. Hayakawa K, Meng Y, Hiramatsu N, et al. Priming of glomerular mesangial cells by activated macrophages causes blunted responses to proinflammatory stimuli. J Immunol, 2006,176:2529-2537.
    2. Coimbra R, Melbostad H, Loomis W, et al. LPS-induced acute lung injury is attenuated by phosphodiesterase inhibition: effects on proinflammatory mediators, metalloproteinases, NF-kappaB, and ICAM-1 expression. J Trauma, 2006, 60:115-125.
    3. Zhang Y, DeWitt DL, McNeely TB, et al. Secretory leukocyte protease inhibitor suppresses the production of monocyte prostaglandin H synthase-2, prostaglandin E2, and matrix metalloproteinases. J Clin Invest, 1997, 99:894-900.
    4. van der Poll T, Coyle SM, Kumar A, et al. Down-regulation of surface receptors for TNF and IL-1 on circulating monocytes and granulocytes during human endotoxemia: effect of neutralization of endotoxin-induced TNF activity by infusion of a recombinant dimeric TNF receptor. J Immunol, 1997, 158:1490-1497.
    5. West MA, Clair L, Bellingham J. Role of calcium in lipopolysaccharide stimulated tumor necrosis factor and interleukin-1 signal transduction in naive and endotoxin-tolerant murine macrophages. J Trauma, 1996, 41:647-652.Black RA, Rauch CT, Kozlosky CJ, et al. A metalloproteinase disintegrin that releases tumour-necrosis factor-alpha from cells. Nature, 1997, 385:729-733.
    7. Moss ML, Jin SL, Becherer JD, et al. Structural features and biochemical properties of TNF-alpha converting enzyme (TACE). J Neuroimmunol, 1997, 72:127-129.
    8. Li Y, Brazzell J, Herrera A, Walcheck B. ADAM17 deficiency by mature neutrophils has differential effects on L-selectin shedding. Blood, 2006, 108: 2275-2279.
    9. Shao MX, Nadel JA. Neutrophil elastase induces MUC5AC mucin production in human airway epithelial cells via a cascade involving protein kinase C, reactive oxygen species, and TNF-alpha-converting enzyme. J Immunol, 2005, 175: 4009-4016.
    10. Li N, Boyd K, Dempsey PJ, et al. Non-Cell Autonomous Expression of TNF-{alpha} Converting Enzyme ADAM17 Is Required for Normal Lymphocyte Development. J Immunol, 2007,178:4214-4221.
    11. Solomon A, Akabayov B, Frenkel A, et al. Key feature of the catalytic cycle of TNF-alpha converting enzyme involves communication between distal protein sites and the enzyme catalytic core. Proc Natl Acad Sci U S A, 2007, 104: 4931-4936.
    14. Serhan CN, Savill J. Resolution of inflammation: the beginning programs the end. Nat Immunol, 2005, 6:1191-1197.
    13. Gilroy DW, Lawrence T, Perretti M, et al. Inflammatory resolution: new opportunities for drug discovery. Nat Rev Drug Discov, 2004,3:401-416.
    14. Serhan CN. Lipoxins and aspirin-triggered 15-epi-lipoxin biosynthesis: an update and role in anti-inflammation and pro-resolution. Prostaglandins Other Lipid Mediat, 2002 ,68-69:433-455.
    15. Ariel A, Chiang N, Arita M, et al. Aspirin-triggered lipoxin A4 and B4 analogs block extracellular signal-regulated kinase-dependent TNF-alpha secretion from human T cells. J Immunol, 2003, 170:6266-6272.
    16. Godson C, Mitchell S, Harvey K, et al. Lipoxins rapidly stimulate nonphlogistic phagocytosis of apoptotic neutrophils by monocyte-derived macrophages. J Immunol, 2000, 164:1663-1667.
    17. Mullberg J, Althoff K, Jostock T, et al. The importance of shedding of membrane proteins for cytokine biology. Eur Cytokine Netw, 2000, 11:27-38.
    18. McMahoenal B, Stenson C, McPhillips F, et al. Lipoxin A4 antagonizes the mitogenic effects of leukotriene D4 in human mesangial cells. Differential activation of MAP kinases through distinct recepters. J Biol Chem, 2000, 275:27566-27575.
    19. Fiorucci S, Wallace JL, Mencarelli A, et al. A beta-oxidation-resistant lipoxin A4 analog treats hapten-induced colitis by attenuating inflammation and immune dysfunction. Proc Natl Acad Sci U S A, 2004, 101:15736-15741.
    20.金胜威,张力,叶笃筠,等.脂氧素对脂多糖诱导的巨噬细胞游离钙浓度变化及活性氧的影响.中国病理生理杂志,2005,21:1651.
    21. Gewirtz AT, Collier-Hyams LS, Young AN, et al. Lipoxin A4 analogs attenuate induction of intestinal epithelial proinflammatory gene expression and reduce the severity of dextran sodium sulfate-induced colitis. J Immunol, 2002, 168:5260-5267.
    22. Fierro IM, Colgan SP, Bernasconi G, et al. Lipoxin A4 and aspirin-triggered 15-epi-lipoxin A4 inhibit human neutrophil migration: comparisons between synthetic 15 epimers in chemotaxis and transmigration with microvessel endothelial cells and epithelial cells. J Immunol, 2003,170:2688-2694.
    23. Jozsef L, Zouki C, Petasis NA, et al. Lipoxin A4 and aspirin-triggered 15-epi-lipoxin A4 inhibit peroxynitrite formation, NF-kappa B and AP-1 activation, and IL-8 gene expression in human leukocytes. Proc Natl Acad Sci U S A, 2002, 99:13266-13271.
    24. Sodin-Semrl S, Spagnolo A, Mikus R, et al. Opposing regulation of interleukin-8 and NF-kappaB responses by lipoxin A4 and serum amyloid A via the common lipoxin A receptor. Int J Immunopathol Pharmacol, 2004,17:145-156.
    25. Wu SH, Lu C, Dong L, et al. Lipoxin A4 inhibits TNF-alpha-induced production of interleukins and proliferation of rat mesangial cells. Kidney Int, 2005, 68:35-46.
    26. Nascimento-Silva V, Arruda MA, Barja-Fidalgo C, et al. Aspirin-triggered lipoxin A4 blocks reactive oxygen species generation in endothelial cells: a novel antioxidative mechanism. Thromb Haemost, 2007,97:88-98.
    27.李凌波,朱孔黎,杨渝珍,等. LPS刺激对TACE基因表达和对TNF-α前体加工的影响.中国免疫学杂志, 2002,18:690-693.
    28. Koff JL, Shao MX, Kim S, et al. Pseudomonas lipopolysaccharide accelerates wound repair via activation of a novel epithelial cell signaling cascade. J Immunol, 2006,177:8693-8700.
    29. Wang Y, Sul HS. Ectodomain shedding of preadipocyte factor 1 (Pref-1) by tumor necrosis factor alpha converting enzyme (TACE) and inhibition of adipocyte differentiation. Mol Cell Biol, 2006, 26:5421-5435.
    30. Tsakadze NL, Sithu SD, Sen U, et al. Tumor necrosis factor-alpha-converting enzyme (TACE/ADAM-17) mediates the ectodomain cleavage of intercellular adhesion molecule-1 (ICAM-1). J Biol Chem, 2006, 281:3157-3164.
    31. Jarry A, Bach-Ngohou K, Masson D, et al. Human colonic myocytes are involved in postischemic inflammation through ADAM17-dependent TNF alpha production. Br J Pharmacol, 2006, 147:64-72.
    1. Liu D, Zeng BX, Zhang SH, et al. Rosiglitazone, a peroxisome proliferator-activated receptor-gamma agonist, reduces acute lung injury in endotoxemic rats. Crit Care Med, 2005, 33:2309–2316.
    2. Ware LB, Matthay MA. The acute respiratory distress syndrome. N Engl J Med, 2000, 342:1334–1349.
    3. Nagase T, Uozumi N, Ishii S, et al. Acute lung injury by sepsis and acid aspiration: a key role for cytosolic phospholipase A2. Nat Immunol, 2000, 1:42–46.
    4. Maines, MD. The heme oxygenase system: a regulator of second messenger gases. Annu Rev Pharmaco Toxicol, 1997, 37:517–554.
    5. Ponka, P. Cell biology of heme. Am J Med Sci, 1999, 318:241–256.
    6. Otterbein, LE. Choi, AMK. Heme oxygenase: colors of defense against cellular stress. Am J Physiol, 2000, 279:L1029–1037.
    7. Otterbein LE, Bach FH, Alam J, et al. Carbon monoxide has anti-inflammatory effects involving the mitogen-activated protein kinase pathway. Nature Med, 2000, 6:422–428.
    8. Otterbein LE, Soares MP, Yamashita K, et al. Heme oxygenase-1: unleashing the protective properties of heme. Trends Immunol, 2003, 24:449–455.
    9. Lee TS, Chau LY. Heme oxygenase-1 mediates the anti-inflammatory effect of interleukin-10 in mice. Nat Med, 2002, 8:240–246.
    10. Otterbein L, Sylvester SL, Choi AM. Hemoglobin provides protection against lethal endotoxemia in rats: the role of heme oxygenase-1. Am J Respir Cell Mol Biol, 1995, 13:595–601.
    11. Otterbein LE, Lee PJ, Chin BY, et al. Protective effects of heme oxygenase-1 in acute lung injury. Chest, 1999, 116:61S–63S.
    12. Suttner DM, Sridhar K, Lee CS, et al. Protective effects of transient HO-1 overexpression on susceptibility to oxygen toxicity in lung cells. Am J Physiol, 1999, 276:L443–451.
    13. Hayashi S, Takamiya R, Yamaguchi T, et al. Induction of heme oxygenase-1 suppresses venular leukocyte adhesion elicited by oxidative stress: role of bilirubin generated by the enzyme. Circ Res, 1999, 85:663–671.
    14. Chiang N, Arita M, Serhan CN. Anti-inflammatory circuitry: lipoxin, aspirin-triggered lipoxins and their receptor ALX. Prostaglandins Leukot Essent Fatty Acids, 2005, 73:163–177.
    15. Serhan CN. Lipoxin biosynthesis and its impact in inflammatory and vascular events. Biochim Biophys Acta, 1994, 1212:1–25.
    16. Godson C, Mitchell S, Harvey K, et al. Cutting edge: lipoxins rapidly stimulate nonphlogistic phagocytosis of apoptotic neutrophils by monocyte-derived macrophages. J Immunol, 2000, 164:1663–1667.
    17. Claria J, Serhan CN. Aspirin triggers previously undescribed bioactive eicosanoids by human endothelial cell-leukocyte interactions. Proc Natl Acad Sci U S A, 1995, 92:9475–9479.
    18. Clish CB, O’Brien JA, Gronert K, et al. Local and systemic delivery of a stable aspirin triggered lipoxin prevents neutrophil recruitment in vivo. Proc Natl Acad Sci USA, 1999, 96:8247–8252.
    19. Bannenberg G, Moussignac RL, Gronert K, et al. Lipoxins and novel 15-epi-lipoxin analogs display potent anti-inflammatory actions after oral administration. Br J Pharmacol, 2004, 143:43–52.
    20. Chiang N, Bermudez EA, Ridker PM, et al. Aspirin triggers anti-inflammatory 15-epi-lipoxin A4 and inhibits thromboxane in a randomized human trial. Proc Natl Acad Sci USA, 2004, 101:15178–15183.
    21. Grosser N, Abate A, Oberle S, et al. Heme oxygenase-1 induction may explain theantioxidant profile of aspirin. Biochem Biophys Res Commun, 2003, 308: 956–960.
    22. Nascimento-Silva V, Arruda MA, Barja-Fidalgo C, et al. Novel lipid mediator aspirin-triggered lipoxin A4 induces heme oxygenase-1 in endothelial cells. Am J Physiol Cell Physiol, 2005, 289:C557–563.
    23. Leonard MO, Hannan K, Burne MJ, et al. 15-Epi-16-(para-fluorophenoxy) lipoxin A4 methyl ester, a synthetic analogue of 15-epi-lipoxin A(4), is protective in experimental ischemic acute renal failure. J Am Soc Nephrol, 2002, 13:1657–1662.
    24. Levy BD, De Sanctis GT, Devchand PR, et al. Lipoxins and aspirin-triggered lipoxins in airway responses. Adv Exp Med Biol, 2003, 525:19–23.
    25. Gewirtz AT, Collier-Hyams LS, Young AN, et al. Lipoxin A4 analogs attenuate induction of intestinal epithelial proinflammatory gene expression and reduce the severity of dextran sodium sulfate-induced colitis. J Immunol, 2002, 168:5260–5267.
    26. Jeyaseelan S, Chu HW, Young SK, et al. Transcriptional profiling of lipopolysaccharide-induced acute lung injury. Infect Immun, 2004,72:7247–7256.
    27. Nick JA, Young SK, Brown KK, et al. Role of p38 mitogen-activated protein kinase in a murine model of pulmonary inflammation. J Immunol, 2000, 164:2151–2159.
    28. Gray KD, Simovic MO, Chapman WC, et al. Endotoxin potentiates lung injury in cerulein-induced pancreatitis. Am J Surg, 2003,186:526–530.
    29. Draper HH, Hadley M. Malondialdehyde determination as index of lipid peroxidation. Methods Enzymol, 1990, 86:421–431.
    30. Bernard GR, Wheeler AP, Russell JA, et al. The effects of ibuprofen on the physiology and survival of patients with sepsis. The Ibuprofen in Sepsis Study Group. N Engl J Med, 1997, 336:912–918.
    31. The ARDS Network. Ketoconazole for early treatment of acute lung injury and acute respiratory distress syndrome: a randomized controlled trial. JAMA, 2000, 283:1995–2002.
    32. Gilroy DW, Lawrence T, Perretti M, et al. Inflammatory resolution: new opportunitiesfor drug discovery. Nat Rev Drug Discov, 2004, 3:401–416.
    33. Lawrence T, Gilroy DW, Colville-Nash PR, et al. Possible new role for NF-kappaB in the resolution of inflammation. Nat Med, 2001, 7:1291–1297.
    34. Serhan CN, Savill J. Resolution of inflammation: the beginning programs the end. Nat Immunol, 2005, 6:1191–1197.
    35. Fukunaga K, Kohli P, Bonnans C, et al. Cyclooxygenase 2 plays a pivotal role in the resolution of acute lung injury. J Immunol, 2005, 174:5033–5039.
    36. Reville K, Crean JK, Vivers S, et al. Lipoxin A4 redistributes myosin IIA and Cdc42 in macrophages: implications for phagocytosis of apoptotic leukocytes. J Immunol, 2006, 176:1878–1888.
    37. Mitchell S, Thomas G, Harvey K, et al. Lipoxins, aspirin triggered epi-lipoxins, lipoxin stable analogues, and the resolution of inflammation: stimulation of macrophage phagocytosis of apoptotic neutrophils in vivo. J Am Soc Nephrol, 2002, 13:2497–2507.
    38. Takai D, Nagase T, Shimizu T. New therapeutic key for cystic fibrosis: a role for lipoxins. Nat Immunol, 2004, 5:357–358.
    39. Willis DMA, Frederick R, Willoughby DA. Heme oxygenase: a novel target for the modulation of the inflammatory response. Nature Med, 1996, 2:87–90.
    40. ABRAHAM E. Neutrophils and acute lung injury. Crit Care Med, 2003, 31: S195–S199.
    41. Serhan CN. Lipoxins and novel aspirin-triggered 15-epi-lipoxin biosynthesis: an update and role in anti-inflammation and proresolution. Prostaglandins Other Lipid Mediators, 2002, 68-69:433–455.
    42. McMahon B, Godson C. Lipoxins: endogenous regulators of inflammation. Am J Physiol Renal Physiol, 2004, 286:F189–F201.
    43. Pouliot M, Serhan CN. Lipoxin A4 and aspirin-triggered 15-epi-LXA4 inhibit tumor necrosis factor-alpha-initiated neutrophil responses and trafficking: novel regulators of a cytokine-chemokine axis relevant to periodontal diseases. J Periodontal Res, 1999,34:370–373.
    44. Sittipunt C, Steinberg KP, Ruzinski JT, et al. Nitric oxide and nitrotyrosine in the lungs of patients with acute respiratory distress syndrome. Am J Respir Crit Care Med, 2001, 163:503–510.
    45. Bhatia M, Moochhala S. Role of inflammatory mediators in the pathophysiology of acute respiratory distress syndrome. J Pathol, 2004, 202:145–156.
    46. Numata M, Suzuki S, Miyazawa N, et al. Inhibition of inducible nitric oxide synthase prevents LPS-induced acute lung injury in dogs. J Immunol, 1998, 160:3031–3037.
    47. Gao J, Zeng BX, Zhou LJ, et al. Protective effects of early treatment with propofol on endotoxin-induced acute lung injury in rats. Br J Anaesth, 2004, 92:277–279.
    48. Jozsef L, Zouki C, Petasis NA, et al. Lipoxin A4 and aspirin-triggered 15-epi-lipoxin A4 inhibit peroxynitrite formation, NF-kappa B and AP-1 activation, and IL-8 gene expression in human leukocytes. Proc Natl Acad Sci U S A, 2002, 99:13266–13271.
    49. Lin HY, Juan SH, Shen SC, et al. Inhibition of lipopolysaccharide-induced nitric oxide production by flavonoids in RAW264.7 macrophages involves heme oxygenase-1. Biochem Pharmacol, 2003, 66:1821–1832.
    50. Levy BD, Fokin VV, Clark JM, et al. Polyisoprenyl phosphate (PIPP) signaling regulates phospholipase D activity: a“stop”signaling switch for aspirin-triggered lipoxin A4. FASEB J, 1999, 13:903–911.
    1. Robert A. Lewis, K. Frank A. Mediation of local homeostasis and inflammation by leukotrienes and other mast cell-dependent compounds. Nature, 1981,293: 103– 108.
    2. Nathan C. Points of control in inflammation. Nature, 2002, 420: 846– 852.
    3. Gilroy DW, Lawrence T, Perretti M, Rossi AG. Inflammatory resolution: new opportunities for drug discovery. Nat Rev Drug Discov, 2004 , 3:401– 416.
    4. Dumonde DC, Glynn LE. The production of arthritis in rabbits by an immunological reaction to fibrin. Br J Exp Pathol, 1962, 43: 373.
    5. Dumonde DC, Glynn LE. The reaction of guinea-pigs to autologous and heterologous fibrin implants. J Pathol Bacteriol, 1965, 90:649–657.
    6. Weissmann G, Becher G, Wiedermann G, Bernheimer AW. Studies on lysozymes. VII. Acute and chronic arthritis produced by intra-articular injections of streptolysin‘S’in rabbits. Am J Pathol, 1965, 46:129.
    7. Willoughby DA, Ryan GB. Evidence for a possible endogenous antigen in chronic inflammation. J Pathol, 1970, 101: 233–239.
    8. Recommendations for the prevention and treatment of glucocorticoid-induced osteoporosis: 2001 update. American College of Rheumatology Ad Hoc Committee on Glucocoritcoid-Induced Osteoporosis. Arthritis Rheum, 2001, 44:1496–1503.
    9. Beer HD, Fassler R, Werner S. Glucocorticoidregulated gene expression during cutaneous wound repair. Vitam Horm, 2000, 59: 217–239.
    10. Mukherjee D. Selective cyclooxygenase-2 (COX-2) inhibitors and potential risk of cardiovascular events. Biochem Pharmacol, 2002, 63:817–821.
    11. Serhan CN, Savill J. Resolution of inflammation: the beginning programs the end. Nat Immunol, 2005, 6:1191–1197.
    12. 12Urade Y, Hayaishi O. Prostaglandin D synthase: structure and function. Vitam Horm,2000, 58:89–120.
    13. Straus DS, Glass CK. Cyclopentenone prostaglandins:new insights on biological activities and cellular targets. Med Res Rev, 2001, 21:185–210.
    14. Jiang C, Ting AT, Seed B. PPAR-gamma agonists inhibit production of monocyte inflammatory cytokines. Nature, 1998, 391: 82–86.
    15. Ricote M, Li AC, Willson TM, Kelly CJ, Glass CK. The peroxisome proliferator-activated receptor-gamma is a negative regulator of macrophage activation. Nature, 1998, 391:79–82.
    16. Nosjean O, Boutin JA. Natural ligands of PPARgamma: are prostaglandin J(2) derivatives really playing the part? Cell Signal, 2002,14:573–583.
    17. Straus DS. et al. 15-deoxy-delta 12,14-prostaglandin J2 inhibits multiple steps in the NF-kappa B signaling pathway. Proc Natl Acad Sci USA, 2000, 97: 4844–4849.
    18. Perez-Sala D, Cernuda-Morollon E, Canada FJ. Molecular basis for the direct inhibition of AP-1 DNA binding by 15-deoxy-Delta 12,14-prostaglandin J2. J Biol Chem, 2003, 278:51251–51260.
    19. Chen C W, Chang YH, Tsi CJ, Lin WW. Inhibition of IFN-gamma-mediated inducible nitric oxide synthase induction by the peroxisome proliferator-activated receptor gamma agonist, 15-deoxy-delta 12, 14-prostaglandin J2, involves inhibition of the upstream Janus kinase/STAT1 signaling pathway. J Immunol, 2003,171: 979–988.
    20. Maggi LB. Anti-inflammatory actions of 15-deoxydelta 12,14-prostaglandin J2 and troglitazone: evidence for heat shock-dependent and-independent inhibition of cytokine-induced inducible nitric oxide synthase expression. Diabetes, 2000, 49:346–355.
    21. Petrova TV, Akama KT, Van Eldik LJ. Cyclopentenone prostaglandins suppress activation of microglia: down-regulation of inducible nitric-oxide synthase by 15-deoxy-Delta12,14-prostaglandin J2. Proc Natl Acad Sci USA, 1999, 96:4668–4673.
    22. Bernardo A, Levi G, Minghetti L. Role of the peroxisome proliferator-activatedreceptor-gamma (PPAR-gamma) and its natural ligand 15-deoxy-Delta12, 14-prostaglandin J2 in the regulation of microglial functions. Eur J Neurosci, 2000, 12: 2215–2223.
    23. Gosset P. Prostaglandin D2 affects the maturation of human monocyte-derived dendritic cells: consequence on the polarization of naive Th cells. J Immunol, 2003, 170: 494349–494352.
    24. Jackson SM. Peroxisome proliferator-activated receptor activators target human endothelial cells to inhibit leukocyte-endothelial cell interaction. Arterioscler Thromb Vasc Biol, 1999, 19: 2094–2104.
    25. Pasceri V, Wu HD, Willerson JT, Yeh ET. Modulation of vascular inflammation in vitro and in vivo by peroxisome proliferator-activated receptor-gamma activators. Circulation, 2000, 101:235–238.
    26. Zhang X, Wang JM, Gong WH, Mukaida N, Young HA. Differential regulation of chemokine gene expression by 15-deoxy-delta 12,14 prostaglandin J2. J Immunol, 2001, 166: 7104–7111.
    27. Zernecke A, Erl W, Fraemohs L, Lietz M, Weber C. Suppression of endothelial adhesion molecule up-regulation with cyclopentenone prostaglandins is dissociated from IkappaB-alpha kinase inhibition and cell death induction. FASEB J, 2001, 17: 1099–1101.
    28. Gilroy DW, Tomlinson A, Willoughby DA. Differential effects of inhibitors of cyclooxygenase (cyclooxygenase 1 and cyclooxygenase 2) in acute inflammation. Eur J Pharmacol, 1998, 355: 211–217.
    29. Gilroy DW. Inducible cyclooxygenase may have antiinflammatory properties. Nature Med, 1999, 5:698–701.
    30. Gilroy DW, Newson J, Sawmynaden P, Willoughby DA, Croxtall JD. A novel role for phospholipase A2 isoforms in the checkpoint control of acute inflammation. FASEB J, 2004, 18: 489–498.
    31. Gilroy DW. Inducible cyclooxygenase-derived 15-deoxy(Delta)12-14PGJ2 brings about acute inflammatory resolution in rat pleurisy by inducing neutrophil and macrophage apoptosis. FASEB J, 2003,17: 2269–2271.
    32. Ward C. Prostaglandin D2 and its metabolites induce caspase-dependent granulocyte apoptosis that is mediated via inhibition of I kappa B alpha degradation using a peroxisome proliferator-activated receptor-gamma-independent mechanism. J Immunol, 2002, 168: 6232–6243.
    33. Castrillo A. Potentiation of protein kinase C zeta activity by 15-deoxy-delta(12,14)-prostaglandin J(2) induces an imbalance between mitogen-activated protein kinases and NF-kappaB that promotes apoptosis in macrophages. Mol Cell Biol, 2003, 23: 1196–1208.
    34. Li L. 15-deoxy-delta 12,14-prostaglandin J2 induces apoptosis of human hepatic myofibroblasts. A pathway involving oxidative stress independently of peroxisomeproliferator-activated receptors. J Biol Chem, 2001, 276:38152–38158.
    35. Scher JU, Pillinger MH. 15d-PGJ2: the anti-inflammatory prostaglandin? Clin Immunol, 2005, 114: 100–109.
    36. Setoguchi K. Peroxisome proliferator-activated receptor-gamma haploinsufficiency enhances B cell proliferative responses and exacerbates experimentally induced arthritis. J Clin Invest, 2001,108: 1667–1675.
    37. Ando, M. Retrovirally introduced prostaglandin D2 synthase suppresses lung injury induced by bleomycin. Am J Respir Cell Mol Biol, 2003, 28: 582–591.
    38. Cuzzocrea S. The cyclopentenone prostaglandin 15-deoxy-delta(12,14)-PGJ2 attenuates the development of colon injury caused by dinitrobenzene sulphonic acid in the rat. Br J Pharmacol, 2003, 138: 678–688.
    39. Chatterjee P K. The cyclopentenone prostaglandin 15-deoxy-Delta(12,14)-prostaglandin J(2) ameliorates ischemic acute renal failure. Cardiovasc Res, 2004, 61: 630–643.
    40. Kawahito Y. 15-deoxy-delta(12,14)-PGJ(2) induces synoviocyte apoptosis and suppresses adjuvantinduced arthritis in rats. J Clin Invest, 2000, 106: 189–197.
    41. Diab, A. Peroxisome proliferator-activated receptorgamma agonist 15-deoxy-delta(12,14)-prostaglandin J(2) ameliorates experimental autoimmune encephalomyelitis. J Immunol, 2002, 168: 2508–2515.
    42. Matsuoka T. Prostaglandin D2 as a mediator of allergic asthma. Science, 2000, 287:2013–2017.
    43. Honda K. Prostaglandin D2 reinforces TH2 type inflammatory responses of airways to low-dose antigen through bronchial expression of macrophage-derived chemokine. J Exp Med, 2003, 198: 533–543.
    44. Lee TS, Tsai HL, Chau LY. Induction of heme oxygenase-1 expression in murine macrophages is essential for the anti-inflammatory effect of low dose 15-deoxy-delta 12,14-prostaglandin J2. J Biol Chem, 2003, 278: 19325–19330.
    45. Ryter SW, Otterbein LE, Morse D, Choi AM. Heme oxygenase/carbon monoxide signaling pathways: regulation and functional significance. Mol Cell Biochem, 2002, 234–235:249–263.
    46. Willis DM, Frederick R, Willoughby DA. Heme oxygenase: a novel target for the modulation of the inflammatory response. Nature Med, 1996, 2:87–90.
    47. Graca-Souza AV, Arruda MA, de Freitas MS, Barja-Fidalgo C, Oliveira PL. Neutrophil activation by heme: implications for inflammatory processes. Blood, 2002, 99: 4160–4165.
    48. Alcaraz MJ, Fernandez P, Guillen MI. Antiinflammatory actions of the heme oxygenase-1 pathway. Curr Pharm Des, 2003, 9: 2541–2551.
    49. Sato K. Carbon monoxide generated by heme oxygenase-1 suppresses the rejection of mouse-to-rat cardiac transplants. J Immunol, 2001, 166: 4185–4194.
    50. Hancock WW, Buelow R, Sayegh MH, Turka LA. Antibody-induced transplant arteriosclerosis is prevented by graft expression of anti-oxidant and anti-apoptoticgenes. Nature Med, 1998, 4: 1392–1396.
    51. Woo J, Iyer S, Mori N, Buelow R. Alleviation of graft-versus-host disease after conditioning with cobalt-protoporphyrin, an inducer of heme oxygenase-1. Transplantation, 2000, 69: 623–633.
    52. Serhan CN, Hamberg M, Samuelsson B. Trihydroxytetraenes: a novel series of compounds formed from arachidonic acid in human leukocytes. Biochem Biophys Res Commun , 1984,118:943–949.
    53. Serhan CN, Hamberg M, Samuelsson B. Lipoxins: novel series of biologically active compounds formed from arachidonic acid in human leukocytes. Proc Natl Acad Sci U S A, 1984,81:5335–5339.
    54. Palmblad J, Gyllenhammar H, Ringertz B. Effects of lipoxins A and B on functional responses of human granulocytes. Adv Exp Med Biol, 1988, 229:137–145.
    55. Fierro IM, Colgan SP, Bernasconi G, et al. Lipoxin A4 and aspirin-triggered 15-epi-lipoxin A4 inhibit human neutrophil migration: comparisons between synthetic 15 epimers in chemotaxis and transmigration with microvessel endothelial cells and epithelial cells. J Immunol, 2003,170:2688–2694.
    56. Jozsef L, Zouki C, Petasis NA, et al. Lipoxin A4 and aspirin-triggered 15-epi-lipoxin A4 inhibit peroxynitrite formation, NF-kappa B and AP-1 activation, and IL-8 gene expression in human leukocytes. Proc Natl Acad Sci U S A, 2002,99:13266–13271.
    57. Goh J, Baird AW, O'Keane C, et al. Lipoxin A(4) and aspirin-triggered 15-epi-lipoxin A(4) antagonize TNF-alpha-stimulated neutrophil-enterocyte interactions in vitro and attenuate TNF-alpha-induced chemokine release and colonocyte apoptosis in human intestinal mucosa ex vivo. J Immunol, 2001,167:2772–2780.
    58. Hachicha M, Pouliot M, Petasis NA, et al. Lipoxin (LX)A4 and aspirin-triggered 15-epi-LXA4 inhibit tumor necrosis factor 1alpha-initiated neutrophil responses and trafficking: regulators of a cytokine-chemokine axis. J Exp Med, 1999,189:1923–1930.
    59. Sodin-Semrl S, Spagnolo A, Mikus R, et al. Opposing regulation of interleukin-8 andNF-kappaB responses by lipoxin A4 and serum amyloid A via the common lipoxin A receptor. Int J Immunopathol Pharmacol, 2004,17:145–156.
    60. Jin SW, Zhang L, Lian QQ, et al. Posttreatment with aspirin-triggered lipoxin A4 analog attenuates lipopolysaccharide-induced acute lung injury in mice: the role of heme oxygenase-1. Anesth Analg, 2007,104:369–377.
    61. Romano M, Maddox JF, Serhan CN. Activation of human monocytes and the acute monocytic leukemia cell line (THP-1) by lipoxins involves unique signaling pathways for lipoxin A4 versus lipoxin B4: evidence for differential Ca2+ mobilization. J Immunol, 1996,157:2149–2154.
    62. Aliberti J, Hieny S, Reis e Sousa C, et al. Lipoxin-mediated inhibition of IL-12 production by DCs: a mechanism for regulation of microbial immunity. Nat Immunol, 2002,3:76–82.
    63. Ariel A, Chiang N, Arita M, et al. Aspirin-triggered lipoxin A4 and B4 analogs block extracellular signal-regulated kinase-dependent TNF-alpha secretion from human T cells. J Immunol JT - Journal of immunology (Baltimore, Md: 1950), 2003,170:6266-72.
    64. Ramstedt U, Ng J, Wigzell H, et al. Action of novel eicosanoids lipoxin A and B on human natural killer cell cytotoxicity: effects on intracellular cAMP and target cell binding. J Immunol, 1985,135:3434–3438.
    65. Van Dyke TE, Serhan CN. Resolution of inflammation: a new paradigm for the pathogenesis of periodontal diseases. J Dent Res, 2003, 82: 82–90.
    66. Claria J. Altered biosynthesis of leukotrienes and lipoxins and host defense disorders in patients with cirrhosis and ascites. Gastroenterology, 1998, 115:147–156.
    67. Levy BD. Multi-pronged inhibition of airway hyperresponsiveness and inflammation by lipoxin A(4). Nature Med, 2002, 8:1018–1023.
    68. Munger KA. Transfection of rat kidney with human 15-lipoxygenase suppresses inflammation and preserves function in experimental glomerulonephritis. Proc NatlAcad Sci USA, 1999, 96: 13375–13380.
    69. Leonard MO. 15-Epi-16-(para-fluorophenoxy)-lipoxin A(4)-methyl ester, a synthetic analogue of 15-epi-lipoxin A(4), is protective in experimental ischemic acute renal failure. J Am Soc Nephrol, 2002,13:1657–1662.
    70. Schottelius AJ. An aspirin-triggered lipoxin A4 stable analog displays a unique topical anti-inflammatory profile. J Immunol, 2002, 169:7063–7070.
    71. Souza MH, de Lima OM, Zamuner SR, Fiorucci S, Wallace JL. Gastritis increases resistance to aspirininduced mucosal injury via COX-2-mediated lipoxin synthesis. Am J Physiol Gastrointest Liver Physiol, 2003, 285: G54–G61.
    72. Serhan CN. Novel functional sets of lipid-derived mediators with antiinflammatory actions generated from omega-3 fatty acids via cyclooxygenase 2-nonsteroidal antiinflammatory drugs and transcellular processing. J Exp Med, 2000, 192: 1197–1204.
    73. Serhan CN. Resolvins: a family of bioactive products of omega-3 fatty acid transformation circuits initiated by aspirin treatment that counter proinflammation signals. J Exp Med, 2002, 196: 1025–1037.
    74. Hughes J. Neutrophil fate in experimental glomerular capillary injury in the rat. Emigration exceeds in situ clearance by apoptosis. Am J Pathol, 1997, 150: 223–234.
    75. Martin C. Chemokines acting via CXCR2 and CXCR4 control the release of neutrophils from the bone marrow and their return following senescence. Immunity, 2003,19:583–593.
    76. Bellingan GJ, Caldwell H, Howie SE, Dransfield I, Haslett C. In vivo fate of the inflammatory macrophage during the resolution of inflammation: inflammatory macrophages do not die locally, but emigrate to the draining lymph nodes. J Immunol, 1996, 157: 2577–2585.
    77. Bellingan GJ. Adhesion molecule-dependent mechanisms regulate the rate of macrophage clearance during the resolution of peritoneal inflammation. J Exp Med,2002, 196:1515–1521.
    78. Savill J, Fadok V. Corpse clearance defines the meaning of cell death. Nature, 2000, 407: 784–788.
    79. Nicholson DW. From bench to clinic with apoptosis-based therapeutic agents. Nature, 2000, 407: 810–816.
    80. Brunner T, Mueller C. Apoptosis in disease: about shortage and excess. Essays Biochem, 2003, 39: 119–130.
    81. Fadok VA, Bratton DL, Henson PM. Phagocyte receptors for apoptotic cells: recognition, uptake, and consequences. J Clin Invest, 2001, 108: 957–962.
    82. Savill J, Dransfield I, Gregory C, Haslett C. A blast from the past: clearance of apoptotic cells regulates immune responses. Nature Rev Immunol, 2002, 2: 965–975.
    83. Ward C, Dransfield I, Chilvers ER, Haslett C, Rossi A. Pharmacological manipulation of granulocyte apoptosis: potential therapeutic targets. Trends Pharmacol Sci, 1999, 20: 503–509.
    84. Giles KM, Hart SP, Haslett C, Rossi AG, Dransfield I. An appetite for apoptotic cells? Controversies and challenges. Br J Haematol, 2000, 109: 1–12.
    85. Rossi AG. Regulation of macrophage phagocytosis of apoptotic cells by cAMP. J Immunol, 1998, 160: 3562–3568.
    86. Liu Y. Glucocorticoids promote nonphlogistic phagocytosis of apoptotic leukocytes. J Immunol, 1999, 162: 3639–3646.
    87. Giles KM. Glucocorticoid augmentation of macrophage capacity for phagocytosis of apoptotic cells is associated with reduced p130Cas expression, loss of paxillin/pyk2 phosphorylation, and high levels of active Rac. J Immunol, 2001,167:976–986.
    88. Ren Y, Savill J. Proinflammatory cytokines potentiate thrombospondin-mediated phagocytosis of neutrophils undergoing apoptosis. J Immunol, 1995,154:2366–2374.
    89. McCutcheon JC. Regulation of macrophage phagocytosis of apoptotic neutrophils by adhesion to fibronectin. J Leukoc Biol, 1998, 64:600–607.
    90. Hart S P, Dougherty GJ, Haslett C, Dransfield I. CD44 regulates phagocytosis of apoptotic neutrophil granulocytes, but not apoptotic lymphocytes, by human macrophages. J Immunol, 1997, 159: 919–925.
    91. Teder P. Resolution of lung inflammation by CD44. Science, 2002, 296:155–158.
    92. Woolley KL. Eosinophil apoptosis and the resolution of airway inflammation in asthma. Am J Respir Crit Care Med, 1996, 154:237–243.
    93. Murphy FJ, Seery LT, Hayes I. Therapeutic approaches to the modulation of apoptosis. Essays Biochem, 2003, 39: 131–153.
    94. Degterev A, Boyce M, Yuan J. A decade of caspases. Oncogene, 2003, 22: 8543–8567.
    95. Earnshaw WC, Martins LM, Kaufmann SH. Mammalian caspases: structure, activation, substrates, and functions during apoptosis. Annu Rev Biochem, 1999, 68: 383–424.
    96. Graczyk PP. Caspase inhibitors as anti-inflammatory and antiapoptotic agents. Prog Med Chem, 2002, 39:1–72.
    97. Iwata A, Harlan JM, Vedder NB, Winn RK. The caspase inhibitor z-VAD is more effective than CD18 adhesion blockade in reducing muscle ischemia-reperfusion injury: implication for clinical trials. Blood, 2002, 100: 2077–2080.
    98. Natori S, Higuchi H, Contreras P, Gores GJ. The caspase inhibitor IDN-6556 prevents caspase activation and apoptosis in sinusoidal endothelial cells during liver preservation injury. Liver Transpl, 2003, 9:278–284.
    99. Hoglen NC. Characterization of IDN-6556: a livertargeted caspase inhibitor. J Pharmacol Exp Ther, 2004, 309:634–640 .
    100.Valentino KL, Gutierrez M, Sanchez R, Winship MJ, Shapiro DA. First clinical trial of a novel caspase inhibitor: anti-apoptotic caspase inhibitor, IDN-6556, improves liver enzymes. Int J Clin Pharmacol Ther, 2003, 41: 441–449.
    101.Karin, M. New twists in gene regulation by glucocorticoid receptor: is DNA binding dispensable? Cell, 1998, 93:487–490.
    102.Ghosh S, May MJ, Kopp EB. NF-kappa B and Rel proteins: evolutionarily conservedmediators of immune responses. Annu Rev Immunol, 1998, 16: 225–260.
    103.Khoshnan A. The NF-kappa B cascade is important in Bcl-xL expression and for the anti-apoptotic effects of the CD28 receptor in primary human CD4+ lymphocytes. J Immunol, 2000, 165: 1743–1754.
    104.Ward C. NF-kappaB activation is a critical regulator of human granulocyte apoptosis in vitro. J Biol Chem. 1999, 274: 4309–4318.
    105.Lawrence T, Gilroy DW, Colville-Nash PR, Willoughby DA. Possible new role for NF-kappaB in the resolution of inflammation. Nature Med, 2001, 7:1291–1297.
    106.Ishikawa H. Chronic inflammation and susceptibility to bacterial infections in mice lacking the polypeptide (p)105 precursor (NF-kappaB1) but expressing p50. J Exp Med, 1998, 187:985–996.
    107.Bohuslav J. Regulation of an essential innate immune response by the p50 subunit of NF-kappaB. J Clin Invest. 1998, 102:1645–1652.
    108.Chen LW. The two faces of IKK and NF-kappaB inhibition: prevention of systemic inflammation but increased local injury following intestinal ischemia-reperfusion. Nature Med, 2003, 9:575–581.
    109.Munck A, Guyre PM, Holbrook NJ. Physiological functions of glucocorticoids in stress and their relation to pharmacological actions. Endocr Rev, 1984, 5:25–44.
    110.Flower RJ, Parente L, Persico P, Salmon JA. A comparison of the acute inflammatory response in adrenalectomised and sham-operated rats. Br J Pharmacol, 1986, 87:57–62.
    111.Perretti M, Becherucci C, Scapigliati G, Parente L. The effect of adrenalectomy on interleukin-1 release in vitro and in vivo. Br J Pharmacol, 1989, 98: 1137–1142.
    112.Adcock IM. Molecular mechanisms of glucocorticosteroid actions. Pulm Pharmacol Ther, 2000, 13: 115–126.
    113.Auphan N, DiDonato JA, Rosette C, Helmberg A, Karin M. Immunosuppression by glucocorticoids: inhibition of NF-kappa B activity through induction of I kappa B synthesis. Science, 1995, 270:286–290.
    114.Buttgereit F, Burmester GR, Brand MD. Bioenergetics of immune functions: fundamental and therapeutic aspects. Immunol Today, 2000, 21: 192–199.
    115.Nissen RM, Yamamoto KR. The glucocorticoid receptor inhibits NFkappaB by interfering with serine-2 phosphorylation of the RNA polymerase II carboxy-terminal domain. Genes Dev, 2000, 14, 2314–2329.
    116.Flower RJ. Eleventh Gaddum memorial lecture. Lipocortin and the mechanism of action of the glucocorticoids. Br J Pharmacol, 1988, 94: 987–1015.
    117.Croxtall JD, Flower RJ. Lipocortin 1 mediates dexamethasone-induced growth arrest of the A549 lung adenocarcinoma cell line. Proc Natl Acad Sci USA, 1992, 89: 3571–3575.
    118.De Caterina R. Macrophage-specific eicosanoid synthesis inhibition and lipocortin-1 induction by glucocorticoids. J Appl Physiol, 1993, 75: 2368–2375.
    119.Mancuso F, Flower RJ, Perretti M. Leukocyte transmigration, but not rolling or adhesion, is selectively inhibited by dexamethasone in the hamster post-capillary venule. Involvement of endogenous lipocortin 1. J Immunol, 1995, 155: 377–386.
    120.Perretti M, Ahluwalia A, Harris JG, Goulding NJ, Flower RJ. Lipocortin-1 fragments inhibit neutrophil accumulation and neutrophil-dependent edema in the mouse. A qualitative comparison with an anti-CD11b monoclonal antibody. J Immunol, 1993, 151: 4306–4314.
    121.Gerke V, Moss SE. Annexins: from structure to function. Physiol Rev, 2002, 82: 331–371.
    122.Perretti M. Endogenous lipid- and peptide-derived anti-inflammatory pathways generated with glucocorticoid and aspirin treatment activate the lipoxin A4 receptor. Nature Med, 2002, 8:1296–1302.
    123.Le Y, Murphy PM, Wang JM. Formyl-peptide receptors revisited. Trends Immunol, 2002, 23:541–548.
    124.Perretti M, Gavins FN. Annexin 1: an endogenous antiinflammatory protein. NewsPhysiol Sci, 2003, 18: 60–64.
    125.Perretti M. Acute inflammatory response in the mouse: exacerbation by immunoneutralization of lipocortin 1. Br J Pharmacol, 1996, 117: 1145–1154.
    126.Hannon R. Aberrant inflammation and resistance to glucocorticoids in annexin 1–/– mouse. FASEB J, 2003, 17: 253–255.
    127.Yang YH. Modulation of inflammation and response to dexamethasone by Annexin 1 in antigen-induced arthritis. Arthritis Rheum, 2004, 50: 976–984.
    128.Perretti M. The annexin 1 receptor(s): is the plot unravelling? Trends Pharmacol Sci, 2003, 24:574–579.
    129.Hasko G, Cronstein BN. Adenosine: an endogenous regulator of innate immunity. Trends Immunol, 2004, 25: 33–39.
    130.Sitkovsky MV. Use of the A(2A) adenosine receptor as a physiological immunosuppressor and to engineer inflammation in vivo. Biochem Pharmacol, 2003, 65: 493–501.
    131.Cronstein BN, Eberle MA, Gruber HE, Levin RI. Methotrexate inhibits neutrophil function by stimulating adenosine release from connective tissue cells. Proc Natl Acad Sci USA, 1991, 88, 2441–2445.
    132.Cronstein BN, Naime D, Ostad E. The antiinflammatory mechanism of methotrexate. Increased adenosine release at inflamed sites diminishes leukocyte accumulation in an in vivo model of inflammation. J Clin Invest, 1993, 92:2675–2682.
    133.Montesinos MC. Adenosine A2A or A3 receptors are required for inhibition of inflammation by methotrexate and its analog MX-68. Arthritis Rheum, 2003, 48: 240–247.
    134.Xaus J. IFN-gamma up-regulates the A2B adenosine receptor expression in macrophages: a mechanism of macrophage deactivation. J Immunol, 1999, 162: 3607–3614.
    135.Hasko G, Deitch EA, Szabo C, Nemeth ZH, Vizi ES. Adenosine: a potential mediatorof immunosuppression in multiple organ failure. Curr Opin Pharmacol, 2002, 2: 440–444.
    136.Blackburn MR. Too much of a good thing: adenosine overload in adenosine-deaminase-deficient mice. Trends Pharmacol Sci, 2003, 24: 66–70.
    137.Catania A, Gatti S, Lipton JM. Targeting melanocortin receptors as a novel strategy to control inflammation. Pharmacol Rev, 2004, 6: 1–29.
    138.Fadok VA, Bratton DL, Frasch SC, Warner ML, Henson PM. The role of phosphatidylserine in recognition of apoptotic cells by phagocytes. Cell Death Differ, 1998, 5: 551–562.
    139.Arur S. Annexin I is an endogenous ligand that mediates apoptotic cell engulfment. Dev Cell, 2003, 4: 587–598.
    140.Huynh ML, Fadok VA, Henson PM. Phosphatidylserine-dependent ingestion of apoptotic cells promotes TGF-beta1 secretion and the resolution of inflammation. J Clin Invest, 2002, 109:41–50.
    141.Blom AB, van Lent PL, Holthuysen AE, van den Berg WB. Skewed balances in regulation of stimulating and inhibitory FC gamma receptors on macrophages of CIA sensitive mice. Inflamm Res, 2001, 50 (Suppl. 3): S155.

© 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