肺间质树突状细胞在多器官功能障碍综合征中的免疫激活与免疫耐受作用研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
目的:1.探讨肺间质树突状细胞(DCs)的免疫表型及功能变化在MODS模型早期过度炎症反应和晚期免疫抑制中的作用。2.证实MODS期肺间质DCs存在免疫耐受现象,探讨DCs负向调节作用对肺组织损伤的影响。3.应用N-乙酰-L-半胱氨酸(NAC)和Flt3配体(Flt3L)分别针对MODS早期过度炎症和晚期免疫抑制中DCs功能进行实验性干预调节,观察减轻肺组织损伤和改善脏器功能的效果。
     方法:腹腔注射酵母多糖复制Balb/c小鼠MODS模型。主要实验分组:急性损伤组(n=30):酵母多糖注射后48h取材;NAC治疗组(n=30):酵母多糖注射后3h、6h、12h、18h、24h、36h皮下注射NAC(100mg/kg体重),48h取材;MODS组(n=30):酵母多糖注射后10d取材;Flt3L治疗组(n=20):酵母多糖注射后48h开始每日皮下注射rFlt3L(10μg/只,连续注射7d),第10d取材;对照组(n=20):以等量生理盐水替代酵母多糖和NAC或rFlt3L按上述时间注射。每组小鼠按不同时间点麻醉后摘眼球取血,肺血管灌注,摘取双侧肺,按不同实验分别进行固定、超低温冻存、酶消化或匀浆处理。自动生化分析仪检测CO2、ALT、AST、Cr等血生化指标。生化法检测肺组织MPO活性。ELISA法检测血清和肺组织匀浆TNF-α含量。光镜观察肺组织病理变化,CD205免疫组化和CD11c免疫荧光标记肺间质DCs。密度梯度离心分离肺低密度单个核细胞,MACS磁珠分选DCs,流式细胞术进行DCs鉴定和免疫表型分析(I-Ad、CD86、PD-L1)。扫描和透射电镜观察DCs的超微形态。凝胶电泳迁移率变动分析法检测DCs胞核内NF-κB含量和活性。Real-time PCR检测DCs的CCR5/CCR7mRNA表达。体外LPS刺激DCs,ELISA法检测DCs孵育上清液细胞因子IL-12p70和IL-10含量。
     结果:1.正常Balb/c小鼠肺间质DCs分为三个亚群:髓系CD11c+CD11b+DCs、类浆系CD11c+CD45R\B220+DCs、淋巴系CD11c+CD11b-/CD45R\B220-DCs,分别占肺单个核细胞比例的6.35%、1.96%、3.21%。肺间质DCs高表达髓系标志物CD11b(39.59%),低表达MHC-Ⅱ/Ⅰ-Ad(8.97%)和共刺激分子CD86(3.16%)。DCs表面见长1-2μm树枝状突起,细胞器不发达,未见各级溶酶体和吞噬颗粒,细胞核形状不规则。2.急性损伤组(与对照组比较),血CO2含量降低,肺组织和血清TNF-α水平显著上升。肺含水量增加,肺组织MPO活性显著增高。肺间质CD11c+DCs数量增加,DCs体积增大,表面突起增多、变长。DCs表面I-Ad和共刺激分子CD86表达显著增高,趋化因子受体CCR5表达显著增加,CCR7表达轻度升高。DCs的NF-κB胞核内转移和活性增加。3.NAC治疗组(与急性损伤组比较),肺含水量、肺组织MPO活性和TNF-α水平明显降低,血CO2含量升高,DCs的NF-κB细胞核内转移和活性降低,DCs表面I-Ad和CD86表达降低,CCR5和CCR7表达下降。实验鼠急性损伤期死亡率降低20%。4.MODS组,血清ALT、AST、脂肪酶、淀粉酶和肌苷水平较对照组显著升高,血葡萄糖水平明显下降。肺间质髓系与类浆系DCs亚群比例及CD205+DCs数量较对照组明显降低,而淋巴系DCs比例明显升高。DCs表达I-Ad和CD86较对照组和急性损伤组明显下降,DCs表达耐受性分子PD-L1增高十分显著。电镜下DCs多呈退变与凋亡改变。在体外实验LPS刺激后,DCs分泌IL-12p70水平较对照组和急性损伤组明显下降而IL-10水平显著升高。5.Flt3L治疗组(与MODS组比较),髓系和类浆系DCs亚群比例较MODS组明显增加,淋巴系DCs比例降低。DCs表达I-Ad+、CD86及CD205+DCs数量较MODS组显著增加,DCs表达PD-L1明显下降。体外LPS刺激后,DCs分泌IL-12p70水平显著回升。肺组织损伤明显减轻,血清ALT、AST和肌苷水平较MODS组均有明显下降,血糖水平上升。实验鼠MODS期死亡率降低12%。
     结论:1.利用密度梯度离心和免疫磁珠分离、纯化肺间质DCs,获得DCs纯度高,活性良好,免疫表型稳定,但操作步骤复杂,技术要求较高。2.MODS急性损伤期,肺间质DCs大量成熟活化导致免疫应答反应亢进和炎症反应失控。肺间质DCs功能紊乱是引起肺过度炎症损伤的主要免疫细胞学因素。3.N-乙酰-L-半胱氨酸体内干预能够降低肺间质DCs的NF-κB胞核内转移和活性,从而抑制DCs分化成熟和趋化迁移,减弱炎症反应,进而减轻MODS急性损伤期的肺组织损伤。4.MODS期,肺间质DCs活性减低(表达I-Ad和CD86比例下降),发生免疫耐受(DCs表达PD-L1显著升高),并通过大量分泌IL-10发挥负向免疫调节作用。DCs的免疫耐受作用参与诱导和促进了免疫抑制与肺功能衰竭。5.Flt3L体内扩增DCs作用能够增加肺间质髓系和类浆系DCs亚群及成熟DCs比例,致DCs分泌IL-12p70增加,正向免疫调节作用增强。Flt3L可以明显逆转MODS期肺间质DCs的免疫耐受状态,进而减轻肺组织损伤和改善脏器功能。
Objective:1. To explore the relationship between immunophenotype and immunologic function of lung dendritic cells (DCs) and excessive inflammation and immunosuppression in multiple organs dysfunction syndrome (MODS).2. To confirm the immunologic tolerance of lung DCs in the stage of MODS and to explore its effect on immunosuppression.3. To observe the effects of alleviating lung damage and improving lung function by applying N-acetyl-L-cysteine (NAC) and fms-like tyrosine kinase 3 ligand(Flt3L) to regulate DCs'function in excessive inflammation and immunosuppression in MODS respectively.
     Methods:The model of MODS was replicated by zymosan injecting into the peritoneal cavity of Balb/c mice. The experimental groups were mainly divided as below:(1) acute damage groups (n=30). (2)NAC treating groups (n=30). (3)MODS groups (n=30). (4)Flt3L treating groups (n=20). (5)Control groups (n=20). The subcutaneous administration of NAC in dose of 100mg/kg body weight was at 3h, 6h,12h,18h,24h and 36h after zymosan injection. The subcutaneous administration of Flt3L in dose of 10μg/per day and per mouse, consecutive injection in 7 days was at 48h after zymosan injection. Symptom and mortality of the mice were observed. Biochemical indicator of CO2, ALT, AST and creatinine were detected by automatic biochemical analysator. MPO in lung tissue was measured by biochemical analysis. Pathological changes of lung were observed by light microscope. Specific surface markers CD205 and CDllc of lung DCs were detected by immunohistochemistry and immunofluorescence. Lung low density mononuclear cells were separated by density gradient centrifugation and then DCs were isolated by MACS microbeads. Purity identification and DCs'phenotypes of I-Ad, CD86 and PD-L1 were analyzed by flow cytometry. Ultrastructure of DCs was observed by scanning and transmission electron microscope. The protein and activity of NF-κB in the DCs' nuclear were detected electrophoretic mobility shift assay. The expression of CCR5/CCR7mRNA in DCs was measured by by real-time quantitative PCR. TNF-a from lung tissue homogenate and serum and IL-12p70 and IL-10 ELISA in supernatant were determined by ELISA kits.
     Results:In acute damage groups(comparing with control groups), the proportion of CO2 in blood decreased. The level of TNF-a, MPO and water content in lung increased dramatically. The numbers of CD11c+DCs rise. The significantly elevated expression of I-Ad, CD86 and CCR5 in DCs were detected. The activity of NF-κB in the DCs' nuclear increased notably. In NAC treating groups (comparing with acute damage groups), the level of TNF-a, MPO and water content in lung decreased notably, and the proportion of CO2 in blood increased. The remarkable decreased expression of I-Ad, CD86, CCR5 and CCR7 in DCs were measured. The activity of NF-κB in the DCs'nuclear declined. The mortality of the mice declined 20%. In MODS groups, the level of ALT, AST, lipase, amylase and creatinine increased dramatically and glucose decreased in serum comparing with control groups. The proportion myeloid and plasmacytoid DCs and CD205+DCs declined, but lymphoid DCs increased with comparison to that of control groups. The remarkable decreased expression of I-Ad and CD86 in DCs were measured, and DCs expressed high-level tolerant molecule PD-L1 markedly. DCs appeared morphologic change of apoptosis and degeneration. DCs secreted more IL-10 and less IL12-p70 after LPS stimulation in vitro contrast to control and acute damage groups. In Flt3L treating groups (comparing with MODS groups), the proportion myeloid and plasmacytoid DCs and CD205+DCs increased, and lymphoid DCs decreased. The remarkable increased expression of I-Ad and CD86 in DCs were detected, and DCs expressed low-level tolerant molecule PD-L1. DCs secreted more IL12-p70 after LPS stimulation in vitro. The mortality of the mice declined 12%.
     Conclusions:1.Isolated by density gradient centrifugation and MACS microbeads, Lung DCs acquired high purity and activity and stable phenotypes.2. In the stage of acute damage, the activation and maturation of lung DCs induced excessive immune response and inflammation, which caused inflammatory damage in lung.3. NAC had the effect on lightening lung damage in the stage of acute damage by inhibiting the maturation and migration of DCs with less NF-κB shift to nuclear.4. In the stage of MODS, lung DCs presented inactivition and induce immunologic tolerance. DCs excreted overmuch IL-10 to have effect on negative immunologic regulation and participated in immunosuppression.5. After Flt3L administration in vivo, lung DCs presented amplification and induce positive immunologic regulation. Flt3L reversed DCs'immunologic tolerance, which lightening lung injury and protect organ function.
引文
1. Sheng ZY, Yang HM. The concept and diagnosis of multiple system organ failure. Chin Med J,1994,107:563-569.
    2. Bone RC. Immunologic dissonance:a continuing evolution in our understanding of the systemic inflammatory response syndrome (SIRS) and the multiple organ dysfunction syndrome (MODS). Ann Intern Med,1996, 125:680-687.
    3. Angus DC, Linde WT, Lidicker J, et al. Epidemiology of severe sepsis in the United States:analysis of incidence, outcome, and associated costs of care. Crit Care Med,2001,29(7):1303-1310.
    4.盛志勇.严重创、烧伤后脓毒症与多器官功能障碍综合征的防治.中华创伤杂志,2005,21(1):11-14.
    5.姚咏明.免疫功能紊乱在脓毒症发病中的作用与意义.中国危重病急救医学,2007,19(6):138-141.
    6. Barie PS, Hydo LJ, Shou J, et al. Decreasing magnitude of multiple organ dysfunction syndrome despite increasingly severe critical surgical illness:a 17-year longitudinal study. J Trauma,2008,65(6):1227-35.
    7. Perl M, Chung CS, Garber M, et al. Contribution of anti-inflammatory/immune suppressive processes to the pathology of sepsis. Front Biosci,2006, 11:272-299.
    8. Joffre O, Nolte MA, Spirri R, et al. Inflammatory signals in dendritic cell activation and the induction of adaptive immunity. Immunol Rev,2009, 227(1):234-47.
    9. Dubsky P, Ueno H, Piqueras B, et al. Human dendritic cell subsets for vaccination. J Clin Immunol,2005,25(6):551-572.
    10. Tinsley KW, Grayson MH, Swanson PE, et al. Sepsis induces apoptosis and profound depletion of splenic interdigitating and follicular dendritic cells. J Immunol,2003,171(2):909-914.
    11.王宏伟,陆江阳.树突状细胞在脓毒症中变化与作用的研究进展.解放军医学杂志,2009,34(6):778-780.
    12. Ding YL, Chung CS, Newton S, et al. Polymicrobial sepsis induces divergent effects on splenic and peritoneal dendritic cell function in mice. Shock,2004, 22(2):137-144.
    13.曲冰杰,王宏伟,陆江阳,等.改善树突状细胞功能干预性治疗脓毒症的研究进展.感染、炎症、修复,2009,10(4):239-242.
    14.陆江阳,王晓虹,梁延杰,等.MODS大鼠胸腺树突状细胞病理改变及作用的研究.中国危重病急救医学,2001,13(11):675-677.
    15.陆江阳,李志宏,王晓虹,等.脾脏树突状细胞在多器官功能障碍综合征的变化及意义.中国危重病急救医学,2006,18(1):24-27.
    16.李志宏,陆江阳,王晓虹,等.多脏器功能不全综合征小鼠IL12的表达及其在免疫失衡中的作用.解放军医学杂志,2003,28(10):887-889.
    17.赵敏,陆江阳,吕艺.多器官功能障碍综合征小鼠脾脏中高迁移率族蛋白-1的表达.临床与实验病理学杂志,2005,21(5):600-603.
    18. Efron PA, Martins A, Minnich D, et al. Characterization of the systemic loss of dendritic cells in murine lymph nodes during polymicrobial sepsis. J Immunol, 2004,173(5):3035-3043.
    19. Wen H, Hogaboam CM, Gauldie J, et al. Severe sepsis exacerbates cell-mediated immunity in the lung due to an altered dendritic cell cytokine profile. Am J Pathol,2006,168(6):1940-1950.
    20. Hotchkiss RS, Tinsley kw, Swanson PE, et al. Depletion of dendritic cells, but not macrophages, in patients with sepsis. J Immunol,2002,168(5):2493-2500.
    21. Guisset O, Dilhuydy MS, Thiebaut R, et al. Decrease in circulating dendritic cells predicts fatal outcome in septic shock. Intensive Care Med,2007, 33(1):148-152.
    22.王宏伟,陆江阳,王晓虹,等.肾脏间质树突状细胞在多器官功能障碍综合
    征中的变化与作用.中华急诊医学杂志,2007,16(11):1146-1150.
    23.王宏伟,陆江阳,王晓虹,等.肝脏间质树突状细胞在多器官功能障碍综合征中的变化与意义.中国危重病急救医学,2007,19(10):596-599.
    24. Julie A, Lorraine B, Gordon R. The role of the coagulation cascade in the continuum of sepsis and acute lung injury and acute respiratory distress syndrome. Semin Respir Crit Care Med,2006,27(4):365-376.
    25. Robert M. A brief overview of acute respiratory distress syndrome. World J Surg,2006,30(8):1829-1834.
    26. Lorraine B. Pathophysiology of acute lung injury and the acute respiratory distress syndrome. Semin Respir Crit Care Med,2006,27(4):337-349.
    27. Jonathan E, Mitchell M, John J. Mechanical ventilation in sepsis-induced acute lung injury/acute respiratory distress syndrome:An evidence-based review. Crit Care Med,2004,32(11):s548-s553.
    28. Donald NC, Kim B. Innate immune control of pulmonary dendritic cell trafficking. Proc Am Thorac Soc,2007,4:234-239.
    29.王宏伟,田光,陆江阳,等.多器官功能障碍综合征肺IL-10的变化对树突状细胞功能的影响与意义.解放军医学杂志,2008,33(12):1409-1412.
    30.王宏伟,陆江阳,王晓虹,等.多器官功能障碍综合征小鼠肺脏间质树突状细胞的病理学观察.中华病理学杂志,2007,36(5):307-312.
    31. Goris RJ, Boekholtz WK, van Bebber IP, et al. Multiple organ failure and sepsis without bacteria. An experimental model Arch Surg,1986,124:897-901.
    32.一种改进的酵母多糖所致多器官衰竭实验模型.陆江阳,梁延杰,胡森,等.解放军医学杂志.1991,16(6):451-455.
    33.孙宇,陆江阳,王晓虹,等.小鼠迟发型MODS模型复制及病理学观察.中国危重病急救医学,2003,15:15-18.
    34. Yong-Jun Liu. Dendritic cell subsets and lineages, and their functions in innate and adaptive immunity. Cell,2001,106:259-262.
    35. Lu L, Woo J, Rao AS, ect. Propagation of dendritic cell progenitors from normal mouse liver using granulocyte/macrophage colony-stimulating factor and their
    maturational development in the presence of type-1 collagen. J Exp Med, 1994,179:1823.
    36. Barbara JM, Gwyneth KO, Donna FK, et al. Flt3 ligand preferentially increases the number of functionally active myeloid dendritic cells in the lungs of mice. J Immunol,2004,172:4077-4083.
    37. Ingel K.D., Ken R.B., Tania M., Different roles for human lung dendritic cell subsets in pulmonary immune defense mechanisms. Am J Respir Cell Mol Biol, 2006,35:387-393.
    38.侯国存,陆江阳,王宏伟,等.CCR7在多器官功能障碍综合征小鼠脾脏树突状细胞迁移变化中的作用.中国免疫学杂志,2009,25(12):1063-1066.
    39.王宏伟,陆江阳.核转录因子-κB信号途径调节树突状细胞功能的研究进展.感染、炎症、修复,2009,10(4):236-238.
    40. Hayden MS, Ghosh S. Signaling to NF-κB. Genes Dev,2004, 18(12):2195-2224.
    41.祝恒成,陈江华,何强,等.IκBα突变体对树突状细胞成熟及其信号通路的影响[J].中华实验外科杂志,2006,23(12):1574.
    42. Paula RK, Terrance WB, Larry P, et al. Cigarette smoke-induced oxidative stress suppresses generation of dendritic cell il-12 and il-23 through ERK-dependent pathways. J Immunol,2008,181:1536-1547.
    43. Lyudmila A, Lyakh, GK, Koski HA, et al. Adenovirus type 5 vectors induce dendritic cell differentiation in human CD14+ monocytes cultured under serum-free conditions. Blood,2002,99(2):600-608.
    44. Benjamim CF, Lundy SK, Lukacs NW, et al. Reversal of long-term sepsis-induced immunosuppression by dendritic cells. Blood,2005, 105(9):3588-3595.
    45. Davidson WJ, Dorscheid D, Spragg R, et al. Exogenous pulmonary surfactant for the treatment of adult patients with acute respiratory distress syndrome: results of a meta-analysis. Critical Care,2006,10(2):R41.
    46. Kronin V, Winkel K, Suss G, et al. A subclass of dendritic cells regulates the response of naive CD8 T cells by limiting their IL-2 production. J Immunol 1996,157:3819-27.
    47. Patrick GH. Antigen Presentation in the Lung. Am J Respir Crit Care Med, 2000,162:S151-S156.
    48.刘茜,陆江阳,王晓红,等.MODS小鼠脾脏树突状细胞对PD-L1、PD-1表达变化与意义.解放军医学杂志,2010,35(1):15-17.
    49. Greenwald RJ, Freeman GJ, Sharpe AH. The B7 family revisited. Annu Rev Immunol,2005:23:515-548.
    50. Barber DL, et al. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature,2006:439:682-687.
    51. Day CL, et al. PD-1 expression on HIV specific T cells is associated with T-cell exhaustion and disease progression. Nature,2006:443:350-354.
    52. Trautmann L, et al. Upregulation of PD-1 expression on HIV-specific CD8t T cells leads to reversible immune dysfunction. Nat Med 2006,12:1198-1202.
    53. Akbari O, Dekruyff RH, Umetsu DT. Pulmonary dendritic cells producing IL-10 mediate tolerance induced by respiratory exposure to antigen. Nat Immunol, 2001,2(8):725-731.
    54. Lyman SD, James L, Vanden BT, et al. Molecular cloning of a ligand for the flt3/flk-2 tyrosine kinase receptor. A proliferative factor for primitive hematopoietic cells. Cell,1993,75(6):1157-1167.
    55. Teleshova N, Jones J, Kenney J, et al. Short-term Flt3L treatment effectively mobilizes functional macaque dendritic cells. J Leukoc Biol,2004,75(6): 1102-1110.
    56. Toby PH, Coates SM, Barratt B, et al. Dendritic cell subsets in blood and lymphoid tissue of rhesus monkeys and their mobilization with Flt3 ligand. Blood,2003,102(7):2513-2521.
    57. Eugene M, Elizabeth D, Eileen R, et al. In vivo generation of human dendritic cell subsets by Flt3 ligand. Blood,2000,96(3):878-884.
    58. Maureen LD, Lu L, Vladimir MS, et al. In vivo administration of flt3 ligand markedly stimulates generation of dendritic cell progenitors from mouse liver. J Immunol,1997,159:4268-4278.
    59. Barbara JM, Gwyneth KO, Donna FK, et al. Flt3 ligand preferentially increases the number of functionally active myeloid dendritic cells in the lungs of mice. J Immunol,2004,172:4077-4083.
    60. Pulendran JL, Smith MJ, Schoenborn M. Prevention of peripheral tolerance by a dendritic cell growth factor:Flt3 ligand as an adjuvant. J Exp Med,1998, 188(11):2075-2082.
    61. Wysocka M, Montaner LJ, Karp CL.Flt3 ligand treatment reverses endotoxin tolerance-related immunoparalysis. Immunol,2005,174(11):7398-7402.
    62.田光,陆江阳,王宏伟,等.Flt3配体对多器官功能障碍综合征小鼠脾脏树突状细胞及细胞免疫功能的修复作用.中国危重病急救医学,2008,20(1):45-48.
    1. ANGUS DC, LINDE WT, LIDICKER J, et al. Epidemiology of severe sepsis in the United States:analysis of incidence, outcome, and associated costs of care. Crit Care Med,2001,29(7):1303-1310.
    2. VINCENT JL, SUN Q, DUBOIS MJ. Clinical trials of immunomodulatory therapies in severe sepsis and septic shock. Clin Infect Dis,2002, 34(8):1084-1093.
    3. PERL M, CHUNG CS, GARBER M, et al. Contribution of anti-inflammatory/immune suppressive processes to the pathology of sepsis Front Biosci,2006,11:272-299.
    4. DUBSKY P, UENO H, PIQUERAS B, et al. Human dendritic cell subsets for vaccination. J Clin Immunol,2005,25(6):551-572.
    5. LIU YJ. Dendritic cell subsets and lineages, and their functions in innate and adaptive immunity (J).Cell,2001,106 (3):259-262.
    6. TINSLEY KW, GRAYSON MH, SWANSON PE, et al. Sepsis induces apoptosis and profound depletion of splenic interdigitating and follicular dendritic cells [J]. J Immunol,2003,171(2):909-914.
    7. DING YL, CHUNG CS, NEWTON S, et al. Polymicrobial sepsis induces divergent effect on splenic and peritoneal dendritic cell function in mice [J]. Shock,2004,22(2):137-144.
    8. EFRON PA, MARTINS A, MINNICH D, et al. Characterization of the systemic loss of dendritic cells in murine lymph nodes during polymicrobial sepsis [J]. J Immunol,2004,173(5):3035-3043.
    9. WEN H, HOGABOAM CM, GAULDIE J, et al. Severe sepsis exacerbates cell-mediated immunity in the lung due to an altered dendritic cell cytokine profile. Am J Pathol,2006,168(6):1940-1950.
    10. HOTCHKISS RS, TINSLEY KW, SWANSON PE, et al. Depletion of dendritic cells, but not macrophages, in patients with sepsis[J]. J Immunol,2002,168(5): 2493-2500.
    11. GUISSET O, DILHUYDY MS, THIEBAUT R, et al. Decrease in circulating dendritic cells predicts fatal outcome in septic shock. Intensive Care Med,2007, 33(1):148-152.
    12.孙宇,陆江阳,王晓虹,等.酵母多糖致伤模型小鼠的胸腺树突状细胞病理观察.临床与实验病理学杂志,2005,21(1):95-99.
    13.陆江阳,李志宏,王晓虹,等.脾脏树突状细胞在多器官功能障碍综合征中的变化及意义.中国危重病急救医学,2006,18(1):24-27.
    14.王宏伟,陆江阳,王晓虹,等.多器官功能障碍综合征小鼠肺脏间质树突状细胞的病理学观察.中华病理学杂志,2007,36(5):307-312.
    15.王宏伟,陆江阳,王晓虹,等.肾脏间质树突状细胞在多器官功能障碍综合征中的变化与作用.中华急诊医学杂志,2007,16(11):1146-1150.
    16.陆江阳,王晓虹,刘茜,等.多器官功能障碍综合征52例尸检脾及脾树突状细胞的病理学观察.诊断病理学杂志,2007,14(3):172-175.
    17. SCUMPIA PO, MCAULIFFE PF, O'MALLEY KA, et al. CD11c+dendritic cells are required for survival in murine polymicrobial sepsis [J]. J Immunol, 2005,175(5):3282-3286.
    18. BENJAMIM CF, LUNDY SK, LUKACS NW, et al. Reversal of long-term sepsis-induced immunosuppression by dendritic cells [J]. Blood,2005, 105(9):3588-3595.
    19. HOTCHKISS RS, SWANSON PE, FREEMAN BD, et al. Apoptotic cell death in patients with sepsis, shock, and multiple organ dysfunction. Crit Care Med,
    1999,27(7):1230-1251.
    20. HOTCHKISS RS, NICHOLSON DW. Apoptosis and caspases regulate death and inflammation in sepsis. Nat Rev Immunol,2006,6(11):813-822.
    21. FLOHE SB, AGRAWAL H, SCHMITZ D, et al. Dendritic cells during polymicrobial sepsis rapidly mature but fail to initiate a protective Thl-type immune response. J Leukoc Biol,2006,79(3):473-481.
    22. MONNERET G, DEBARD AL, VENET F, et al. Marked elevation of human circulating CD4+CD25+ regulatory T cells in sepsis-induced immunoparalysis. Crit Care Med,2003,31(7):2068-2071.
    23. WYSOCKA M, ROBERRTSON S, RIEMANN H, et al. IL-12 suppression during experimental endotoxin tolerance:dendritic cell loss and macrophage hyporesponsiveness. J Immunol,2001,166(12):7504-7513.
    24. KAWASAKI T, HUBBARD WJ, CHOUDHRY MA, et al. Trauma-hemorrhage induces depressed splenic dendritic cell functions in mice. J Immunol,2006, 177(7):4514-4520.
    1. Liu YJ. Dendritic cell subsets and lineages, and their functions in innate and adaptive immunity [J].Cell,2001,106(3):259-262.
    2. Sen R and Baltimoer D. Multiple nuclear factors interact with the immunoglobulin enhancer sequences[J]. Cell,1986,46(5):705-716.
    3. Hayden, MS and Ghosh S. Signaling to NF-κB [J]. Genes Dev,2004, 18(12):2195-2224.
    4.林季,颜光涛.急性炎症反应中真核细胞转录因子-kB的信号转导作用[J].中国危重病急救医学,2003,15(1):45-48.
    5. Wu L, D'Amico A,Winkel KD,et al. RelB is essential for the development of myeloid-related CD8alpha-dendritic cells but not of lymphoid-related CD8alpha+dendritic cells[J]. Immunity,1998,9(6):839-847.
    6. Ouaaz F, Arron J, Zheng Y,et al.Dendritic cell development and survival require distinct NF-KappaB subunits[J]. Immunity,2002,16(2):257-270.
    7. Guermonprez P,Valladeau J,Zitvogel L,et al. Antigen presentation and T cell stimulation by dendritic cells[J].Annu Rev Immunol,2002,20(4):621-667.
    8. O'Sullivan BJ, Thomas R. CD40 ligation conditions dendritic cell antigen-presenting function through sustained activation of NF-κB [J]. J Immunol,2002,168(11):5491-5498.
    9. Sato T, Odagiri H, Ikenaga SK,et al. Chemosensitivity of human pancreatic carcinoma cells is enhanced by IkappaBalpha super-repressor[J]. Cancer Sci,2003,94(5):467-472.
    10.祝恒成,陈江华,何强,等.IκBα突变体对树突状细胞成熟及其信号通路的影响[J].中华实验外科杂志,2006,23(12):1574.
    11. Suresh R, Loc TN, Bogoljub C, et al. B7-DC/PD-L2 cross-linking induces NF-KB-dependent protection of dendritic cells from cell death [J]. J Immunol, 2007,178(3):1426-1432.
    12. Frasca L, Scotta C, Lombardi G, et al. Human anergic CD4+T cells can act as suppressor cells by affecting autologous dendritic cell conditioning and survival [J]. J Immunol.2002,168(3):1060-1068.
    13. Kriehuber E, Bauer W, Charbonnier AS, et al. Balance between NF-kappaB and JNK/AP-1 activity controls dendritic cell life and death[J]. Blood,2005,106(1):175-183.
    14. Ouaaz F, Arron J, Zheng Y, et al. Dendritic cell development and survival require distinct NF-kappaB subunits[J]. Immunity,2002,16(2):257-270.
    15. Dokter WH, Koopmans SB, Vellenga E. Effects of IL-10 and IL-4 on LPS-induced transcription factors (AP-1,NF-IL6 and NF-kappaB) which are involved in IL-6 regulation[J]. Leukemia,1996,10(8):1308-1316.
    16. Kadowaki N, Ho S, Antonenko S, et al. Subsets of human dendritic cell precursors express different Toll-like receptors and respond to different microbial antigens[J]. J Exp Med,2001,194(6):863-869.
    17. Zhang H, Tay PN, Cao W, et al. Integrin-nucleated Toll-like receptor(TLR) dimerization reveals subcellular targeting of TLRs and distinct mechanisms of TLR4 activation and signaling[J]. FEBS Lett,2002,532(1-2):171-176.
    18. Artis D, Kane CM, Fiore J, et al. Dendritic cell-intrinsic expression of NF-kappa B1 is required to promote optimal Th2 cell differentiation [J]. J Immunol 2005,174(11):7154-7159.
    19.叶蔚,项柏康.中医药调节核因子-kB活化的研究进展[J].中国中西医结合杂志,2004,24(10):951-954.
    20. Liu Q, Chen T, Chen G, et al. Triptolide impairs dendritic cell migration by inhibiting CCR7 and COX-2 expression through PI3-K/Akt and NF-kappaB pathways[J]. Mol Immunol,2007,44(10):2686-2696.
    21. Liu Q, Shu X, Sun A, et al. Plant-derived small molecule albaconol suppresses LPS-triggered proinflammatory cytokine production and antigen presentation of dendritic cells by impairing NF-kappaB activation[J]. Int Immuno pharmacol, 2008,8(8):1103-1111.
    22. Tas SW, Jong EC, Hajji N, et al. Selective inhibition of NF-kappaB in dendritic cells by the NEMO-binding domain peptide blocks maturation and prevents T cell proliferation and polarization [J]. Eur J Immunol 2005,35(4):1164-1174.
    23. Vale' rie V, Wim VB, Nathalie V, et al. N-Acetyl-L-cysteine inhibits primary human T cell responses at the dendritic cell level:association with NF-kB Inhibition [J]. J Immunol,1999,162(5):2569-2574.
    24. Fairchild PJ and Waldmann H. Dendritic cells and prospects for transplantation tolerance [J].Curr Opin Immunol,2000,12(5):528-535.
    25.俞静,林茂芳,牟海波,等.抗CD47单抗对人树突状细胞的免疫调控作用及其机制探讨[J].中国病理生理杂志,2004,20(12):2248-2252.

© 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