烧伤后高迁移率族蛋白B1对调节性T细胞免疫功能影响的实验与临床研究
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:
     实验一:(1)采用严重烫伤延迟复苏动物模型,明确高迁移率族蛋白B1(HMGB1)的变化及其与调节性T细胞(Treg)免疫功能的关系。(2)通过严重烫伤延迟复苏动物模型,阐明HMGB1对Treg分化成熟的受体作用机制。(3)探讨严重烫伤延迟复苏后HMGB1介导的Treg对树突状细胞(DC)免疫功能的影响及调节作用。(4)探讨严重烫伤延迟复苏后HMGB1介导的Treg对T淋巴细胞免疫功能的影响及调节途径。(5)观察丙酮酸乙酯(EP)对重度烫伤延迟复苏动物免疫功能及脏器损害的可能保护效应。
     实验二:(1)对临床严重烧伤患者进行动态监测,明确严重烧伤后病人外周血HMGB1、Treg分化成熟以及T淋巴细胞免疫功能的变化规律,探讨HMGB1、Treg及T淋巴细胞免疫功能改变在患者烧伤后发生脓毒症及不良预后中所起的作用及临床意义。(2)探讨严重烧伤后HMGB1、Treg及T淋巴细胞免疫指标间的相互关系及可能的作用机制。
     方法:
     1.采用重度烫伤延迟复苏动物模型,即雄性清洁级Wistar大鼠,浸于沸水中造成30%体表面积Ⅲ度烫伤。伤后延迟6小时抗休克复苏,在伤后6小时从腹腔给予林格液(40 ml/kg)抗休克治疗,此外在烫伤后12、24、36、48小时分别给予4 ml/次林格液腹腔内注射。实验分组:136只大鼠随机分为5组,(1)正常对照组(n=8):麻醉后活杀;(2)假烫伤组(n=32);(3)烫伤组(n=32);(4)EP治疗组(n=32):EP加入林格液中随补液给药(EP为28 mM);(5)晚期糖基化终末产物受体(RAGE)抗体(1 mg/ml)治疗组(n=32):烫伤后6、24小时从阴茎背静脉给予RAGE抗体(1 mg/kg)治疗。以上后4组实验动物再分为4个亚组,分别于烫伤后1、3、5、7天处死动物,无菌留取血和脾脏标本。分离血清,-20℃贮存。脾脏一部分无菌、无酶液氮冻存,另一部分用于提取Treg、DC和T淋巴细胞。Treg、DC的分离纯化采用MiniMACS免疫磁性分离系统进行。利用B细胞和单核细胞具有易粘附于尼龙纤维表面的特性,分离T淋巴细胞。采用流式细胞术检测细胞免疫表型和细胞表面受体,MTT法检测脾T淋巴细胞增殖,酶联免疫吸附试验(ELISA)检测血清、细胞孵育上清及组织匀浆液的细胞因子和T淋巴细胞活化的核因子(NF)-kB。采用荧光定量PCR检测目的基因表达水平。
     2.对106例烧伤总面积大于或等于30%的患者进行动态观察,对所采集临床病历资料进行分析。实验分组:(1)按烧伤总面积将患者分为三组:Ⅰ组41例(烧伤总面积30%~49%),Ⅱ组34例(烧伤总面积50%~69%),Ⅲ组31例(烧伤总面积70%~99%);(2)根据烧伤脓毒症的诊断标准,将患者进一步分为脓毒症组(59例)与非脓毒症组(47例);(3)根据脓毒症患者的预后情况,将其分为死亡组(17例)与存活组(42例)。同时设正常对照组(25位健康献血员)。分别于烧伤后1、3、7、14、21天采集患者外周静脉血,分离外周血T淋巴细胞,采用MiniMACS免疫磁性分离系统对外周血Treg进行分离纯化,同时分离血清,-20℃贮存。采用流式细胞术检测细胞免疫表型,MTT法检测T淋巴细胞增殖活性,ELISA检测血清HMGB1及细胞孵育上清的细胞因子水平,采用荧光定量PCR检测目的基因表达水平。
     结果:
     实验一:(1)烫伤延迟复苏导致动物脾脏HMGB1基因、蛋白表达和血清HMGB1水平在伤后1~7天显著升高,脾组织中HMGB1含量第1天达峰值,血清HMGB1水平于第3天达峰值。EP干预后烫伤动物脾脏HMGB1基因、蛋白表达和血清HMGB1水平1~7天均明显降低。RAGE抗体干预对烫伤动物脾脏和血清HMGB1无明显影响。(2)严重烫伤后脾脏Treg表面分子CTLA-4的表达1~5天明显增强,表面标记物Foxp3的表达1~7天明显增高,与假烫伤动物比较差异显著,表明严重烫伤可促使Treg成熟。EP及RAGE抗体干预组动物脾脏Treg表面CTLA-4和Foxp3的表达明显降低,与烫伤组比较均有统计学差异,提示EP处理和RAGE抗体干预可抑制Treg成熟。(3)严重烫伤后1~7天动物脾脏Treg表面RAGE的表达明显增强。EP干预后脾脏Treg表面RAGE的表达于1~7天显著降低,但仍明显高于假烫伤组。RAGE抗体干预后Treg表面RAGE表达1~7天显著降低。(4)烫伤延迟复苏后动物脾脏IL-10 mRNA表达以及Treg孵育液中IL-10含量明显增高。EP或RAGE抗体干预后,烫伤动物脾脏IL-10 mRNA表达和Treg孵育液中IL-10含量在伤后1~7天明显降低。(5)严重烫伤后1~7天脾脏DC表面CD80的表达与假烫伤组比较无统计学差异,CD86表达明显增强,MHCⅡ的表达仅在伤后第1天明显增高,DC摄取葡聚糖的能力与假烫伤组比较1~7天明显减低,表明DC向成熟发展,但表型表达不完全。EP或RAGE抗体干预后烫伤动物脾脏DC表面CD80、MHCⅡ的表达1~7天均明显升高,CD86的表达仍保持较高,DC摄取葡聚糖的能力在1~7天仍较低,表明DC成熟,表型表达趋于正常。(6)烫伤延迟复苏后1~7天脾T淋巴细胞增殖反应受抑制,IL-2mRNA表达1、3天无改变,5、7天明显降低,分泌IL-2的量1~7天显著下降,IL-2RαmRNA、IL-2Rα蛋白表达1~5天明显降低。EP或RAGE抗体干预能明显减轻1~7天烫伤对脾T淋巴细胞增殖的抑制,并且IL-2 mRNA表达、IL-2的分泌和IL-2RαmRNA、IL-2Rα表达明显上升。(7)严重烫伤后动物脾T淋巴细胞分泌IL-4的量比假烫伤组明显增加,而分泌IFN-γ的量显著降低,提示严重烫伤后T淋巴细胞向Th2漂移。用EP或RAGE抗体干预后,烫伤动物脾T淋巴细胞分泌IL-4的量明显下降,分泌IFN-γ水平则明显升高,提示EP或RAGE抗体干预可使烫伤后T淋巴细胞向Th1漂移。(8)烫伤组与假烫伤组比较,大鼠脾脏T淋巴细胞NF-kB活性伤后1~7天明显降低。应用EP或RAGE抗体干预均可明显提高1~7天烫伤动物脾T淋巴细胞NF-kB活性。(9)烫伤组与假烫伤组比较,大鼠血中丙氨酸转氨酶(ALT)、天门冬氨酸转氨酶(AST)及心肌激酶(CK-MB)伤后1~7天明显升高,血中肌酐(Cr)、尿素氮(BUN)1~5天明显升高。EP或RAGE抗体干预可明显降低烫伤后动物血清ALT、AST、Cr、BUN及CK-MB含量。(10)脾组织中HMGB1含量与Treg表面CTLA-4、Foxp3、RAGE、IL-10的表达呈正相关。脾组织中HMGB1含量与脾T淋巴细胞增值反应、表面IL-2Rα表达、孵育上清中IL-2、IFN-γ含量、NF-kB活性呈负相关,与血清sIL-2R含量、孵育上清中IL-4含量呈正相关。血清HMGB1含量与血清生化指标ALT、AST、Cr、BUN及CK-MB水平均呈正相关。
     实验二:(1)与正常对照组比较,严重烧伤患者不同烧伤面积组T淋巴细胞HMGB1基因表达及血浆HMGB1水平均明显升高,Ⅰ组与Ⅲ组比较存在明显差异;严重烧伤患者伤后各时间点HMGB1基因表达及血浆HMGB1水平均明显升高,脓毒症组HMGB1基因表达及血浆HMGB1水平在伤后7~21天显著高于非脓毒症组;脓毒症患者死亡组HMGB1基因表达及血浆HMGB1水平在伤后3~21天显著高于存活组。(2)与正常对照组比较,严重烧伤患者各烧伤面积组Treg表面分子CTLA-4及标记物FOXP3表达均明显增强,各组间比较存在明显差异;严重烧伤患者伤后不同时间点CTLA-4及FOXP3表达均明显升高,脓毒症组CTLA-4及FOXP3表达在伤后3~21天显著高于非脓毒症组;脓毒症患者死亡组CTLA-4及FOXP3表达在伤后3~21天显著高于存活组。(3)与正常对照组比较,严重烧伤患者不同烧伤面积组Treg IL-10基因/蛋白表达及TGF-β1水平均显著升高,各组间比较存在明显差异;严重烧伤患者伤后各时间点Treg IL-10基因/蛋白表达及TGF-β1水平均明显升高,脓毒症组Treg IL-10基因/蛋白表达及TGF-β1水平在伤后3~21天显著高于非脓毒症组;脓毒症患者死亡组Treg IL-10基因/蛋白表达及TGF-β1水平在伤后3~21天显著高于存活组。(4)与正常对照组比较,严重烧伤患者各烧伤面积组外周血T淋巴细胞增殖反应活性及IL-2基因/蛋白表达水平均明显降低,Ⅰ组与Ⅲ组比较存在明显差异;严重烧伤患者伤后T淋巴细胞增殖反应活性及IL-2基因/蛋白表达水平均明显降低,脓毒症组T淋巴细胞增殖反应活性及IL-2基因/蛋白表达水平在伤后3~21天显著低于非脓毒症组;脓毒症患者死亡组T淋巴细胞增殖反应活性及IL-2基因/蛋白表达水平在伤后3~21天显著低于存活组。(5)与正常对照组比较,严重烧伤患者各烧伤面积组T淋巴细胞分泌IL-4水平明显升高,而分泌IFN-γ水平显著降低;与Ⅰ组比较,Ⅲ组IL-4分泌水平明显升高,而IFN-γ分泌水平显著降低;严重烧伤患者伤后各时间点T淋巴细胞分泌IL-4水平明显升高,而分泌IFN-γ水平显著降低;脓毒症组T淋巴细胞分泌IL-4/IFN-γ水平在伤后3~21天显著高/低于非脓毒症组;脓毒症患者死亡组T淋巴细胞分泌IL-4/IFN-γ水平在伤后3~21天显著高/低于存活组。(6)血浆HMGB1含量与Treg及T淋巴细胞各实验组相关免疫指标均无明显相关性。Treg各实验组免疫指标(部分或全部)与T淋巴细胞分泌IL-4水平呈正相关,与T淋巴细胞增值反应活性、IL-2及IFN-γ分泌水平呈负相关。
     结论:
     实验一:(1)HMGB1在严重烫伤后具有升高较晚、持续时间较长的特征。HMGB1可能参与了烫伤后脓毒症所致多器官损害的发病过程。(2)HMGB1的持续升高可刺激Treg向成熟发展,从而介导T淋巴细胞增殖反应低下,并向Th2漂移,免疫功能抑制。(3)烫伤后大量的HMGB1可能主要通过受体RAGE介导Treg表型表达、Treg功能成熟。(4)HMGB1介导的Treg功能成熟可影响烫伤后DC细胞向成熟发育,但其表型表达异常,功能出现障碍。(5)HMGB1介导的Treg功能成熟还可通过下调T淋巴细胞NF-kB活性而减少了IL-2基因表达和蛋白合成,进而影响T淋巴细胞的增殖反应和免疫功能。(6)EP可能主要通过抑制HMGB1的合成释放,改善烫伤延迟复苏动物细胞免疫功能紊乱,保护动物的主要脏器功能。
     实验二:(1)HMGB1在严重烧伤后具有增高较晚、持续时间较长的特征,其含量与烧伤严重程度、并发脓毒症及患者预后相关,HMGB1参与了严重烧伤后机体免疫紊乱、发生脓毒症并致患者死亡的病理过程。(2)严重烧伤后Treg功能向成熟发展,使其免疫抑制功能充分发挥,从而可能导致机体的免疫功能紊乱。其表面分子表达及细胞因子分泌在不同烧伤面积、是否并发脓毒症及是否存活等不同组间存在显著差异,Treg可通过分泌抑制性细胞因子参与了严重烧伤后机体免疫失衡、诱发脓毒症甚至最终造成患者死亡的病理过程。(3)严重烧伤后T淋巴细胞免疫功能受到抑制,并引起T淋巴细胞向Th2漂移。烧伤程度越重、脓毒症发生率和患者死亡率越高,机体免疫抑制状态也越明显。(4)HMGB1含量与Treg及T淋巴细胞相关免疫指标均无明显相关,而Treg免疫指标与T淋巴细胞免疫功能指标间存在显著相关性(正相关或负相关)。说明Treg可能对严重烧伤后T淋巴细胞免疫功能低下,并出现Th1向Th2极化现象具有重要影响。
Objective:
     Part 1. The present study was performed: (1) to investigate in vivo the changes in high mobility group box 1 protein (HMGB1) and its effect on the maturation of regulatory T cell (Treg) after severe thermal injury; (2) to identify if RAGE is the possible receptor on surface of Treg to bind HMGB1; (3) to study in vivo the effect of HMGB1 stimulated Treg on splenic dendritic cell (DC) and its potential mechanisms after severe thermal injury; (4) to investigate the effect of the HMGB1 stimulated Treg on splenic T lymphocyte-mediated immunity and its potential mechanism; and (5) to observe the protective effect of ethyl pyruvate (EP) on immune function and organ function in burn rats with delayed resuscitation.
     Part 2. The clinical study was performed to investigate the systemic release and kinetics of HMGB1, and to observe the potential roles of the maturation of regulatory T cell and T lymphocyte-mediated immunity in severely burned patients. Meanwhile, the significance of changes in plasma HMGB1 levels, Treg and T lymphocyte immunity and their relationship with sepsis as well as outcome of the patients were investigated in patients after major burns.
     Methods:
     1. A widely used technique for induction of full-thickness scald injury was used in the present study. For rats with burn injury, and the dorsal and lateral surfaces of animals were shaved under anesthesia. After being secured in a protective template with an opening corresponding to 30% of the total body surface area, and the exposed skin of the back was immersed in 99°C water for 12 s. Sham-injured rats were subjected to the same procedure except the temperature of the bath was of room temperature. 40 ml/kg lactated Ringer's solution was administered i.p 6 hours after the injury for delayed resuscitation, followed by i.p injection of 4 ml at 12,24,36 and 48 h after burn injury, respectively.
     One hundred and thirty six male Wistar rats were randomly divided into five groups as follows: normal control group (8 rats), sham burn group (32 rats), burn group (32 rats), burn with ethyl pyruvate (EP) treatment group (32 rats), and burn with anti-RAGE (advanced glycation end-products) antibody treatment group (32 rats), and the later four groups were further divided into four subgroups of 8 rats each, which were sacrificed on postburn days (PBD) 1, 3, 5 and 7 respectively. EP was added to lactated Ringer's solution (EP 28 mM) in EP treatment group.. Anti-RAGE antibody lmg/kg was given via dorsal penile vein at 6 h and 24 h respectively after burn injury in anti-RAGE antibody treatment group. Animals of all groups were sacrificed at designated time points, and blood as well as spleen samples were harvested aseptically to determine organ damage related variables and levels of various cytokines. Spleen was divided into two portions as following: one portion was used to detect gene and protein expression levels of HMGB1 and IL-10, and the other portion was used to procure Treg and DC by MACS microbeads and T cell by using column of nylon wool. Cells were cultured, and phenotypes were analyzed by flow cytometry and the contents of cytokines released into supernatants were also determined. All cytokines in blood, supernatant and tissue, as well as activated NF-kB of T cell were determined by ELISA kits for rats. Gene expression was measured by real-time quantitative PCR taken GAPDH as the internal standard.
     2. One hundred and six patients with total burn surface area larger than 30% were included in the present study, and they were divided into three burn size groups: 30%-49% total body surface area (TBSA) burn (group I, n=41), 50%-69% TBSA burn (group II, n=34), and >70% TBSA burn (groupIII, n=31). According to wheather there was development of sepsis or not, patients were divided into sepsis group (n=59) and none-sepsis group (n=47); then the patients with sepsis were further divided into non-survival group (n=17) and survival group (n=42). Healthy volunteers served as normal controls (n=25). The periphery blood samples were collected on PBD 1,3,7,14, and 21. The blood samples were divided into two portions as following: one portion was used to procure T cell and Treg by MACS microbeads, and the other portion was used to detect levels of HMGB1 in serum. Cells were cultured, and phenotypes were analyzed by flow cytometry and the contents of cytokines released into supernatants were also determined. All cytokines in blood, supernatant and tissue were determined by ELISA kits for human. Gene expression was assessed by real-time quantitative PCR taken GAPDH as the internal standard.
     Results:
     Part 1. (1) The significantly elevated expression levels of splenic HMGB1 gene and protein and levels of serum HMGB1 were detected on PBD 1-7, and the elevation was significantly inhibited by the treatment of EP, but not by anti-RAGE antibody. (2) The expression levels of CTLA-4 on PBD 1-5 and levels of Foxp3 in Treg on PBD 1 -7 were significantly enhanced in burn rats in comparison to Treg from sham-injured rats. Treatment with EP or anti-RAGE antibody to inhibit HMGB1 could significantly decrease the expression levels of CTLA-4 and Foxp3 of Treg. (3) The expression of RAGE on the surface of Treg from burned rats was found to be markedly elevated on PBD 1-7 compared with sham-injured rats, and it was not influenced by treatment of EP, but was blocked by treatment of anti-RAGE antibody. (4) The expression levels of splenic IL-10 mRNA and levels of IL-10 produced by Treg in supernatant were found to be significantly enhanced on PBD 1-7, and they were significantly inhibited by treatment of EP, but not by anti-RAGE antibody. (5) It was noted that DC expressed similar levels of CD80, strongly enhanced levels of CD86 and slightly enhanced levels of MHC class II on PBD 1-7 compared with DC from sham-injured rats. The capacity of DC to engulf dextran was decreased markedly after burn injury. Treatment with EP or anti-RAGE antibody to inhibit HMGB1 could significantly raise the expression levels of CD80, MHC class II of DC, but not the capacity of DC to engulf dextran. (6) The T cell proliferative activity in response to ConA in burn-injured rats was significantly suppressed on PBD 1-7 as compared with sham-injured rats, and gene/protein expressions of IL-2 and expressions of IL-2Ra of T cells in burn-injured rats were simultaneously suppressed after burn injury to different extent. EP or anti-RAGE antibody treatment could restore T cell proliferative activity response to Con A, gene/protein expressions of IL-2, and expression of IL-2Ra after burn injury. (7) Levels of IL-4 produced by T cell as a response to Con A increased markedly after burn injury, whereas levels of IFN-γlowered markedly, indicating that Th cells might shift into Th2 cells. Intervention with EP or anti-RAGE antibody could significantly inhibit the release of IL-4 and enhance the production of IFN-γby T cell response to Con A after thermal injury, indicating that EP or anti-RAGE antibody intervention might influence the polarization of T cells in animals subjected to thermal injury and induced Th cells to drift to Th1 cells. (8) The NF-kB activation of splenic T cell was downregulated significantly on PBD 1-7. Treatment with EP or anti-RAGE antibody could completely restored the NF-kB activity of splenic T cell after burn injury. (9) The serum ALT, AST, Cr, BUN and CK-MB levels were significantly elevated after burns, and treatment with EP or anti-RAGE antibody could inhibit these increase to different extent. (10) The levels of splenic HMGB1 were positively correlated with the levels of CTLA-4, Foxp3, RAGE, IL-10, sIL-2R, IL-4, and were negatively correlated with the levels of IL-2Rα, IL-2, IFN-γand T cell proliferative activity. Significant positive correlations were also found between the levels of serum HMGB1 and levels of serum ALT, AST, Cr, BUN and CK-MB.
     Part 2. (1) The gene/protein levels of blood HMGB1 were significantly elevated on PBD 1-21 in patients with various burn sizes compared with normal controls, and there were obvious differences between group I and group III. The blood HMGB1 mRNA and plasma HMGB1 levels were significantly higher in septic patients than those without sepsis on PBD 7-21. Among septic patients, the HMGB1 levels in the survival group were markedly lower than those with fatal outcome on PBD 3-21. (2) Increased expressions of CTLA-4 and Foxp3 on the surface of Treg from burned patients were found on PBD 1-21 compared with normal control group, and there were obvious differences among patients with various burn sizes. The expressions of CTLA-4 and Foxp3 were significantly higher in patients with serious burns at all time points, and they were even higher septic patients than those without sepsis on PBD 3-21. Among septic patients, the expressions of CTLA-4 and Foxp3 in the survival group were obviously lower than those with fatal outcome on PBD 3-21. (3) Elevated gene/protein expression of IL-10 and TGF-β1 in Treg from burned patients were detected on PBD 1-21 in comparison to normal controls, and there was obvious difference among patients with different extent of burn injury. The gene/protein expression of IL-10 and TGF-β1 in Treg were significantly higher in septic patients than those without sepsis on PBD 3-21. Among septic patients, expression levels of IL-10 and TGF-β1 in the survival group were obviously lower than those with fatal outcome on PBD 3-21. (4) The T cell proliferative activity in response to PHA and gene/protein expression of IL-2 of T cell in burn-injured patients were significantly suppressed on PBD 1-21 compared with normal control group, and there were obvious differences between group I and group III. The T cell proliferative activity in response to PHA and gene/protein expression of IL-2 of T cell were significantly lower in septic patients than those without sepsis on PBD 3-21. Among septic patients, the T cell proliferative activity in response to PHA and gene/protein expression of IL-2 in the survival group were markedly higher than those with fatal outcome on PBD 3-21. (5) Levels of IL-4 produced by T cell response to PHA increased markedly after burn injury, whereas levels of IFN-γdecreased markedly as compared with normal control group, and there were obvious differences between group I and groupIII, indicating that Th cells might have shifted to Th2 cells. The same results were found in non-septic patients and the survival group as compared with those with sepsis and the non-survival group on PBD 3-21. (6) No significant correlations were found between serum HMGB1 levels and the immune indexes of Treg and T cells. In addition, the immune indexes of Treg were positively correlated with levels of IL-4 produced by T cells, but were negatively correlated with levels of IL-2, IFN-γand T cell proliferative activity.
     Conclusion:
     Part 1. Serum and splenic HMGB1 were markedly up-regulated for a prolonged period but delayed after severe thermal injury. HMGB1 might play an important role in the development of excessive inflammatory response and subsequent multiple organ dysfunction syndrome. The excessive release of HMGB1 might stimulate splenic Treg to mature (which might mainly induced by binding RAGE on the surface of Treg), and further stimulate splenic DC to mature abnormally, thereby inducing suppression of proliferative activity of T lymphocytes and drifting of Th1 to Th2 after burn injury. NF-κB signaling might be involved in the normal Treg mediating suppression of T lymphocyte associated with excessive release of HMGB1. EP has a therapeutic potential for suppressing HMGB1-induced immune dysfunction and ameliorating multiple organ injury.
     Part 2. (1) Plasma HMGB1 was markedly elevated for a delayed but prolonged period after severe thermal injury in patients. Extensive burn injury could result in increased serum HMGB1 levels, and this phemenon was found to be associated with the development of sepsis and fatal outcome of burned patients.. (2) Severe burn injury per se could lead to activation and maturation of Treg cells, thus invoking its immunodepressive activity to full extent, resulting in immunosuppression. Similar to HMGB1, different degree of elevated levels of cytokines produced by Treg and activation markers on Treg surface might also take a role involved in the pathogenesis of sepsis and mortality in burn patients. (3) Suppression of T lymphocyte immune function and drifting of Th1 to Th2 were induced by severe burn injury. The immunosuppressive state of T lymphocytes was related to the extent of burn injury, development of sepsis and poor outcome in burned patients. (4) Plasma HMGB1 levels showed no significant correlations with the immune indexes of Treg and T lympholeukocyte, but the immune indexes of Treg were significantly correlated with T lymphocyte immune function, suggesting that Treg might have a potential effect on suppression of the proliferative activity, cytokine release of T cells and drifting Th1 to Th2 following major burns.
引文
1.姚咏明,柴家科,林洪远主编.现代脓毒症理论与实践,第1版,北京:科学出版社,2005,96-130.
    2.Mantell LL,Parrish WR,Ulloa L.HMGB-1 as a therapeutic target for infectious and inflammatory disorders.Shock,2006,25(1):4-11.
    3.Fang WH,Yao YM,Shi ZG,et al.The significance of changes in high mobility group-1 protein mRNA expression in rats after thermal injury.Shock,2002,17(4):329-333.
    4.李红云,姚咏明,施志国,等.高迁移率族-1蛋白在烫伤后金葡菌脓毒症中的改变与意义.中华实验外科杂志,2001,18(3):239-241.
    5.Zhang LT,Yao YM,Lu JQ,et al.Sodium butyrate prevents lethality of severe sepsis in rats.Shock,2007,27(6):672-677.
    6.姚咏明,刘辉.对高迁移率族蛋白B1作用的新认识.中国危重病急救医学,2005,17(7):385-387.
    7.Wang H,Ward MF,Fan XG,et al.Potential role of high mobility group box 1in viral infectious diseases.Viral Immunol,2006,19(1):3-9.
    8.Chatita TA.Role of regulatory T cells in human diseases.J Allergy Clin Immunol,2005,116(5):949-959.
    9.Belkaid Y.The role of CD4+CD25+ regulatory T cells in Leishmania infection.Exp Opin Biol Ther,2003,3(6):875-885.
    10.Zheng SG,Meng LZ,Wang JH,et al.Transfer of regulatory T cells generated ex vivo modifies graft rejection through induction of tolerogenic CD4+CD25+ cells in the recipient.Int Immunol,2006,18(2):279-289.
    11.Le Gall F,Reusch U,Moldenbauer G,et al.Immunosuppression properties of anti-CD3 single-chain Fv and diabody.J Immunol Methods,2004,285(1):111-127.
    12.Zoltan F.Development and function of CD25+CD4+ regulatory T cells.Curr Opin Immunol,2004,16(2):203-208.
    13.Zheng SG,Wang JH,Stohl W,et al.TGF-beta requires CTLA-4 early after T cell activation to induce FoxP3 and generate adaptive CD4+CD25+ regulatory cells.J Immunol,2006,176(6):3321-3329.
    14.Shevach EM.CD4+CD25+ suppressive T cells:more questions than answers.Nat Rev Immunol,2002,2(6):389-400.
    15.Fehervari Z,Sakaguchi S.Development and function of CD25+CD4+regulatory T cells.Curr Opin lmmunol,2004,16(2):203-208.
    16.Jonulei H,Schmitt E.The Regulatory T cell Family:distinct subsets and their interrelations.J Immunol,2003,171(12):6323-6327.
    17.张立天,姚咏明,陆家齐,等.高迁移率族蛋白-1在脓毒症所致多器官功能损伤中的作用.中华外科杂志,2003,41(4):303-306.
    18.王忠堂,姚咏明,盛志勇,等.休克期切痂对烫伤大鼠肝、肺组织高迁移率族蛋白B1表达及促炎/抗炎平衡的影响.中华外科杂志,2004,42(14):839-844.
    19.Wang H,Bloom O,Zhang M,et al.HMGB1 as a late mediator of endotoxin lethality in mice.Science,1999,285(5425):248-251.
    20.Perry SE,Mostafa SM,Wenstone R,et al.Is low monocyte HLA-DR expression helpful to predict outcome in severe sepsis? Intensive Care Med,2003,29(8):1245-1252.
    21.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.
    22.徐姗,姚咏明,董宁,等.高迁移率族蛋白B1对大鼠脾脏树突状细胞表面共刺激分子表达的影响.中华创伤杂志,2006,22(8):574-578.
    23.Krook H,Hagberg A,Song Z,et al.A distinct Th1 immune response precedes the described Th2 response in islet xenograft rejection.Diabetes,2002,51(1):79-86.
    24.Kim HY,Kim HJ,Min HS,et al.NKT cells promote antibody-induced joint inflammation by suppressing transforming growth factor betal production.J Exp Med,2005,201(1):41-47.
    25.Bradford MM.A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding.Anal Biochem,1976,72(27):248-254.
    26.姚咏明,盛志勇.高迁移率族蛋白-1在脓毒症发病中的作用与意义.解放军医学杂志,2002,27(9):753-756.
    27.Zayed H,Izsvak Z,Khare D,et al.The DNA-bending protein HMGB1 is a cellular cofactor of Sleeping Beauty transposition.Nucleic Acids Res,2003,31(9):2313-2322.
    28.Calogero S,Grassi F,Aguzzi A,et al.The lack of chromosomal protein Hmgl does not disrupt cell growth but causes lethal hypoglycaemia in newborn mice.Nat Genet,1999,22(3):276-280.
    29.Andersson U,Wang H,Palmblad K,et al.High mobility group 1 protein (HMG-1)stimulates proinflammatory cytokine synthesis in human monocytes.J Exp Med,2000,192(4):565-570.
    30.Sunden-Cullberg J,Norrby-Teglund A,Treutiger CJ.The role of high mobility group box-1 protein in severe sepsis.Curr Opin Infect Dis,2006,19(3):231-236.
    31.Yang H,Ochani M,Li J,et al.Reversing established sepsis with antagonists of endogenous high-mobility group box 1.Proc Natl Acad Sci USA,2004,101(1):296-301.
    32.Sunden-Cullberg J,Norrby-Teglund A,Rouhiainen A,et al.Persistent elevation of high mobility group box-1 protein(HMGB1)in patients with severe sepsis and septic shock.Crit Care Med,2005,33(3):564-573.
    33.Ulloa L,Ochani M,Yang H,et al.Ethyl pyruvate prevents lethality in mice with established lethal sepsis and systemic inflammation.Proc Natl Acad Sci U S A,2002,99(19):12351-12356.
    34.王文江,姚咏明,咸力明,等.丙酮酸乙酯对烫伤延迟复苏大鼠多器官功能及死亡率的影响.中国危重病急救医学,2006,18(3):132-135.
    35.Yasuda T,Ueda T,Takeyama Y,et al.Significant increase of serum high-mobility group box chromosomal protein 1 levels in patients with severe acute pancreatitis.Pancreas,2006,33(4):359-363.
    36.Sakamoto Y,Mashiko K,Matsumoto H,et al.Effect of direct hemoperfusion with a polymyxin B immobilized fiber column on high mobility group box-1(HMGB-1)in severe septic shock:report of a case.Asaio J,2006,52(6):e37-39.
    37.Suda K,Kitagawa Y,Ozawa S,et al.Anti-high-mobility group box chromosomal protein 1 antibodies improve survival of rats with sepsis.World J Surg,2006,30(9):1755-1762.
    38.Ronchetti S,Zollo O,Bruscoli S,et al.Frontline:GITR,a member of the TNF receptor superfamily,is co-stimulatory to mouse T lymphocyte subpopulations.Eur J Immunol,2004,34(3):613-622.
    39.Dumitriu IE,Baruah P,Bianchi ME,et al.Requirement of HMGB1 and RAGE for the maturation of human plasmacytoid dendritic ceils.Eur J Immunol,2005,35(7):2184-2190.
    40.张莹,姚咏明,常青,等.不同细胞刺激剂对小鼠调节性T细胞功能活化的影响.中国危重病急救医学,2007,19(3):142-145.
    41.Scaffidi P,Misteli T,Bianchi ME.Release of chromatin protein HMGB1 by necrotic cells triggers inflammation.Nature,2002,418(6894):191-195.
    42.Bonaldi T,Talamo F,Scaffidi P,et al.Monocytic cells hyperacetylate chromatin protein HMGB1 to redirect it towards secretion.EMBO J,2003,22(20):5551-5560.
    43.Gardella S,Andrei C,Ferrera D,et al.The nuclear protein HMGB1 is secreted by monocytes via a non-classical,vesicle-mediated secretory pathway.EMBO Rep,2002,3(10):995-1001.
    44.Schmidt AM,Yan SD,Yan SF,et al.The multiligand receptor RAGE as a progression factor amplifying immune and inflammatory responses.J Clin Invest,2001,108(7):949-955.
    45.Kokkola R,Andersson A,Mullins G,et al.RAGE is the major receptor for the proinflammatory activity of HMGB1 in rodent macrophages.Scand J Immunol,2005,61(1):1-9.
    46.Park JS,Arcaroli J,Yum HK,et al.Activation of gene expression in human neutrophils by high mobility group box 1 protein.Am J Physiol Cell Physiol,2003,284(4):C870-879.
    47.Liliensiek B,Weigand MA,Bierhaus A,et al.Receptor for advanced glycation end products(RAGE)regulates sepsis but not the adaptive immune response.J Clin Invest,2004,113(11):1641-1650.
    48.Neumann M,Fries H,Scheicher C,et al.Differential expression of Rel/NF-kappaB and octamer factors is a hallmark of the generation and maturation of dendritic cells.Blood,2000,95(1):277-285.
    49.Tanaka N,Yonekura H,Yamagishi S,et al.The receptor for advanced glycation end products is induced by the glycation products themselves and tumor necrosis factor-alpha through nuclear factor-kappa B,and by 17beta-estradiol through Sp-1 in human vascular endothelial cells.J Biol Chem,2000,275(33):25781-25790.
    50. Ouaaz F, Li M, Beg AA. A critical role for the RelA subunit of nuclear factor kappaB in regulation of multiple immune-response genes and in Fas-induced cell death. J Exp Med, 1999,189(6):999-1004.
    51. Hazzalin CA, Mahadevan LC. MAPK-regulated transcription: a continuously variable gene switch? Nat Rev Mol Cell Biol, 2002,3(1):30-40.
    52. Arrighi JF, Rebsamen M, Rousset F, et al. A critical role for p38 mitogen-activated protein kinase in the maturation of human blood-derived dendritic cells induced by lipopolysaccharide, TNF-alpha, and contact sensitizers. J Immunol, 2001,166(6):3837-3845.
    53. Park JS, Svetkauskaite D, He Q, et al. Involvement of toll-like receptors 2 and 4 in cellular activation by high mobility group box 1 protein. J Biol Chem, 2004,279(9):7370-7377.
    54. Yu M, Wang H, Ding A, et al. HMGB1 signals through toll-like receptor (TLR) 4 and TLR2. Shock, 2006,26(2):174-179.
    55. Adams S, O'Neill DW, Bhardwaj N. Recent advances in dendritic cell biology. J Clin Immunol, 2005,25(3): 177-188.
    56. Oderup C, Cederbom L, Makowska A, et al. Cytotoxic T lymphocyte antigen-4-dependent down-modulation of costimulatory molecules on dendritic cells in CD4+CD25+ regulatory T-cell-mediated suppression. Immunology, 2006,118(2): 240-249.
    57. Wang Z, Larregina AT, Shufesky WJ, et al. Use of the inhibitory effect of apoptotic cells on dendritic cells for graft survival via T-cell deletion and regulatory T cells. Am J Transplant, 2006, 6(6): 1297-1311.
    58. Dumitriu IE, Baruah P, Bianchi ME, et al. Requirement of HMGB1 and RAGE for the maturation of human plasmacytoid dendritic cells. Eur J Immunol, 2005, 35(7):2184-2190.
    59. Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature, 1998, 392(6673):245-252.
    60. Flohe SB, Agrawal H, Schmitz D, et al. Dendritic cells during polymicrobial sepsis rapidly mature but fail to initiate a protective Th1-type immune response. J Leukoc Biol, 2006,79(3):473-481.
    61. Ding Y, 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.
    62. Messmer D, Yang H, Telusma G, et al. High mobility group box protein 1: an endogenous signal for dendritic cell maturation and Th1 polarization. J Immunol, 2004,173(1):307-313.
    63. Yang D, Chen Q, Yang H, et al. High mobility group box-1 protein induces the migration and activation of human dendritic cells and acts as an alarmin. J LeukocBiol,2007,81(1):59-66.
    64. 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.
    65. Chen W. Dendritic cells and (CD4+)CD25+ T regulatory cells: crosstalk between two professionals in immunity versus tolerance. Front Biosci, 2006, 11(3):1360-1370.
    66. Heidecke CD, Hensler T, Weighardt H, et al. Selective defects of T lymphocyte function in patients with lethal intraabdominal infection. Am J Surg, 1999,178(4):288-292.
    67. Pellegrini JD, De AK, Kodys K, et al. Relationships between T lymphocyte apoptosis and anergy following trauma. J Surg Res, 2000, 88(2):200-206.
    68. Bozza S, Perruccio K, Montagnoli C, et al. A dendritic cell vaccine agaist invasive aspergillosis in allogeneic hematopoietic transplantation. Blood, 2004,102 (10):3807-3814.
    69. Patenaude J, D'Elia M, Hamelin C, et al. Burn injury induces a change in T cell homeostasis affecting preferentially CD4+ T cells. J Leukoc Biol, 2005, 77(2):141-150.
    70. Bocca AL, Hayashi EE, Pinheiro AG, et al. Treatment of Paracoccidioides brasiliensis-infected mice with a nitric oxide inhibitor prevents the failure of cell-mediated immune response. J Immunol, 1998,161(6):3056-3063.
    71. DeMarco RA, Fink MP, Lotze MT. Monocytes promote natural killer cell interferon gamma production in response to the endogenous danger signal HMGB1. Mol Immunol, 2005,42(4):433-444.
    72. O'Riordain DS, Mendez MV, O'Riordain MG, et al. Molecular mechanisms of decreased interleukin-2 production after thermal injury. Surgery, 1993, 114(2):407-414; discussion 414-405.
    73. Lederer JA, Rodrick ML, Mannick JA. The effects of injury on the adaptive immune response. Shock, 1999,11(3):153-159.
    74.Spolarics Z,Siddiqi M,Siegel JH,et al.Depressed interleukin-12-producing activity by monocytes correlates with adverse clinical course and a shift toward Th2-type lymphocyte pattern in severely injured male trauma patients.Crit Care Med,2003,31(6):1722-1729.
    75.Ayala A,Chung CS,Song GY,et al.IL-10 mediation of activation-induced TH1 cell apoptosis and lymphoid dysfunction in polymicrobial sepsis.Cytokine,2001,14(1):37-48.
    76.Ono S,Ueno C,Aosasa S,et al.Severe sepsis induces deficient interferon-gamma and interleukin-12 production,but interleukin-12 therapy improves survival in peritonitis.Am J Surg,2001,182(5):491-497.
    77.Fukuzuka K,Rosenberg JJ,Gaines GC,et al.Caspase-3-dependent organ apoptosis early after burn injury.Ann Surg,1999,229(6):851-858;discussion 858-859.
    78.Song GY,Chung CS,Chaudry IH,et al.MAPK p38 antagonism as a novel method of inhibiting lymphoid immune suppression in polymicrobial sepsis.Am J Physiol Cell Physiol,2001,281(2):C662-669.
    79.Iwasaka H,Noguchi T.[Th1/Th2 balance in systemic inflammatory response syndrome(SIRS)].Nippon Rinsho,2004,62(12):2237-2243,
    80.Pachot A,Monneret G,Voirin N,et al.Longitudinal study of cytokine and immune transcription factor mRNA expression in septic shock.Clin Immunol,2005,114(1):61-69.
    81.Trinchieri G.Interleukin-10 and its role in the generation of TH2 cells.Immunol Today,1993,14(7):335-338.
    82.董月青,姚咏明,刘辉,等.烫伤后高迁移率族蛋白B1的变化及其对脾淋巴细胞周期的影响.解放军医学杂志,2005,30(10):861-863.
    83.Hotchkiss RS,Karl IE.The pathophysiology and treatment of sepsis.N Engl J Med,2003,348(2):138-150.
    84.Sun Z,Andersson R.NF-kappaB activation and inhibition:a review.Shock,2002,18(2):99-106.
    85.Maksimow M,Soderstrom TS,Jalkanen S,et al.Fas costimulation of naive CD4 T cells is controlled by NF-kappaB signaling and caspase activity.J Leukoc Biol,2006,79(2):369-377.
    86.Maksimow M,Miiluniemi M,Marttila-Ichihara F,et al.Antigen targeting to endosomal pathway in dendritic cell vaccination activates regulatory T cells and attenuates tumor immunity.Blood,2006,108(4):1298-1305.
    87.O'Suilleabhain CB,Kim S,Rodrick MR,et al.Injury induces alterations in T-cell NFkappaB and AP-1 activation.Shock,2001,15(6):432-437.
    88.Phan HH,Cho K,Nelson HA,et al.Downregulation of NF-kappaB activity associated with alteration in proliferative response in the spleen after burn injury.Shock,2005,23(1):73-79.
    89.Venkataraman R,Kellum JA,Song M,et al.Resuscitation with Ringer's ethyl pyruvate solution prolongs survival and modulates plasma cytokine and nitrite/nitrate concentrations in a rat model of lipopolysaccharide-induced shock.Shock,2002,18(6):507-512.
    90.Song M,Kellum JA,Kaldas H,et al.Evidence that glutathione depletion is a mechanism responsible for the anti-inflammatory effects of ethyl pyruvate in cultured lipopolysaccharide-stimulated RAW 264.7 cells.J Pharmacol Exp Ther,2004,308(1):307-316.
    91.Dobsak P,Courderot-Masuyer C,Zeller M,et al.Antioxidative properties of pyruvate and protection of the ischemic rat heart during cardioplegia.J Cardiovasc Pharmacol,1999,34(5):651-659.
    92.Czura CJ,Tracey KJ.Targeting high mobility group box 1 as a late-acting mediator of inflammation.Crit Care Med,2003,31(1 Suppl):S46-50.
    93.姚咏明,姚胜,陈劲松,等.NF-κB抑制剂对烫伤脓毒症大鼠致炎/抗炎细胞因子表达的影响.解放军医学杂志,2004,29(1):33-35.
    94.Rahman I,MacNee W.Regulation of redox glutathione levels and gene transcription in lung inflammation:therapeutic approaches.Free Radic Biol Med,2000,28(9):1405-1420.
    95.姚咏明,鄢小建,董宁,等.脓毒症大鼠高迁移率族蛋白-1对组织Toll样受体2基因表达的影响.中华创伤杂志,2003,19(1):13-16.
    96.董月青,姚咏明,魏鹏,等.丙酮酸乙酯对烫伤延迟复苏大鼠细胞免疫功能的影响.中国危重病急救医学,2005,17(1):24-27.
    97.Masson Ⅰ,Mathieu J,Nolland XB,et al.Role of nitric oxide in depressed lymphoproliferative responses and altered cytokine production following thermal injury in rats.Cell Immunol,1998,186(2):121-132.
    98.Valenti L,Mathieu J,Chancerelle Y,et al.Nitric oxide inhibits spleen cell proliferative response after burn injury by inducing cytostasis,apoptosis,and necrosis of activated T lymphocytes:role of the guanylate cyclase.Cell Immunol,2003,221(1):50-63.
    99.Das UN.Is pyruvate an endogenous anti-inflammatory molecule? Nutrition,2006,22(9):965-972.
    100.Fink MP.Ethyl pyruvate:a novel treatment for sepsis and shock.Minerva Anestesiol,2004,70(5):365-371.
    101.Woo YJ,Taylor MD,Cohen JE,et al.Ethyl pyruvate preserves cardiac function and attenuates oxidative injury after prolonged myocardial ischemia.J Thorac Cardiovasc Surg,2004,127(5):1262-1269.
    102.Taylor MD,Grand TJ,Cohen JE,et al.Ethyl pyruvate enhances ATP levels,reduces oxidative stress and preserves cardiac function in a rat model of off-pump coronary bypass.Heart Lung Circ,2005,14(1):25-31.
    103.Yang R,Han X,Delude RL,et al.Ethyl pyruvate ameliorates acute alcohol-induced liver injury and inflammation in mice.J Lab Clin Med,2003,142(5):322-331.
    104.Su F,Wang Z,Cai Y,et al.Beneficial effects of ethyl pyruvate in septic shock from peritonitis.Arch Surg,2007,142(2):166-171.
    105.Kim JB,Yu YM,Kim SW,et al.Anti-inflammatory mechanism is involved in ethyl pyruvate-mediated efficacious neuroprotection in the postischemic brain.Brain Res,2005,1060(1-2):188-192.
    106.Harada T,Moore BA,Yang R,et al.Ethyl pyruvate ameliorates ileus induced by bowel manipulation in mice.Surgery,2005,138(3):530-537.
    107.Aneja R,Fink MP.Promising therapeutic agents for sepsis.Trends Microbiol,2007,15(1):31-37.
    108.董宁,姚咏明,董月青,等.丙酮酸乙酯对烫伤延迟复苏大鼠脾淋巴细胞增殖及凋亡影响.中国危重病急救医学,2005,17(7):393-396.
    1.Poser Ⅰ,Golob M,Buettner R,et al.Upregulation of HMG1 leads to melanoma inhibitory activity expression in malignant melanoma cells and contributes to their malignancy phenotype.Mol Cell Biol,2003,23(8):2991-2998.
    2.Wei HM,Sun R,Xiao W,et al.Type two cytokines predominance of human lung cancer and its reverse by traditional Chinese medicine TTMP.Cell Mol Immunol,2004,1(1):63-70.
    3.Kurokawa CS,Araujo JP Jr,Soares AM,et al.Pro- and anti-inflammatory cytokines produced by human monocytes challenged in vitro with Paracoccidioides brasiliensis.Microbiol Immunol,2007,51(4):421-428.
    4.Lyng MB,Laenkholm AV,Pallisgaard N,et al.Identification of genes for normali zation of real-time RT-PCR data in breast carcinomas.BMC Cancer,22,8(1):20.
    5.姚咏明,柴家科,林洪远,主编.现代脓毒症理论与实践.第1版.北京:科学出版社,2005.1-10.
    6.Wang H,Bloom O,Zhang M,et al.HMG-1 as a late mediator of endotoxin lethality in mice.Science,1999,285(5425):248-251.
    7.Sunden-Cullberg J,Norrby-Teglund A,Rouhiainen A,et al.Persistent elevation of high mobility group box-1 protein(HMGB1)in patients with severe sepsis and septic shock.Crit Care Med,2005,33(3):564-573.
    8.姚咏明,盛志勇.我国创伤脓毒症基础研究新进展.中华创伤杂志,2003,19(1):9-12.
    9.郭振荣,盛志勇,高维谊,等.休克期切痂有助于控制或减轻感染并发症.中华外科杂志,1993,33(7):406-408.
    10.张立天,姚咏明,陆家齐,等.脓毒症大鼠高迁移率族蛋白-1基因表达及其与内毒素血症的关系.中华普通外科杂志,2002,17(9):556-558.
    11.张立天,姚咏明,陆家齐,等.高迁移率族蛋白-1在脓毒症所致多器官功能损害中的作用.中华外科杂志,2003,41(4):303-306.
    12.Wang H,Yang H,Tracey KJ.Extracellular role of HMGB1 in inflammation and sepsis.J Intern Med,2004,255(3):320-331.
    13.Yang H,Ochani M,Li J,et al.Reversing established sepsis with antagonists of endogenous high-mobility group box 1.Proc Natl Acad Sci USA,2004,101(1):296-301.
    14.Wang H,Yang H,Czura CJ,et al.HMGB1 as a late mediator of lethal systemic inflammation.Am J Respir Crit Care Med,2001,164(10 Pt 1):1768-1773.
    15.Yang H,Wang H,Czura CJ,et al.HMGB1 as a cytokine and therapeutic target.J Endotoxin Res,2002,8(6):469-472.
    16.刘峰,姚咏明.高迁移率族蛋白1-新的“晚期”促炎性细胞因子.国际病理科学与临床杂志,2005,25(5):377-380.
    17.董宁,姚咏明.大面积烧伤脓毒症患者T淋巴细胞免疫功能的改变及临床意义.中华烧伤杂志,2007,23(2):84-87.
    18.董宁,姚咏明,于燕,等.严重烧伤后高迁移率族蛋白B1的变化及其与脓毒症的关系.中国医学科学院院报,2007,29(4):466-470.
    19.Fang WH,Yao YM,Shi ZG,et al.The significance of changes in high mobility groupo-1 protein mRNA expression in rats after thermal injury.Shock,2002,17(4):329-333
    20.姚咏明,张立天,陆家齐,等.脓毒症大鼠内毒素增敏系统改变与高迁移率族蛋白-1表达的关系.中华创伤杂志,2002,18(9):550-553.
    21.Chen G,Li J,Ochani M,et al.Bacterial endotoxin stimulates macrophages to release HMGB1 partly through CD14- and TNF-dependent mechanisms.J Leukoc Biol,2004,76(5):994-1001.
    22.Park JS,Gamboni-Robertson F,He Q,et al.High mobility group box 1 protein interacts with multiple Toll-like receptors.Am J Physiol Cell Physiol,2006,290(3):C917-924.
    23.Yang H,Tracey KJ.High mobility group box 1(HMGB1).Crit Care Med,2005,33(12 Suppl):S472-S474.
    24.Suda K,Kitagawa Y,Ozawa S,et al.Anti-high-mobility group box chromosomal protein 1 antibodies improve survival of rats with sepsis.World J Surg,2006,30(9):1755-1762.
    25.Mantell LL,Parrish WR,Ulloa L.HMGB-1 as a therapeutic target for infectious and inflammatory disorders.Shock,2006,25(1):4-11.
    26.Belkaid Y.Rouse BT.Natural regulatory T cells in infectious disease.Nat Immunol,2005,6(4):353-360.
    27.Dumitriu IE,Baruah P,Bianchi ME,et al.Requirement of HMGB1 and RAGE for the maturation of human plasmacytoid dendritic cells.Eur J Immunol,2005,35(7):2184-2190.
    28.Monneret G,Debard AL,Venet F,et al.Marked elevation of human circulating CD4CD25 regulatory T cells in sepsis-induced immunoparalysis.Crit Care Med,2003,31(7):2068-2071.
    29.Venet F,Pachot A,Debard AL,et al.Increased percentage of CD4+CD25+regulatory T cells during septic shock is due to the decrease of CD4+CD25-lymphocytes.Crit Care Med,2004,32(11):2329-2331.
    30.Purcell EM,Dolan SM,Kuhn J,et al.Burn injury induces an early activation response by lymph node CD4+T cell.Shock,2006,25(2):135-140.
    31.Chatila TA.Role of regulatory T cells in human diseases.J Allergy Clin Immunol,2005,116(5):949-959.
    32.Cook DN,Hollingsworth JW Jr,Schwartz DA.Toll-like receptors and the genetics of innate immunity.Curr Opin Allergy Clin Immunol,2003,3(6):523-529.
    33.Caramalho Ⅰ,Lopes-Carvalho T,Ostler D,et al.Regulatory T cells selectively express Toll-Like receptors and activated by lipopolysaccharide.J Exp Med,2003,197(4):403-411.
    34.Vigouroux S,Yvon E,Biagi E,et al.Antigen-induced regulatory T cells.Blood,2004,104(1):26-33.
    35.王忠堂,姚咏明,盛志勇,等.休克期切痂对烫伤大鼠肝、肺组织高迁移率族蛋白B1表达及促炎/抗炎平衡的影响.中华外科杂志,2004,42(14):839-844.
    36.庞伟,郭振荣,帅秀蓉,等.休克期切痂对烫伤大鼠γ干扰素、白细胞介素4 mRNA表达的影响.中华外科杂志,2004,42(18):1142-1145.
    37.Guo ZR,Sheng CY,Diao L,et al.Extensive wound excision in the acute shock stage in patients with major burns.Burns,1995,21(2):139-142.
    38.Hart DW,Wolf SE,Chinkes DL,etal.Effects of early excision and aggressive enteral feeding on hypermetabolism,catabolism,and sepsis after severe burn.J Trauma,2003,54(4):755-761.
    39.Hietbrink F,Koenderman L,Rijkers GT,et al.Trauma:the role of the innate immune system.World J Emerg Surg,2006,20(1):15.
    40 董月青,黄军华,姚咏明.脓毒症中T淋巴细胞调亡及其机制研究进展.中国危重病急救医学,2004,16(6):381-384.
    41.Oberholzer A,Oberholzer C,Moldawer LL,et al.Sepsis syndromes:understanding the role of innate and acquired immunity.Shock,2001,16(1):83-96.
    42.Wesche DE,Lomas-Neira JL,Perl M,et al.Leukocyte apoptosis and its signifycance in sepsis and shock.J Leukoc Biol,2005,78(2):325-337.
    43.姚咏明,黄立锋,林洪远.进一步提高对脓毒症免疫机制及调理策略的认识.创伤外科杂志,2007,9(1):4-7.
    44.Hotchkiss RS,Coopersmith CM,Karl IE,et al.Prevention of lymphocyte apoptosis-a potential treatment of sepsis? Clin Infect Dis,2005,15(7):465-469.
    45.董月青,姚咏明,董宁,等.烫伤大鼠高迁移率族蛋白B1的变化及其对脾淋巴细胞免疫反应的影响.创伤外科杂志,2007,9(1):24-27.
    46.Wesche-Soldato DE,Lomas-Neira JL,Perl M,et al.The role and regulation of apoptosis in sepsis.J Endotoxin Res,2005,11(6):375-382.
    47.Zhang LT,Yao YM,Dong Ning,et al.Relationship between high-mobility group box 1 protein release and T-cell suppression in rats after thermal injury.Shock,2008,[Epub ahead of print].
    48.Hotchkiss RS,Karl JE.The Pathophysiology and treatment of sepsis.N Engl J Med,2003,348(2):138-150.
    49.董月青,姚咏明.脓毒症中细胞免疫紊乱的机制.中国危重病急救医学,2004,16(10):636-638.
    50.van Parijs L,Abbas AK.Homeostasis and self-tolerance in the immune system:turning lymphocytes off.Science,1998,280(5361):243-248.
    51.Jobin N,Garrel D,Bernier J.Increased serum-soluble interleukin-2 receptor in burn patients:characterization and effects on the immune system.Hum Immunol,2000,61(3):233-246.
    52.Sayeed MM.Signaling mechanisms of altered cellular responses in trauma,burn,and sepsis:role of Ca2+.Arch Surg,2000,135(12):1432-1442.
    53.Choudhry MA,Haque F,Khan M,et al.Enteral nutritional supplementation prevents mesenteric lymph node T-cell suppression in burn injury.Crit Care Med,2003,31(6):1764-1770.
    54.Choudhry M,Mao AH,Haque F,et al.Role of NFAT and AP-1 in PGE2-mediated T cell suppression in burn.Shock,2002,18(3):212-216.
    55.Patenaude J,D'Elia M,Hamelin C,et al.Burn injury induces a change in T cell homeostasis affecting preferentially CD4+ T cell.J Leukoc Biol,2005,77(2):141-150.
    56.姚咏明,盛志勇.重视对脓毒症本质的探讨.中华急诊医学杂志,2005,14(3):185-186.
    57.Vigouroux S,Yvon E,Biagi E,et al.Antigen-induced regulatory T cells.Blood,2004,104(1):26-33.
    58.Fehervari Z,Sakaguchi S.Control of Foxp3+CD25+CD4+ regulatory cell activation and function by dendritic cells.Int Immunol,2004,16(12):1769-1780.
    59.Amsen D,Blander JM,Lee GR,et al.Instruction of distinct CD4+T helper cell fates by different notch ligands on antigen presenting cells.Cell,2004,117(4):515-526.
    60.张莹,姚咏明,盛志勇.调节性T细胞研究进展.生理科学进展,2007,38(1):83-88.
    61.Capron A,Dombrowicz D,Capron M.Helminth infections and allergic diseases:from the Th2 paradigm to regulatory networks.Clin Rev Allergy Immunol,2004,26(1):25-34.
    62.Venet F,Lepape A,Debard AL,et al.The Th2 response as monitored by CRTH2or CCR3 expression is severely decreased during septic shock.Clin Immunol,2004,113(3):278-284.
    63.张莹,姚咏明.调节性T细胞与脓毒症关系的研究进展.中国危重病急救医学,2006,18(11):695-698.
    64.Iwasaka H,Noguchi T.Th1/Th2 balance in systemic inflammatory response syndrome(SIRS).Nippon Rinsho,2004,62(12):2237-2243.
    65.Huang LF,Yao YM,Meng HD,et al.The effect of high mobility group box 1protein(HMGB1)on immune function of human T lymphocytes in vitro.Chin Crit Care Med,2008,20(1):7-13.
    1.Chatila TA.Role of regulatory T cells in human diseases[J].Allergy Clin Immunol,2005,116(5):949-959.
    2.曹雪涛.树突状细胞的研究热点及其与疾病的防治[J].中华医学杂志,1999,79(3):163-164.
    3.Hock BD,Roberts G,McKenzie JL,et al.Exposure to the electrofusion process can increase the immunogenicity of human cells[J].Cancer Immunol Immunother,2005,54(9):880-890.
    4.Yagi H,Nomura T,Nakamura K,et al.Crucial role of FOXP3 in the development and function of human CD25+CD4+ regulatory T cells[J].Int Immunol,2005,16(11):1643-1656.
    5.Fehervari Z,Sakaguchi S.Development and function of CD25+CD4+regulatory T cells[J].Curr Opin lmmunol,2004,16(2):203-208.
    6.Oderup C,Cederbom L,Makowska A,et al.Cytotoxic T lymphocyte antigen-4dependent down-modulation of costimulatory molecules on dendritic cells in CD4+CD25+ regulatory T-cell-mediated suppression[J].Immunology,2006,118(2):240-249.
    7.Wang Z,Larregina AT,Shufesky WJ,et al.Use of the inhibitory effect of apoptotic cells on dendritic cells for graft survival via T-cell deletion and regulatory T cells[J].Am J Transplant,2006,6(6):1297-1311.
    8.Houot R,Perrot Ⅰ,Garcia E,et al.Human CD4+CD25high regulatory T cells modulate myeloid but not plasmacytoid dendritic cells activation[J]. J Immunol, 2006,176(9): 5293-5298.
    9. Van Meirvenne S, Dullaers M, Heirman C, et al. In vivo depletion of CD4+CD25+ regulatory T cells enhances the antigen-specific primary and memory CTL response elicited by mature mRNA-electroporated dendritic cells[J]. Mol Ther, 2005,12(5):922- 932.
    10. Barchet W, Price JD, Cella M, et al. Complement-induced regulatory T cells suppress T-cell responses but allow for dendritic-cell maturation[J].. Blood, 2006,107(4): 1497-1504.
    11. Veldhoen M, Moncrieffe H, Hocking RJ, et al. Modulation of dendritic cell function by naive and regulatory CD4+ T cells[J]. J Immunol, 2006, 176(10): 6202-6210.
    12. Tadokoro CE, Shakhar G, Shen S, et al. Regulatory T cells inhibit stable contacts between CD4+ T cells and dendritic cells in vivo[J]. J Exp Med, 2006, 203(3):505-511.
    13. Nagatani K, Sagawa K, Komagata Y, et al. Peyer's patch dendritic cells capturing oral antigen interact with antigen-specific T cells and induce gut-homing CD4(+)CD25(+) regulatory T cells in Peyer's patches[J]. Ann N Y Acad Sci, 2004,1029(1):366-370.
    14. Watanabe N, Wang YH, Lee HK, et al. Hassall's corpuscles instruct dendritic cells to induce CD4+CD25+ regulatory T cells in human thymus[J]. Nature, 2005,436(7054):1181-1185.
    15. Tarbell KV, Yamazaki S, Steinman RM. The interactions of dendritic cells with antigen-specific, regulatory T cells that suppress autoimmunity[J]. Semin Immunol, 2006,18(2):93-102.
    16. Banerjee D, Dhodapkar MV, Matayeva E, et al. Expansion of FOXP3 high regulatory T cells by human dendritic cells (DCs) in vitro and after DC injection of cytokine matured DCs in myeloma patients[J]. Blood, 2006, 107(4): 1134-1043.
    17. Maksimow M, Miiluniemi M, Marttila-Ichihara F, et al. Antigen targeting to endosomal pathway in dendritic cell vaccination activates regulatory T cells and attenuates tumor-immunity[J]. Blood, 2006,107(4): 1211-1219.
    18. Kawamura K, Kadowaki N, Kitawaki T, et al. Virus-stimulated plasmacytoid dendritic cells induce CD4+ cytotoxic regulatory T cells[J]. Blood, 2006, 107(3): 1031-1038.
    19. Zhang X, Huang H, Yuan J, et al. CD4-8- dendritic cells prime CD4+T regulatory 1 cells to suppress antitumor immunity[J]. J Immunol, 2005, 175(5):2931-2937.
    20. Chen W. Dendritic cells and (CD4+)CD25+ T regulatory cells: crosstalk between two professionals in immunity versus tolerance[J]. Front Biosci, 2006, 11(3):1360-1370.
    21. Leithauser F, Meinhardt-Krajina T, Fink K, et al. Foxp3-expressing CD103+ regulatory T cells accumulate in dendritic cell aggregates of the colonic mucosa in murine transfer colitis[J]. Am J Pathol, 2006,168(6):1898-1909.
    22. Ghiringhelli F, Puig PE, Roux S, et al. Tumor cells convert immature myeloid dendritic cells into TGF-beta-secreting cells inducing CD4+CD25+ regulatory T cell proliferation[J]. J Exp Med, 2005,202(7):919-929.
    23. Wei S, Kryczek I, Zou L, et al. Plasmacytoid dendritic cells induce CD8+ regulatory T cells in human ovarian carcinoma[J]. Cancer Res, 2005, 65(12): 5020-5026.
    24. Romagnani C, Delia Chiesa M, Kohler S, et al. Activation of human NK cells by plasmacytoid dendritic cells and its modulation by CD4+ T helper cells and CD4+ CD25hi T regulatory cells[J]. Eur J Immunol, 2005, 35(8):2452-2558.
    25. Kared H, Masson A, Adle-Biassette H, et al. Treatment with granulocyte colony-stimulating factor prevents diabetes in NOD mice by recruiting plasmacytoid dendritic cells and functional CD4(+)CD25(+) regulatory T-cells[J]. Diabetes, 2005, 54(1):78-84.
    26. Krathwohl MD, Schacker TW, Anderson JL. Abnormal presence of semimature dendritic cells that induce regulatory T cells in HIV-infected subjects[J]. J Infect Dis, 2006,193(4):494-504.
    27. Gonzalez-Rey E, Chorny A, Fernandez-Martin A, et al. Vasoactive intestinal peptide generates human tolerogenic dendritic cells that induce CD4 and CD8 regulatory T cells[J]. Blood, 2006,107(9):3632-3638.
    28. Haas J, Hug A, Viehover A, et al. Reduced suppressive effect of CD4+CD25high regulatory T cells on the T cell immune response against myelin oligodendrocyte glycoprotein in patients with multiple sclerosis[J]. Eur J Immunol, 2005,35(11):3343-3352.
    29. Navarro J, Aristimuno C, Sanchez-Ramon S, et al. Circulating dendritic cells subsets and regulatory T-cells at multiple sclerosis relapse: differential short-term changes on corticosteroids therapy[J]. J Neuroimmunol, 2006, 79(9):367-378.
    30. Dombrecht EJ, Aerts NE, Schuerwegh AJ, et al. Influence of anti-tumor necrosis factor therapy (Adalimumab) on regulatory T cells and dendritic cells in rheumatoid arthritis[J]. Clin Exp Rheumatol, 2006,24(1):31-37.

© 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