可溶性ST2融合蛋白对小鼠肝脏缺血/再灌注损伤的保护作用及其机制
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
一、ST2与缺血/再灌注损伤相关
     缺血/再灌注损伤(I/R, Ischemia and reperfusion)是指缺血缺氧的组织器官重新获得血液供应后,往往并不能使损伤减轻或恢复,反而加重组织器官功能障碍、结构损伤和细胞死亡的现象。在肝脏移植、外伤、休克以及部分肝切除手术过程中,通常会发生短时间肝脏缺血、缺氧。肝脏缺血/再灌注损伤所引发的复杂病理生理学改变包括缺血直接导致的细胞损伤,以及相关炎症信号通路激活所致组织器官失功和破坏。肝脏缺血/再灌注损伤发生机制十分复杂,尚未完全明确,近年来,与组织器官损伤相关的炎症反应引起广泛关注。
     目前已知,肝脏巨噬细胞(Kupffer细胞)激活后产生ROS,可诱导肝细胞NOS活性上升、活化JNK信号途径、上调炎性细胞因子表达并伴随中性粒细胞局部浸润等,由此促进炎症反应相关的组织细胞损伤。初始细胞损伤导致炎症反应扩大,继而促进组织细胞损伤加重,其确切机制尚未完全明确。近年对此提出新观点:组织细胞起始破坏及炎症信号途径激活,可能与局部缺血组织或细胞间基质损伤所释放(内源性)损伤/危险相关分子模式信号(damage/danger associated molecular pattern,DAMP)有关。
     天然免疫细胞表面Toll样受体(Toll like receptor, TLR)是脊柱动物识别病原体感染最原始的一种方式,其通过识别、结合特异性分子模式而启动胞内信号转导途径,进而激活核转录因子NF-κB,诱导炎性细胞因子表达和多种生物学效应。TLR在天然免疫和适应性免疫应答间发挥重要的桥梁作用。现已发现,TLR识别的分子模式不仅包括病原微生物(及其产物)所共有的某些非特异性、高度保守的病原相关分子模式(Pathogen associated molecular pattern,PAMP),如脂多糖(LPS)、磷壁酸(LTA)、肽聚糖、细菌DNA、双链RNA等,还包括组织细胞损伤破坏时所释放的一系列内源性“危险信号”,如热休克蛋白(HSP)、氧自由基、核甘酸、神经介质、白介素等。
     肝脏由实质细胞(肝细胞)和非实质细胞(包括Kupffer细胞、DC、内皮细胞和星状细胞)组成。近年报道,肝细胞和肝脏非实质细胞表面普遍表达低水平TLR4。相对其他TLR,TLR4处于病原微生物感染和无菌性炎症反应的交界面,更易接受细菌内毒素(LPS)和内源性配体信号的刺激。Billiar等证实,肝脏缺血/再灌注过程中,肝脏非实质细胞表面TLR4可识别损伤/坏死细胞所释放的多种内源性配体,如HSP 60/70、纤维蛋白原、纤粘连蛋白、肝磷脂硫酸盐和高迁移率家族蛋白(high-mobility group box-1,HMGB1)等,并在介导肝脏再灌注损伤中发挥重要作用。
     ST2(又命名为T1、DER4)最初被发现是由小鼠成纤维细胞所分泌,属IL-1受体超家族。st2基因定位于小鼠1号染色体和人类2号染色体并与IL-1R区基因紧密相邻。ST2分子结构类似于IL-1受体,但其不与IL-1家族成员(如IL- 1α、IL- 1β或IL- 1R拮抗剂)结合。st2基因在mRNA转录水平经不同方式剪接修饰,生成(2.7 kb和5kb)两种mRNA序列,翻译后生成相应蛋白分子,即短序列分泌型ST2(可溶性ST2或sST2)和长序列跨膜型ST2(ST2L)。可溶性ST2与跨膜型ST2胞外段序列基本一致,仅在C末端存在9个氨基酸序列差异。可溶性ST2由成纤维细胞分泌,跨膜型ST2表达于肥大细胞和Th2型细胞表面,参与Th2型应答。近年在胃、小肠、结肠等组织发现一种新的剪接体,编码一类新的跨膜型ST2蛋白,但其功能特征有待进一步研究。
     ST2参与细胞生长调控和胚胎发育,其在造血细胞系中表达并与Toll/IL-1受体结构相似,提示可能参与免疫应答及其调控。应用抗ST2抗体处理BABL/C小鼠,能诱导Th2型应答向Th1型应答偏移,从而抑制利什曼原虫感染;胶原质诱导的小鼠关节炎主要为Th1型应答,经抗ST2抗体处理可加重炎性细胞浸润和关节炎损伤;抗ST2抗体、ST2融合蛋白处理过敏性肺炎、血吸虫卵感染和呼吸道合胞病毒感染等动物模型,以及ST2基因敲除小鼠体内,均发现ST2L可促进Th2型应答,主要表现为特征性嗜酸粒细胞局部浸润和Th2型细胞因子IL-
     4、IL- 5和IL- 13表达水平上升。
     Xu等探讨可溶性ST2-IgG融合蛋白对LPS激活巨噬细胞的影响,发现可溶性ST2可下调巨噬细胞所介导炎症反应,显著降低炎性细胞因子TNF-α、IL- 6和IL- 12等分泌,其机制是ST2与细胞表面相应ST2配体(ST2 BP)结合,向胞内传递抗炎信号,进而抑制炎性细胞因子产生;体内实验发现,给予ST2融合蛋白可降低小鼠内毒素休克死亡率。可溶性ST2通过与相应配体结合而下调炎性信号的分子机制尚未明确,但其抗炎作用涉及细胞表面受体识别LPS和相应炎性信号通路。TLR 4是LPS特异性受体,可通过胞内转接蛋白MyD88依赖性和非依赖性途径传递炎性信号,在激活巨噬细胞中发挥关键作用,并参与识别组织细胞损伤/破坏而释放的多种内源性配体。
     上述研究进展提示:肝脏缺血/再灌注所引发无菌性炎症反应和组织细胞损伤过程中,ST2可能对肝脏Kupffer细胞活性发挥重要调节作用,该效应可能涉及TLR4信号转导途径。本课题研究目标为:克隆小鼠可溶性ST2基因,构建其真核表达载体,制备可溶性ST2-Fc融合蛋白;体外观察sST2-Fc融合蛋白对LPS激活巨噬细胞释放炎性细胞因子的影响;体内转染sST2-Fc真核表达载体,观察sST2-Fc对小鼠肝脏缺血/再灌注损伤的影响,并探讨其机制,为今后应用于临床治疗组织器官缺血/再灌注损伤和相关炎症性疾病提供实验依据。
     二、小鼠可溶性ST2和人IgG Fc基因的克隆及真核表达载体的构建分别从小鼠脾脏和人外周血单个核细胞提取总RNA。利用RT-PCR技术获得编码小鼠可溶性ST2和人IgG Fc的cDNA,克隆入真核表达载体pcDNA 3.1,构建重组载体pcDNA 3.1- sST2- IgG Fc(psST2-Fc)。经酶切鉴定及测序分析证实,所克隆和构建的sST2-Fc cDNA阅读框及连接部位序列正确,表明小鼠可溶性ST2和人IgG Fc融合基因的重组真核表达质粒构建成功。
     三、可溶性ST2-Fc融合蛋白的制备及其体外效应的研究
     1.可溶性ST2-Fc基因表达和鉴定
     采用Lipofectamine TM 2000将psST2-Fc载体转染CHO细胞,稳定表达可溶性ST2-Fc融合蛋白;采用Western blot、ELISA检测CHO细胞培养上清中所表达sST2-Fc融合蛋白,用人IgG作为标准品对sST2-Fc进行定量检测;采用RT-PCR检测psST2-Fc融合基因mRNA表达。结果显示:转染psST2-Fc的CHO细胞可表达sST2-Fc融合基因mRNA,在约929 bp处呈现一明显条带,与预期结果一致;收集转染psST2-Fc的CHO细胞培养上清液,借助Protein A sepharose CL-4B亲和层析柱纯化sST2-Fc融合蛋白,可与抗小鼠ST2单克隆抗体特异性结合形成一明显条带,其分子量约100 kDa,与预期结果相一致。
     2. FACS检测sST2-Fc融合蛋白与相应配体结合
     为检测sST2-Fc融合蛋白与相应配体结合能力,小鼠腹腔巨噬细胞和sST2-Fc融合蛋白作用后,用FITC标记的羊抗人IgG Fc抗体染色。FACS结果显示,巨噬细胞表面表达ST2的特异性配体,并在LPS刺激下表达升高。
     3. sST2-Fc对巨噬细胞分泌细胞因子的影响
     采用ELISA检测sST2-Fc融合蛋白对LPS刺激巨噬细胞分泌细胞因子的影响。结果表明,sST2-Fc融合蛋白能明显抑制LPS刺激腹腔巨噬细胞产生TNF-α和IL-6的作用。
     四、sST2-Fc对肝脏缺血/再灌注损伤保护作用的实验研究
     建立BALB/C小鼠肝脏缺血/再灌注模型,在预定时间点取材进行检测,同时设立假手术对照组。
     1. psST2-Fc在小鼠体内表达
     借助ELISA检测psST2-Fc基因在体内表达水平。psST2-Fc基因转染36小时后,sST2-Fc融合蛋白水平达84.8±11.1 ng/ml,但pcDNA 3.1基因转染对照组未检出sST2-Fc融合蛋白表达。
     2. sST2-Fc对小鼠肝脏缺血/再灌注损伤的影响
     与pcDNA 3.1和生理盐水处理组相比,psST2-Fc基因处理组肝脏缺血/再灌注损伤明显改善:血清谷丙转氨酶(ALT)、IL-6和TNF-α水平均明显降低;(HE染色显示),肝细胞水肿变性坏死和肝血窦淤血均明显较轻。
     3. sST2-Fc抑制肝脏巨噬细胞活性
     免疫组织化学检测肝脏巨噬细胞表面特异性F4/80抗原表达,分析再灌注6小时后损伤肝组织局部巨噬细胞数量变化。结果发现:sST2-Fc处理组,肝组织F4/80+细胞数量(156±30个/高倍视野)明显低于pcDNA 3.1(193±26个/高倍视野)和生理盐水(189±28个/高倍视野)组,差异有显著性(P < 0.05)。
     肝脏缺血/再灌注3小时后,采用荧光定量RT-PCR检测肝组织中细胞因子mRNA水平。结果发现:psST2-Fc处理组肝组织中TNF-α和IL- 6 mRNA转录水平显著低于pcDNA 3.1和生理盐水组,差异有显著性(P < 0.05)。
     4.缺血/再灌注肝脏组织中TLR4 mRNA表达
     肝脏缺血/再灌注3小时后,采用荧光定量RT-PCR检测肝组织中TLR4 mRNA水平。结果显示:与pcDNA 3.1和生理盐水组相比,psST2-Fc处理组肝组织TLR4 mRNA表达水平明显下降,差异有显著性(P < 0.05)。
     5.缺血/再灌注肝脏组织中NF-κB活性
     肝脏缺血/再灌注3小时后,提取肝组织细胞核蛋白,采用凝胶迁移率实验(EMSA)检测NF-κB转录活性。结果发现:psST2-Fc处理组肝组织中NF-κB活性明显低于pcDNA 3.1和生理盐水组。
     以上研究结果表明:可溶性ST2可直接与巨噬细胞表面相应受体结合,且能抑制LPS刺激巨噬细胞分泌炎性细胞因子的作用;体内转染psST2-Fc能有效表达,并明显降低肝脏缺血/再灌注所致损伤,其机制与抑制巨噬细胞活性及下调TLR4 mRNA表达有关。
     五、结论
     1.缺血/再灌注动物模型中,可溶性ST2可有效抑制肝脏巨噬细胞活化和炎性细胞因子TNF-α、IL- 6产生,此效应与其抑制TLR4信号途径及核转录因子NF-κB激活有关。
     2.转染psST2-Fc基因能明显改善肝脏缺血/再灌注损伤,提示sST2是一种有效的免疫抑制剂,可望在治疗组织器官缺血/再灌注损伤和相关炎症疾病中具有应用前景。
1. Introduction
     Ischemia/reperfusion (I/R) injury is a complicated pathophysiologic process where- by hypoxic organ damage, tissue injury is accentuated following return of blood flow and oxygen delivery to the compromised tissue. Transient episodes of ischemia occur in procedures such as solid organ transplantation, trauma, shock, partial hepatectomy, and diverse surgical procedures, when inflow occlusion or total vascular exclusion is used to minimize blood loss. The pathophysiology of liver I/R injury includes direct cellular damage as the result of ischemic insult as well as delayed dysfunction and le- sion that results from activation of inflammatory pathways. The distal interaction el- ements in the cascade of inflammatory responses resulting in organ damage following hepatic I/R injury have been extensively studied. Activation of Kupffer cells with pr- oduction of reactive oxygen species, up-regulation of the inducible NOS in hepatocy- tes, activation of JNK, up-regualtion proinflammatiory cytokines, and neutrophil acc- umulation have all been identified as contributing events to the inflammation associa- ted damage. The extent to which the initial cellular injury contributes to propagation of the inflammatory response and leads to further tissue damage is poorly understood. Most recent studies show that a key link between the initial damage to cells and the subsequent activation of inflammatory signaling involves release of endogenous dan- ger signals from ischemic cells and disruption of the tissue matrix.
     TLRs are present on the surface of innate immune cells, including dendritic cells, macrophages, and natural killer cells in vertebrates. TLRs has been found to be the primary form of pattern recognition receptors for the recognition invasion of pathogenic microorganisms, mediating intracellular signal transduction pathways, leading to the activation of transcriptional factors such as NF-κB, and the expression of genes related to inflammatory and immune response. To date, the TLR family at least consists of 11 members, and is found to be an important link between innate immunity and adaptive immunity. Recent evidence suggests that the innate immune system use the TLRs for the recognition of specific molecular patterns that are present in microbial products, such as LPS, LTA, peptidoglycan, bacterial DNA, dsRNA and endogenous danger signals, such as hot shock protein (HSPs), oxygen free radicals (OFR), deoxyribonucleic acid, neurotransimitters, and interleukin too. The liver consists of parenchymal cells (hepatocytes) and nonparenchymal cells (NPC), including Kupffer cells, sinusoidal endothelial cells, stellate cells, and hepatic dendritic cells. Recent studies found that TLR4 is expressed on hepatocytes and NPC, however the levels of TLR4 expression are lower in the liver than other organs.
     Perhaps more than any of the other TLR family members, TLR4 located on the interface of microbial and sterile inflammation by responding to both bacterial endotoxin and multiple other endogenous ligands. Billiar and his collaborators demonstrated that the NPC, not the hepatocytes, are responsible for recognizing the initial damage including HSPs, fibrinogen, fibronectin, heparin sulfate, high mobility group box 1(HMGB1) from ischemic cells and for activating TLR4-dependent signaling required for I/R- induced injury.
     ST2 (also known as T1 or DER4) was originally defined in murine fibroblasts as a late response gene induced by serum, and is a member of the interleukin-1 (IL-1) rec- eptor superfamily. ST2 gene is tightly linked to the IL-1R genes region on mouse chr- omosome 1 and human chromosome 2. Despite its similarity to the IL-1R, ST2 does not bind IL-1α, IL- 1β, or IL- 1R antagonist. In the mouse, differential mRNA proce- ssing within the ST2 gene produces two mRNAs of 2.7 Kb and 5 Kb, corresponding to shorter secreted form (soluble ST2 or sST2) and longer, membrane anchored form (ST2L) of the protein. Hence, sST2 is identical with the extracelluar region of ST2L except for an additional nine amino acids, which are present at the C terminus of the molecule. sST2 is expressed in embryonic tissues, mammary tumors and fibroblasts, while ST2L expressed on mast cells and on T helper 2 (Th2) cells but not on T helper 1 (Th1) cells. Recently, another alternatively spliced transcript that is expressed in the stomach, small intestine, and colon, and encodes a novel membrane form of ST2 has been identified in humans. However, its functional role remains to be identified. In addition to the proposed involvement of ST2 in cell growth regulation and embryoge- nesis, the presence of ST2 in hematopoetic cell lines and its homology with the mem- bers of Toll/IL-1 receptor family indicates its potential functional role in immune res- ponses. Treatment with anti-ST2 antibody could induce resistance to L.major infecti- on in the susceptible BABL/C mouse by swithching a detrimental Th2 to a protective Th1 response. However, Anti-ST2 antibody pretreatment exacerbated murine collag- en-induced arthritis (CIA), in which Th1 responses predominate within rheumatoid arthritis synovial T cell subsets. In the conditions of allergic airway inflammation and infection with Shistosoma mansoni eggs or respiratory syncytial virus, anti-ST2 anti- bodies, ST2 fusion proteins or ST2-deficient mice were used to show that ST2L is re- quired for development of effective Th2 responses, characterised by eosinophil recru- itment and production of proinflammatory cytokines IL- 4, IL- 5, and IL-13.
     Basis on the study of the influence of soluble ST2-IgG fusion protein on LPS res- ponses of mouse macrophages, Xu and his collaborators found that soluble ST2 play an important role in down-regulation macrophage proinflammatory activity. Treatm- ent with ST2 fusion protein before LPS stimulation markedly suppressed the produc- tion of the proinflammatory cytokines TNF-α, IL- 6 and IL-12 from macrophages. Moreover, administration of sST2 significantly reduced LPS-mediated mortality. Re- cent studies showed that through the ST2 binding protein, ST2 was able to directly bind to the surface of murine macrophages and subsequently inhibited the production of inflammatory cytokines. The molecular mechanism of this down-regulation rema- ins to be investigated, however, it appears likely that ST2 serves as a negative regul- ation factor that acts at the cell surface. Among the well-known Toll-like receptors (TLRs), TLR4 has been found to accept LPS as a ligand and subsequently triggers LPS-mediated signaling. Moreover, it also has been found that TLR4 participates in the recognition of endogenous ligands from damaged/ stressed cells, such as heat sh- ock proteins, hyaluronic acid, heparin sulfate, and high-mobility group box 1. Increa- sing evidence shows that ischemia/reperfusion-induced liver inflammation and hepat- ocellular damage is partially dependent on TLR4 signaling.
     These above findings suggested that sST2 may also be an important negative regu- lation factor of sterile inflammation and cell injury in warm hepatic I/R and the action of sST2 may be also related to TLR4 signaling. To test this hypothesis, we construct- ed a recombinant eukaryotic expression plasmid encoding murine sST2- human IgG1 Fc (sST2-Fc) fusion protein, and investigated its effect on hepatic I/R injury. We de- monstrated here that the expression of this plasmid in vivo effectively prevents hepa- tocellular damage and suppresses the activation of inflammatory cascades. Furtherm- ore, we found that the protection of sST2-Fc in hepatic I/R was also involved in dow- nregulation of TLR4 mRNA expression. These studies may provide a new route for the therapy of ischemic liver injury and other clinical setting involved inflammation diseases.
     2. Cloning of mouse soluble ST2 and human IgG1αFc gene, and construction of expression vector
     The cDNA of mouse soluble ST2 was reverse transcribed from the total RNA of mouse spleen by RT-PCR, and human IgG1αFc gene cDNA was reverse transcribed from the total RNA of human PBMC by RT-PCR too. The fragment of hIg gene was cloned into the eukaryotic expression system pcDNA 3.1 to construct pcDNA 3.1-Fc vector, then the fragment of soluble ST2 gene was inserted into pcDNA 3.1-Fc vector upstream hIg Fc to construct eukaryotic expression vector pcDNA 3.1- sST2- Fc (psST2- Fc). The recombinant plasmid psST2-Fc is identified by restriction endoniclease digestion and sequence analysis.
     3. Preparation and biological function of soluble ST2-Fc fusion protein
     The expression and identification of soluble ST2-Fc fusion protein
     By using LipofectamineTM 2000, psST2-Fc plasmid was transfected into CHO cells, and the expression of soluble sST2-Fc fusion protein in CHO cells was confirmed by ELISA and Western blot analysis. The relative amount of soluble ST2-Fc was determined with ELISA. mRNA for recombinant sST2-Fc expression was analyzed by RT-PCR. The soluble ST2-Fc fusion protein from the cultural supernatants was purified by protein A-Sepharose affinity chromatography. The results showed that CHO cells transfected with psST2-Fc secreted sST2-Fc fusion protein into cultural medium. Western blot analysis using a monoclonal antibody specific for ST2 detected a 100 kDa protein, which is in agreement with the recombinant molecular weight of sST2 and hIg Fc. A 929 bp fragment was amplified from the total RNA of psST2-Fc transfected CHO cells by RT-PCR corresponding to anticipated size. The fusion protein display a single band at 100 kDa in SDS-PAGE analysis after purified by protein A Sepharose 4B affinity chromatography. The results showed that the quality of soluble ST2-Fc fusion is suitable for further experiment.
     Soluble ST2 binds to the surface of peritoneal macrophages
     Using sST2-Fc fusion protein, we analyzed the ability of peritoneal macrophages to bind ST2. The results show that cells incubated with sST2-Fc were markedly stained with anti-hIgG. However, cells incubated with normal hIgG had a staining pattern identical with the isotype control. Importanly, this binding activity was significantly enhanced by overnight exposure to peritoneal macrophages to LPS. Soluble ST2-Fc protein inhibits TNF-a and IL- 6 production from LPS- indu- ced macrophages
     Basis on binding of sST2-Fc to peritoneal macrophages was upregulated by LPS tr- eatment, we investigated the reciprocal involvement of sST2-Fc in regulation of the LPS response. By using ELISA, we found that production of TNF-αand IL- 6 in res- pond to LPS in the peritoneal macrophages was markedly suppressed by sST2-Fc tre- atment.
     4. The warm hepatic I/R injury was attenuated by pretreatment with soluble ST2-Fc
     The expression of psST2-Fc in vivo
     By using a specific ELISA, the concentration of serum sST2-Fc protein was analy- zed in pcDNA 3.1-treated mice and psST2-Fc- transfected mice. Results show that sST2-Fc fusion protein was not detected in non-transfected mice, nor detected in pcDNA3.1- treated mice. However, the serum from sST2-Fc-transfected mice showed a significant increase in the sST2-Fc level (84.8±11.1 ng/ml) at 36 h after psST2-Fc transfection.
     The effect of psST2-Fc on warm hepatic I/R injury
     Serum ALT levels and necrotic areas in liver tissue were measured on BALB/C mice subjected to 60 min of liver ischemia and 6 h of reperfusion. Results show that serum ALT, TNF-α, and IL- 6 levels were significantly increased in control mice treated with pcDNA 3.1 or saline underwent ischemia/reperfusion injury. In contrast, mice pretreated with psST2-Fc exhibited lower levels of serum ALT compared with control animals subjected to liver I/R. Liver histological examination was consistent with the serum ALT estimation of liver damage.
     In vivo expression of psST2-Fc inhibits the activation of Kupffer cells To determine the effect of sST2 on the activation of Kupffer cells in the sinusoidal space of liver after I/R, we first observed its influence on the infiltration of Kupffer cells. By staining Kupffer cells with F4/80 on liver sections, results show that the nu- mber of F4/80+ Cells in psST2-Fc- treated group (156±30 positive cells per high power field ) was significantly reduced as compared with that in pcDNA3.1- treated group (193±26) or saline group (189±28) (P < 0.05). By using real-time RT-PCR, we found that compared with pcDNA 3.1 and salined-treated animals, the expression of TNF-α, IL- 6 was significantly suppressed in treatment with psST2-Fc.
     TLR4 levels were suppressed in the I/R liver by sST2
     TLR4 mRNA levels were examined by using real-time RT-PCR in liver tissues after I/R. Compared with sham-treated animals, liver I/R in pcDNA 3.1 or saline- treated animals resulted in an enhanced expression of TLR4 mRNA 3 h after reper- fusion. However, psST2-Fc- treated mice showed minimal increase in TLR4 mRNA levels compared with sham-treated mice after I/R.
     sST2 modulates inflammatory signaling pathway
     To investigate the possible molecular mechanism of sST2 to down-regulate the gene expression of TNF- a and IL- 6, levels of NF-κB activation were measured by EMSA. We found that NF-κB DNA-binding activity was increased in the ischemic liver 3 h after reperfusion in pcDNA 3.1 or saline-treated control mice, when compa- red with sham- treated mice. Mice pretreated with psST2-Fc exhibited less NF-κB DNA-binding activity.
     All these results show that soluble ST2 could directly bind to the surface of macro- phages, and then represses the production of proinflammatory cytokine from LPS- induced macrophages. Moreover, we successfully attenuated hepatic reperfusion in- jury after short term warm ischemia by in vivo transfection of plasmid encoding sST2 gene. The protective effect is associated with inactivation of Kupffer cells and the pr- oduction of TNF- a and IL- 6, possibly through the down-regulation of TLR4 expres- sion.
     5. Conclusion
     sST2 effectively inhibit the activation of Kupffer cells and the production of TNF-a and IL- 6, possibly through the down-regulation of TLR4 expression. Furthermore, the hepatic reperfusion injury after short term warm ischemia was significantly atten- uated by in vivo transfection of plasmid encoding sST2 gene. It was suggested that pretreatment with sST2 may be effective in settings of ischemic liver injury to mini- mize organ damage and may be useful in other clinical settings related to inflamma- tion diseases.
引文
1 . Jaeschke H. Molecular mechanisms of hepatic ischemia-reperfusion injury and preconditioning. Am J Physiol Gastrointest Liver Physiol. 2003; 284: G15 - G26.
    2 . Fondevila C, Busuttil RW, Kupiec-Weglinski JW. Hepatic ischemia/reperfusion injury– a fresh look. Exp Mol Pathol. 2003; 74: 86 - 93.
    3 . Bui AH, Kupiec-Weglinski JW, Lassman C. Recent developments in ischemic reperfusion injury in liver transplantation. Curr Opin Organ Transplant. 2006; 11: 271- 276.
    4 . Zwacka RM, Zhou W, Zhang Y, et al. Redox gene therapy for ischemia/reperfusi- on injury of the liver reduces AP1 and NF-kappa B activation. Nat Med. 1998; 4: 698 -704.
    5 . Dulkanchainun TS, Goss JA, Imagawa DK, et al. The reduction of hepatic ischemia/reperfusion injury by a soluble P-selectin glycoprotein ligand-1. Ann Surg. 1998; 227: 832-840.
    6 . Matsuda T, Yamaguchi Y, Matsumura F, et al. Immunosuppressants decrease neutrophil chemoattractant and attenuate ischemia/reperfusion injury of the liver in rats. J Trauma. 1998; 44: 475 - 484.
    7 . Zwacka RM, Zhang Y, Halldorson J, et al. CD4+ T lymphocytes mediate ischemi- a/reperfusion-induced inflammatory responses in mouse liver. J Clin Invest. 1997; 100: 279 - 289.
    8 . Anselmo DM, Amersi FF, Shen XD, et al. FTY720 pretreatment reduces warm hepatic ischemia reperfusion injury through inhibition of T-lymphocyte infiltr- ation. Am J Transplant. 2002; 2: 843 - 849.
    9 . Clavien PA, Rudiger HA, Selzner M. Mechanism of hepatocyte death after isch- emia: apoptosis versus necrosis. Hepatology. 2001; 33:1555 - 1557.
    10. Selzner N, Rudiger H, Graf R, et al. Protective strategies against ischemic injury of the liver. Gastroenterology. 2003; 125: 917- 936.
    11. Tsung A, Hoffman RA, Izuishi K, et al. Hepatic ischemia/reperfusion injuryinvolves functional TLR4 signaling in nonparenchymal cells. J. Immunol. 2005; 175: 7661- 7668.
    12. Medzhitov R. Toll-like receptors and innate immunity. Nature Rev Immunol. 2001; 1 (2): 135-145.
    13. Roach JC, Glusman G, Rowen L, et al. The evolution of vertebrate Toll-like rece- ptors. Proc Natl Acad Sci USA. 2005; 102 (27): 9577- 9581.
    14. Takeda K, Kaisho T, Akira S. Toll-like receptor. Annut Rev Immunol. 2003; 21: 335- 376.
    15. Iwasaki A, Medzhitov R. Toll-like receptor control of the adaptive immune resp- onses. Nat Immunol. 2004; 5(10): 987-995.
    16 Obhrai J, Goldstein DR. The role of Toll-like receptors in solid organ trans- plantation. Transplantation. 2006; 81(4):497-502.
    17. Akira S, Takeda K. Toll-like receptor signalling. Nat Rev Immunol. 2004; 4 (7): 499 - 511.
    18. Yarovinsky F, Zhang D, Andersen JF, et al. TLR11 activation of dendritic cells by a protozoan profilin-like protein. Science. 2005; 308 (5728): 1626 -1629.
    19. Vabulas RM, Ahmad-Nejad P, Ghose S, et al. HSP70 as endogenous stimulus of the Toll/interleukin-1 receptor signal pathway. J Biol Chem. 2002; 277: 15107- 15112.
    20. Smiley ST, King JA, Hancock WW. Fibrinogen stimulates macrophage chemok- ine secretion through Toll-like receptor 4. J Immunol. 2001;167: 2887 - 2894.
    21. Johnson GB, Brunn GJ, Kodaira Y, et al. Receptor-mediated monitoring of tissue well-being via detection of soluble heparan sulfate by Tolllike receptor 4. J Imm- unol. 2002;168: 5233 - 5239.
    22. 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: 7370 - 7371.
    23. Werenskiold AK, Hoffmann S, Klemenz R. Induction of a mitogenresponsive gene after expression of the Ha-ras oncogene in NIH 3T3 fibroblasts. Mol Cell Biol. 1989; 9:5207 - 5214.
    24. Tominaga S. A putative protein of a growth specific cDNA from BALB/c-3T3 cells is highly similar to the extracellular portion of mouse interleukin 1 receptor. FEBS Lett. 1989; 258: 301 - 304.
    25. Dunne A, O’Neill LAJ. The interleukin-1 receptor/Toll-like receptor superfamily: signal transduction during inflammation and host defense. Sci STKE. 2003; 171: re3.
    26. Tominaga S, Jenkins NA, Gilbert DJ, et al. Molecular cloning of the murine ST2 gene: characterization and chromosomal mapping. Biochim Biophys Acta. 1991; 1090: 1- 8.
    27. Dale M, Nicklin MJ. Interleukin-1 receptor cluster: gene organization of IL1R2, IL1R1, IL1RL2 (IL-1Rrp2), IL1RL1 (T1/ST2), and IL18R1 (IL-1Rrp) on human chromosome 2q. Genomics. 1999; 57: 177 - 179.
    28. Kumar S, Minnich MD, Young PR. ST2/T1 protein functionally binds to two secreted proteins from Balb/c 3T3 and human umbilical vein endothelial cells but does not bind interleukin 1. J Biol Chem. 1995; 270: 27905 - 27913.
    29. Gayle MA, Slack JL, Bonnert TP, et al. Cloning of a putative ligand for the T1/ ST2 receptor. J Biol Chem. 1996; 271: 5784 - 5789.
    30. Moritz DR, Rodewald HR, GheyselinckJ, et al. The IL-1 receptor-related T1 anti- gen is expressed on immature and mature mast cells and on fetal blood mast cell progenitors. J Immunol. 1998; 161: 4866 - 4874.
    31. Xu D, Chan WL, Leung BP, et al. Selective expression of a stable cell surface molecule on type 2 but not type 1 helper T cells. J Exp Med 1998; 187:787-794.
    32. Lohning M, Stroehmann A, Coyle AJ, et al. T1/ST2 is preferentially expressed on murine Th2 cells, independent of interleukin 4, interleukin 5, and interleukin 10, and important for Th2 effector function. Proc Natl Acad Sci USA. 1998; 95: 6930 - 6935.
    33. Tominaga S, Kuroiwa K, Tago K, et al. Presence and expression of a novel variant form of ST2 gene product in human leukemic cell line UT-7/GM. Biochem Biop- hys Res Commun. 1999; 264: 14 -18.
    34. Kropf P, Bickle Q, Herath S, et al. Organ-specific distribution of CD4+ T1/ST2+Th2 cells in Leishmania major infection. Eur J Immunol. 2002; 32: 2450 - 2459.
    35. Coyle AJ, Lloyd C, Tian J, et al. Crucial role of the interleukin 1 receptor family member T1/ST2 in T helper cell type 2-mediated lung mucosal immune responses. J Exp Med. 1999; 190: 895- 902.
    36. Lambrecht BN, De Veerman M, Coyle AJ, et al. Myeloid dendritic cells induce Th2 responses to inhaled antigen, leading to eosinophilic airway inflammation. J Clin Invest. 2000;106: 551–559.
    37. Lohning M, Grogan JL, Coyle AJ, et al. T1/ST2 expression is enhanced on CD4+ T cells from schistosome egg-induced granulomas: analysis of Th cell cytokine coexpression ex vivo. J Immunol. 1999; 162: 3882 - 3889.
    38. Walzl G, Matthews S, Kendall S, et al. Inhibition of T1/ST2 during respiratory syncytial virus infection prevents T helper cell type 2 (Th2)- but not Th1-driven immunopathology. J Exp Med. 2001; 193: 785 - 792.
    39. Trajkovic V, Sweet MJ, Xu D. T1/ST2-an IL-1 receptor-like modulator of imm- une response. Cytokine Growth F R. 2004; 15: 87-95.
    40. Kumar S, Tzimas MN, Griswold DE, et al. Expression of ST2, an interleukin-1 receptor homologue, is induced by pro-inflammatory stimuli. Biochem Biophys Res Commun. 1997; 235: 474 - 478.
    41. Kuroiwa K, Arai T, Okazaki H, et al. Identification of human ST2 protein in the sera of patients with autoimmune diseases. Biochem Biophys Res Commun. 2001; 284: 1104 - 1108.
    42. Oshikawa K, Yanagisawa K, Tominaga S, et al. ST2 protein induced by inflamm- atory stimuli can modulate acute lung inflammation. Biochem Biophys Res Com- mun. 2002; 299: 18 - 24.
    43. Sweet M, Leung B, Kang D, et al. A novel pathway regulating lipopolysacchar- ide-induced shock by ST2/T1 via inhibition of Toll-like receptor 4 expression, J Immunol. 2001; 166: 6633 - 6639.
    44. Nomura F, Akashi S, Sakao Y, et al. Endotoxin tolerance in mouse peritoneal macrophages correlates with down-regulation of surface Toll-like receptor 4 ex- pression. J Immunol. 2000; 164: 3476 -3479.
    45. Gachter T, Werenskiold AK, Klemenz R. Transcription of the interleukin-1 recep- tor-related T1 gene is initiated at different promoters in mast cells and fibroblasts. J Biol Chem. 1996; 271: 124-129.
    46. Sambrook J, Fritishi EF, Maniatis.分子克隆实验指南.金冬雁,黎孟枫,译.第二版.北京:科学出版社, 1999.
    47. Steinhorn DM, Cerra FB. Comparative effects of lipopolysaccharide on newborn versus adult rat hepatocyte and nonparenchymal cell co-cultures. Crit Care Med. 1997; 25 :121-127.
    48. Wanner GA, Muller PE, ErtelW, et al. Differential effect of anti-TNF-alpha anti- body on proinflammatory cytokine release by Kupffer cells following liver ische- mia and reperfusion. Shock. 1999; 11: 391- 395.
    49. Colletti LM, Kunkel SL, Walz A, et al. The role of cytokine networks in the local liver injury following hepatic ischemia/reperfusion in the rat. Hepatology. 1996; 23: 506 - 514.
    50. Suzuki S, Toledo-Pereyra LH, Rodriguez FJ, et al. Neutrophil infiltration as an important factor in liver ischemia and reperfusion injury. Modulating effects of FK506 and cyclosporine. Transplantation. 1993; 55: 1265 - 1272.
    51. Jaeschke H, Farhood A. Neutrophil and Kupffer cell-induced oxidant stress and ischemia–reperfusion injury in rat liver. Am J Physiol. 1991; 260 : G355 - G362.
    52. Yamamoto T, Naito M, Moriyama H, et al. Repopulation of murine Kupffer cells after intravenous administration of liposome-encapsulated dichloromethylene dip- hosphonate. Am J Pathol. 1996; 149: 1271- 1286.
    53. Kukan M, Vajdova K, Horecky J, et al. Effects of blockade of Kupffer cells by gadolinium chloride on hepatobiliary function in cold ischemia-reperfusion injury of rat liver.Hepatology. 1997; 26: 1250 -1257.
    54. Frankenberg M, Weimann J, Fritz S, et al. Gadolinium chloride-induced impro- vement of postischemic hepatic perfusion after warm ischemia is associated with reduced hepatic endothelin secretion. Transplant International. 2005; 18 : 429 - 436.
    55. Hoffmann JA, Reichhart JM. Drosophila innate immunity: an evolutionary pers-pective. Nat Immunol. 2002; 3(2): 121-126.
    56. Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, et al. Recognition of comm- ensal microflora by Toll-like receptors is required for intestinal homeostasis. Cell. 2004; 118(2): 229 - 241.
    57. Gordon S. Pattern recognition receptors: doubling up for the innate immune resp- onse. Cell. 2002; 111: 927 - 930.
    58. Schnare M, Barton GM, Holt AC, et al. Toll-like receptors control activation of adaptive immune responses. Nat Immunol. 2001; 2 (10): 947 - 950.
    59. Andrade C, Waddell TK, Keshavijee S, et al. Innate immunity and organ transp- lantation:the potential role of Toll-like receptors. Am J Transplant. 2005; 5: 969- 975.
    60. Dabbagh K, Lewis DB. Toll-like receptors and T-helper-1/Thelper-2 responses. Curr Opin Infect Dis. 2003; 16: 199-204.
    61. Kapsenberg ML. Dendritic-cell control of pathogen-driven T-cell polarization. Nat Rev Immunol. 2003; 3: 984 - 993.
    62. Fitzgerald KA, Palsson-McDermott EM, Bowie AG, et al. Mal (MyD88-adapter- like) is required for Toll-like receptor-4 signal transduction. Nature. 2001; 413: 78 - 83.
    63. Horng T, Barton GM, Flavell RA, et al. The adaptor molecule TIRAP provides signalling specificity for Toll-like receptors. Nature. 2002; 420: 329 - 333.
    64. Zhai Y, Shen X, Connell RO, et al. Cutting edge: TLR4 activation mediates liver ischemia/reperfusion inflammatory response via IFN regulatory factor 3-depend- ent MyD88-independent pathway. J Immunol. 2004; 173: 7115 - 7119.
    65. Hancock WW, Gao W, Csizmadia V, et al. Donor-derived IP-10 initiates develo- pment of acute allograft rejection. J Exp Med. 2001; 193(8): 975 - 980.
    66. Romics L, Dolganiuc A, Kodys K, et al. Selective priming to Toll-like receptor 4 (TLR4), not TLR2, ligands by P. acnes involves up-regulation of MD-2 in mice. Hepatology. 2004; 40: 555 - 564.
    67. Ohashi K, Burkart V, Flohe S, et al. Cutting edge: heat shock protein 60 is a puta- tive endogenous ligand of the Toll-like receptor-4 complex. J Immunol. 2000; 164:558 - 561.
    68. Asea A, Kraeft SK, Kurt-Jones EA, et al. HSP70 stimulates cytokine production through a CD14-dependant pathway, demonstrating its dual role as a chaperone and cytokine. Nat Med. 2000; 6: 435 - 442.
    69. Oyama J, Blais C, Liu X, et al. Reduced myocardial ischemia-reperfusion injury in Toll-like receptor 4-deficient mice. Circulation. 2004; 109: 784–789.
    70. Gajewska BU, Swirski FK, Alvarez D, et al. Temporal-spatial analysis of the im- mune response in a murine model of ovalbumin-induced airways inflammation. Am J Respir Cell Mol Biol. 2001; 25: 326–334.
    71. Townsend MJ, Fallon PG, Matthews DJ, et al. T1/ST2-deficient mice demonstrate the importance of T1/ST2 in developing primary T helper cell type 2 responses. J Exp Med. 2000; 191: 1069– 1076.
    72. Han J, Ulevitch RJ. Limiting inflammatory responses during activation of innate immunity. Nat Rev Immunol. 2005; 6: 1198 - 1205.
    73. Fagundes CT, Amaral F A, Souza ALS, et al. ST2, an IL-1R family member, attenuates inflammation and lethality after intestinal ischemia and reperfusion. J Leukoc Biol. 2007; 81: 492–499.
    74. Schmitz J, Owyang A, Oldham E, et al. IL-33, an interleukin-1-like cytokine that signals via the IL-1 receptor-related protein st2 and induces T helper type 2- asso- ciated cytokines. Immunity. 2005; 23: 479- 490.
    75. Carriere V, Roussel L, Ortega N, et al. IL-33, the IL-1-like cytokine ligand for ST2 receptor, is a chromatin-associated nuclear factor in vivo. Proc Natl Acad Sci USA. 2007; 104(1): 282- 287.
    76. Chomarat P, Banchereau J. Interleukin-4 and interleukin-13: their similarities and discrepancies. Int Rev Immunol. 1998; 17: 1- 52.
    77. Corry DB. IL-13 in allergy: home at last. Curr Opin Immunol. 1999;11: 610 - 614.
    78. Karlen S, De Boer ML, Lipscombe RJ, et al. Biological and molecular character- istics of interleukin-5 and its receptor. Int Rev Immunol. 1998; 16: 227- 247.
    79. Karin M, Lin A. NF-κB at the crossroads of life and death. Nat Immunol. 2002; 3:221- 227.
    80. Kato A, Edwards MJ, Lentsch AB. Gene deletion of NF-kB p50 does not alter the hepatic inflammatory response to ischemia/reperfusion. J Hepatol. 2002; 37: 48 - 55.
    81. Yoshidome H, Kato A, Edwards MJ, et al. Interleukin-10 suppresses hepatic isch- emia/reperfusion injury in mice: implications of a central role for nuclear factorκB. Hepatology. 1999; 30: 203 - 208.
    82. Iimuro Y, Nishiura T, Hellerbrand C, et al. NF-κB prevents apoptosis and liver dysfunction during liver regeneration. J Clin Invest. 1998; 101: 802 - 811.
    83. Takahashi Y, Ganster RW, Gambotto A, et al. Role of NF-κB on liver cold ische- mia reperfusion injury. Am J Physiol. 2002; 283: G1175- G1184.
    84. Suetsugu H, Iimuro Y, Uehara T, et al. Nuclear factor-κB inactivation in the rat liver ameliorates short term total warm ischaemia/reperfusion injury. Gut. 2005; 54: 835 - 842.
    85. Yin H, Huang B, Yang H, et al. pretreatment with soluble ST2 reduces warm hepatic ischemia/reperfusion injury. Biochem Biophys Res Commun. 2006; 351: 940- 946.
    1 . ?zkaynak E, Gao W, Shemmeri N, et al. Importance of ICOS- B7RP-1 costimu- lation in acute and chronicallograft rejection. Nat Immunol. 2001; 2: 591- 596.
    2 . Wells AD, Li XC, Li YS, et al. Requirement for T-cell apoptosis in the induction of peripheral transplantation tolerance. Nat Med. 1999; 5: 1303 -1307.
    3 . Rulifsona IC, Szota GL, Palmerb E, et al. Inability to induce tolerance through direct antigen presentation. Am J Transplant. 2002; 2: 510- 519.
    4 . Orosz CG. Advances in the study of chronic allograft rejection. Curr Opin Organ Transplant. 2003; 8: 167- 171.
    5 . Palmer SM, Burch LH, Mir S, et al. Donor polymorphisms in Toll-like receptor-4 influence the development of rejection after renal transplantation. Clin Transplant. 2005; 20: 30- 36.
    6 . Pierson RN. Lung Transplantation: Current Status and Challenges. Transplantation. 2006; 81: 1609 - 1615.
    7 . Medzhitov R, Janeway CA. Decoding the patterns of self and nonself by the innate immune system. Science. 2002; 296: 298- 300.
    8 . Matzinger P. The danger model: a renewed sense of self. Science. 2002; 296: 301- 305.
    9 . Matzinger P. Tolerance, danger, and the extended family. Annu Rev Immunol. 1994; 12: 991- 1045.
    10. Goldstein DR. Toll-like receptors and other links between innate and acquired all- oimmunity. Curr Opin Immunol. 2004; 16: 538- 544.
    11. Andrade CF, Waddell TK, Keshavjee S, et al. Innate immunity and organ transpl- antation: the potential role of Toll-like receptors. Am J Transplant. 2005; 5: 969 - 975.
    12. Barton GM, Medzhitov R. Control of adaptive immune responses by Toll-like receptors. Curr Opin Immunol. 2002; 14: 380- 383.
    13. Beg AA. Endogenous ligands of Toll-like receptors: implications for regulatinginflammatory and immune responses. Trends Immunol. 2002; 23: 509- 512.
    14. Moine AL, Goldman M. Contributions of innate immunity to allograft rejection and survival. Curr Opin Organ Transplant. 2003; 8: 2- 6.
    15. Bui AH, Kupiec-Weglinski JW, Lassman C. Recent developments in ischemic re- perfusion injury in liver transplantation. Curr Opin Organ Transplant. 2006; 11: 271-276.
    16. Huang Y, Yin H, Han J, et al. Extracellular hmgb1 functions as an innate immune- mediator implicated in murine cardiac allograft acute rejection. Am J Transplant. 2007; 7: 1-10.
    17. Obhrai J, Goldstein DR. The role of Toll-like receptors in solid organ transplanta- tion. Transplantation. 2006; 81: 497- 502.
    18. Hoffmann JA, Reichhat JM. Drosophila innate immunity: an evolutionary perspe- ctive. Nat Immunol. 2002; 3(2): 121 - 126.
    19. Hoffmann JA. The immune response of Drosophila. Nature. 2003; 426: 33 - 38.
    20. Takeda K, Akira S. TLR signaling pathways. Semin Immunol. 2004; 16: 3 - 9.
    21. O’Neill LAJ, Fitzgerald KA, Bowie AG. The Toll-IL-1 receptor adaptor family grows to five members. Trends Immunol. 2003; 24 (6): 286 - 290.
    22.Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, et al. Recognition of commensal microflora by Toll-like receptors is required for intestinal homeostasis. Cell. 2004; 118 (2): 229 - 241.
    23. Yarovinsky F, Zhang D, Andersen JF, et al. TLR 11 activation of dendritic cells by a protozoan profilin-like protein. Science. 2005; 308 (5728): 1626 -1629.
    24. Roach JC, Glusman G, Rowen L, et al. The evolution of vertebrate Toll-like rece- ptors. Proc Natl Acad Sci U S A. 2005; 102 (27): 9577 - 9582.
    25. Zhang D, Zhang G, Hayden MS, et al. A Toll-like receptor that prevents infection by uropathogenic bacteria. Science. 2004; 303:1522 - 1526.
    26. Cook C, Pisetsky D, Schwartz D. Toll-like receptors in the pathogenesis of human disease. Nat Immunol. 2004; 5 (10): 975 - 979.
    27. Gordon S. Pattern recognition receptors: doubling up for the innate immune respo- nse. Cell. 2002; 111 (7): 927- 930.
    28. Takeda K, Kaisho T, Akira S. Toll-like receptors. Annu Rev Immunol. 2003; 21: 335 - 376.
    29. Krutzik SR, Tan B, Li H, et al. TLR activation triggers the rapid differentiation of monocytes into macrophages and dendritic cells. Nat Med. 2005; 11: 653 - 660.
    30. Akira S. Mammalian Toll-like receptors. Curr Opin Immunol. 2003; 15: 5 - 11.
    31. Hertz CJ,Wu Q, Porter EM, et al. Activation of Toll-like receptor 2 on human tracheobronchial epithelial cells induces the antimicrobial peptide human {beta} defensin-2. J Immunol. 2003; 171 (12): 6820 - 6826.
    32. Eisenbarth SC, Piggott DA, Huleatt JW, et al. Lipopolysaccharide-enhanced, Toll- like receptor 4-dependent t helper cell type 2 responses to inhaled antigen. J Exp Med. 2002; 196 (12): 1645-1651.
    33. Barton GM, Medzhitov R. Toll-like receptor signaling pathways. Science. 2003; 300 (5625): 1524 - 1525.
    34. Tauszig-Delamasure S, Bilak H, Capovilla M, et al. Drosophila MyD88 is required for the response to fungal and gram-positive bacterial infections. Nat Immunol. 2002; 3: 91-97.
    35. Paterson HM, Murphy TJ, Purcell EJ, et al. Injury primes the innate immune syst- em for enhanced Toll-like receptor reactivity. J Immunol. 2003; 171: 1473 - 1483.
    36. Han J. MyD88 beyond Toll. Nat Immunol. 2006; 7:370-371.
    37. Horng T, Barton GM, Medzhitov R. TIRAP: an adapter molecule in the Toll signaling pathway. Nat Immunol. 2001; 2 (9): 835-841.
    38. Horng T, Barton GM, Flavell RA, et al. The adaptor molecule TIRAP provides signalling specificity for Toll-like receptors. Nature. 2002; 420 (6913): 329- 333.
    39. Hoebe K, Janssen EM, Kim SO, et al. Upregulation of costimulatory molecules induced by lipopolysaccharide and double-stranded RNA occurs by Trif- depe- ndent and Trif-independent pathways. Nat Immunol. 2003; 4 (12): 1223 - 1229.
    40. Yamamoto M, Sato S, Hemmi H, et al. Role of adaptor TRIF in the MyD88- in- dependent Toll-like receptor signaling pathway. Science. 2003; 301 (5633): 640- 643.
    41. Yamamoto M, Sato S, Hemmi H, et al. TRAM is specifically involved in the Toll-like receptor 4-mediated MyD88-independent signaling pathway. Nat Immunol. 2003; 4 (11): 1144 - 1150.
    42. Han J, Ulevitch RJ. Limiting inflammatory responses during activation of innate immunity. Nat Immunol. 2005; 6: 1198-1205.
    43. Kobayashi K, Hernandez LD, Galan JE, et al. IRAK-M is a negative regulator of Toll -like receptor signaling. Cell. 2002; 110 (2): 191-202.
    44. Wald D, Qin J, Zhao Z, et al. SIGIRR, a negative regulator of Toll-like receptor- interleukin 1 receptor signaling. Nat Immunol. 2003; 4 (9): 920-927.
    45. Iwasaki ARM. Toll-like receptor control of the adaptive immune responses. Nat Immunol. 2004; 5(10): 987-995.
    46. Shortman K, Liu Y. Mouse and human dendritic cell subtypes. Nat Rev Immunol. 2002; 2: 151-161.
    47. Yin H, Huang B, Yang H, et al. Pretreatment with soluble ST2 reduces warm hepatic ischemia/reperfusion injury. Biochem Biophys Res Commun. 2006; 351: 940-946.
    48. Li M, Carpio DF, Zheng Y, et al. An essential role of the NF-kappa B/Toll-like rece- ptor pathway in induction of inflammatory and tissue-repair gene expression by necrotic cells. J Immunol. 2001; 166: 7128-7135.
    49. Fischer S, Maclean AA, Liu M, et al. Dynamic changes in apoptotic and necrotic cell death correlate with severity of ischemia/reperfusion injury in lung transplant- ation. Am J Respir Crit Care Med. 2000; 162: 1932-1939.
    50. Lucas M, Stuart LM, Savill J, et al. Apoptotic cells and innate immune stimuli combine to regulate macrophage cytokine secretion. J Immunol. 2003; 171: 2610- 2615.
    51. Asea A, Kraeft SK, Kurt-Jones EA, et al. HSP70 stimulates cytokine production through a CD14-dependant pathway, demonstrating its dual role as a chaperone and cytokine. Nat Med. 2000; 6 (4): 435 - 442.
    52. Brown BD, Lillicrap D. Dangerous liaisons: the role of“danger”signals in the im- mune response to gene therapy. Blood. 2002; 100: 1133-1140.
    53. Izuishi K, Tsung A, Jeyabalan G, et al. Cutting edge: high-mobility group box 1preconditioning protects against liver ischemia- reperfusion injury. J Immunol. 2005; 176: 7154 - 7158.
    54. Tsan MF, Gao B. Endogenous ligands of Toll-like receptors. J Leukoc Biol. 2004; 76: 514 - 519.
    55. Kariko K, Ni H, Capodici J, et al. mRNA is an endogenous ligand for Toll-like re- ceptor 3. J Biol Chem. 2004; 279: 12542 - 12550.
    56. Tabeta K, Georgel P, Janssen E, et al. Toll-like receptors 9 and 3 as essential com- ponents of innate immune defense against mouse cytomegalovirus infection. Proc Natl Acad Sci U S A. 2004; 101: 3516 - 3521.
    57. Heil F, Hemmi H, Hochrein H, et al. Species-specific recognition of single-stran- ded RNA via toll-like receptor 7 and 8. Science. 2004; 303: 1526 - 1529.
    58. Walker WE, Nasr I, Camira. Absence of innate MyD88 signaling promotes indu- cible allograft acceptance1. J Immunol. 2006; 177: 5307- 5316.
    59. Schnare M, Barton GM, Holt AC, et al. Toll-like receptors control activation of adaptive immune responses. Nat Immunol. 2001; 2: 947- 950.
    60. Jankovic D, Kullberg MC, Hieny S, et al. In the absence of IL-12, CD4(+) T cell responses to intracellular pathogens fail to default to a Th2 pattern and are host protective in an IL-10(-/-) setting. Immunity. 2002; 16 (3): 429 - 439.
    61. Goldstein DR, Tesar BM, Akira S, et al. Critical role of the Toll-like receptor sig- nal adaptor protein MyD88 in acute allograft rejection. J Clin Invest. 2003; 111: 1571- 1578.
    62. Tesar BM, Zhang J, Li Q, et al. TH1 immune responses to fully MHC mismatched allografts are diminished in the absence of MyD88, a Toll-like receptor signal adaptor protein. Am J Transplant. 2004; 4: 1429 - 1439.
    63. Saleem S, Konieczny BT, Lowry RP, et al. Acute rejection of vascularized heart allografts in the absence of IFN gamma. Transplantation. 1996; 62: 1908 - 1911.
    64. Palmer SM, Burch LH, Davis RD, et al. The role of innate immunity in acute allo- graft rejection after lung transplantation. Am J Respir Crit Care Med. 2003; 168 (6): 628 - 632.
    65. Palmer SM, Burch LH, Trindade AJ, et al. Innate immunity influences long-termoutcomes after human lung transplant. Am J. Respir Crit Care Med. 2005; 171 (7): 780 - 785.
    66. Kiechl S, Lorenz E, Reindl M, et al. Toll-like receptor 4 polymorphisms and athe- rogenesis. N Engl J Med. 2002; 347 (3): 185 - 192.
    67. Methe H, Zimmer E, Grimm C, et al. Evidence for a role of Toll-like receptor 4 in development of chronic allograft rejection after cardiac transplantation. Transpla- ntation. 2004; 78(9): 1324- 1331.
    68. Tailleux L, Schwartz O, Herrmann JL, et al. DC-SIGN is the major Mycobacteri- um tuberculosis receptor on human dendritic cells. J Exp Med. 2003; 197 (1): 121 - 127.
    69. Gantner BN, Simmons RM, Canavera SJ, et al. Collaborative induction of inflam- matory responses by dectin-1 and Toll-like receptor 2. J Exp Med. 2003; 197 (9): 1107 - 1117.
    70. Philpott DJ, Girardin SE. The role of Toll-like receptors and Nod proteins in bac- terial infection. Mol Immunol. 2004; 41 (11): 1099 - 1108.
    71. Kondo T, Morita K, Watarai Y, et al. Early increased chemokine expression and production in murine allogeneic skin grafts is mediated by natural killer cells. Tra- nsplantation. 2000; 69 (5): 969- 977.
    72. Fox A, Koulmanda M, Mandel TE, et al. Evidence that macrophages are required for T-cell infiltration and rejection of fetal pig pancreas xenografts in nonobese diabetic mice. Transplantation. 1998; 66 (11): 1407- 1416.
    73. Maier S, Tertilt C, Chambron N, et al. Inhibition of natural killer cells results in acceptance of cardiac allografts in CD28-/- mice. Nat Med. 2002; 7 (5): 557 - 562.
    74. Pratt JR, Basheer SA, Sacks SH. Local synthesis of complement component C3 regulates acute renal transplant rejection. Nat Med. 2002; 8 (6): 582 - 587.