Toll-Like Receptor-9 Agonist Inhibits Airway
TLRs信号通路和TLRs的Cross-talk在炎症性疾病中作用的研究进展
第48卷第3期2022年5月吉林大学学报(医学版)Journal of Jilin University(Medicine Edition)Vol.48No.3May2022DOI:10.13481/j.1671‑587X.20220334TLRs信号通路和TLRs的Cross-talk在炎症性疾病中作用的研究进展Progress research in role of TLRs signaling pathway and Cross-talk of TLRs in inflammatory diseases蒋孙班1,康思思2,赵利娜1,王朝1,蒋丽娜1(1.河北北方学院医学检验学院免疫教研室,河北张家口075000;2.河北省张家口市第二医院患者回访中心,河北张家口075000)[摘要]Toll样受体(TLRs)是一种重要的模式识别受体(PRR),主要通过2条信号通路向下游传递信号以发挥免疫学效应。
经过下游分子诱导的TLR通过和其他PRR(包括其他TLRs)、免疫分子和蛋白酶类的交叉作用,即Cross-talk,与炎症性疾病的发生发展过程密切相关。
TLR信号通路包括MyD88依赖信号通路和MyD88非依赖性信号通路(TRIF通路),其下游的信号分子肿瘤坏死因子受体相关因子3(TRAF3)和肿瘤坏死因子受体相关因子6(TRAF6),在引导信号传导方向的过程中起重要作用。
TLRs信号通路能完全激活炎症,而TLRs的Cross-talk参与各种炎症性疾病的预后和转归。
TLRs的Cross-talk在系统性红斑狼疮、急性肺损伤和脓毒症等炎症性疾病的发生过程中通过增加细胞因子的分泌、激活蛋白酶使免疫细胞过度活化和增强免疫细胞的趋化作用加速相关疾病进程,甚至在炎症末期因机体免疫分子及免疫细胞消耗过度而引发免疫抑制,这阻碍了机体免疫稳态的维持。
现对炎症性疾病进程中组织和细胞中TLRs信号通路分子的表达变化及其Cross-talk作用的分子机制进行综述,深入了解TLRs的Cross-talk在炎症发生发展中的作用机制,为治疗炎症性疾病提供新的策略和靶标。
大鼠Toll样受体9(TLR-9CD289)ELISA试剂盒
YM-S0007人白细胞活化黏附因子(ALCAM)ELISA kit ELISA试剂盒96T/48T进口/国产
YM-S0008人活化素A(Activin-A)ELISA kit ELISA试剂盒96T/48T进口/国产
YM-S0015人内分泌腺来源的血管内皮生长因子(EG-VEGF)ELISA kit ELISA试剂盒96T/48T进口/国产
YM-S0016人睫状神经营养因子(CNTF)ELISA kit ELISA试剂盒96T/48T进口/国产
YM-S0017人白细胞分化抗原30(CD30)ELISA kit ELISA试剂盒96T/48T进口/国产
YM-S0021人E钙粘着蛋白/上皮性钙黏附蛋白(E-Cad)ELISA kit ELISA试剂盒96T/48T进口/国产
YM-S0003人角化细胞内分泌因子(KAF)/双调蛋白(AR)ELISA kit ELISA试剂盒96T/48T进口/国产
YM-S0004人血管紧张素Ⅱ(ANG-Ⅱ)ELISA kit ELISA试剂盒96T/48T进口/国产
YM-S0005人血管紧张素转化酶(ACE)ELISA kit ELISA试剂盒96T/48T进口/国产
大鼠Toll样受体9(TLR-9/CD289)ELISA试剂盒相关详情来电咨询。
联系方式: 电话:021-51829239 在 线QQ:953483277
YM-S0001人血管生成素1(ANG-1)ELISA kit ELISA试剂盒96T/48T进口/国产
YM-S0002人血管生长素(ANG)ELISA kit ELISA试剂盒96T/48T进口/国产
黄芪多糖免疫作用的基础与临床研究进展
黄芪多糖免疫作用的基础与临床研究进展杨志霞;王琦【摘要】黄芪迄今已有2000多年的药用历史,其主要药效成分黄芪多糖在中医或中西医结合防治多种疾病的基础与临床应用研究中,发挥着重要的免疫调节药理作用.但目前对黄芪多糖的临床配伍及单体效价的研究尚处于探索阶段,故利用先进的科技手段,开发黄芪多糖新的作用靶点和功效的研究工作亟待加强.【期刊名称】《世界中医药》【年(卷),期】2013(008)007【总页数】4页(P833-836)【关键词】黄芪;黄芪多糖;免疫调节;黄芪制剂【作者】杨志霞;王琦【作者单位】北京中医药大学基础医学院中医体质与生殖中心,北京,100029;北京中医药大学基础医学院中医体质与生殖中心,北京,100029【正文语种】中文黄芪为豆科植物蒙古黄芪Astragalusmembranaceus(Fisch.)Bgevar.monghlicus(Bge.)Hisao或膜荚黄芪Astragalusmembranaceus(Fisch.)Bge.的干燥根,主产于内蒙古、山西、黑龙江、甘肃等地,《中华人民共和国药典》2010年版规定只有膜荚黄芪和蒙古黄芪是黄芪正品药材,后者是前者的变种[1]。
黄芪味甘,性温,入脾肺经,生用能补气固表,利尿托毒,排脓,敛疮生肌之效[2],“可治一切气衰血虚之症”[3]。
其药用历史迄今已有2000多年,始载于《神农本草经》,明《本草纲目》记载“耆长也,黄芪色黄,为补老之长故名”,《兰室秘藏·卷上》亦将黄芪之名冠于方剂之首。
秦汉以前多用治痈疽等外科疾病,秦汉两晋时期则更突出“补虚”功效[4]。
黄芪药效成分主要包括黄芪多糖(Astragalus Polysaccharide,APS)、黄酮类及皂苷类,黄酮类主要表现为心血管活性,皂苷类主要与抗衰老和免疫调节活性有关[5],而APS则是黄芪中含量最多并且免疫活性最强的一类物质[6],具有免疫调节、抗炎、提高巨噬细胞活性、抗肿瘤[7]、保护心肌[8]及抗氧化辐射[9]等多方面作用,但也有其有效性问题。
每周只需注射一次,3个月即可轻松减掉10斤肥肉能让你管住嘴的减肥神药真的来了 临床大发现
每周只需注射一次,3个月即可轻松减掉10斤肥肉。
能让你管住嘴的减肥神药真的来了临床大发现“管住嘴,迈开腿”简简单单六个字,就道出了减肥的真谛。
然而,面对那么多的美食诱惑,光这前三个字就足以让无数人的减肥大业半途而废了。
不过,好消息来了!最近,肥胖研究领域中的著名期刊《糖尿病,肥胖和代谢》杂志刊登的一项临床研究[1]显示,诺和诺德公司开发的索马鲁肽,可以抑制食欲,让你轻松“管住嘴”。
只需一周注射1次,连续注射12周后,就可减重10斤!而且,在这减轻的体重中,主要还是体内的脂肪组织,药物对除脂肪以外的去脂体重影响很小。
不光有效,还很安全!这项研究的通讯作者,来自英国利兹大学的John Blundell 教授表示,“索马鲁肽的作用是非常令人惊讶的,我们在12周内就观察到了其他减肥药物需要6个月才能达到的效果。
它减少了饥饿感和食欲,让患者能更好地控制饮食摄入。
”[2] John Blundell教授索马鲁肽(Semaglutide)本身是一款针对2型糖尿病的降糖药,主要成分为胰高血糖素样肽-1(GLP-1)类似物。
GLP-1是一种由小肠分泌的激素,在血液中葡萄糖水平升高时促进胰岛素的合成和分泌。
GLP-1进入人体后很容易被酶降解,天然的GLP-1半衰期仅有几分钟,所以,为了让它更长久的工作,研究人员会对它进行一些结构上的改造,在保留功能的同时不那么容易被酶降解。
这样得到的GLP-1类似物药物,比如大名鼎鼎的利拉鲁肽,可以将注射频率减缓到每天1~2次。
而索马鲁肽可以说是它们的“升级版”,在经过改造后,它的半衰期可延长至大约1周,因此注射一次的效果可以维持大约一周的时间[3],对于患者来说更方便。
在不久前公布的全球大型III期临床试验中,索马鲁肽表现优秀,既能控制血糖,还可以保护心血管,这为它在上周赢得了FDA内分泌及代谢药物专家咨询委员会16:0的支持率,不出意外的话,索马鲁肽上市在即[4]。
不少分析人士预测它未来十年内的销售峰值将超百亿,成为治疗2型糖尿病中最好的降糖药。
Toll样受体9(TLR9)在鱼类中的研究进展
2021年第02期Toll 样受体9(TLR9)在鱼类中的研究进展王冠杰1,2,胡国斌1,2★(1.中国海洋大学海洋生物科学学院,山东青岛266003;2.中国海洋大学进化与海洋生物多样性研究所,山东青岛266003)1免疫背景先天免疫系统是宿主保护机体对抗微生物病原体入侵时主要的防御机制。
与高等脊椎动物如,鸟类和哺乳类等具有适应性免疫系统不同,低等脊椎动物如鱼类主要依赖于先天免疫系统,在抵抗病原微生物中赋予更快但特异性较低的免疫应答。
黏膜免疫系统是机体先天免疫的最重要组成部分之一,是机体抵御感染的第一道防线。
由于生活在病原体丰富的水环境中,鱼类的体粘膜表面直接暴露在外部病原微生物中,由多种病原体不断开垦。
因此,为了防止病原体的附着和侵袭,首先要对粘膜表面的病原体进行检测、筛选和识别。
胚系基因编码的抗原识别受体(PRRs ),分布在细胞表面、细胞内或血液和组织中,能识别由细菌、病毒、真菌和原生动物表达而触发的保守的病原相关分子模式(PAMPs ),包括脂蛋白、dsRNA 、鞭毛蛋白和微生物的CpG ODN 。
因此在粘膜免疫中被视为一个识别病原体和激活免疫信号通路的重要组成部分。
根据特征不同,迄今为止报道了PRRs 的三大主要集合,包括TLRs ,NLRs 和RLRs 。
2TLRs 基本特征TLRs 基因在宿主防御系统第一防线中起到防止原体附着和进入的角色,具有物种特定性。
TLRs 具有LRRs 结构域的胞外区,能促进PAMPs 的识别,跨膜区,便于与膜体附着,作者简介:王冠杰(1993~),女,天津人,硕士研究生,研究方向:海水养殖动物病害与免疫。
★通信作者:胡国斌(1971~),湖北武汉人,博士,教授,研究方向:海水养殖动物疾病与免疫。
摘要:Toll 样受体(TLRs )是一类细胞外N-端具有富含亮氨酸重复(LRRs )结构域和细胞内C-端具有Toll/白介素(IL )-1受体(TIR )结构域的跨膜蛋白,是连接天然免疫和获得性免疫的桥梁。
碧云天MTT细胞增殖及细胞毒性检测试剂盒说明书.pdf_1694034687.4777226
MTT 细胞增殖及细胞毒性检测试剂盒产品编号 产品名称包装 C0009S MTT 细胞增殖及细胞毒性检测试剂盒 500次 C0009MMTT 细胞增殖及细胞毒性检测试剂盒2500次产品简介:MTT 细胞增殖及细胞毒性检测试剂盒(MTT Cell Proliferation and Cytotoxicity Assay Kit)是一种非常经典的细胞增殖和细胞毒性检测试剂盒,被广泛应用于细胞增殖和细胞毒性的检测。
MTT 可以被线粒体内的一些脱氢酶还原生成结晶状的深紫色产物formazan (图1A)。
在特定溶剂存在的情况下,可以被完全溶解(图1B)。
然后通过酶标仪可以测定570nm 波长附近的吸光度(图2)。
细胞增殖越多越快,则吸光度越高;细胞毒性越大,则吸光度越低。
图1. MTT 细胞增殖及细胞毒性检测试剂盒实测效果图。
A. HeLa 细胞加入使用本试剂盒配制的MTT 溶液,在细胞培养箱内孵育4小时,显微镜下可见大量结晶状的深紫色产物formazan 生成。
B. 不同数量HeLa 细胞在MTT溶液(MTT solution)加入后4小时的效果图(上图)及深紫色产物formazan 生成后加入Formazan 溶解液(Formazan solvent),充分溶解后的效果图(下图)。
图2. 本试剂盒检测不同数量HeLa 细胞的效果图。
不同的检测条件下,实际读数会因标准品的配制、检测仪器等的不同而存在差异,图中数据仅供参考。
本试剂盒采用了独特的Formazan 溶解液配方,无需去除原有的培养液,可以直接加入Formazan 溶解液溶解formazan 。
从而避免了由于去除培养液时formazan 被部分去除而引起的误差。
本试剂盒本底低,灵敏度高,线性范围宽,使用方便。
碧云天各种细胞增殖和细胞毒性检测试剂盒的比较和选择,请参考/support/cell-proliferation.htm 。
本试剂盒C0009S 包装可以测定500个样品,C0009M 包装可以测定2500个样品。
Toll样受体
• 1988年,Hashimoto等人发现Toll基因编码一种跨膜 蛋白质,并阐明了Toll蛋白的结构。
• 1991年,Gay等人发现,Toll蛋白在结构上与哺乳动 物中一种天然免疫功能分子——白细胞介素受体1 (IL-1)具有同源性:二者的细胞质部分相似。这第 一次提示了人们Toll可能和免疫有关。
• 第二,Toll样受体对获得性免疫应答类型具有调控作 用。多数TLRs活化后可以诱导抗微生物防御系统,产 生IL1β、IL6和TNF以及趋化型细胞因子,从而调节机 体Th1和Th2两种方面的平衡。
第14页,共15页。
谢谢!
第15页,共15页。
第8页,共15页。
Toll样受体 在天然免疫中识别作用
• TLR如同天然免疫的眼睛,监视与识别各种不同的疾病 相关分子模式,是机体抵抗感染性疾病的第一道屏障。
• TLR4不但可识别外源的病原体,还可识别内源性物质及
降解物,识别革兰氏阴性菌脂多糖(LPS),还可识别宿 主坏死细胞释放的热休克蛋白(HSP),体内类肝素硫酸 盐和透明质酸盐降解的多糖部分以及局部的内源性酶 的级联活化反应也可激活TLR4。
• 翌年,Charles Janeway和Ruslan Medzhitov阐明了一种 Toll样受体(后来被命名为TLR4)能够激活与适应性免 疫有关的基因。Bruce A. Beutle随后发现TLR4能够探测 LPS的存在。后来他们又发现,如果使小鼠中的TLR4突 变而丧失功能,小鼠不会对LPS起反应。
• TLR5可以识别鞭毛蛋白,鞭毛蛋白是目前发现的 TLR5的惟一配体。识别具有鞭毛蛋白的L型细菌、 铜绿假单胞菌、枯草芽孢杆菌和鼠伤寒沙门菌等。
TOLL样受体抗体对鼠疟疾病理进程的影响
TOLL样受体抗体对鼠疟疾病理进程的影响摘要:利用TOLL样受体抗体(TLRs)尾静脉注射并封闭对应TLR的方法,观察疟疾病理进程的变化。
结果表明,TLR2、9、11抗体处理后小鼠的存活率在第10天分别为70%、0、0,对照组为50%;TLR2、9、11抗体处理后小鼠的红细胞感染率峰值分别为55%、61%、58%,对照组为52%。
抗体封闭TLR2后,可提高小鼠的存活率;抗体封闭TLR9、11后,小鼠红细胞感染率显著升高而存活率显著降低。
证明TLR是一类有潜力的抗疟药物或治疗辅助药物的靶点。
关键词:疟疾;尾静脉注射;TOLL样受体;抗体Abstract:Thetoll-likereceptors(TLRs)inricewereblockedbytailveininjectionwithcorrespondinganti-TLRantibodiesandthenGIMSAstainingwasusedtoobservethepathologicalcourseofmalaria.TheresultsshowedthataftertreatedwithTLR2,9and11antibody,thesurvivingratesofmiceatday10were70%,0and0,respectivelyandthepeakofparasitemiawere55%,61%,58%,respectively,whichwere50%and52%incontrolgroup,respectively.Itwas indicatedthatbytreatingwithTLR2antibody,thesurvivingrateofinfectedmicecouldbeincreased.AftertreatedwithTLR9and11antibodies,theparasitemiaandmortalityweresignificantlyincreased.Itwas provedthatTLRswerepotentialtargetsofnewanti-malarialdrugs.Keywords:malaria;tailveininjection;TLRs;antibody疟疾是一种在热带和亚热带地区造成大量死亡的恶性传染病,以雌性按蚊(Anopheles)为传播媒介[1]。
toll-样受体-讲座
associated factor (TRAF) family • TRAF6 interacts with IRAK-1 and gets
IRAK and TRAF6
• 4 IRAKs known, IRAK1, IRAK-2, IRAK-M and IRAK-4
• IRAK are serine/threonine kinases • IRAK-4 phosphorylates IRAK-1 • IRAK-M plays an inhibitory role in TLR
➢ TLR4 has a MyD88 independent pathway as well
Cytoplasmic TIR domain
MyD88 Adaptor
➢ MyD88 knockout mice have no response to LPS
➢ MyD88 is essential to all inflammatory signaling pathways
• Poly I:C is an agonist for TLR3 that mimics binding of dsRNA to TLasmic tails of TLRs show similarities to IL-1 receptor (TIR)
• Found in intracellular membranes since viral nucleic acids are endogenously generated
• Note the TIR domains of these TLRs face the cytosol of the cell
toll样受体
Toll样受体Toll样受体(也称作TLR,即Toll-like receptor)是一类重要的受体蛋白,它可以识别和结合一些特定的分子,从而激活免疫系统的应答。
这些分子通常是一种称为模式识别受体(PRRs)的受体所能识别的,它们在宿主防御机制中起着至关重要的作用。
概述Toll样受体是一类跨膜蛋白,属于PRRs家族的一种。
它们通过其外在结构上的高度保守的Leucine-rich repeat(LRR)结构域来识别和结合特定的分子,例如细菌的毒素、细菌的表面成分以及病毒的核酸。
目前已经发现了多种TLR,它们在不同的细胞类型和组织中表达,并参与多种的生理和病理过程。
TLR的结构每个TLR蛋白含有多个LRR结构域,这些结构域通过丰富的β折叠片层连接在一起,形成蛋白的外在结构。
这种结构不仅赋予了TLR以警戒细菌入侵的能力,还可以通过结合配体从而触发下游信号通路的激活。
此外,TLR还包含一个胞浆端的富含精氨酸的TIR(Toll/interleukin-1 receptor)结构域,这个结构域在TLR的信号转导中起着重要的作用。
当TLR与其配体结合后,TIR 结构域会与下游信号分子结合,从而激活信号转导通路。
TLR的功能TLR的主要功能是发挥免疫系统的免疫应答。
当TLR识别到特定的分子(也称为TLR配体)时,它可以通过激活下游信号通路来启动免疫应答。
这种应答包括产生和释放一系列的细胞因子、促发炎因子和抗病毒因子,进一步引发炎症反应和免疫细胞的活化。
此外,TLR还可以调节免疫系统的平衡,以对抗侵袭性病原体的攻击。
通过TLR的激活,免疫系统可以更加快速和有效地识别和清除病原体,从而维护机体的健康。
TLR与疾病由于TLR在免疫应答中的重要作用,TLR功能的异常调节可能会导致多种疾病的发生和发展。
例如,TLR的过度激活可能导致慢性炎症的产生,进而导致一些自身免疫性疾病的发生,如类风湿关节炎和系统性红斑狼疮。
此外,一些病原体也可以通过干扰TLR的功能来逃避宿主的免疫应答。
髓样分化因子88多态性的研究进展
髓样分化因子88多态性的研究进展胡玉懿, 陈 朴 综述, 郭 玮, 潘柏申 审校 (复旦大学附属中山医院检验科,上海 200032)摘要:髓样分化因子88(MYD88)是细胞内传递信号的关键衔接蛋白,可介导多种Toll 样受体(TLR )、白细胞介素1受体(IL-1R )及白细胞介素-18受体(IL-18R )的信号传递,在固有免疫中发挥显著作用。
MYD88依赖通路在多种病原体致病过程中发挥作用,与肿瘤、感染性疾病、自身免疫性疾病等密切相关。
MYD88依赖通路被认为是这些疾病治疗的关键靶点。
MYD 88基因 L265P 突变导致MYD88蛋白Toll-白细胞介素1受体域(TIR )的265位氨基酸发生改变。
该突变可通过加强核因子-κB (NF-κB )等转录因子促进酪氨酸激酶-信号转导子和转录激活子3(JAK-STA T3)信号通路传导,介导白细胞介素(IL )-6、IL-10及β-干扰素(IFN-β)等炎症因子的产生。
文章从MYD88的结构、基本功能、在信号传导通路中的作用以及MYD 88基因L265P 突变与疾病的关联等方面进行综述。
关键词:髓样分化因子88;Toll 样受体;淋巴浆细胞淋巴瘤Research progress of myeloid differentiation factor 88 polymorphism HU Yuyi ,CHEN Pu ,GUO Wei ,P AN Baishen. (Department of Clinical Laboratory ,Zhongshan Hospital ,Fudan University ,Shanghai 200032,China )Abstract :Myeloid differentiation factor 88(MYD88)is an important adaptor protein mediating the signal transduction of Toll-like receptors (TLR ),interleukin 1 receptor (IL-1R )and interleukin 18 receptor (IL-18R ),which plays a key role in innate immunity. MYD88-dependent pathways play roles in the pathogenesis of multiple pathogens and have relations with tumors ,infectious diseases ,autoimmune diseases and so on ,which are considered to be important targets for the intervention of these diseases. The L265P mutant in MYD 88,an amino acid change (L265P )in the MYD88 Toll-like receptor/interleukin 1 receptor (TIR ),promotes Janus kinase-signal transducer and activator of transcription 3(JAK-STAT3) signaling pathway by enhancing transcription factors such as nuclear factor kappa B (NF-κB ),and mediates the production of inflammatory factors such as interleukin (IL )-6,IL-10 and interferon-beta (IFN-β). The structure and fundamental function of MYD88 and its role in signal transduction are reviewed. Furthermore ,the association between MYD 88 L265P and diseases are also illustrated.Key words :Myeloid differentiation factor 88;Toll-like receptor ;Lymphoplasmacytoid lymphoma基金项目:国家自然科学基金资助项目(81572064);国家自然科学基金资助项目(81772263);上海市卫生计生系统重要薄弱学科 建设项目(2015ZB0201)作者简介:胡玉懿,女,1991年生,学士,技师,主要从事临床检验工作。
Interferon-beta Inhibits Toll-Like Receptor 9 Processing inMultiple Sclerosis
INTRODUCTIONMultiple sclerosis (MS) is considered to be an immune-mediated disorder of the centralnervous system 1-2. Although the immunopathogenesis of the disease is not completelyunderstood, both polygenic and environmental factors contribute to disease onset and/orclinical exacerbation 3-5. Viral pathogens have been implicated in the etiology andpathogenesis of MS (reviewed in 6). Among those, strong data implicates Epstein-Barr Virus(EBV), a human DNA virus 7-11. For established MS patients, the risk of diseaseexacerbation was found to be increased at the time or shortly after clinical viral infections 12-14 (reviewed by Rutschmann et al 15). The antiviral protein IFN-gamma, a T helper type 1(Th1)-type cytokine produced mainly by NK and T cells, was found to trigger multiplesclerosis in vivo 16-17.Plasmacytoid dendritic cells (pDCs), characterized as CD11c -CD123++Lin -DR ++ cells,produce large amounts of type I IFN in response to viral infection 18-19. Compared to otherperipheral blood mononuclear cells, pDCs express a high level of Toll-like receptor 9(TLR9) 20 which recognizes viral DNA within the early endosomes at the initial phase ofviral infection. It has been recently discovered that the full-length TLR9 has to be cleavedfrom the N-terminal to generate a functional (processed) TLR9 C-terminal 21-22. Activatedvia TLR9, pDCs can secrete 100-1,000-fold higher levels of interferon (IFN)-alpha, than anyother blood cell type 23.PDCs link innate and adaptive immunity via a number of cytokines implicated in thepathogenesis of demeylination 24. IFN-Type I cytokines induce intracellular signaling inlymphocytes via a transcription factor STAT4 leading to Th1 cell differentiation (reviewedin 25). IL-6 promotes myelin antigen-specific Th17- and Th1-responses in experimentalautoimmune encephalomyelitis (EAE) 26. TNF-alpha directly induces oligodendrocyteapoptosis 27 and mediates human neuronal injury after activation with TLR9 agonists 28.Activation of antigen-presenting cells through TLR9 can overcome tolerance and precipitateEAE 29-31. The generation of Th17 cells is decreased in pDC-depleted mice and isassociated with less severe clinical and histopathological signs of EAE 32. PDCs are foundin the CSF of MS patients 33-36 and accumulate in MS lesions 8,37. Thus, pDCs may serveas a strong link between viral infection and MS exacerbation.We hypothesize that pDCs may trigger MS exacerbation in response to viral pathogens butare inhibited by disease-modifying therapy such as IFN-beta, consequently decreasing thefrequency of MS attacks. Here we describe a new immunodulatory effect of IFN-beta in MSinvolving the inhibition of TLR9 processing.MATERIALS AND METHODS1. Subjects Patients and healthy donors, 18-60 years old, were enrolled in the study. Patients werediagnosed with clinically definite relapsing-remitting MS (RR MS) or clinically isolatedsyndrome (CIS) as described 38, and not taking any immuno-modulatory drugs other thanIFN-beta based treatment. The typical clinical presentations of patients with CIS wereunilateral optic neuritis, hemiparesis or unilateral sensory deficit (confirmed by asymptomatic spinal cord lesion on MRI). Patients with secondary progressive MS andprimary progressive MS, patients with EDSS score 6 or higher, or patients who received IVsteroids or any other non-IFN-beta immunomodulatory drugs less than 2 months prior toblood drawing were excluded. The patients at the time of clinical attack of MS wereexcluded. The patients were treated with IFN-beta 1a or IFN-beta 1b in doses approved byFDA and recommended by the drug manufacturers. The patients and healthy donors wereNIH-PA Author Manuscript NIH-PA Author ManuscriptNIH-PA Author Manuscriptenrolled at two MS Centers. The patients and healthy subjects presented in Figures 1 anddescribed in Table 1 were enrolled at the Partners MS Center, Boston, MA. The patients andhealthy subjects presented in other figures and described in Table 2 and appropriate figurelegends were enrolled at the RWJ Center for MS, New Brunswick, NJ. The participation ofpatients and healthy subjects in the study was approved by institutional review boards.Informed consent was obtained from all subjects.2. Blood samples and pDC separationPeripheral blood mononuclear cells (PBMC) were isolated from 80-100 ml of heparinizedblood samples within 4 hours after venipuncture as described 39. Fresh pDCs were separatedfrom PBMC by immunomagnetic sorting and BDCA-4 cell isolation kit (Miltenyi Biotec)with 2 steps of pDC enrichment on magnetic columns. Such sample yields approximately200,000-250,000 pDCs. More than 90% of separated cells were CD11c -CD123+DR ++ asmeasured by flow cytometry. The separated cells had approximately 100 fold higher level ofTLR9 gene expression compared to PBMC (Table 3). The separated cells were immediatelyused for cell culture experiments, suspended in the RNAlater stabilization reagent (Qiagen,Santa Clarita, CA) for gene expression analysis or frozen at -80°C for Western blotexperiments.3. Cytokine secretion by pDCs Freshly isolated pDC (100,000 cells/ml) were cultured in round-bottomed 96 well plates in a total volume of 0.2 ml/well. Parallel cultures of pDCs were treated with the TLR9 agonist CpG ODN class A (ODN#2336, Coley Pharmaceutical), or control ODN (#2243, Coley Pharmaceutical) at 5 mcg/ml (0.735 mcMol). Cell supernatants were collected after 16 hours and tested by multi-subtype ELISA kit (Cat # 41105-2, PBL Biomedical Laboratories) to measure the total IFN-alpha secretion. The kit is able to recognize 13 different IFN-alpha species. In addition, detection of selected cytokines (IFN α2, TNF α, and IL-6) was providedby the BioMarker Services (Millipore Corporation, St. Charles, MO) using MILLIPLEXMulti-Analyte Profiling with Luminex xMAP Multiplexing Technology(/drugdiscovery/dd3/map_portfolio) with a standard curve rangefrom 3.2 to 10,000 pg/ml.4. RT-qPCRTotal RNA was prepared from 1.0 × 105 cell sample using the RNeasy Mini Kit (Qiagen,Santa Clarita, CA) according to the manufacturer’s protocol 40 and processed for cDNAsynthesis using the TaqMan Reverse Transcription Kit (Cat No N808-0234, AppliedBiosystems, Foster City, CA). For TaqMan RT-PCR, Master Mix Kit (Cat No 4304437) andTaqMan® Gene Expression Assays (the set of primers and probes) for human genes such asGAPDH, IFN-alpha1, IRF7, TLR9, were purchased from Applied Biosystems. RT-qPCRwas performed according to the manufacturer’s protocol using iCycler iQ Real-Timedetection System (Bio-Rad Laboratories, Hercules, CA). The human GAPDH, housekeepinggene, was used to normalize each sample and each gene.5. Affymetrix microarray analysis of TLR9 gene expressionTotal RNA was prepared from 1.0 × 105 pDC sample using Trizol reagent (Invitrogen,USA) and further purified with Qiagen RNAeasy columns with DNase treatment (Qiagen,Valencia, CA). RNA quality was assessed by capillary electrophoresis using the AgilentBioanalyzer 2100 and spectrophotometric analysis prior to cDNA synthesis. Fiftynanograms of total RNA from each sample were amplified to cDNA, fragmentated andbiotinylated using the Nugen kits WT Ovation Pico (NuGen, San Carlos, CA) and FLOvation Biotin (NuGen). The labeled samples were hybridized to Affymetrix GeneChip®NIH-PA Author ManuscriptNIH-PA Author ManuscriptNIH-PA Author ManuscriptHuman Genome U133A 2.0 Arrays according to manufacturer’s recommendations forhybridization, washing and scanning. Data analysis was done for each pair of patient byGeneSpring software to get the fold change of relative gene expression.6. Western Blot100,000 cells were sonicated in Nupage LDS Sample buffer (4X) (Invitrogen) with 50mMDTT and heated at 70°C for 10 minutes. Cell lysates were loaded on Nupage 4-12% Bis-Trisgels (Invitrogen), electrophoresed and then transferred to PVDF membrane (Biorad) byelectroblotting. The membranes were blocked in Tris/Tween-20 buffer with 5% Non-fat drymilk (Biorad) for 1hr at room temperature and then probed with 1.5 mcg/ml of rabbitpolyclonal anti-human TLR9 antibodies (Ab) (Santa Cruz Biothechnology) for overnight at4°C to detect the C–terminal of TLR9 (processed TLR9). Membranes were washed threetimes with 50mM Tris/Tween-20 buffer (pH7.4) with 150mM NaCl and 0.1% Tween-20and probed with anti-rabbit IgG-HRP (1:25,000 final dilution, Sigma-Aldrich) for one hourat room temperature. The Pierce ECL Western Blotting Substrate was used to detect proteinsby chemiluminescence. The intensities of protein bands were measured with Image Jsoftware (NIH). Membranes were stripped with Restore Western Blot Stripping Buffer(Pierce) and re-probed with 2 mcg/ml of mouse monoclonal anti-human TLR9 Ab(Imgenex) to detect the full length TLR9 followed by anti-mouse IgG-HRP (1:1,000 finaldilution, Sigma-Aldrich). Membranes were stripped again and re-probed with mousemonoclonal anti-Beta-actin Ab (1:5,000 final dilution, Sigma-Aldrich) followed by anti-mouse IgG-HRP (1:10,000 final dilution, Sigma-Aldrich) for protein normalization.6. Statistical analysisWithin patient change from baseline (Figure 2), laboratory measures were tested by a pairedt -test. The difference between groups (Figures 1 and 4) was tested by unpaired t -test.RESULTS1. Cytokine production by activated pDCs in RR MSa) Increased level of IFN-alpha production in RR MS patients is inhibited inIFN-beta treated patients—PDCs are the major source of the Th1-promoting cytokineIFN-alpha. We compared IFN-alpha production by activated pDCs in healthy donors (HD),untreated and treated RR MS patients. As shown in Figure 1, IFN-alpha production wassignificantly elevated in untreated RR MS patients as compared to healthy subjects, p =0.0164. RR MS patients treated with IFN-beta based medications had significantlydecreased IFN-alpha production as compared to untreated RR MS patients, p = 0.027. Therewas no statistically significant difference between healthy subjects and IFN-beta treated MSpatients, p > 0.05.b) IFN-beta inhibits TLR9-dependent intracellular signaling pathways—Twomajor TLR9-dependent intracellular signaling pathways were described. Both pathwaysshare a key adaptor MyD88 (myeloid differentiation primary-responsible gene 88). The firstpathway leads to production of IFN-type I cytokines and requires phosphorylation andtranslocation of IRF7 to the nucleus promoting IFN-alpha and IFN-beta transcription. Incontrast, the gene expression of pro-inflammatory cytokines, such as TNF-alpha and IL-6, isdependent on activation of NF-κB and ATF2-c-Jun 19 41. Therefore we studied IL-6, TNF-alpha and IFN-alpha production in the same cultures of pDCs activated with TLR9 agonists(Figure 2). In addition, to avoid genetic heterogeneity between tested groups and tominimize the possible effect of IFN-beta neutralizing antibodies which usually do not appearuntil 6 months after IFN-beta is initiated 42, each MS patient was tested prior to and after a3month course of IFN-beta treatment. The patients had no prior history of being treated withNIH-PA Author Manuscript NIH-PA Author ManuscriptNIH-PA Author ManuscriptIFN-beta-based drugs. As shown on Figure 2, compared to pre-treatment, the averagecytokine production was significantly reduced by 49% (IFN-alpha2), 55% (IL-6) and 63%(TNF-alpha) in IFN-beta treated patients. Among fourteen patients tested, we could identifythree groups of patients based on their response to IFN-beta. The first group (ten patients)had all cytokines decreased after treatment with IFN-beta. The second group (two patients)had increased IFN-alpha2 secretion and decreased IL-6 and TNF-alpha secretion. The thirdgroup (two patients) had both IFN-alpha and IL-6 increased while TNF-alpha secretion wasdecreased. In part, it could be linked to different intracellular signaling mechanismsinvolved in regulation of IFN-type I cytokines versus other pro-inflammatory cytokines,such as TNF-alpha and IL-6 as described above.Although disease modifying treatment was offered to all patients diagnosed with MS or CIS,two patients declined treatment after being diagnosed with CIS. They agreed to provide 2blood samples 3 months apart. The mean cytokine production for those patients at twodifferent time points was 2873±1707 and 3220±1045 pg/ml (IFN-alpha2), 150±93 and172.5±74.5 pg/ml (IL-6), 418±212 and 380±87 pg/ml (TNF-alpha). In summary, the resultspresented in Figures 1 and 2 strongly suggest that IFN-beta treatment inhibits TLR9-mediated intracellular signaling pathways in activated pDCs from MS patients.2. IFN-beta does not affect TLR9 gene expressionTLR9, a unique receptor able to recognize DNA sequences typical of DNA viruses, isexpressed mostly by pDCs and B cells in human peripheral blood 43. We have comparedTLR9, IRF7 and IFN-alpha gene expression in PBMC and pDCs by QRT-PCR. Asexpected, expression of TLR9 was approximately 100 fold elevated in pDCs compared toPBMC (Table 3). Please note that TLR9 is expressed in the population of PBMC depletedfrom pDCs due to the presence of TLR9-positive B cells. IRF7 is constitutively expressed inpDCs at high levels 44-45.Next we studied if IFN-beta treatment affects TLR9 gene expression in MS patients. TLR9gene expression was studied by oligo microarrays in paired pDC samples separated from 8MS patients before and after a 3 month course of treatment with recombinant human IFN-beta-1b (subcutaneous, n=4) or recombinant human IFN-beta-1a (intramuscular, n=4). Theaverage age of patients was 37 ± 12 years old (Mean ±SD), 6 females and 2 males. Based onpaired t test, there was no statistically significant effect of IFN-beta treatment on TLR9 geneexpression. The fold change of relative TLR9 gene expression after treatment was 1.205 ±0.286 (Mean ±SD), p = 0.082. The analysis of the same oligo microarrays before and aftertreatment with the imposed minimal threshold of 1.5 fold change from the baseline and the pvalue of 0.05 or less did not show a significant effect of IFN-beta treatment on geneexpression for the following molecules involved in TLR9 intracellular pathway as depictedin Kegg Toll-like receptor signaling pathway(http://www.genome.jp/kegg-bin/show_pathway?hsa04620): MyD88, IRAK-1, IRAK-4,TRAF6, IRF5, IRF7, CHUK (IKK α), CHUK (IKK α), CHUK (IKK α), NFKB1 (NFkB),NFKBIA(IkB α) (data not shown).3. IFN-beta treatment inhibits TLR9 processing in MS patientsIt has been recently discovered that a full length TLR9 (100 to 150 kDa depending on thelevel of protein glycosylation) is in fact non-functional and needs to be cleaved to generate afunctional C-terminal TLR9 with a molecular weight between 65 to 80 kDa depending onthe source of TLR9-positive cells 21-22. Therefore, to study the processing of TLR9, weapplied Western Blot analysis and probing with the C terminal-specific Abs. We were ableto detect a 65 kDa band corresponding to the processed C-terminal TLR9 in all pDCsamples (Figure 3). As expected, the processed TLR9 was expressed mostly in pDCs. The NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscriptexpression of the processed TLR9 in B cells was approximately 20-50 fold less as comparedto pDCs and was extremely low in the total population of PBMC (Figure 3). We also provedthe specific interaction between a 65 kDa protein and CpG oligonucleotide (TLR9 agonist)by co-culture of biotinylated CpG oligonucleotides with pDCs followed by proteinimmunoprecipitation with Streptavidin Dynabeads followed by Western Blot of theprecipitate with C-terminal anti-TLR9 Abs (Supplementary Figure 1). We then analyzedTLR9 processing in pDCs isolated from healthy subjects and MS patients. As shown inFigure 4A, there was no significant difference in the level of full-length unprocessed TLR9between treated (n = 14) and untreated (n = 12) patients. However, IFN beta-treated CIS/RRMS patients had significantly decreased level of the processed (functionally active) TLR9 C-terminal as compared to untreated patients, p = 0.0005. Therefore, it was not the full-lengthTLR9 protein synthesis but the processing of the TLR9 protein which was significantlyinhibited in IFN-beta treated patients. TLR9 processing was also analyzed separately inpatients with CIS and patients with clinically definite RR MS. Among patients with RR MS,TLR9 processing was also significantly decreased (p = 0.003) in IFN-beta treated patients (n= 7) as compared to untreated patients (n=9) but did not reach statistical significance for CISpatients (n = 5 in each group), which could be due to the small number of subjects tested.DISCUSSION Although many IFN-alpha species and IFN-beta bind to the same Type I IFN receptor, it is not quite clear why there is so much variation in their biological activity 46.Our initial results (Figure 1) suggested that activated pDCs produce increased level of IFN-alpha, a Th1 promoting cytokine, in non-treated MS patients as compared to healthy subjects. In addition, IFN-alpha secretion was significantly decreased in MS patients treated with recombinant IFN-beta as compared to non-treated patients. Several studies examined the function of pDCs in MS 35,36. IFN-beta treatment did not affect the concentration ofpDCs in peripheral blood from MS patients 35,37, but inhibited expression of MHC class Iand BDCA-2 and upregulated CD38, B7H1 37 and CD123 35 on pDCs. TLR9 ligand-induced IFN-alpha production by unfractionated peripheral blood mononuclear cells wasdecreased in MS patients but was not studied in enriched pDCs 36. Recently, Bayas et al.studied TLR9-agonist activated IFN-alpha secretion by pDCs in MS. Bayas et al. usedmethods for pDC separation, culture condition and IFN-alpha detection different from ourprotocol. The difference between MS patients and healthy subjects was not statisticallysignificant for the strong IFN-alpha inducer (CpG Type A oligo which is similar to TLR9agonist used in our study) 47. IFN-beta treatment inhibited IFN-alpha production by pDCsseparated from healthy subjects but was not studied in MS patients 37.In our experiments, IFN-beta treated patients had decreased production of IFN-alpha, IL-6and TNF-alpha by activated pDCs (Figure 2). Those cytokines are regulated by two differentTLR9-dependent pathways 19,41. Therefore, it led us to hypothesize that IFN-beta maymodulate TLR9 expression.However, IFN-beta did not affect TLR9 gene expression or the level of unprocessed TLR9protein (Figure 4a). Therefore, we hypothesized that IFN-beta treatment may inhibit theeffect of viral agonists on pDCs by reducing the processing of TLR9. Ex-vivo experimentswith pDCs separated from CIS/MS patients confirmed this hypothesis (Figure 4b).TLR9 processing has been recently discovered in two independent laboratories 21-22. To ourknowledge, TLR9 processing has not been studied in human diseases. The exact mechanismof modulation of TLR9 processing is not clear at this time and may be influenced by theeffect of IFN-beta on other immunomodulatory proteins. Protease inhibitors were shown toNIH-PA Author ManuscriptNIH-PA Author ManuscriptNIH-PA Author Manuscriptprevent TLR9 processing; however, proteases responsible for cleavage of TLR9 have notbeen definitively identified 21-22. Matsumoto et al reported that cathepsins B and L inhibitorblock TLR9 responses 48. Interestingly, recombinant human IFN-beta was also shown tosuppress the cathepsin B activity in a dose-dependent manner 49. However, Ewald et al.suggested that in addition to cathepsins, other proteases are capable to cleave TLR9 asselective cathepsin inhibitors could not abolishTLR9 processing completely 21-22.The finding of decreased TLR9 processing in IFN-beta treated MS patients establishes anew mechanism of disease-modifying treatment in MS. TLR9 is the only member of thefamily of TLRs able to sense DNA viruses implicated in the pathogenesis of MS. Ifconfirmed by others and linked to clinical disease activity, modulation of TLR9 processingmay prove to be an important target for a new generation of MS immunomodulatory drugssuch as TLR antagonists.Supplementary Material Refer to Web version on PubMed Central for supplementary material.Acknowledgments K.E.B. is supported by grant number K23NS052553 from the National Institute of Neurological Disorders and Stroke and grants from the National Multiple Sclerosis Society and Bayer Healthcare. We thank Joan Moore and Sheila Reaves for assistance in preparing this manuscript.References 1. McFarland HF, Martin R. Multiple sclerosis: a complicated picture of autoimmunity. Nat Immunol.2007; 8(9):913–9. [PubMed: 17712344]2. Sospedra M, Martin R. Immunology of multiple sclerosis. Annu Rev Immunol. 2005; 23:683–747.[PubMed: 15771584]3. Hafler DA, Compston A, Sawcer S, Lander ES, Daly MJ, De Jager PL, et al. Risk alleles formultiple sclerosis identified by a genomewide study. N Engl J Med. 2007; 357(9):851–62.[PubMed: 17660530]4. Noseworthy J, Lucchinetti C, Rodriguez M, Weinshenker B. Multiple Sclerosis. New EnglandJournal of Medicine. 2000; 343(13):938–52. [PubMed: 11006371]5. Compston A, Coles A. Multiple Sclerosis. Lancet. 2002; 359:1221–31. [PubMed: 11955556]6. Cook, S.; Rohowsky-Kochan, C.; Bansil, S.; Dowling, P. Evidence for a viral etiology of multiplesclerosis. In: Cook, S., editor. Handbook of multiple sclerosis. 2. New York: Marcel Dekker, Inc;1996. p. 97-118.7. Haahr S, Hollsberg P. Multiple sclerosis is linked to Epstein-Barr virus infection. Rev Med Virol.2006; 16(5):297–310. [PubMed: 16927411]8. Serafini B, Rosicarelli B, Franciotta D, Magliozzi R, Reynolds R, Cinque P, et al. DysregulatedEpstein-Barr virus infection in the multiple sclerosis brain. J Exp Med. 2007; 204(12):2899–912.[PubMed: 17984305]9. Giovannoni G, Cutter GR, Lunemann J, Martin R, Munz C, Sriram S, et al. Infectious causes ofmultiple sclerosis. Lancet Neurol. 2006; 5(10):887–94. [PubMed: 16987736]10. Levin LI, Munger KL, Rubertone MV, Peck CA, Lennette ET, Spiegelman D, et al. Multiplesclerosis and Epstein-Barr virus. Jama. 2003; 289(12):1533–6. [PubMed: 12672770]11. Ascherio A, Munger K. Epidemiology of multiple sclerosis: from risk factors to prevention. SeminNeurol. 2008; 28(1):17–28. [PubMed: 18256984]12. Edwards S, Zvartu M, Clarke H, Irving W, Blumhardt L. Clinical relapses and disease activity onmagnetic resonance imaging associated with viral upper respiratory tract infections in multiplesclerosis. Journal of Neurology, Neurosurgery and Psychiatry. 1998; 64:736–41.NIH-PA Author ManuscriptNIH-PA Author ManuscriptNIH-PA Author Manuscript13. Sibley W, Bamford C, Clark L. Clinical viral infections and multiple sclerosis. Lancet. 1985;1:1313–15. [PubMed: 2860501]14. Panitch HS. Influence of infection on exacerbations of multiple sclerosis. Ann Neurol. 1994;36(Suppl):S25–8. [PubMed: 8017885]15. Rutschmann OT, McCrory DC, Matchar DB. Immunization and MS: a summary of published evidence and recommendations. Neurology. 2002; 59(12):1837–43. [PubMed: 12499473]16. Panitch HS, Hirsch RL, Schindler J, Johnson KP. Treatment of multiple sclerosis with gamma interferon: exacerbations associated with activation of the immune system. Neurology. 1987;7:1097–102. [PubMed: 3110648]17. Panitch HS, Hirsch RL, Haley AS, Johnson KP. Exacerbations of multiple sclerosis in patients treated with gamma interferon. Lancet. 1987; 1(8538):893–5. [PubMed: 2882294]18. Colonna M, Trinchieri G, Liu Y. Plasmacytoid dendritic cells in immunity. Nature Immunology.2004; 5(12):1219–26. [PubMed: 15549123]19. Gilliet M, Cao W, Liu YJ. Plasmacytoid dendritic cells: sensing nucleic acids in viral infection and autoimmune diseases. Nat Rev Immunol. 2008; 8(8):594–606. [PubMed: 18641647]20. Liu YJ. IPC: professional type 1 interferon-producing cells and plasmacytoid dendritic cell precursors. Annu Rev Immunol. 2005; 23:275–306. [PubMed: 15771572]21. Ewald SE, Lee BL, Lau L, Wickliffe KE, Shi GP, Chapman HA, et al. The ectodomain of Toll-like receptor 9 is cleaved to generate a functional receptor. Nature. 2008; 456(7222):658–62. [PubMed:18820679]22. Park B, Brinkmann MM, Spooner E, Lee CC, Kim YM, Ploegh HL. Proteolytic cleavage in an endolysosomal compartment is required for activation of Toll-like receptor 9. Nat Immunol. 2008;9(12):1407–14. [PubMed: 18931679]23. Siegal F, Kadowaki N, Shodell M, Fitzgerald-Bocarsly P, Shah K, Ho S, et al. The nature of the principal type 1 interferon-producing cells in human blood. Science. 1999; 284:1835–37.[PubMed: 10364556]24. McKenna K, Beignon AS, Bhardwaj N. Plasmacytoid dendritic cells: linking innate and adaptive immunity. J Virol. 2005; 79(1):17–27. [PubMed: 15596797]25. Korman BD, Kastner DL, Gregersen PK, Remmers EF. STAT4: genetics, mechanisms, andimplications for autoimmunity. Curr Allergy Asthma Rep. 2008; 8(5):398–403. [PubMed:18682104]26. Serada S, Fujimoto M, Mihara M, Koike N, Ohsugi Y, Nomura S, et al. IL-6 blockade inhibits theinduction of myelin antigen-specific Th17 cells and Th1 cells in experimental autoimmuneencephalomyelitis. Proc Natl Acad Sci U S A. 2008; 105(26):9041–6. [PubMed: 18577591]27. Akassoglou K, Bauer J, Kassiotis G, Pasparakis M, Lassmann H, Kollias G, et al. Oligodendrocyteapoptosis and primary demyelination induced by local TNF/p55TNF receptor signaling in the central nervous system of transgenic mice: models for multiple sclerosis with primaryoligodendrogliopathy. Am J Pathol. 1998; 153(3):801–13. [PubMed: 9736029]28. Iliev AI, Stringaris AK, Nau R, Neumann H. Neuronal injury mediated via stimulation ofmicroglial toll-like receptor-9 (TLR9). Faseb J. 2004; 18(2):412–4. [PubMed: 14688201]29. Segal BM, Chang JT, Shevach EM. CpG oligonucleotides are potent adjuvants for the activation ofautoreactive encephalitogenic T cells in vivo. J Immunol. 2000; 164(11):5683–8. [PubMed:10820244]30. Ichikawa HT, Williams LP, Segal BM. Activation of APCs through CD40 or Toll-like receptor 9overcomes tolerance and precipitates autoimmune disease. J Immunol. 2002; 169(5):2781–7.[PubMed: 12193753]31. Waldner H, Collins M, Kuchroo VK. Activation of antigen-presenting cells by microbial productsbreaks self tolerance and induces autoimmune disease. Journal of Clinical Investigation. 2004;113(7):990–97. [PubMed: 15057305]32. Isaksson M, Ardesjo B, Ronnblom L, Kampe O, Lassmann H, Eloranta ML, et al. PlasmacytoidDC promote priming of autoimmune Th17 cells and EAE. Eur J Immunol. 2009; 39(10):2925–35.[PubMed: 19637225]33. Pashenkov M, Huang Y, Kostulas V, Haglund M, Soderstrom M, Link H. Two subsets of dendriticcells are present in human cerebrospinal fluid. Brain. 2001; 124:480–92. [PubMed: 11222448]NIH-PA Author ManuscriptNIH-PA Author ManuscriptNIH-PA Author Manuscript34. Pashenkov M, Teleshova N, Kouwenhoven M, Kostulas V, Huang Y, Soderstrom M, et al.Elevated expression of CCR5 by myeloid (CD11c+) blood dendritic cells in multiple sclerosis and acute optic neuritis. Clinical and Experimental Immunology. 2002; 127:519–26. [PubMed:11966770]35. Lopez C, Comabella M, Al-zayat H, Tintore M, Montalban X. Altered maturation of circulating dendritic cells in primary progressive MS patients. J Neuroimmunol. 2006; 175(1-2):183–91.[PubMed: 16698091]36. Stasiolek M, Bayas A, Kruse N, Wieczarkowiecz A, Toyka KV, Gold R, et al. Impaired maturation and altered regulatory function of plasmacytoid dendritic cells in multiple sclerosis. Brain. 2006;129(Pt 5):1293–305. [PubMed: 16513684]37. Lande R, Gafa V, Serafini B, Giacomini E, Visconti A, Remoli ME, et al. Plasmacytoid dendritic cells in multiple sclerosis: intracerebral recruitment and impaired maturation in response to interferon-beta. J Neuropathol Exp Neurol. 2008; 67(5):388–401. [PubMed: 18431257]38. Jacobs LD, Beck RW, Simon JH, Kinkel RP, Brownscheidle CM, Murray TJ, et al. Intramuscular interferon beta-1a therapy initiated during a first demyelinating event in multiple sclerosis.CHAMPS Study Group. N Engl J Med. 2000; 343(13):898–904. [PubMed: 11006365]39. Balashov KE, Rottman JB, Weiner HL, Hancock WW. CCR5(+) and CXCR3(+) T cells are increased in multiple sclerosis and their ligands MIP-1alpha and IP-10 are expressed in demyelinating brain lesions. Proc Natl Acad Sci U S A. 1999; 96(12):6873–8. [PubMed:10359806]40. Wandinger K, Sturzebecher C, Bielekova B, Detore G, Rosenwald A, Staudt L, et al. Complex immunomodulatory effects of interferon-beta in multiple sclerosis include the upregulation of T helper 1-associated marker genes. Annals of Neurology. 2001; 50:349–57. [PubMed: 11558791]41. Kawai T, Akira S. Innate immune recognition of viral infection. Nat Immunol. 2006; 7(2):131–7.[PubMed: 16424890]42. Giovannoni G, Goodman A. Neutralizing anti-IFN-beta antibodies: how much more evidence do we need to use them in practice? Neurology. 2005; 65(1):6–8. [PubMed: 16009876]43. Ito T, Wang YH, Liu YJ. Plasmacytoid dendritic cell precursors/type I interferon-producing cells sense viral infection by Toll-like receptor (TLR) 7 and TLR9. Springer Semin Immunopathol.2005; 26(3):221–9. [PubMed: 15592841]44. Izaguirre A, Barnes BJ, Amrute S, Yeow WS, Megjugorac N, Dai J, et al. Comparative analysis ofIRF and IFN-alpha expression in human plasmacytoid and monocyte-derived dendritic cells. J Leukoc Biol. 2003; 74(6):1125–38. [PubMed: 12960254]45. Dai J, Megjugorac NJ, Amrute SB, Fitzgerald-Bocarsly P. Regulation of IFN regulatory factor-7and IFN-alpha production by enveloped virus and lipopolysaccharide in human plasmacytoid dendritic cells. J Immunol. 2004; 173(3):1535–48. [PubMed: 15265881]46. Ortaldo JR, Herberman RB, Harvey C, Osheroff P, Pan YC, Kelder B, et al. A species of humanalpha interferon that lacks the ability to boost human natural killer activity. Proc Natl Acad Sci U S A. 1984; 81(15):4926–9. [PubMed: 6589637]47. Bayas A, Stasiolek M, Kruse N, Toyka KV, Selmaj K, Gold R. Altered innate immune response ofplasmacytoid dendritic cells in multiple sclerosis. Clin Exp Immunol. 2009; 157(3):332–42.[PubMed: 19664140]48. Matsumoto F, Saitoh S, Fukui R, Kobayashi T, Tanimura N, Konno K, et al. Cathepsins arerequired for Toll-like receptor 9 responses. Biochem Biophys Res Commun. 2008; 367(3):693–9.[PubMed: 18166152]49. Rollag H, Morland B. The effect of recombinant interferons on cathepsin B activity in humanmonocytes. Scand J Rheumatol Suppl. 1988; 76:79–83. [PubMed: 2472002]NIH-PA Author ManuscriptNIH-PA Author ManuscriptNIH-PA Author Manuscript。
toll-like receptor signaling pway
toll-like receptor signaling pwaytoll-like receptor signaling pathway是一种重要的免疫系统信号传导途径,它在宿主抵御感染和炎症调节中起着关键作用。
本文将逐步介绍toll-like receptor(TLR)信号传导途径,并探讨其在免疫应答中的意义。
第一部分:导言(200-300字)免疫系统是人体的防御机制,负责识别和清除有害的病原体。
免疫应答依赖于免疫细胞和分子之间的复杂通信网络,在这个网络中,TLR信号传导途径具有重要的调节作用。
本文将详细介绍TLR的结构、功能及与其他免疫细胞之间的相互作用。
第二部分:TLR的结构与功能(400-600字)TLR是一类跨膜受体,主要分布在免疫细胞的细胞膜上。
人类有10种TLR (TLR1到TLR10),它们可以识别和结合不同的微生物分子,如细菌的表面成分和病毒的核酸。
TLR1、TLR2、TLR4、TLR5和TLR6主要识别细菌感染,而TLR3、TLR7、TLR8和TLR9则参与病毒感染的识别。
当TLR与它们特异性的配体结合时,它们会激活干扰素调节因子(IRFs)和核因子κB(NF-κB)等信号传导分子。
这些信号分子起到了信号放大和免疫应答程度的调节作用。
在TLR信号传导途径中,TLR与适配蛋白MyD88、TIRAP、TRIF和TRAM等结合,形成一个复杂的信号转导网络。
第三部分:TLR信号传导途径的步骤(800-1000字)在TLR信号传导途径中,首先是配体结合TLR受体并引起共聚集。
这个过程激活了TLR的胞浆末端,使其与适配蛋白结合。
适配蛋白的结合激活了下游信号传导途径,促使IRFs和NF-κB等信号分子入核从而调节基因表达。
TLR2和TLR4通常通过MyD88适配蛋白信号传导。
MyD88与TLR结合后,物理上与转录因子IRFs和NF-κB交叉结合,激活它们并促进核入。
另一适配蛋白TRIF是TLR3和TLR4信号传导的重要介质。
二丙酸倍氯米松和糖皮质激素类药物灌肠治疗溃疡性结肠炎的疗效对比分析
二丙酸倍氯米松和糖皮质激素类药物灌肠治疗溃疡性结肠炎的疗效对比分析董治华;隋典朋;王娜【摘要】目的:比较二丙酸倍氯米松(BDP)和糖皮质激素类药物灌肠治疗溃疡性结肠炎(UC)的疗效及其影响因素。
方法选取以2010年3月至2013年6月于该院消化内科门诊或者住院就诊的80例 U C患者为研究对象,按照治疗方式的不同以随机数字表法分为传统组和BD P治疗组,每组各40例,对比两组患者的临床疗效,采取多因素Logistic分析影响治疗疗效的因素。
结果传统组患者治疗后显效12例,有效16例,总体有效率为70.00%(28/40),而BD P治疗组患者的总体治疗有效率为90.00%(36/40),明显高于传统组( P<0.05);多因素Logistic回归结果显示,C反应蛋白(OR=3.81,P=0.002),血小板(OR=2.01,P=0.031),药物不良反应(OR=2.34,P=0.017)和重型(OR=2.54,P=0.014)是影响传统组治疗疗效的风险因素;而BDP治疗组治疗效果的风险因素分别是重型(OR=3.11,P=0.025)和C反应蛋白(OR=2.86,P=0.004)。
结论 BDP对 UC的疗效明显优于传统糖皮质激素类药物,影响其疗效的因素主要是疾病的严重程度和C反应蛋白水平。
%Objective To compare the effect of enema of beclomethasone dipropionat(BDP) and glucocorticoid drugs in treating ulcerative colitis(UC) and its influencing factors .Methods 80 cases of UC in our hospital from March 2010 to June 2013 were selected as the research subjects ,and randomly divided into the conventional therapy group and the BDP treatment group according to the different treatment modes ,40 cases in each group .The clinical curative effects were compared between the twogroups and the influencing factors were analyzed by Logistic multiva‐riate analysis .Results In the conventional therapy group after treatment ,12 cases were significantly effective and 16 cases were effective with the total effective rate of 70 .00% (28/40) ,while the total effective rate in the BDP treat‐ment group was 90 .00% (36/40) ,which was significantly higher than that in the conventional therapy group (P<0 .05);the multivariate Logistic regression analysis results showed that ,C‐reactive protein(OR=3 .81 ,P=0 .002) , platelet (OR=2 .01 ,P=0 .031) ,adverse drug reactions(OR= 2 .34 ,P=0 .017) and severe type (OR= 2 .54 ,P=0 .014) were the risk factors affecting the therapeutic effect in the conventional therapy group;severe type (OR=3 .11 ,P=0 .025) and C‐reactiveprotein(OR=2 .86 ,P=0 .004) were the risk factors in the BDP treatment group . Conclusion The effect of BDP in treating UC is obviously superior to the traditional glucocorticoid drugs ,and the main factors affecting effect are the severity of disease and C‐reaction protein level .【期刊名称】《检验医学与临床》【年(卷),期】2015(000)017【总页数】4页(P2494-2496,2499)【关键词】二丙酸倍氯米松;糖皮质激素;溃疡性结肠炎;影响因素【作者】董治华;隋典朋;王娜【作者单位】重庆市綦江区人民医院消化内科 401420;重庆市綦江区人民医院科教部 401420;重庆市綦江区人民医院科教部 401420【正文语种】中文溃疡性结肠炎(UC)是一种特发性、反复发作的慢性肠道炎性反应疾病,临床上多表现为血性腹泻、恶性呕吐等消化道症状[1]。
- 1、下载文档前请自行甄别文档内容的完整性,平台不提供额外的编辑、内容补充、找答案等附加服务。
- 2、"仅部分预览"的文档,不可在线预览部分如存在完整性等问题,可反馈申请退款(可完整预览的文档不适用该条件!)。
- 3、如文档侵犯您的权益,请联系客服反馈,我们会尽快为您处理(人工客服工作时间:9:00-18:30)。
O riginal Paper I nt Arch Allergy Immunol 2010;151:285–296 D OI :10.1159/000250437 Toll-Like Receptor-9 Agonist Inhibits Airway Inflammation, Remodeling and Hyperreactivity in Mice Exposed to Chronic Environmental Tobacco Smoke and AllergenD ae Jin Song a, b Myung Goo Min a Marina Miller a Jae Youn Cho a Hye Yung Yum a, c David H. Broide aaD epartment of Medicine, University of California San Diego, S an Diego, Calif. , USA; b D epartment of Pediatrics,College of Medicine, Korea University, and cA topy Clinic,Seoul Medical Center,S eoul ,Koreaer, and AHR. The reduced airway remodeling in mice treated with the TLR-9 agonist was associated with significantly re-duced numbers of peribronchial MBP+ and peribronchial TGF-  1 + cells, and with significantly reduced levels of lung Th2 cytokines [interleukin-5 and interleukin-13] and TGF-  1.C onclusion: These studies demonstrate that TLR-9-based therapies inhibit airway inflammation, remodeling and AHR in mice coexposed to ETS and allergen who exhibit en-hanced airway inflammation and remodeling.C opyright © 2009 S. Karger AG, Basel I ntroduc tion Asthma is a disease characterized by airway inflam-mation and airway hyperreactivity (AHR)[1] . A variety of environmental triggers can aggravate asthma includ-ing allergens, viruses, pollutants and tobacco smoke[1].In terms of tobacco smoke exposure acting as a trigger for asthma, several studies have demonstrated that exposure to either high levels of tobacco smoke in active smokers [2–4]or low levels of tobacco smoke exposure in non-smokers passively exposed to environmental tobaccosmoke (ETS)[5–7] are associated with adverse asthma outcomes including increased prevalence of asthma, in-creased severity of asthma symptoms, increased frequen-Key Words T oll-like receptor-9 ؒ Airway hyperreactivity ؒAirway inflammation ؒ Airway remodeling ؒEosinophilsA bstrac tB ackground: As passive environmental tobacco smoke (ETS) exposure in nonsmokers can increase both asthma symp-toms and the frequency of asthma exacerbations, we uti-lized a mouse model, in which ovalbumin (OVA) + ETS induce significantly increased levels of eosinophilic airway inflam-mation and remodeling compared to either stimulus alone, to determine whether a Toll-like receptor-9 (TLR-9) agonist could reduce levels of airway inflammation, airway remodel-ing and airway hyperreactivity (AHR). M ethods: Mice treated with or without a TLR-9 agonist were sensitized to OVA and challenged with OVA + ETS for 1 month. AHR to methacho-line was assessed in intubated and ventilated mice. Lung Th2 cytokines and TGF-  1 were measured by ELISA. Lungs were processed for histology and immunohistology to quantify eosinophils, mucus, peribronchial fibrosis and smooth mus-cle changes using image analysis. R esults: Administration of a TLR-9 agonist to mice coexposed to chronic ETS and chron-ic OVA allergen significantly reduced levels of eosinophilic airway inflammation, mucus production, peribronchial fi-brosis, the thickness of the peribronchial smooth muscle lay-Received: January 27, 2009 A ccepted after revision: July 2, 2009P ublished online: October 22, 2009 6:31 A Mcy of asthma medication use and increased emergency room visits by asthmatic children. The importance of ETS exposure to asthma is further suggested from recent gene association studies demonstrating a link between a region on chromosome 17q21, combined with ETS, and asthma [8] . Experimental ETS challenge studies in hu-mans also indicate that such exposure has adverse effects on airflow and/or airway responsiveness in asthma [9, 10] . Thus, to reduce the adverse impact of ETS on asthma requires strategies that include reducing the number of smokers and, consequently, of ETS-exposed asthmatics. However, in the USA, although 45% of smokers each year make an attempt to quit, less than 5% of the general pop-ulation are successful [11] . As up to 68% of nonsmoking children with asthma in the inner cities of the USA are also exposed to ETS as assessed by salivary cotinine levels [7] , more immediate strategies are needed to reduce ad-verse asthma outcomes in ETS-exposed asthmatics in ad-dition to the long-term strategy of reducing the number of smokers.S tudies in mouse models suggest a potential immuno-logic mechanism for the interaction of ETS and allergen resulting in adverse asthma outcomes. For example, stud-ies by our group [12] and others [13, 14] have demonstrat-ed that exposure of mice to the combination of ETS and ovalbumin (OVA) allergen induces significantly higher levels of Th2 cytokines, eosinophilic airway inflamma-tion, mucus expression, peribronchial fibrosis, thickness of the smooth muscle layer, and AHR compared to levels induced by exposure to either OVA alone or ETS alone. As recent studies suggest that inhaled and oral cortico-steroids, currently our most effective anti-inflammatory therapies in asthma, are not as effective in asthmatics who smoke [15–18] , there is a need to identify novel ther-apeutic interventions that inhibit Th2 immune responses both in asthmatics exposed and not exposed to ETS. We have previously demonstrated that administration of a Toll-like receptor-9 (TLR-9) ligand [i.e. immunostimula-tory sequences (ISS) of DNA containing a CpG motif] inhibits Th2 cytokine responses [19] , eosinophilic airway inflammation [19, 20] , airway remodeling [21–24]and AHR [19] in mouse models of asthma. In addition, con-jugation of the TLR-9 ligand to the major ragweed aller-gen A mb a 1 significantly reduces seasonal ragweed rhi-nitis symptoms in ragweed-allergic human subjects [25]. Based on the ability of TLR-9 ligands to inhibit Th2 cy-tokine responses [19] , as well as on our demonstration that mice coexposed to ETS and OVA allergen have en-hanced Th2 cytokine responses [12] , we have investigated in the present study whether a TLR-9 ligand would be ef-fective in reducing the enhanced airway inflammation, remodeling and AHR in mice induced by chronic coex-posure to ETS and allergen.M ethodsT herapeutic Intervention with TLR-9 Ligand in Chronic ETS+ OVA-Exposed MiceI n this study we have examined whether administration of aTLR-9 ligand (i.e. ISS) inhibits airway inflammation, airway re-modeling and AHR in mice exposed to chronic ETS in combina-tion with chronic OVA allergen for 1 month. Different groups of8- to 10-week-old BALB/c mice (12 mice/group; The Jackson Lab-oratory, Bar Harbor, Me., USA) were chronically exposed to ETSas well as to either no OVA, OVA or OVA + ISS. As controls wealso included mice not exposed to ETS (no OVA and OVA). Theresults of an OVA + corticosteroid intervention in ETS + OVA-exposed mice are reported elsewhere [26] , and the present study focuses on the effect of ISS on airway inflammation, airway re-modeling, and AHR in ETS-exposed mice. Both interventionstudies (ISS or corticosteroid) used the same control groups tolimit the number of control mice required for these experiments.The group of ETS-exposed mice that received the OVA + ISS were administered intraperitoneally endotoxin-free phosphorothioateISS-oligodeoxynucleotides (5 -TG ACTG TG AA C G T T C G A G A-TGA-3 ; Trilink, San Diego, Calif., USA; 100 g in 100 l of ster-ile, endotoxin-free PBS), starting 1 day before the first intranasalOVA challenge and then continued every other week for the dura-tion of the 1-month period of combined ETS exposure and twice-weekly OVA challenges. Previous studies in our laboratory have demonstrated that ISS inhibits OVA-induced eosinophilic in-flammation, airway remodeling and AHR when administered 1day before OVA challenge [19, 21–24] , and that this inhibitory ef-fect lasts at least 4 weeks [27] .C hronic ETS + Chronic OVA ExposureW e have previously demonstrated that chronic ETS exposurealone does not increase airway inflammation, airway remodelingor AHR in mice [12] . In contrast, coexposure of mice to chronicETS and chronic OVA allergen significantly increases levels of eosinophilic inflammation, airway remodeling and AHR as com-pared to mice exposed to chronic OVA allergen with no ETS ex-posure [12] .C hronic ETS Exposure( f ig. 1 ). Three groups of mice (no OVA,OVA and OVA + ISS) were exposed to chronic ETS (side-streamsmoke from 6 cigarettes/day each administered over approxi-mately 5 min with a 15-min break between cigarettes, 5 days/week) generated by burning 2R4F reference cigarettes (2.45 mg nicotine/cigarette; Tobacco Research Institute, University of Ken-tucky, Lexington, Ky., USA) using a smoking machine (McChes-ney-Jaeger CSM-SSM Single Cigarette Machine, CH Technolo-gies USA, Inc., Westwood, N.J., USA) regulated by programma -b le controls provided with JASPER Windows 9x/2000 softwareover RS-232 communication ports (CH Technologies USA, Inc.)as previously described in this laboratory [12].Each smoldering cigarette is puffed for approximately 2 s, once every 25 s, for a to-tal of 12 puffs/cigarette, at a flow rate of 5 liters/min. The outflowfrom the smoking machine was adjusted to mimic an exposure to6:31AMETS by producing a mixture of room air (98%) and mainstream smoke (2%). The mice were exposed to the ETS in a 12-port, nose-only, directed flow inhalation exposure system (Jaeger-NYU 12 port). Nose ports were monitored for total suspended particulates which we have previously reported to be 173 85.3 g/m3using a gravimetric method [12] . All animal experimental protocols were approved by the University of California, San Diego Animal Subjects Committee.C hronic OVA Protocol(f ig.1).I n these studies,mice were im-munized subcutaneously on days 0, 7, 14 and 21 with 25 g of OVA (OVA, grade V; Sigma Chemicals, St. Louis, Mo., USA) adsorbed to 1 mg of alum (Aldrich) in 200 l normal saline as previously described [28] . OVA-challenged mice received intranasal OVA challenges on days 27, 29 and 31 under isoflurane (Vedco, Inc., St. Joseph, Mo., USA) anesthesia, which were then repeated twice a week for 1 month. The no-OVA age- and sex-matched control mice were sensitized but not challenged with OVA during the 1-month study. The groups of mice that were exposed to ETS had their first ETS exposure on day 33 after they had been sensitized with OVA subcutaneously, and received intranasal OVA challenges on days 27, 29 and 31 as previously described in this laboratory [28] .Chron-ic ETS was continued daily for the subsequent 1-month period of twice-weekly intranasal OVA challenges.P rocessing of Lungs for ImmunohistologyT he mice were sacrificed 24 h after the final chronic OVA and/ or chronic ETS challenge and bronchoalveolar lavage (BAL) fluid and lungs were analyzed as previously described [28] . The lungs in the different groups of mice were equivalently inflated with an in-tratracheal injection of a similar volume of 4% paraformaldehyde solution (Sigma Chemicals) to preserve the pulmonary architec-ture. These lungs were then processed as a batch for either histo-logic staining or immunostaining under identical conditions. Stained and immunostained slides were all quantified under iden-tical light microscope conditions, including magnification (!20), gain, camera position and background illumination. The quantita-tive histologic and image analysis of all coded slides was performed by research associates blinded to the coding of all the slides.B AL and Peribronchial Eosinophils.Total BAL eosinophil counts and the number of peribronchial MBP+ cells were quanti-tated as previously described [28] . In brief, lung sections were pro-cessed for MBP immunohistochemistry using an anti-mouse MBP antibody (kindly provided by James Lee, PhD, Mayo Clinic, Scottsdale, Ariz., USA). The number of individual cells staining positive for MBP in the peribronchial space was counted using a light microscope. Results are expressed as the number of peri-bronchial cells staining positive for MBP/bronchiole with 150–200 m of internal diameter. At least 10 bronchioles were count-ed in each slide.M ucus.The number of PAS-positive and PAS-negative airway epithelial cells in individual bronchioles were counted as previ-ously described in this laboratory [28] . At least 10 bronchioles were counted in each slide. Results are expressed as the percentage of PAS+ cells/bronchiole which is calculated from the number of PAS+ epithelial cells/bronchus divided by the total number of ep-ithelial cells of each bronchiole.P eribronchial Fibrosis.The area of peribronchial trichrome staining in the paraffin-embedded lungs was outlined and quan-tified using a light microscope (Leica DMLS; Leica Microsystems, Inc., New York, N.Y., USA) attached to an image analysis system (Image-Pro Plus; Media Cybernetics, Bethesda, Md., USA) as pre-viously described [28] . Results are expressed as the area of tri-chrome staining/ m length of the basement membrane of bron-chioles 150–200 m of internal diameter.P eribronchial TGF-1+ Cells.The number of peribronchial cells expressing TGF- 1were assessed in lung sections processed for immunohistochemistry using an anti-TGF- 1primary anti-body (Santa Cruz, Calif., USA), the immunoperoxidase method and image analysis quantitation as previously described [28].Re-sults are expressed as the number of TG F- 1+cells/bronchus [28] .F ig. 1.Mice were immunized s.c. on days 0, 7, 14 and 21 with OVA (arrows pointing up). Intranasal OVA challenges were adminis-tered on days 27, 29 and 31, and then repeated twice a week for 1 month. Different groups of mice were either administered ETS alone or ETS in combination with OVA challenges. ETS was start-ed on day 33 (after mice had been sensitized and challenged on 3 occasions with intranasal OVA). The ETS was continued 5 days/week for 1 month. ISS was administered on 3 occasions i.p. (ar-rows pointing down) starting on day 26 with repeat doses on days32 and 46. The mice were sacrificed 24 h after the final OVA chal-lenge on day 60 and BAL fluid and lungs were analyzed.6:31AMT hickness of the Peribronchial Smooth Muscle Layer.The thick-ness of the airway smooth muscle layer (the transverse diameter) was measured from the innermost aspect to the outermost aspect of the smooth muscle layer [28] . The smooth muscle layer thick-ness in at least 10 bronchioles of similar size (150–200 m) was calculated on each slide. Lung sections were also immunostained with an anti- ␣-smooth muscle actin primary antibody (Sigma-Aldrich). The area of ␣-smooth muscle actin staining was out-lined and quantified using a light microscope attached to an im-age analysis system as previously described [28] . Results are ex-pressed as the area of ␣-smooth muscle actin staining/ m length of the basement membrane of bronchioles 150–200 m of inter-nal diameter.A irway Hyperreactivity.AHR to methacholine (Mch) was as-sessed 24 h after the final chronic OVA and/or chronic ETS chal-lenge (after 1 month of repetitive OVA 8ETS challenges) in in-tubated and ventilated mice (flexiVent ventilator; Scireq, Mon-treal, Que., Canada) as previously described in this laboratory [29] . The frequency-independent airway resistance (R a w) was de-termined in mice exposed to nebulized PBS and Mch (3, 24 and 48 mg/ml) [29] . In addition to measuring R a w, the Scireq software also recorded tissue elastance (cm H 2Oؒs/ml) and compliance (ml/cm H 2O).L ung Levels of Th2 Cytokines and TGF- 1L evels of Th2 cytokines (IL-5, IL-13) and TGF- 1were mea-sured in BAL by ELISA (R&D Systems, Inc., Minneapolis, Minn., USA). The IL-5 assay has a sensitivity of 15 pg/ml while the IL-13 and TGF- 1assays each have a sensitivity of 31 pg/ml.P ercentage Reduction in Inflammation and Remodeling inResponse to ISS TherapyT o calculate the percentage reduction in individual indices of airway inflammation and remodeling in response to ISS therapy, the absolute increase in each of these indices in response to OVA + ETS was calculated according to the formula: (OVA + ETS) – (no OVA + ETS). This value is the maximum increase induced by OVA + ETS above baseline values. The reduction of this value in-duced by ISS therapy was calculated as a percentage.S tatistical AnalysisR esults in the different groups of mice were compared by ANOVA using the nonparametric Kruskal-Wallis test followed by posttesting using Dunn’s multiple comparison of means. All re-sults are presented as mean 8SEM. A statistical software pack-age (G raphPad Prism; G raphPad Software, San Diego, Calif., USA) was used for the analysis. p !0.05 was considered statisti-cally significant.R esultsE ffect of ETS on OVA-Induced Airway Inflammation,Airway Remodeling and AHRE xposure of mice to chronic ETS alone did not induce an increase in BAL eosinophils ( f ig. 2 a), MBP+ peribron-chial eosinophils ( f ig. 2 b), TGF- 1+cells ( f ig. 3 a), peri-bronchial fibrosis ( f ig. 3 b, 4), thickness of the peribron-chial smooth muscle layer ( f ig. 5 ), AHR ( f ig. 6 a) and mu-cus production ( f ig. 7 ) compared to non-ETS-exposed mice as previously reported in this laboratory [26].In contrast, chronic ETS in combination with chronic OVA allergen significantly increased all the indices of airway inflammation ( f ig. 2 ), airway remodeling ( f ig. 3 b, 4,5) and AHR ( f ig. 6 a) compared to chronic OVA allergen alone as previously demonstrated in this laboratory [26] .F ig. 2.Eosinophils were quantitated by either Wright-G iemsa staining in BAL fluid ( a) or by immunostaining lung sections with anti-MBP antibody ( b). In ETS-exposed mice challenged with OVA, ISS significantly reduced the number of BAL eosino-phils (p !0.0005; ETS + OVA + ISS vs. ETS + OVA) ( a) and peri-bronchial eosinophils (p !0.0005; ETS + OVA + ISS vs. ETS +OVA) ( b).6:31AME ffect of TLR-9 Ligand on ETS + OVA-InducedEosinophilic Airway InflammationA dministration of ISS to mice exposed to chronic ETS + OVA allergen significantly reduced levels of BAL eo-sinophils by approximately 85% compared to chronic ETS + OVA allergen-challenged mice that did not receive ISS (ETS + OVA vs. ETS + OVA + ISS; p !0.0005) ( f ig. 2 a). Similarly, administration of ISS to mice exposed to chron-ic ETS + OVA allergen significantly reduced levels of peribronchial MBP+ eosinophils by approximately 78% compared to chronic ETS + OVA allergen-challenged mice that did not receive ISS (ETS + OVA vs. ETS + OVA + ISS; p !0.0005)(f ig.2b).E ffect of TLR-9 Ligand on ETS + OVA-InducedPeribronchial TGF- 1+CellsA dministration of ISS to mice exposed to chronic ETS + OVA allergen significantly reduced the number of peri-bronchial TGF- 1+ cells by approximately 72% compared to chronic ETS + OVA allergen-challenged mice that did not receive ISS (ETS + OVA vs. ETS + OVA + ISS; p !0.0001) ( f ig. 3 a).E ffect of TLR-9 Ligand on ETS + OVA-InducedPeribronchial FibrosisA dministration of ISS to mice exposed to chronic ETS + OVA allergen significantly reduced the area of peri-bronchial trichrome staining by approximately 44% com-pared to chronic ETS + OVA allergen-challenged mice that did not receive ISS (ETS + OVA vs. ETS + OVA + ISS; p !0.0001)(f ig.3b,4).E ffect of TLR-9 Ligand on ETS + OVA-InducedThickness of Peribronchial Smooth Muscle LayerA dministration of ISS to mice exposed to chronic ETS + OVA allergen significantly reduced the thickness of the peribronchial smooth muscle layer by approximately 53% compared to chronic ETS + OVA allergen-challenged mice that did not receive ISS (ETS + OVA vs. ETS + OVA + ISS; p !0.0001) ( f ig. 5 a).I n addition to measuring the thickness of the smooth muscle layer, we also determined the area of peribron-chial ␣-smooth muscle actin immunostaining. Adminis-tration of ISS to mice exposed to chronic ETS + OVA al-lergen significantly reduced the area of peribronchial ␣-smooth muscle actin immunostaining by approximately 49% compared to chronic ETS + OVA allergen-chal-lenged mice that did not receive ISS (ETS + OVA vs. ETS + OVA + ISS; p !0.0001) ( f ig. 5 b).E ffect of TLR-9 Ligand on ETS + OVA-Induced AHRA dministration of ISS to mice exposed to chronic ETS + OVA allergen significantly reduced AHR compared to chronic ETS + OVA allergen-challenged mice that did not receive ISS (ETS + OVA vs. ETS + OVA + ISS; Mch 48 mg/ ml; p = 0.04) ( f ig. 6 a). Administration of ISS also reduced tissue elastance in OVA + ETS-challenged mice (p !0.05; Mch 48 mg/ml; ETS + OVA + ISS vs. ETS + OVA)F ig. 3.a The number of peribronchial cells immunostaining pos-itive for TGF- 1in mouse lungs was quantitated by image analy-sis. In ETS-exposed mice challenged with OVA, ISS significantly reduced the number of peribronchial cells immunostaining posi-tive for TGF- 1(p !0.0001; ETS + OVA + ISS vs. ETS + OVA).b The area of peribronchial trichrome staining in mouse lungswas quantitated in m2/m length of bronchus by image analysis.In ETS-exposed mice challenged with OVA, ISS significantly re-duced the area of peribronchial trichrome staining (p !0.0001;ETS + OVA + ISS vs. ETS + OVA).6:31AMTrichrome PASNo OVA + No ETSNo OVA + ETSOVA + No ETSOVA + ETSOVA + ETS + ISSF ig. 4. Lungs from the 5 groups of mice (no OVA, no OVA + ETS, OVA, OVA + ETS, OVA + ETS + ISS) were processed for tri-chrome staining to detect peribronchial fi-brosis (blue) and for PAS staining to detect epithelial mucus expression.6:31 A MF ig. 5.a The thickness of peribronchial smooth muscle layer was assessed by image analysis. b The area of the peribronchial region immunostaining positive with ␣-smooth muscle actin antibody was quantitated by image analysis ( m2/m length of the base-ment membrane of the bronchus). In ETS-exposed mice chal-lenged with OVA, ISS significantly reduced both the thickness of the peribronchial smooth muscle layer (p !0.0001; ETS + OVA + ISS vs. ETS + OVA) ( a) and the area of the peribronchial region immunostaining positive with ␣-smooth muscle actin antibody (p !0.0001; ETS + OVA + ISS vs. ETS + OVA) ( b). SMA = Smooth muscle actin.F ig. 6.a AHR to Mch was assessed 24 h after final chronic OVAand/or chronic ETS challenge in intubated and ventilated mice.Results are expressed as R a w in mice exposed to nebulized Mch (3,24 and 48 mg/ml). In ETS-exposed mice challenged with OVA,ISS significantly reduced AHR (p !0.04; Mch 48 mg/ml; ETS +OVA + ISS vs. ETS + OVA). ISS also reduced tissue elastance inOVA + ETS-challenged mice (p !0.05; Mch 48 mg/ml) ( b)andimproved compliance (p !0.001; Mch 48 mg/ml; ETS + OVA +ISS vs. ETS + OVA) ( c).6:31AM( f ig. 6 b ) and improved compliance (p ! 0.001; Mch 48 mg/ml; ETS + OVA + ISS vs. ETS + OVA) ( f ig. 6 c ). E ffect of TLR-9 Ligand on ETS + OVA-Induced Mucus ExpressionA dministration of ISS to mice exposed to chronic ETS + OVA allergen significantly reduced levels of mucus ex-pression by approximately 58% compared to chronic ETS + OVA allergen-challenged mice that did not receive ISS (ETS + OVA vs. ETS + OVA + ISS; p ! 0.0001) ( f ig. 7 ).E ffect of TLR-9 Ligand on ETS + OVA-Induced Lung Th2 Cytokines (IL-5, IL-13) and TGF- 1 A dministration of ISS to mice exposed to chronic ETS + OVA allergen significantly reduced levels of BAL IL-5 ( f ig. 8 a ), BAL IL-13 ( f ig. 8 b ) and TG F-  1( f ig. 8 c ) com-pared to chronic ETS + OVA allergen-challenged mice that did not receive ISS (ETS + OVA vs. ETS + OVA + ISS; p !0.05).D isc ussion I n this study we demonstrated that a TLR-9 agonist (i.e. ISS) is very effective in reducing ETS-enhanced eo-sinophilic airway inflammation, airway remodeling and AHR in mice exposed to the combination of chronic ETS and chronic OVA allergen. In mice coexposed to ETS and OVA, the TLR-9 agonist inhibited the expression of cyto-kines that contribute to eosinophilic inflammation (i.e. IL-5), AHR (i.e. IL-13) and airway remodeling (i.e. TGF-  1 ). Previous studies from our [12, 26] and other labora-tories [13, 14] have demonstrated an enhanced Th2 re-sponse to the combination of ETS and allergen as opposed to either stimulus alone. However, the demon-stration that a TLR-9 agonist inhibits enhanced airway inflammation, remodeling and AHR in mice exposed to OVA + ETS is novel and most likely due to the ability of TLR-9-based therapies to inhibit Th2 responses to aller-gen[19] , which are enhanced in mice coexposed to aller-gen and ETS[12, 26] and reduced in those mice admin-istered the TLR-9 ligand. As corticosteroids exhibit a reduced anti-inflammatory effectiveness in smokers[15–18] , it is important to identify therapeutic interven-tions that inhibit Th2 responses in the presence of ETS. In addition to having anti-inflammatory properties, ISS also inhibited airway remodeling in ETS-exposed mice. Studies have shown that smokers with asthma have a more rapid decline in lung function as compared to non-smokers with asthma[2] . Thus, there is also a need to identify novel therapeutic interventions that inhibit air-way remodeling in smokers. In mice exposed to allergen + ETS, the increased numbers of cells expressing TGF-  1may contribute to airway remodeling as several[30–32],but not all [33] , studies in mice in which TGF-  1signal-ing is inhibited demonstrate reduced airway remodeling.Thus, the ability of ISS to significantly reduce levels of lung TGF-  1 as well as the numbers of peribronchial cellsexpressing TGF-  1, such as eosinophils, in mice exposed to ETS + allergen may contribute to a reduction in levels of airway remodeling and airway responsiveness. How-ever, as TLR-9-based therapies influence several different cell types which express TLR-9, the resultant inhibit ef-fects on remodeling and airway responsiveness may, or may not, be linked.T his study extends previous observations that ISS is able to inhibit inflammation, remodeling and AHR inmice exposed to allergen alone[19–24] to demonstrate that ISS can inhibit these asthma outcomes in mice ex-posed to the combination of ETS and allergen who have enhanced inflammation, remodeling and AHR. As we have previously demonstrated that ISS inhibits these air-way inflammation and remodeling outcomes in non-ETS-exposed OVA-challenged mice[21] , we did not in-clude a group of ISS-treated, OVA-challenged, non-ETS-exposed mice in this study. Although the lack of a control ISS + OVA group is a limitation of our study, in compar-ing results of ISS therapy in this ETS + OVA exposure study to previous studies of ISS in non-ETS-exposedF ig. 7. The percentage of bronchial epithelial cells staining posi-tive for PAS was quantitated by light microscopy. In ETS-exposed mice challenged with OVA, ISS significantly reduced the percent-age of PAS+ airway epithelial cells (p ! 0.0001; ETS + OVA + ISS vs. ETS + OVA).6:31 A Mmice, we demonstrated that ISS inhibited airway inflam-mation and remodeling in mice exposed to ETS + OVA as effectively as in previous studies in which OVA-ex-posed mice were treated with ISS in the absence of ETSexposure [21]. For example, comparing the levels of inhi-bition of the indices of airway inflammation and airway remodeling in the current study to those of previous stud-ies by our group [21] demonstrates that ISS induced com-parable levels of inhibition of BAL eosinophilia (85 vs.63%)[21] , a reduction in smooth muscle thickness (53 vs. 45%) and a reduction in the number of PAS+ cells (58 vs. 64%). In addition, in the present study ISS reduced OVA + ETS-induced airway inflammation and remodeling to levels significantly below that of untreated mice exposed to OVA alone (OVA + no ETS + no ISS vs. OVA + ETS + ISS), suggesting that ISS was impacting both the OVA as well as the OVA + ETS effect on inflammation and re-modeling. These observations, demonstrating the poten-tial therapeutic utility of ISS in ETS-exposed mice, arepotentially important to the large number of asthmatic children and adults exposed to ETS, in whom ETS expo-sure is associated with adverse asthma outcomes[6, 7, 34].The large number of asthmatics exposed to ETS has beennoted in pediatric and adult studies[6, 7, 34] . For exam-ple, approximately 68% of nonsmoking urban children aged 8–14 years had evidence of ETS exposure as assessed by salivary cotinine levels [7] while approximately 29% of nonsmoking adult asthmatics aged 18–50 years in Cali-fornia reported some regular ETS exposure (defined as most days or nights) during an 18-month study period[34] . The ETS-exposed asthmatics had increased asthma severity, increased health care utilization for asthma (emergency department visits, urgent physician visits and hospitalizations) and worse asthma-specific quality of life [34] . The asthmatics who reported cessation of ETS exposure over the 18-month study experienced a reduc-F ig. 8. Levels of Th2 cytokines (IL-5, IL-13) and TGF-  1 in BAL were measured by ELISA. Administration of ISS to mice exposed to chronic ETS + OVA allergen significantly reduced levels of IL-5 ( a ), IL-13 ( b ) and TFG-  1 (c ) compared to chronic ETS + OVA-challenged mice that did not receive ISS (p ! 0.05; ETS + OVA vs. ETS + OVA + ISS).6:31 A M。