TLR4 TLR4在人冠脉内皮细胞胞内发挥作用 LBP和sCD14在介导LPS-反应中的作用
- 格式:pdf
- 大小:4.16 MB
- 文档页数:25
冠心病病人外周血sCD14、TLR4表达与冠状动脉粥样硬化斑块稳定性的关系李海龙;林英子【期刊名称】《中西医结合心脑血管病杂志》【年(卷),期】2022(20)6【摘要】目的探讨冠心病病人外周血可溶性白细胞分化抗原14(sCD14)、Toll样受体4(TLR4)表达水平与冠状动脉粥样硬化斑块稳定性的相关性。
方法选取2018年10月—2020年2月我院心血管内科收治的冠心病病人146例(冠心病组),其中,稳定型心绞痛(SAP)38例、不稳定型心绞痛(UAP)55例、急性心肌梗死(AMI)53例;采用Gensini积分评估冠心病病人冠状动脉病变程度。
另选取同期体检健康者146名作为对照组。
采用流式细胞仪检测研究对象外周血TLR4表达水平,酶联免疫吸附法(ELISA)测定血清中sCD14水平。
Pearson相关分析法分析sCD14、TLR4表达水平与Gensini评分相关性。
采用二元Logistic回归分析影响冠心病病人冠状动脉粥样硬化斑块不稳定的因素。
结果与对照组比较,SAP组、AMI组、UAP组sCD14、TLR4表达水平均升高(P<0.05);与SAP组比较,AMI组、UAP组Gensini评分、外周血sCD14、TLR4表达水平均升高(P<0.05);与UAP组比较,AMI组Gensini评分升高(P<0.05)。
与硬斑块组比较,软斑块组、混合型斑块组sCD14、TLR4表达水平升高(P<0.05);与混合型斑块组比较,软斑块组sCD14、TLR4表达水平升高(P<0.05)。
sCD14、TLR4表达水平均与Gensini评分呈正相关(P<0.05)。
Logistic回归分析显示,高水平TLR4、高水平sCD14、高Gensini 评分是冠心病病人冠状动脉粥样硬化斑块不稳定的危险因素(P<0.05)。
结论冠心病病人外周血TLR4表达水平及血清sCD14水平明显升高,与冠状动脉粥样硬化斑块不稳定有密切关系,且TLR4和sCD14与冠状动脉病变严重程度存在相关性。
TLR4激动上调内皮细胞氧化低密度脂蛋白受体LOX-1表达王虹艳;曲鹏;富晶;姜华【期刊名称】《高血压杂志》【年(卷),期】2005(13)7【摘要】目的近年研究表明介导先天免疫反应的受体Toll样受体4(TLR4)参与了动脉硬化的发生发展。
业已证明氧化低密度脂蛋白受体LOX1介导内皮细胞活化和功能失调,激发炎症过程,在动脉粥样硬化的发生和发展中起着极为重要作用。
本研究观察TLR4激动是否调节内皮细胞LOX1表达。
方法应用脂多糖(LPS)刺激体外培养的人脐静脉内皮细胞(HUVECs)24h。
采用RTPCR和流式细胞术分别检测TLR4、LOX1mRNA和蛋白表达水平。
为了观察转录因子NFκB在调节LOX1表达中的作用,应用NFκB特异性抑制剂咖啡酸苯乙酯(CAPE)预处理细胞,然后以LPS 刺激,检测LOX1mRNA和蛋白变化。
结果LPS(10~1000ng/mL)上调HUVECsTLR4和LOX1mRNA表达,LPS(1000ng/mL)上调TLR4和LOX1蛋白表达,CAPE(20μg/mL)可抑制LPS介导的LOX1表达上调。
结论TLR4/NFκB信号途径可能通过上调内皮细胞LOX1表达参与动脉粥样硬化的发生及发展。
【总页数】5页(P422-426)【关键词】Toll样受体4;脂多糖;人脐静脉内皮细胞;氧化低密度脂蛋白受体;动脉粥样硬化【作者】王虹艳;曲鹏;富晶;姜华【作者单位】大连医科大学附属第二医院心血管内科;大连医科大学附属第二医院实验中心【正文语种】中文【中图分类】R543.5【相关文献】1.氧化型低密度脂蛋白诱导肝窦内皮细胞植物血凝素样氧化型低密度脂蛋白受体1的表达 [J], 刘静;王云芳;牛瑞兰;张琦;权金星;田利民2.Toll样受体4/核因子κB和氧化低密度脂蛋白受体LOX-1对单核内皮细胞黏附的影响 [J], 王虹艳;曲鹏;吕申;刘敏;姜华3.银杏叶提取物对脂多糖介导的血管内皮细胞低密度脂蛋白受体LOX-1表达的影响 [J], 富晶;王虹艳;曲鹏4.氧化低密度脂蛋白促进内皮细胞MMP-9 mRNA、LOX-1 mRNA的表达及LOX-1在其中的作用研究 [J], 祝河忠;申晓彧;陈佳娟;潘庆敏;严洁5.氧化应激和植物凝集素样氧化型低密度脂蛋白受体1在氧化低密度脂蛋白上调内皮细胞自噬水平中的作用 [J], 韩侨;张艳林;尤寿江;刘慧慧;曹勇军;陈锐;刘春风因版权原因,仅展示原文概要,查看原文内容请购买。
四川大掌博士后研究工作报告中文摘要G一细菌是牙周炎的主要致病菌,内毒素是革兰氏阴性菌外膜的主要成分,是一种公认的重要的牙周致病因子,在牙周炎发病中的作用越来越受到重视。
牙龈成纤维细胞是牙周组织的主要细胞群,作为牙周组织中重要炎症反应细胞,在LPS引起的牙周炎组织病理损害过程中具有重要作用。
近年来的研究表明,成纤维细胞也具有一定的免疫细胞活性,可与内毒素直接作用,可作为免疫辅助细胞参与牙周组织的破坏过程,产生炎症细胞因子,介导炎症反应,参与牙周炎的发生和发展。
LPS受体识别、激活细胞内的信号转导途径等方面一直存在争议。
人牙龈成纤维细胞LPS受体的寻找和鉴定一直是该领域研究的热点课题。
Toll样受体(Toll-likereceptors,TLRs)的发现和研究为该领域带来了新的机遇,TLRs极有可能是LPS的识别受体,将刺激信号传入胞内,在介导LPS的病理生理中具有重要作用。
那么,牙龈成纤维细胞是否表达Toll样受体,如果表达,表达何种Toll样受体,其是否介导了LPS对牙龈成纤维细胞的激活,其特异的信号转导通路及调节机制如何,以及何种转录因子在其中起了重要作用,本实验拟对上述问题进行研究,为进一步研究LPS对牙龈成纤维细胞激活/损伤机理奠定基础,也为揭示LPS的膜信号转导机制和牙周炎的防治提供新思路。
基于以上原因,本论文进行了以下实验:1人牙龈成纤维细胞TLR4编码区cDNA克隆、蛋白表达和多克隆抗体的制各首先我们从人牙龈成纤维细胞中成功地克隆到了人TLR4全部编码区序列,约2.4kb,证实了人牙龈成纤维细胞表达TLR4mRNA:根据TLR4的基因结构和读框,我们采用基因重组的方法成功地构建了原核表达载体pRSETA.TLR4,并通过对T7启动子有较好效果的大肠杆菌BL21进行IPTG诱导表达,结果获得了预计的约90KD的TLR4融合蛋白,并用NiNl’A柱亲和层析进行了初步纯化;纯化的TLR4融合蛋白免疫新西兰大白兔,采用双向免疫扩散试验和ELISA法检测效价的方法证明了所制备的抗体是成功的,为进一步的研究奠定了物质基础。
小剂量脂多糖诱导血管内皮细胞TLR4的表达路玲;常文秀;曹书华;王勇强;高红梅【摘要】Objective To investigate the expression of Toll like receptor4(TLR4)on human umbilical vein endothelial cells (HUVEC) induced by small doses of lipopolysaccharide (LPS). Methods The ECV304 belonging to HUVEC was cultured in vitro, and hatched with LPS and LPS + antibody of TLR4(anti-TLR4) respectively. Cell proliferation activity was detected by MTT. The effect of LPS on change of the cell surface of TLR4 was observed with immunohistochemical staining method. The expression of TLR4-mRNA and of IL-8mRNA were evaluated by Fluorescence Detection Quantitative Polymerase Chain Reaction (PCR). Results ECV304 cells stimulated by LPS of(10-50)ng/mL within 24 h, MTT detection showed no apparent change (P> 0.05); but with 100 ng/mL LPS stimulated the cells 24 h, the number of cells reduced significantly (P <T 0.05), under the circumstances, anti-TLR4 didn't withstand evidently. The 10 ng/mL LPS stimulated ECV304 in 24 h, the expression of TLR4 was elevatory; and with 50 ng/mL LPS stimulation, the expression of TLR4 was significantly increased in 6-24 h (P<0.05), with 50 ng/mL LPS stimulating ECV304 cells in 4 h and 8 h. The expression of TLR4-mRNA in LPS group and LPS+anti-TLR4 group increased significantly as compared with the normal control group (P<0.05), but LPS+anti-TLR4 group only in 8 h lower than LPS group;the expression of IL-8mRNA in LPS group and LPS+anti-TLR4 evaluated obviously as compared with the normal control group only in 8h, which did not affect with anti-TLR4. Conclusion Small doses of LPS can induce ECV304 cells expressing TLR4 and cause cell activation, and anti-TLR4 can inhibit TLR4 expression, but can't restrain cell activation.%目的:观察小剂量脂多糖(LPS)对人脐静脉内皮细胞(ECV304)TOLL样受体4(TLR4)表达的影响.方法:体外培养ECV304细胞,分别与LPS及LPS+TLR4抗体进行孵育.MTT法检测细胞的增殖活性,免疫组化染色法检测细胞表面TLR4的表达,实时荧光定量聚合酶链反应(RT-PCR)检测细胞核核内TLR4-mRNA及IL-8mRNA的表达.结果:LPS(10~50 ng/mL)刺激ECV304细胞24 h内,细胞增殖活性无明显变化(P >0.05);而以100 ng/mL 刺激24 h后,细胞增殖活性明显降低(P< 0.05),TLR4抗体对此无明显拮抗作用.LPS能明显上调ECV304表达TLR4、TLR4-mRNA及IL-8mRNA,其中10 ng/mL的LPS在24 h时、50 ng/mL LPS在6~24 h时,TLR4表达具有统计学意义(P< 0.05);50 ng/mL的LPS刺激ECV304细胞在4 h及8 h 时,细胞核内TLR4mRNA 表达均明显升高(P < 0.05),而IL-8mRNA 表达在8 h 时明显升高(P < 0.05).TLR4 抗体对ECV304表达TLR4及TLR4-mRNA有拮抗作用(P< 0.05),对IL-8mRNA表达无明显拮抗作用(P> 0.05).结论:小剂量LPS可诱导ECV304细胞表达TLR4并引起细胞活化,TLR4抗体可抑制TLR4的表达,但不能抑制细胞的活化.【期刊名称】《中国中西医结合外科杂志》【年(卷),期】2012(018)001【总页数】5页(P47-51)【关键词】脂多糖;血管内皮细胞;TOLL样受体4【作者】路玲;常文秀;曹书华;王勇强;高红梅【作者单位】天津医科大学第一中心临床学院ICU,天津300192;天津医科大学第一中心临床学院ICU,天津300192;天津医科大学第一中心临床学院ICU,天津300192;天津医科大学第一中心临床学院ICU,天津300192;天津医科大学第一中心临床学院ICU,天津300192【正文语种】中文【中图分类】Q95-33脂多糖(Lipopolysaccharide,LPS)是革兰阴性菌细胞壁的主要成分与毒力因子,可使内皮细胞活化,促使炎症介质、酶等释放,作用于凝血、纤溶系统,促发弥漫性血管内凝血。
CD14与肿瘤的相互关系及研究进展李康【摘要】CD14为白细胞表面分化抗原,在介导机体免疫应答及炎性反应中具有重要作用.近年来研究发现CD14的基因多态性能够影响多种肿瘤的易患性,同时其免疫调节功能与肿瘤的发生、发展密切相关,但其详细的作用机制仍未明确.该文针对CD14多态性与肿瘤、CD14与肿瘤免疫逃逸、CD14与肿瘤发生、发展的关系研究现状展开综述,期望为CD14在抗肿瘤中的作用研究提供理论支持.【期刊名称】《医学综述》【年(卷),期】2014(020)015【总页数】3页(P2726-2728)【关键词】CD14;肿瘤;基因多态性;免疫逃逸【作者】李康【作者单位】西藏自治区人民医院消化内科,拉萨850000【正文语种】中文【中图分类】R34分化抗原群(cluster of differentiation antigen 14,CD14)为细胞表面糖蛋白家族成员之一,最初被发现为白细胞表面分化抗原,是单核细胞、中性粒细胞和巨噬细胞的特异性表面标志物[1]。
哺乳细胞中CD14为革兰染色阴性菌脂多糖(lipopolysaccharide,LPS)的高亲和受体,能够参与机体对革兰染色阴性菌的吞噬与消化,在介导机体的免疫应答以及炎性反应中均发挥着重要作用[2-3]。
近年来,多项研究指出CD14的基因多态性与肿瘤的易患性密切相关。
随着研究的深入,CD14与肿瘤免疫逃逸以及肿瘤发生、发展的关系也逐渐受到关注。
基于CD14对免疫应答及细胞因子的调控作用,深入探讨CD14的功能及作用机制对于抗肿瘤研究具有非常重要的意义。
1 CD14的结构人CD14基因位于第5号染色体长臂5q23~31区,长约3900 bp,包含2个外显子和较长的5′端以及3′端非编码序列。
CD14蛋白包含375个氨基酸残基,其成熟肽长356个氨基酸,其中包含19个氨基酸长度的信号肽,4个N型糖链黏部位,约占整个相对分子质量的20%。
此外,CD14的氨基酸序列中还包含一个重复10次的富含亮氨酸的特异性序列,可能与CD14分子的膜结合以及与其他蛋白质的结合有关。
TLR4在冠状动脉粥样硬化性心脏病患者单个核细胞炎性反应中的作用黄俊;何隆淼;杨琴;陈云肖【期刊名称】《中国急救医学》【年(卷),期】2016(036)008【摘要】目的探讨冠状动脉粥样硬化性心脏病患者外周血单个核细胞Toll样受体4(Toll-like receptor 4,TLR4)介导的炎性反应及在急性冠状动脉事件中的作用.方法连续入选研究对象,取外周血分离单个核细胞,予以脂多糖(LPS)和(或)阿托伐他汀处理.流式细胞仪检测细胞TLR4表达,ELISA法检测肿瘤坏死因子(TNF)-α、白细胞介素(IL)-6、IL-1、IL-10水平.结果急性冠状动脉综合征(ACS)组单个核细胞TLR4及TNF-α、IL-6、IL-1明显高于稳定型心绞痛(SAP)组、冠状动脉慢血流(CSF)组及健康对照(HC)组,而IL-10低于其他各组(P<0.05).LPS刺激后,与HC组比较,SAP组、CSF组TLR4及TNF-α、IL-6、IL-1明显高于原有水平,IL-10低于原有水平(P<0.05),而阿托伐他汀干预能抑制这一现象.阿托伐他汀干预ACS组可致TLR4和TNF-α、IL-6、IL-1水平下降,IL-10水平上升(P<0.05).结论 TLR4过度表达及其炎症反应可能与ACS的发生密切相关.LPS能激活SAP患者、CSF患者单个核细胞TLR4,介导炎性反应.阿托伐他汀能抑制TLR4的过度表达及其炎性反应.【总页数】4页(P703-706)【作者】黄俊;何隆淼;杨琴;陈云肖【作者单位】330006 江西南昌,南昌大学第一附属医院心内科;330006 江西南昌,南昌大学第一附属医院心内科;330006 江西南昌,南昌大学第一附属医院心内科;330006 江西南昌,南昌大学第一附属医院心内科【正文语种】中文【相关文献】1.外周血单个核细胞TLR4的表达在慢性乙型重型肝炎患者中的意义2.右美托咪定对脂多糖诱导的子宫内膜炎小鼠子宫组织中TLR4-NF-κB/NLRP3炎性小体介导的炎性反应的影响3.优质护理在呼出气一氧化氮测定哮喘患者气道炎性反应评估中的作用研究4.优质护理在呼出气一氧化氮测定哮喘患者气道炎性反应评估中的作用研究5.慢性心力衰竭患者外周血单个核细胞miR-590表达与左心室重构和炎性反应的关系因版权原因,仅展示原文概要,查看原文内容请购买。
TLR4与N-HDL-C在冠心病中的表达及辛伐他汀的干预机制一、TLR4在冠心病中的表达TLR4是Toll样受体家族的一员,是一种重要的免疫受体,可识别和结合细菌脂多糖,引发炎症反应。
研究表明,TLR4在冠心病中的表达水平明显升高,并且与冠心病的发展和进展密切相关。
具体来说,TLR4的高表达能够激活NF-κB信号通路,导致炎症介质的释放增加,进而引发动脉粥样硬化斑块形成以及血管炎症反应的加剧,从而增加冠心病的风险。
研究发现TLR4的激活还可以诱导内皮细胞的黏附分子表达增加,进而促进白细胞的黏附和血管内皮损伤。
TLR4在冠心病的病理生理过程中起着举足轻重的作用。
二、N-HDL-C在冠心病中的作用高密度脂蛋白胆固醇(HDL)被认为是一种“良好”的胆固醇,其具有抗氧化、抗炎和反粘附等多种保护心脏疾病的作用。
N-HDL-C是指对HDL-C进行了修饰的一种脂蛋白,其具有更强的抗氧化和抗炎能力。
研究表明,N-HDL-C的水平与冠心病的发生和发展密切相关。
N-HDL-C水平降低会导致氧化应激和炎症反应的增加,从而加速动脉粥样硬化的发展。
N-HDL-C还可以促进胆固醇从动脉壁中回收到肝脏,从而减少胆固醇在动脉中的沉积,减轻动脉粥样硬化的程度。
N-HDL-C在冠心病的发病机制中扮演着重要的保护作用。
三、辛伐他汀的干预机制辛伐他汀是一种常用的降脂药物,其主要作用是通过抑制HMG-CoA还原酶,降低胆固醇的合成,从而起到降脂的效果。
辛伐他汀还具有多种其他保护心脏的作用。
研究表明,辛伐他汀可以通过抑制TLR4的激活,减少炎症介质的释放,从而减轻血管炎症反应。
辛伐他汀还可以增加N-HDL-C的水平,通过其抗氧化和抗炎作用保护血管内皮功能,减少动脉粥样硬化斑块的形成。
辛伐他汀还可以通过提高内皮细胞NO的合成,扩张血管,减少心血管事件的发生。
辛伐他汀通过降低胆固醇合成和提高HDL-C水平,从而起到保护血管和心脏的作用。
辛伐他汀还可以通过抑制TLR4的激活和增加N-HDL-C的水平,进一步减轻血管炎症反应,从而减少冠心病的发生和发展。
TLR4、CD14在干燥综合征发病中的作用及其机制研究的
开题报告
第一部分:研究背景与意义
干燥综合征是一种常见的自身免疫疾病,其发病机制至今不明确。
临床上常见的症状包括口干、眼干、皮肤干燥等表现。
干燥综合征患者的唾液腺和泪腺等分泌腺体积缩小、腺泡变形,导致分泌液体量和质量下降。
在免疫学方面,干燥综合征是具有自身免疫性质,T细胞介导的体液免疫和细胞免疫功能异常。
尽管许多研究聚焦在免疫失调上,但在干燥综合征中,TLR4、CD14的角色尚未完全认识。
因此,本研究旨在探讨TLR4、CD14在干燥综合征患者中的表达及其参与发病机制的作用,以期为干燥综合征的治疗提供科学依据。
第二部分:研究内容及方法
研究目的:研究TLR4、CD14在干燥综合征发病中的作用及其机制
研究方法:
1.研究对象选择:40例干燥综合征患者和30例健康对照者
2.外周血细胞分离,提取外周血单个核细胞 (PBMCs),RNA/DNA分离后用qPCR法测定TLR4、CD14 mRNA/P蛋白的表达水平,并用免疫荧光法检测TLR4、CD14蛋白的表达水平
3.患者血清中检测自身抗体、炎症因子水平的变化。
包括其抗核抗体、抗SSA、抗SSB自身抗体、白介素10(IL-10)、肿瘤坏死因子α(TNFα)、白细胞介素6(IL-6)等。
第三部分:预期结果及意义
本研究将详细分析TLR4和CD14在干燥综合征发病机制中的作用,验证它们在干燥综合征中的具有重要作用及免疫生物学意义。
预计的实验结果将有助于深入了解干燥综合征的免疫病理学机制,为干燥综合征的深入研究、诊断及治疗提供新的理论基础。
TLR4信号转导通路中胞外分子之间的相互作用
陈伟;史忠
【期刊名称】《医学综述》
【年(卷),期】2006(012)009
【摘要】细菌脂多糖(LPS)介导的炎症信号主要通过一种跨膜蛋白toll样受体
4(toll-like receptor 4,TLR4)传入胞内,引起一系列相应的细胞效应.近年来对该信
号通路的研究取得了一些新的进展,本文综述了该信号通路中胞外的主要分子LPS、脂多糖结合蛋白(LBP)、CD14、TLR4和髓样分化蛋白-2(MD-2),及其各分子之间
的相互作用.通过这些分子之间的相互作用激活TLR4并触发信号转导.
【总页数】3页(P522-524)
【作者】陈伟;史忠
【作者单位】中国人民解放军第三军医大学新桥医院急救部,重庆,400037;中国人
民解放军第三军医大学新桥医院急救部,重庆,400037
【正文语种】中文
【中图分类】R392.11
【相关文献】
1.酸乳体系中乳酸菌胞外多糖与蛋白相互作用研究进展 [J], 曾令鹤;钱方;姜淑娟;
牟光庆
2.克氏锥虫Tc13家族新成员的分子鉴定及其特性--对Tulahuén细胞株的Tc13
抗原与β1-肾上腺素受体胞外第二环相互作用的描述 [J],
3.中性粒细胞胞外诱捕网形成的分子机制及其在脓毒症中病理作用的研究进展 [J],
郑大勇;张天翼;林源希;李真玉;宗晓龙
4.光谱法在分子之间相互作用研究中的应用 [J], 刘峥;夏之宁
5.表面活性剂在药物制剂中的应用及其与药物分子之间的相互作用 [J], 张晋;刘少杰;郑利强;李干佐
因版权原因,仅展示原文概要,查看原文内容请购买。
9 简述TLR4信号通路在炎症中的功能。
TLR4 recognizes lipopolysaccharide (LPS) together with myeloid differentiation factor 2 (MD2) on the cell surface. LPS is a component derived from the outer membrane of Gram-negative bacteria and is known to be a cause of septic shock.The crystal structure of a complex comprising TLR4, MD2, and LPS revealed that two complexes of TLR4-MD2-LPS interact symmetrically to form a TLR4 homodimer (Park et al., 2009).TLR4 is also involved in the recognition of viruses by binding to viral envelope proteins. In addition, TLR4 modulates the patho-genesis of H5N1 avian influenza virus infection by recognizing a DAMP rather than the virus itself (Imai et al., 2008). Acutelung injury caused by avian influenza virus infection produces endogenous oxidized phospholipids, which stimulate TLR4.Mice lacking TLR4 were found to be resistant to avian flu-induced lethality.1TL R4的结构分布及信号通路TLR4是人类发现的第一个TLR 相关蛋白,几乎分布于所有的细胞系,主要表达在参与宿主防御功能的细胞上,如单核巨噬细胞、粒细胞、树突状细胞、淋巴细胞、内皮细胞和上皮细胞,以骨髓单核细胞的表达尤其多[ 5 ],近年来发现,肾小管上皮细胞、心脏、呼吸道上皮细胞和肠上皮细胞也均表达TLR4[ 6 ] 。
The FASEB Journal express article 10.1096/fj.03-1263fje. Published online May 7, 2004.Toll-like receptor 4 functions intracellularly in human coronary artery endothelial cells: roles of LBP and sCD14 in mediating LPS-responsesStefan Dunzendorfer, Hyun-Ku Lee, Katrin Soldau, and Peter S. TobiasThe Scripps Research Institute, Department of Immunology, 10550 North Torrey Pines Road, La Jolla, CA 92037Corresponding author: Peter S. Tobias, The Scripps Research Institute, Department of Immunology IMM-12, 10550 North Torrey Pines Road, La Jolla, CA 92037. E-mail:tobias@ABSTRACTEndothelial cells are activated by microbial agonists through Toll-like receptors (TLRs) to express inflammatory mediators; this is of significance in acute as well as chronic inflammatory states such as septic shock and atherosclerosis, respectively. We investigated mechanisms of lipopolysaccharide (LPS)-induced cell activation in human coronary artery endothelial cells (HCAEC) using a combination of FACS, confocal microscopy, RT-PCR, and functional assays. We found that TLR4, in contrast to TLR2, is not only located intracellularly but also functions intracellularly. That being the case, internalization of LPS is required for activation. We further characterized the HCAEC LPS uptake system and found that HCAEC exhibit an effective LPS uptake only in the presence of LPS binding protein (LBP). In addition to its function as a catalyst for LPS-CD14 complex formation, LBP enables HCAEC activation at low LPS concentrations by facilitating the uptake, and therefore delivery, of LPS-CD14 complexes to intracellular TLR4-MD-2. LBP-dependent uptake involves a scavenger receptor pathway. Our findings may be of pathophysiological relevance in the initial response of the organism to infection. Results further suggest that LBP levels, which vary as LBP is an acute phase reactant, could be relevant to initiating inflammatory responses in the vasculature in response to chronic or recurring low LPS. Key words: innate immunity • endothelium • endotoxinEndothelial cells (EC), which form the inner vascular lining, are important in the regulation of vascular tone, coagulation and fibrinolysis, cellular growth, differentiation, and last but not least, immune and inflammatory responses (1, 2). Moreover, these cells are targets for many endogenous and exogenous agents (e.g., lipopolysaccharide [LPS]), that activate the endothelium and result in production of proinflammatory mediators (3). These mechanisms play roles in Gram-negative sepsis, where the body may be overwhelmed with LPS and also in diseases that are linked to a low, but chronic, LPS burden. There is considerable evidence that endotoxin (LPS) contributes to the propagation of atherosclerosis (4, 5), which shares many features with inflammatory diseases (6).LPS is a major component of the outer surface of Gram-negative bacteria and a potent activator of cells of the immune and inflammatory system, including endothelial cells. In vivo, LPS-induced cell activation depends on the presence of at least four proteins: TLR4 (7–9), MD-2 (10), CD14 (11), and LPS binding protein (LBP) (12). The first three may be present as a complex on the surfaces of myeloid (and transfected) cells (13). Many cells, among them epithelial and endothelial cells, do not express the membrane bound form of CD14 (mCD14); their activation involves soluble CD14 (sCD14) (14). LPS·sCD14 complexes will form slowly in vitro (15), but, in vivo, plasma LBP (16–18) catalyzes LPS·CD14 complex formation (19). LPS·CD14 complexes, whether membrane bound or soluble, are necessary for LPS binding to TLR4-MD-2 (20). In mCD14 expressing cells, most LPS appears to be internalized quite independently of activation (21), and mCD14-negative cells also internalize LPS (22, 23). Although LPS internalization by phagocytes has generally been regarded as a disposal function (21), studies have also suggested that internalization might be required for LPS signaling in myeloid lineage cells (24, 25). However, a very recent publication clearly demonstrates that signaling starts on the surface of TLR4-transfected and TLR4 surface expressing cells when incubated with LPS-sCD14 complexes, even in the absence of LPS uptake (26). Other recent work suggests that internalization of LPS is required for activation of murine intestinal epithelial cells, where mCD14, but not TLR4, is found on the surface (27, 28).MATERIALS AND METHODSAntibodiesAnti-TLR4 polyclonal antibody (pAb) H80 and monoclonal antibody (mAb) HTA125 were purchased from Santa Cruz Biotechnology (Santa Cruz, CA). Monoclonal anti-TLR2 2392 was a gift of Paul Godowski (Genentech), and anti-TLR4 HTA1216, HTA414, and HTA405 were gifts from Kensuke Miyake (Saga Medical School, Japan). Anti-LBP mAb 2B5 and anti-CD14 mAb 28C5 were produced in our laboratory (29). Polyclonal rabbit anti-human MyD88 was from Imgenex (San Diego, CA). The monoclonal mouse anti-human CD14 (clone M5E2), mouse anti-human CD144 (clone 55-7H1), mouse anti-human CD106 (VCAM-1; clone 51-10C9), rat anti-mouse CD31 (PECAM-1) (clone MEC13.3), rat anti-mouse CD144 (VE-cadherin; clone 11D4.1), and rat anti-mouse CD106 (VCAM-1; clone 429-MVCAM.A) used for immunofluorescence staining and PE-conjugated anti-human CD62E (E-selectin/ELAM; clone 68-5H11) were from PharMingen (San Diego, CA). All secondary antibodies used in fluorescence microscopy were Alexa Fluor conjugates (Molecular Probes, Eugene, OR). PE-conjugated secondary antibodies used in flow cytometry were purchased from PharMingen (San Diego, CA).Proteins, LPS, and other reagentsHuman recombinant sCD14 and LBP were expressed in and purified from BTI-TN-5B1-4 cells (Invitrogen, San Diego, CA) as recently described (23). Both proteins can be radiolabeled at an artificially introduced protein kinase A site. Macrophage-activating lipopeptide-2 (MALP-2) was from Alexis Biochemicals (San Diego, CA), recombinant mouse TNF-α was purchased from Serotec (Oxford, UK), and hIL-1β was from Sigma (St. Louis, MO). Compound 406 was generously provided by Professor S. Kusumoto (Osaka University, Japan). All LPS was purchased from List Biological Laboratories (Campbell, CA). Escherichia coli (O55:B5) LPSwas exclusively used for cell stimulation and tritium-labeled LPS (3H-LPS) from E. coli (K12 LCD25) was used in LPS uptake experiments. For use in fluorescence microscopy, LPS from Salmonella minnesota (Re595) was labeled with FITC (Molecular Probes) (19). Complexes of sCD14 with either LPS were preformed without LBP as described previously (30). Under these conditions, no LPS remains unbound. Complexed LPS (LPS·CD14, FITC-LPS·CD14, 3H-LPS·CD14) was prepared freshly each time and used right away.Cell cultureHCAEC and human umbilical vein endothelial cells (HUVEC) were purchased from Clonetics (San Diego, CA) and grown to confluence in full growth medium (EGM-2-MV) from the same company. HCAEC were from healthy young donors and were used for experiments from passages 4–8. There was no obvious difference in TLR2 surface expression or responsiveness to various stimuli between passage 4 and passage 8 cells. Medium was changed every day, and cells were passaged in a 1:3 ratio using trypsin/EDTA (0.025%/0.2 mM).Mouse endothelial cell cultureMouse microvascular and vascular EC were isolated from lung tissue and the aortas of C57/BL6 mice. Halothane-anesthetized animals were killed, and the lungs and the aorta were explanted under sterile conditions, minced, and digested for 1 h at 37°C in 1 mg/ml collagenase type II (Invitrogen). Cells were stained with rat anti-mouse CD31 (10 µg/ml), and goat anti-rat IgG conjugated to paramagnetic beads (Miltenyi Biotec, Auburn, CA) enabled further purification using MACS (magnetic cell sorting). Endothelial cells were then cultured and maintained on fibronectin-coated tissue culture plates in mouse brain microvascular growth medium (Cell Application Incorporation, San Diego, CA) supplemented with 15 ng/ml basic fibroblast growth factor (Sigma). Purity of the cells was >90% as determined by staining for mouse CD31, mouse CD106, and mouse CD144 and by uptake of Alexa Fluor 488-labeled AcLDL (Molecular Probes).Monocyte isolationHuman peripheral blood monocytes were purified by MACS (Miltenyi Biotec) according to the manufacturer’s protocol. Purity of the preparations and viability of the cells regularly exceeded 95% as determined by subsequent FACS analyses using FITC-anti-human CD14 and propidium iodide (2.5 µg/ml). The isolation procedure did not activate the cells and did not diminish their ability to be stimulated.Flow cytometryFor flow cytometry analysis of protein surface expression, HCAEC cultured in six-well plates were harvested with a cell scraper. Freshly prepared monocytes and HCAEC were washed twice and stained with various primary and isotype control antibodies (10 µg/ml) for 30 min at 4°C in PBS/5% FCS containing 0.1% sodium azide. After additional washing steps, cells were incubated with secondary PE-conjugated antibodies for a further 30 min at 4°C, washed and resuspended in PBS, and analyzed on a FACScan (BD Biosciences, San Diego, CA). HCAEC activation was determined by CD62E surface expression, which was quantified using PE-conjugated mouse anti-human CD62E or control PE-conjugated mouse IgG for 30 min at 4°C before FACS analyses.RT-PCRRNA was purified from LPS-stimulated or unstimulated HCAEC or monocytes using the Absolutely RNA RT-PCR Miniprep Kit from Stratagene (La Jolla, CA). mRNA levels of TLR2, TLR4, MD-2, CD14, LBP, and gp96 were determined by RT-PCR. Briefly, the first-strand cDNA was reverse-transcribed from 1 µg total RNA with random primers using the SuperScript first-strand Synthesis System from Invitrogen. The cDNA product was amplified by 30 cycles of 30 s at 94°C, 45 s at 55°C, and 2 min extension at 72°C using specific primer pairs. cDNA from monocytes or from stimulated HepG2 cells was used as control for CD14 or LBP, respectively. The RT-PCR products were separated in 1.2% (wt/vol) agarose gels and visualized with ethidium bromide.Immunofluorescence staining and fluorescence microscopyHCAEC were cultured in eight-well Nunc Lab-Tec II chambered coverglasses (Nalge Nunc, Naperville, IL) and after an incubation period of 4 h with either LPS (100 ng/ml), FITC-LPS (100 ng/ml), sCD14 (1.6 µg), LBP (2.5 µg), or preformed FITC-LPS·CD14 complexes (100 ng/ml), cells were fixed and permeabilized using the Cytofix/Cytoperm kit from PharMingen (San Diego, CA) according to the manufacturer’s protocol. Nonspecific binding sites were blocked with 0.5% BSA for 30 min at room temperature, and thereafter the following primary antibodies were added at the indicated concentration for 1 h at room temperature: H80 (20 µg/ml), HTA125 (10 µg/ml), 2B5 (10 µg/ml), anti-hCD14 (10 µg/ml), anti-hCD144 (10 µg/ml), anti-hVCAM-1 (10 µg/ml). Fluorescent wheat germ agglutinin (Oregon Green 488; Molecular Probes) was used for Golgi staining, and fluorescent phallotoxin was used for F-actin staining (both at 5 µg/ml). Cells were washed, and secondary fluorescence-labeled antibodies were added at 5 µg/ml for 1 h at room temperature. Nuclei were counterstained with DAPI (300 nM) for 3 min before cells were extensively washed and treated with SlowFade Light antifade reagent (Molecular Probes). Monocytes were stained in suspension. Thereafter, they were mounted to a cover glass with ProLong (Molecular Probes) mounting medium, thus maintaining their round shape. Images were acquired using the DeltaVision Optical Sectioning Microscope (Model 283). Following data acquisition using the Photometrics CH350L liquid-cooled CCD camera, which collects low-intensity signals with a linear response to light intensity, the data are then deconvoluted using DeltaVision software SoftWoRx 2.5 based on the Agard/Sadat inverse matrix algorithm. Following computational deconvolution, this system can provide high-resolution 3D images of cells, but each single optical section can still be viewed alone.Cell stimulation experimentsHCAEC and HUVEC were cultured in six-well plates, and after reaching confluence, EGM-2-MV was changed to DMEM/2% FCS or DMEM/2% depleted FCS at least 6 h before an experiment was started. Antibodies were added 30 min before cells were stimulated for 4 h at 37°C with LPS, LPS·CD14, or MALP-2 at various concentrations. Thereafter, the cells were harvested and stained for flow cytometry analyses. Human peripheral blood monocytes were resuspended in DMEM/2% FCS and seeded into 24-well tissue culture plates at 1 × 104cells/well. After addition of antibodies for 30 min, cells were stimulated with either LPS or MALP-2 (1 ng/ml) for 4 h at 37°C. Supernatants were harvested, and hIL-8 was measured with an ELISA (Biosource, Camarillo, CA). Mouse EC were stimulated with LPS, MALP-2, or cytokines for 12 h in culture medium containing 5% FCS. Thereafter, medium was measured for mouse macrophage inflammatory protein-2 (MIP-2), which is the murine homologue of human IL-8, by ELISA (R&D Systems, Minneapolis, MN). For stimulation of HCAEC with low LPS concentrations or with preformed LPS·CD14 complexes, it was necessary to deplete serum of sCD14 or LBP using anti-CD14 (mAb 28C5) and anti-LBP (mAb 2B5) as described previously (21, 23). Depleted FCS contained <1 ng/ml of remaining antibodies (measured by ELISA). Both CD14 and LBP were below the detection limit of Western blotting (1 ng of protein in our experiments).LPS uptake experimentsHCAEC, or mouse EC, were grown to confluence in six-well tissue culture plates, and medium was changed to DMEM/2% FCS before experiments. 3H-LPS (100 ng/ml) or 3H-LPS·CD14 (100 ng/ml) was incubated for 2 h with the cells of one well along with various antibodies, sCD14, and LBP. Thereafter, the adherent cells were washed twice with PBS and further treated with 250 µg/ml proteinase K (Sigma) in HBSS for 30 min at room temperature to remove cell surface proteins/receptors (22). The 3H counts remaining after proteinase K treatment were therefore considered to be intracellular. This treatment also detached the cells, which were harvested, washed twice in PBS, and lysed in 300 µl 2% SDS/50 mM EDTA. Liquid samples were transferred to scintillation vials and assayed for counts in Ecoscint scintillation fluid (National Diagnostics, Atlanta, GA). The same procedure was carried out in experiments where 32P-labeled LBP (250 ng/ml) uptake was investigated.Transient transfectionOne day after seeding into six-well plates, HCAEC (5×105 cells/well) were transiently transfected with cDNA for CD14 or TLR4 in pFLAG-CMV.1 (Sigma) using 6 µl/well Lipofectin Reagent (Invitrogen) and 1.6 µg/well DNA in 100µl Opti-MEM I (Gibco BRL, Gaithersburg, MD). After 3 h under serum-free conditions, EGM-2-MV was added and cells were analyzed the following day for surface expression of CD14 or TLR4. In several experiments, transfection efficiency was typically 5–10%.StatisticsData are expressed as mean and standard error of the mean (SE). Means were compared by Kruskal-Wallis ANOVA and by Mann Whitney U test. A difference with P < 0.05 was considered significant. Statistical analyses were calculated using the StatView software package (Abacus Concepts, Berkeley, CA).RESULTSFACS analyses of HCAEC do not detect surface TLR4To investigate cell surface expression of TLR2 and TLR4 protein, we performed several FACS analyses on monocytes and HCAEC and compared the results with the TLR2 and TLR4 mRNAexpression levels (Fig. 1). Nonpermeabilized HCAEC or monocytes were stained with 5 different anti-TLR4 (monoclonal HTA1216, HTA405, HTA414, HTA125, and polyclonal H80), anti-TLR2 (mAb 2392), or anti-CD14 (M5E2) primary antibodies. Dot blot assays with immobilized primary antibody confirmed that the secondary antibodies used were able to detect their primary partners (not shown). In contrast to human peripheral blood monocytes, which showed clear surface staining for TLR2, TLR4, and CD14, we could not detect TLR4 or CD14 on the surface of HCAEC, although the TLR4 mRNA expression was similar in monocytes and HCAEC. At an expression level of TLR2 mRNA (after induction with LPS-IFNγ or TNF-IFNγ) comparable to basal TLR4 mRNA levels, 92–98% of HCAEC stained positive for surface TLR2; even in unstimulated HCAEC, where almost no TLR2 mRNA was found, FACS still was able to detect surface TLR2. Although LPS-IFNγ enhanced TLR4 mRNA expression in HCAEC, TLR4 was still not detected on the surface. Figure 1 (HCAEC) shows a representative result of many experiments; in this experiment, HTA125 was used for TLR4 and anti-TLR2 2392 for TLR2 detection. Both antibodies worked very well in monocytes (see Fig. 1, Monocytes). However, upon transient transfection and overexpression of TLR4 or CD14 in HCAEC, some surface expression (~1% of cells) was seen in FACS analyses (see Fig. 1, HCAEC transfected), establishing that the detection system works in HCAEC.HCAEC express RNA for TLR2, TLR4, MD-2, and gp96, but not CD14 or LBPTo assess mRNA expression in HCAEC, RT-PCR analyses was performed. Agarose gel electrophoresis revealed clear bands at the expected fragment sizes for TLR2, TLR4, and MD-2. Stimulation of the HCAEC for 12 h with 10 ng/ml of E. coli LPS (O55:B5) did not significantly change TLR4 or MD-2 mRNA expression, but enhanced TLR2 mRNA. More important, neither stimulated nor unstimulated HCAEC were positive for LBP or CD14. As expected, mRNA for the chaperon protein gp96 was found in three different HCAEC preparations and in monocytes (Fig. 2).TLR4 is intracellular in HCAEC and on the surface of monocytesExpression of TLR4 mRNA in the absence of cell surface TLR4 led us to search for intracellular TLR4 in HCAEC by 3D-deconvolution confocal microscopy. In HCAEC, which are nontransfected cells, TLR4 was found exclusively around the nucleus (Fig. 3A–D). After stimulation of the cells, VCAM-1 was found intracellularly and surface expressed (Fig. 3C). Nonimmune rabbit IgG was used as control and revealed uniform background staining (not shown). In monocytes, use of the same anti-TLR4 antibody (H80), resulted in bright surface staining and showed colocalization of TLR4 to membrane CD14 (Fig. 3E).Anti-TLR4 antibodies do not block HCAEC response to LPSHCAEC activation was determined by measuring CD62E surface expression (Fig. 4). Preliminary experiments showed that CD62E is minimally expressed on resting cells, but is rapidly induced and highly expressed upon stimulation. HCAEC were incubated with various antibodies 30 min before stimulation with LPS (10 ng/ml) for 4 h. All anti-TLR4 antibodies used failed to inhibit LPS-induced activation of HCAEC. Even long-term preincubation (up to 12 h) of HCAEC with anti-TLR4 was not able to block LPS activation (not shown). This is in contrast to monocytes, where HTA405 or HTA125 significantly, but incompletely, inhibited LPS-induced IL-8 secretion. The effects of HTA405 on HCAEC or monocyte activation by LPS werefurther explored at a wide range of LPS and mAb concentrations (see Table 1). In no case werethe results qualitatively different from Figure 4. As anticipated from published work, the abilityof LPS to stimulate cells was abolished by anti-LBP (mAb 2B5) or anti-CD14 (mAb 28C5) inendothelial cells as well as in monocytes. Anti-TLR2 (mAb 2392) completely blocked MALP-2-induced cell activation in both cell types (Fig. 4). The fact that anti-TLR2 did not inhibit LPS-induced cell activation excludes the possibility that TLR2 agonist impurities in the LPSpreparation were responsible for activation of the cells. Similarly, lack of an effect of 2B5 onMALP-2-initiated activation suggests that there is not an LPS contaminant in MALP-2. In contrast to anti-TLR4, Compound 406 (lipid 4A) dose-dependently (0.1–100 µg/ml) inhibitedLPS-induced HCAEC activation (not shown).TLR4 is required for LPS signaling but not for LPS uptake in ECWe cultured pulmonary microvascular and aortic EC derived from TLR4 –/– C57/BL6 mice andcompared their response to LPS and other proinflammatory stimuli with cells from wild-typeanimals (Fig. 5). Compared with cultured HCAEC, the mouse cells did not respond well withCD62E expression to any stimulus, and therefore we measured MIP-2 secreted into the medium.Even after 12 h incubation with 10 ng/ml of E. c oli LPS (O55:B5), the TLR4 –/– cells were stillcompletely unresponsive to this stimulus, whereas the response to murine TNF (10 ng/ml),MALP-2 (100 ng/ml), or human IL-1 (10 ng/ml) was independent of TLR4 genotype. The wild-type cells were activated by all stimuli. Results shown (Fig. 5A) are from experiments withmurine aortic EC; lung microvascular EC responded similarly (data not shown). When TLR4 –/–cells were incubated with 3H-LPS (100 ng/ml) for 2 h with various concentrations of LBP and 3H-LPS uptake was measured, no difference was found compared with wild-type cells, suggesting that uptake occurs independently of TLR4 phenotype (Fig. 5B). Moreover, mouse ECLPS uptake mimicked the LPS uptake observed in human cells (HCAEC).LBP facilitates the uptake of LPS and LPS·CD14 complexes into HCAECBecause previous data show that sCD14 and LBP are required for LPS-induced HCAECactivation, we further investigated the role of these proteins in LPS uptake. HCAEC wereincubated for 2 h with 100 ng/ml of 3H-LPS along with an increasing amount of human LBP(Fig. 6A). LBP dose-dependently stimulated 3H-LPS uptake into cells. The maximum of theeffect was seen close to equimolarity of LPS with LBP and resulted in a ~10-fold increase in theamount of LPS transported into the cells. Because LPS·CD14 complexes are necessary for cellactivation, we were curious whether LBP would enhance uptake of complexes preformedwithout LBP, and found that LBP did enhance 3H-LPS·CD14 complex uptake ~5-fold. LBP-enhanced LPS and LPS·CD14 complex uptake was sensitive to an anti-LBP antibody (mAb2B5), but was never blocked completely and a low basal uptake remained. This suggests thatLBP is not absolutely necessary for the uptake of LPS and LPS·CD14 complexes. This basaluptake was insensitive to either anti-TLR4 (mAb HTA405), anti-LBP (mAb 2B5), or anti-CD14(mAb 28C5) (data not shown).Similar patterns of uptake are seen when LPS, sCD14, and LBP were added simultaneously andtherefore CD14 complexation of LPS was catalyzed by LBP (Fig. 6B). Thus LPS in the absenceof LBP, alone or in the presence of sCD14, is only minimally internalized as compared with LPSin the presence of LBP. When LBP catalyzed complexation of LPS to sCD14 is inhibited by anti-CD14 (mAb 28C5), uptake follows the LPS-LBP pattern (10-fold increase). When complexation of LPS to either sCD14 or LBP is inhibited by anti-LBP (mAb 2B5), the effects of both LBP and CD14 are abolished. Finally, data obtained with 32P-LBP (Fig. 6C) show that LBP is internalized with LPS and that the uptake follows the same pattern as observed when LPS uptake was measured.LPS, LBP, and CD14 all colocalize with intracellular TLR4In fact, LPS complexes are found intracellularly. HCAEC were incubated for 4 h with FITC-LPS (100 ng/ml) alone or with sCD14 (1.6 µg/ml), LBP (2.5 µg/ml), or both. Thereafter, cells were stained for TLR4, LBP, or CD14 (Fig. 7). Confocal microscopy showed clear colocalization of FITC-LPS and LBP (Fig. 7A), FITC-LPS and intracellular TLR4 (Fig. 7B), LBP and TLR4 in the presence of sCD14 (Fig. 7C), and finally sCD14 and TLR4 in the presence of LBP (Fig. 7D). Under serum-free conditions (without sCD14), where no cell activation occurs, no colocalization of either FITC-LPS (Fig. 7E) or LBP (Fig. 7F) with intracellular TLR4 was observed. Colocalization of LBP and CD14 could not be shown because both are detected by mouse antibodies and these could not be distinguished by labeled second antibodies. LBP or CD14 were never detected intracellularly when added without LPS; instead, large aggregates were found in the microscopic field outside the cells. When HCAEC were stimulated for 2 h with LPS (10 ng/ml), the intracellular adaptor molecule MyD88, which is involved in TLR4 signaling, was recruited to a perinuclear region (Fig. 7G). This was not the case in the absence of sCD14 (Fig. 7H).LBP enhances HCAEC activation at low LPS concentrations via facilitated LPS·CD14 uptakeBecause the preceding experiments revealed a role for LBP in LPS uptake, we were curious whether this would have an impact on cell activation. To exclude additional LPS complex formation during the experiments, CD14-depleted FCS was used for experiments with preformed LPS·CD14 complexes and, because we wanted to see effects of added recombinant LBP, LBP-depleted FCS was used for experiments with uncomplexed LPS. Neither CD14 nor LBP at the concentrations used in our experiments activated cells without LPS.Figure 8A shows that 10 ng/ml of LPS with 250 ng/ml LBP enabled a full response of the cells and sCD14 (160 ng/ml) showed no additional effect. When LPS and sCD14 were added without LBP under these conditions, a minimal stimulation was observed, probably due to incomplete LBP depletion of the serum. Stimulation under each condition was blocked by an anti-LBP antibody, thus confirming the necessity of LBP for LPS·CD14 complex formation during the 4 h course of these experiments.In contrast, 10 ng/ml of preformed LPS·CD14 complexes stimulated the cells without any effect of anti-LBP or additional LBP (Fig. 8C), suggesting that LBP is not absolutely required (Fig. 6A) for sufficient uptake of preformed LPS·CD14 complexes at 10 ng/ml. However, uptake did occur, because a blocking anti-TLR4 (mAb HTA405), which would block surface TLR4, failed to inhibit activation even when added together with anti-LBP (mAb 2B5).At a low LPS concentration (0.1 ng/ml) (Fig. 8B), addition of sCD14 (100 ng/ml) did not stimulate cells. Providing LBP (200 ng/ml) enabled a response of the cells, which was further enhanced by additional sCD14. Activation was again blocked by anti-LBP (mAb 2B5), which would also inhibit rapid LBP catalyzed formation of stimulatory LPS·CD14 complexes. Preformed LPS·CD14 complexes at 0.1 ng/ml (Fig. 8D) did not stimulate cells in CD14-depleted FCS. When additional LBP was provided, cell stimulation was observed, which cannot be explained by LBP catalyzed LPS·sCD14 complex formation, because preformed complexes were added in CD14 depleted medium. Again, stimulation was insensitive to anti-TLR4 (mAb HTA405) and therefore mediated by intracellular TLR4.Thus, at 0.1 ng/ml of LPS, LBP enhances the uptake of LPS·CD14 complexes, preformed in vitro (Fig. 8D) or formed by LBP-mediated catalysis (Fig. 8B), and this LBP-enhanced uptake results in HCAEC activation.LBP-stimulated LPS uptake into HCAEC involves a scavenger receptor pathwaySome scavenger receptors have been shown to be lipid A or LPS uptake receptors (31–33). Because HCAEC were found to be CD14 negative, we were curious whether scavenger receptor blocking agents would affect the LPS uptake in these cells. HCAEC were pretreated with dextran sulfate (MW 8000), polyinosinic acid (PIA), polyadenylic acid (PAA), or polycytidylic acid (PCA) for 30 min before measurement of 3H-LPS uptake with or without additional LBP. Both dextran and PIA reduced the LBP-enhanced 3H-LPS uptake to the level of basal 3H-LPS uptake, whereas the polycationic substances PAA and PCA lacked this effect (Fig. 9). Unfortunately, both dextran sulfate and PIA activated HCAEC to express surface CD62E even in the absence of added LPS (data not shown).DISCUSSIONTo begin a study of regulation and modification of TLR2 and TLR4 expression in HCAEC under various conditions, we performed FACS analyses on those cells, but despite the use of several different anti-TLR4 antibodies, we never observed positive surface staining for TLR4. Nevertheless, the cells responded well to both TLR2 and TLR4 agonists, and mRNA for both TLR2 and TLR4 was expressed. TLR2 mRNA was only weakly detectable in resting cells, but the TLR2 protein was still found on the HCAEC surface by FACS, confirming the high sensitivity of this method. After stimulation of the cells, TLR2 mRNA levels corresponded to TLR2 surface expression. However, this was not observed for TLR4. Despite high expression of TLR4 mRNA in both monocytes and HCAEC, TLR4 protein was never seen on the surface of HCAEC. The reliability of the detection system was confirmed in monocytes, which stained well for TLR2 and TLR4 on the surface, and in transfected HCAEC, where some TLR4 surface protein could be detected by FACS after transfection and overexpression of human TLR4.With the knowledge that TLR4 has been shown to reside in the Golgi apparatus in murine intestinal epithelial cells (28) and that LPS is transported to the Golgi in human epithelial HeLa cells (34), we further investigated HCAEC using confocal microscopy and found TLR4 located in a perinuclear region but not on the surface, where VCAM-1 was observed after cell stimulation. This is in contrast to monocytes, where TLR4 was clearly seen to colocalize with。