用户名: 密码: 验证码:
oxLDL诱导小鼠足细胞脂质蓄积和CXCL16表达以及辛伐他汀干预的研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
研究背景和目的:
     自1982年,Moorhead等首次提出“脂质肾毒性”学说以来,大量的研究结果表明,高脂血症和脂质在肾脏沉积是发生肾小球硬化的重要危险因素,而肾小球硬化的病理生理和组织学变化,如血脂异常、病变早期表现内皮损害、后期出现系膜细胞增殖及细胞外基质积聚,这些机制与动脉粥样硬化的发病机制相似,因此提出了“肾小球动脉硬化”这一概念。近年来,氧化低密度脂蛋白(oxLDL)在肾小球硬化及间质纤维化中的作用越来越受到关注,成为研究热点。临床和实验研究表明,高脂血症可以使oxLDL在肾脏沉积、炎症细胞浸润、肾脏固有细胞增生和损伤、细胞外基质积聚及泡沫细胞形成,直接或间接地导致肾小球硬化。既往对脂质在肾脏沉积导致肾损害的研究,多集中在肾脏系膜细胞及内皮细胞,而Joles的实验证明oxLDL可以同时对肾脏系膜细胞、内皮细胞和足细胞造成损伤,在诸多损伤中足细胞可能是最主要的受害者。
     足细胞是一种高度分化的肾脏固有细胞,在体内足细胞伸出足突包绕在肾小球基底膜(GBM)上,形成裂孔隔膜(slit diaphragm,SD)复合体,这一结构在维持肾小球滤过屏障中起重要作用。足细胞损伤是肾病蛋白尿形成的重要原因之一,在肾小球硬化的发生、发展中起重要作用。Bussolati等研究发现oxLDL能够介导足细胞SD蛋白nephrin表达缺失而致细胞损伤,但有关oxLDL介导足细胞损伤的具体调控机制,尚未得到深入的研究,因此建立可靠的oxLDL诱导肾小球足细胞脂质蓄积模型,探索影响足细胞摄取oxLDL的机制,对于减轻足细胞损伤以及延缓肾小球硬化而言,可能是一种极具前景的治疗策略。
     Gutwein等的体内和体外实验证明在人肾组织足细胞上存在CXC趋化因子配体16(CXC chemokine ligand16,CXCL16)的表达。CXCL16在体内有两种存在形式:膜结合型CXCL16和可溶型CXCL16。跨膜CXCL16的一个重要作用是oxLDL的清道夫受体。oxLDL通过细胞膜上的清道夫受体介导进入细胞,造成细胞内脂质大量积聚,导致泡沫细胞形成。金属蛋白水解酶10(a disintegrin and metalloprotease10, ADAM-10)是两种形式转化的关键因素,参与诱导CXCL16从细胞膜的脱落。新近的研究认为,炎性细胞因子如干扰素-γ (INF-γ)和肿瘤坏死因子-a (TNF-a)可以增加CXCL16在人足细胞内以及可溶形式的表达。有关跨模型CXCL16是否参与体外培养鼠源性肾小球足细胞oxLDL摄取,以及干扰素-γ和ADAM10抑制剂可能的调控作用,国内外研究甚少。
     他汀类药物是近年来发现的有效的调脂药。大量实验和临床研究资料显示,该类药具有显著的降脂作用。他汀类药物在体内主要的作用是通过选择性阻断胆固醇合成的限速酶结合位点,影响胆固醇生物合成,同时代偿性促进低密度脂蛋白(LDL)受体合成,加速LDL降解,从而降低血脂。目前研究发现,他汀类药物不仅有依赖降血脂的肾脏保护作用,而且还有抗细胞增殖、抗炎症、免疫调节及抑制清道夫受体表达等非依赖降血脂的肾脏保护作用,但确切机制尚不明确。辛伐他汀是最有效、应用最为广泛的一种他汀类药物,目前关于辛伐他汀对清道夫受体介导的鼠源肾小球足细胞脂质蓄积的影响尚未见报道。
     本研究首次采用油红染色观察小鼠肾小球条件性永生足细胞对oxLDL摄取情况,探讨足细胞摄取oxLDL与CXCL16蛋白表达的关系,旨在建立oxLDL诱导足细胞脂质蓄积模型,为研究足细胞摄取oxLDL的机制及其可能存在的调控因素提供可靠的实验基础。通过体外观察INF-γ、CXCL16特异性抗体和ADAM10抑制剂对足细胞摄取oxLDL的影响以及CXCL16蛋白表达的变化,初步探讨可能调控足细胞脂质氧化损伤的因素。此外,通过观察辛伐他汀对oxLDL诱导鼠源肾小球足细胞脂质蓄积及CXCL16、nephrin蛋白表达的影响,探讨辛伐他汀对足细胞的保护作用机制。
     第一部分oxLDL诱导小鼠足细胞脂质蓄积模型的建立
     目的:观察oxLDL诱导小鼠肾小球条件永生性足细胞脂质蓄积情况,旨在建立oxLDL诱导足细胞脂质蓄积模型,为研究足细胞摄取oxLDL的机制及其可能存在的调控因素提供可靠的实验基础。
     方法:采用体外培养的小鼠肾小球条件永生性足细胞株(MPC5)作为研究对象,在37℃无干扰素-γ的1640完全培养基培养10-14d至其分化成熟后,分别加入梯度浓度为20,40,80,160μg/ml的oxLDL,孵育24h及48h后,油红0染色观察细胞内脂质蓄积情况;比色法检测细胞内总胆固醇浓度。结果:油红0染色可见:与对照组相比,20、40μg/ml oxLDL组作用24h及48h,细胞中均未发现明显脂滴;80、160μg/ml oxLDL组作用24h有少许脂滴出现,作用48h均发现明显脂滴。
     细胞内总胆固醇浓度定量分析结果显示,不同浓度oxLDL孵育足细胞24h脂质蓄积不明显,细胞内总胆固醇浓度分别为97.5±29.6μ g/ml、101.3±36.4μ g/ml、110.6±35.1μ g/m1、121.5±26.4u g/ml,与对照组组(90.3±30.1u g/m1)相比,各组细胞内总胆固醇浓度均无显著性差异(P均>0.05)。不同浓度oxLDL孵育足细胞48h,与对照组(102.6±33.5μ g/m1)相比,低浓度oxLDL(20、40μ g/m1)组无明显变化,细胞内总胆固醇浓度分别为98.4±31.2μg/ml、103.6±31.9μg/m1(P>0.05);高浓度oxLDL(80及160μg/m1)组,细胞内总胆固醇浓度显著增高,分别为201.0±20.3u g/ml、278.0±35.1μg/m1(P<0.05,P<0.01);且高浓度oxLDL组孵育48h与24h比较,随着孵育时间延长,细胞内总胆固醇浓度显著增高(P<0.05,P<0.01)。
     结论:首次用油红染色观察足细胞脂质蓄积,方法简便实用。oxLDL诱导鼠源肾小球足细胞脂质蓄积在一定范围内呈剂量依赖性和时间依赖性。80μg/ml oxLDL诱导体外培养小鼠肾小球条件永生足细胞株48h,油红染色可见显著脂质沉积,细胞内胆固醇含量显著增高,成功构建脂质蓄积模型。
     第二部分不同刺激因素对oxLDL诱导足细胞脂质蓄积和CXCL16表达的影响
     目的:观察重组小鼠细胞因子IFN-γ、 CXCL16单克隆抗体和ADAM10抑制剂对体外培养的小鼠肾小球足细胞摄取oxLDL以及足细胞CXCL16蛋白表达的影响,初步探讨足细胞脂质蓄积的影响因素和调控机制。
     方法:体外培养小鼠肾小球条件永生性足细胞(MPC5),在37℃无干扰素-γ的1640完全培养基培养10~14d至其分化成熟后,分别加不同处理因素:1.oxLDL(80ug/ml)与不同浓度干扰素-γ(5.0,10.0,20.OU/ml)共同孵育细胞48h和oxLDL(80ug/ml)与干扰素-γ(20.0U/ml)共同孵育细胞24h及48h;2.oxLDL(80ug/ml)与梯度浓度CXCL16单克隆抗体(0.625,1.25,2.5,5.0,10.0ug/ml)共同孵育细胞48h和oxLDL(80ug/ml)与CXCL16抗体(5.0ug/ml)共同孵育细胞24h及48h;3.oxLDL(80ug/ml)与梯度浓度ADMA10抑制剂(0.125,0.25,0.5,1.0,2.0ug/ml)共同孵育细胞48h。另设Ctrl组(不给药),oxLDL组(只加80ug/ml oxLDL)。油红染色观察足细胞脂质蓄积情况,比色法检测足细胞内总胆固醇浓度,Western blot检测足细胞CXCL16的蛋白表达。
     结果:1.干扰素-γ对oxLDL诱导足细胞脂质蓄积及CXCL16蛋白表达的影响1.1干扰素-γ对oxLDL诱导足细胞脂质蓄积的影响:油红染色结果显示,oxLDL组及oxLDL与各浓度IFN-γ共同孵育组均出现大量脂滴。与Ctrl组相比,oxLDL+各浓度IFN-γ共同孵育组,细胞内总胆固醇含量显著增加,分别为209.2±44.7ug/ml、211.9±22.6ug/ml、273.8±27.1ug/ml(P<0.05;P<0.01);与oxLDL组比较,oxLDL+IFN-γ(20U/ml)组细胞内总胆固醇含量显著增加(P<0.05)。用oxLDL(80μg/ml)与IFN-γ(20U/ml)共同孵育足细胞24h及48h,细胞内总胆固醇含量分别为174.9±22.1ug/ml及275.3±12.4ug/ml,结果显示随着作用时间延长,细胞内总胆固醇含量显著增加(P<0.01)。此外,oxLDL与IFN-γ共同孵育组,细胞内总胆固醇含量均高于同期oxLDL组(P<0.05)。1.2干扰素-γ对足细胞内CXCL16蛋白表达的影响:Western blot定量分析结果显示,oxLDL组及oxLDL+各浓度IFN-γ孵育组足细胞内CXCL16蛋白表达显著高于Ctrl组(P<0.05);与oxLDL组比较,oxLDL+IFN-γ(20U/ml)组CXCL16蛋白表达显著增加(P<0.05)。时效性研究发现,oxLDL组CXCL16的蛋白表达在孵育48h与24h比较无明显变化,不随时间延长而增加;而oxLDL与IFN-γ(20U/ml)共同孵育组,细胞内CXCL16的蛋白水平在48h与24h比较,随作用时间延长而显著增加(P<0.05)。
     2.CXCL16单克隆抗体对oxLDL诱导足细胞脂质蓄积及CXCL16蛋白表达的影响
     2.1CXCL16抗体对足细胞内脂质蓄积的影响:油红染色结果显示,与oxLDL组相比,oxLDL与低浓度(0.625μg/ml、1.25μg/ml) CXCL16抗体共同孵育48h油红染色无明显变化;而oxLDL与高浓度(2.5、5.0和10.0μg/ml) CXCL16抗体共同孵育48h,细胞内脂滴均明显减少。细胞内胆固醇检测结果显示,与oxLDL组比较,oxLDL与低浓度(0.625μ g/m1、1.25μ g/m1)CXCL16抗体共同孵育48h,细胞内总胆固醇含量分别为203.8±24.8u g/ml及174.1±15.1μ g/ml,差异无统计学意义(P>0.05);oxLDL与高浓度(2.5、5.0和10.0μ g/m1)CXCL16抗体共同孵育48h组,细胞内总胆固醇含量呈剂量依赖性降低,分别为147.2±15.1ug/ml、100.7±11.4ug/ml及84.1±9.1ug/ml(P<0.05;P<0.01)。
     2.2CXCL16抗体对足细胞内CXCL16蛋白表达的影响:Western blot定量检测结果显示,足细胞内CXCL16蛋白表达水平随CXCL16抗体浓度增加呈剂量依赖性降低。与oxLDL组比较,oxLDL与低浓度(0.625μg/ml、1.25μg/ml) CXCL16抗体共同孵育48h组,CXCL16蛋白的表达无显著变化;而oxLDL与高浓度(2.5、5.0和10.0μg/ml) CXCL16抗体共同孵育48h组,细胞内CXCL16蛋白的表达显著降低(P<0.05;P<0.01)。以oxLDL(80μg/ml)与同一浓度(5.0μ g/m1)CXCL16抗体共同孵育足细胞24h及48h后,与oxLDL组相比,oxLDL与CXCL16抗体共同孵育组足细胞内CXCL16蛋白表达均显著降低(P<0.05)。
     3. ADAM10抑制剂对oxLDL诱导足细胞脂质蓄积和CXCL16表达的影响
     3.1ADAM10抑制剂对细胞内脂质蓄积的影响:油红染色结果显示,与oxLDL组相比,oxLDL分别与0.125μg/m1及0.25μg/mlADAM10抑制剂共同孵育48h,细胞内脂滴无显著变化;oxLDL分别与0.5μ g/ml,1.0μ g/m1和2.0ug/mlADAM10抑制剂共同孵育48h,细胞内脂滴显著增加。比色法结果显示,与oxLDL组比较,oxLDL与0.125μ g/ml及0.25μg/mlADAM10抑制剂共同孵育48h组,细胞内总胆固醇的含量略增高,分别为211.6±12.6μ g/m1及227.8±18.3μ g/ml,但差异无统计学意义(P>0.05);oxLDL与0.5μ g/ml,1.0μ g/ml和2.0u g/mlADAM10抑制剂共同孵育48h组,细胞内总胆固醇的含量显著增加(P<0.05;P<0.01),分别为258.5±26.8u g/ml、284.0±23.2μg/ml、338.3±26.6μ g/ml。
     3.2ADAM10抑制剂对细胞内CXCL16蛋白表达的影响:Western blot定量分析结果显示,与oxLDL组比较,oxLDL与0.125μg/ml及0.25μg/mlADAM10抑制剂共同孵育组,足细胞内CXCL16蛋白表达略增加,但差异无统计学意义(P>0.05);oxLDL与0.5μg/ml、1.0μg/ml和2.0μg/mlADAM10抑制剂共同孵育组,足细胞内CXCL16蛋白表达显著增加(P<0.05;P<0.01)。结论:1oxLDL诱导体外培养鼠源肾小球足细胞内CXCL16蛋白表达显著增加,提示oxLDL可能上调足细胞内清道夫受体CXCL16的数量并被过度摄取,导致细胞内胆固醇聚集。
     2在体外培养条件下,oxLDL与一定浓度的IFN-γ共同孵育足细胞,可使细胞内脂滴增多,总胆固醇含量增高,同时发现足细胞内CXCL16蛋白表达显著增加。提示干扰素-Y通过诱导鼠源肾小球足细胞CXCL16表达上调,促进足细胞对oxLDL的摄取。
     3在体外培养条件下,oxLDL与一定浓度的CXCL16抗体共同孵育足细胞,可见细胞内脂滴明显减少,总胆固醇含量显著降低,同时发现足细胞内CXCL16蛋白水平显著降低。提示应用CXCL16抗体阻断足细胞CXCL16蛋白表达,可显著降低足细胞对oxLDL的摄取及脂质蓄积。
     4在体外培养条件下,oxLDL与一定浓度的ADAM10抑制剂共同孵育足细胞,可见细胞内脂滴显著增多,总胆固醇含量显著增加,同时发现足细胞内CXCL16蛋白水平显著增加。提示ADAM10抑制剂可显著增加足细胞CXCL16表达,从而促进足细胞对oxLDL的摄取。
     第三部分辛伐他汀对oxLDL诱导鼠源肾小球足细胞脂质蓄积干预机制的探讨
     目的:观察辛伐他汀干预前后oxLDL诱导鼠源肾小球足细胞脂质蓄积及CXCL16、 nephrin蛋白表达的变化,探讨其对小鼠肾小球足细胞的保护机制。
     方法:用80μg/ml oxLDL与不同浓度的辛伐他汀(1.0μg/ml、2.0μg/ml)共同孵育小鼠肾小球条件永生性足细胞48h,另设空白对照组(不给药),oxLDL组(只加80μg/ml oxLDL),辛伐他汀组(只加1.0μg/ml辛伐他汀)。油红0染色观察细胞内脂质蓄积情况,比色法检测细胞总胆固醇浓度,Western blot检测足细胞CXCL16/nephrin蛋白的表达。
     结果:油红0染色可见,与oxLDL组比较,oxLDL+辛伐他汀各组细胞内脂滴明显减少。比色法检测发现,对照组细胞内总胆固醇含量为90.3±30.1μ g/ml,辛伐他汀组为93.3±28.6μg/ml,oxLDL组为226.5±21.6u g/ml,oxLDL+辛伐他汀组分别为151.8±6.8μ g/ml和135.5±26.9μ g/ml。与对照组比较,辛伐他汀组细胞总胆固醇含量无显著变化(P>0.05);与oxLDL组比较,oxLDL+辛伐他汀各组细胞总胆固醇含量显著降低(P<0.05)。Western blot检测足细胞CXCL16蛋白的表达水平,与对照组比较,辛伐他汀组CXCL16蛋白表达无改变(P>0.05);与oxLDL组比较,oxLDL+辛伐他汀各组CXCL16表达显著降低(P<0.05)。nephrin蛋白的表达检测结果显示,与对照组比较,辛伐他汀组nephrin蛋白表达无改变(P>0.05),oxLD组nephrin表达显著降低(P<0.01);与oxLDL组比较,oxLDL+辛伐他汀各组nephrin表达水平显著增加(P<0.05)。结论:1.经辛伐他汀和oxLDL共同孵育的鼠源足细胞内脂滴明显减少,总胆固醇浓度显著降低,足细胞CXCL16蛋白表达水平亦显著降低。提示辛伐他汀下调oxLDL诱导的鼠源肾小球足细胞CXCL16蛋白表达,减少足细胞吞噬脂质,从而减轻足细胞内脂质蓄积。
     2.oxLDL诱导鼠源足细胞nephrin蛋白表达显著下降,经辛伐他汀和oxLDL共同孵育的足细胞内nephrin蛋白表达显著增加,提示oxLDL诱导鼠源足细胞nephrin蛋白表达缺失而致足细胞损伤,辛伐他汀能够上调oxLDL诱导的鼠源肾小球足细胞nephrin蛋白表达,对于稳定足细胞功能意义重大。
     创新和意义:
     1.首次用油红0染色直观地观察oxLDL诱导鼠源肾小球足细胞内脂质蓄积情况,方法简便实用、经济,可重复性强。oxLDL诱导足细胞脂质蓄积模型的建立,为研究足细胞摄取oxLDL的机制及其调控因素提供可靠的实验基础。
     2.通过体外细胞培养,证实CXCL16是介导足细胞脂质损伤的关键分子,其表达可被IFN-γ、ADAM10抑制剂等调控,从而影响足细胞摄取脂质,为临床早期防治足细胞脂质氧化损伤提供重要线索。
     3.首次通过体外实验证实,辛伐他汀干预能够下调oxLDL诱导的鼠源肾小球足细胞CXCL16蛋白表达,减轻足细胞内脂质蓄积;同时上调足细胞nephrin蛋白表达,从而减轻足细胞损伤,初步探讨辛伐他汀对足细胞的保护作用,为临床应用他汀类药物保护肾脏提供参考依据。
Backgroud and objectives
     A lot of study results showed that hyperlipidaemia and lipid accumulation in kidneys were important risk factors caused glomerulosclerosis since Moorhead put forward the theory "lipid nephrotoxicity" for the first time in1982, and these mechanisms of pathophysiology and histological changes of glomerulosclerosis such as dyslipidemia, endothelial damage in the early stage, mesangial cells proliferation and extracellular matrix accumulation in the late stage were similar to the pathogenesis of arteriosclerosis, Therefore, the concept of "Glomerulus Arteriosclerosis" were put forward. In recent years, the effect of oxidized low-density lipoprotein (oxLDL) on glomerulosclerosis and tubulointerstitial fibrosis were more concerned and became research focus. Clinical data and experimental studies have demonstrated that hyperlipidemia resulted in accumulation of oxLDL in kidney, inflammatory cell infiltration, proliferation and damage of renal cells, accumulation of extracellular matrix and formation of foam cell, which directly or indirectly caused glomerular sclerosis. Previous studies about kidneys damage resulted from lipid accumulation in kidneys were more focus on renal mesangial cells and endothelial cells. Joles et al reported that oxLDL caused damage in renal mesangial cells, endothelial cells and podocytes, and podocytes might be the major victim of oxLDL insult..
     Podocytes are a kind of highly differentiated renal cells. The foot processes of podocytes wrap the glomerular basement membrane (GBM) to form slit diaphragm (SD) complex, which plays a crucial role in the glomerular filtration barrier (GFB). Damage of podocytes is one of important causes of proteinuria and also plays a key role in the pathogenesis and development of glomerular sclerosis. Bussolati et al found that oxLDL mediated expression deficiency of SD nephrin in podocytes and which resulted in cells damage. However, the regulatory mechanism of how oxLDL induces the damage of podocytes remains to be elucidated. Therefore, understanding the mechanisms of oxLDL-induced lipid accumulation and the influence factors of oxLDL intake in podocytes may provide a novel therapeutic strategy to alleviate podocytes damage and delay the development of glomerular sclerosis.
     In vitro and in vivo studies of Gutwein et al have reported the presence of CXC chemokine ligand16(CXCL16) in human renal podocytes. CXCL16exists as membrane-bound CXCL16and soluble CXCL16. One of the important roles of transmembrane CXCL16is to act as a scavenger receptor for oxLDL. OxLDL enters cells by mediating of scavenger receptor on cytomembrane, leading to intracellular lipid accumulation and formation of foam cells. A disintegrin and metalloprotease10(ADAM-10) is the key factor of transformation between the two forms, which is involved in the induced shedding of CXCL16from cell membrane. The recent studies confirm that proinflammatory cytokines like interferon-γ (INF-γ) and tumor necrosis factor-a (TNF-a) can increase the expression of cellular CXCL16and the release of its soluble form from human podocytes. To our knowledge, studies on the role of transmembrane CXCL16in oxLDL intake of podocytes in vitro and the possible regulatory effect of IFN-y and ADAM10inhibitor in this process have never been reported.
     Statins are effective lipid drugs found in recent years. A lot experiments and clinical studies show this kind of drugs has significant lipid-lowering function. The main function of Statins in vivo is to affect cholesterol biosynthesis by blocking binding site of rate-limiting enzyme of cholesterol synthesis selectively and vicariously facilitate receptor synthesis of low density lipoprotein(LDL)at the same time, accelerate degradation of LDL, thus reduce blood fat. Studies at present find that Statins have not only the function of protecting kidneys rely on reducing blood fat, but also the function of protecting kidneys not rely on reducing blood fat such as resistant to cells proliferation、anti-inflammatory, immunoregulation and inhibit expression of scavenger receptor, but the exact mechanism is not clear. Simvastatin is the most effective and widely used statins. The effect of statins on lipid accumulation in mouse glomerulus podocytes has not been reported at present.
     This study observed the situation that conditional immortal mouse glomerulus podocytes ingested oxLDL by using oil red staining for the first time, and examed the expression of CXCL16, aiming to establish the model of oxLDL-induced lipid accumulation in podocytes. Therefore it may provide credible experiment basis for studying the mechanism of lipid uptake in mouse podocytes. In vitro the effects of INF-γ、CXCL16specific antibody and AD AM10inhibitor on oxLDL's uptake as well as the variation of CXCL16expression were also explored in order to discuss initially the factors that maybe regulated lipid accumulation in podocytes. Furthermore, the effects of simvastatin on lipid accumulation and on the expression of CXCL16and nephrin were observed aiming to discuss the protective effect of simvastatin in mouse podocytes
     Part1Establishment the model of lipid accumulation in mouse podocytes induced by oxLDL
     Objectives:Observed lipid accumulation in conditional immortal mouse podocytes induced by oxLDL in order to establish the model of lipid accumulation in podocytes, which may provide credible experiment basis for studying the mechanism of oxLDL uptake and the possible factors of regulation.
     Methods:Conditional immortal podocytes of mouse glomerulus (MPC5) was cultured in RPIM1640complete medium without interferon-y at37℃for10-14days until differentiated and matured, added oxLDL of concentration gradient was20,40,80,160μg/ml respectively, incubated for24h and48h. Lipid accumulation in podocytes were assessed by oil red O staining, and measured quantitatively by Colorimetric cholesterol detection kit. Results:Oil red O staining showed:No obvious lipid droplets were found in cells treated with oxLDL at20,40μg/ml for24h and48h compared with control group; A few lipid droplets were found in cells treated with oxLDL at80,160μg/ml for24h, and markedly more lipid droplets were found in cells treated with oxLDL at80,160μg/ml for48h.
     The quantitative analysis of total cholesterol concentration in cells showed, no obvious accumulation of lipid in podocytes incubated with different concentrations of oxLDL for24h, intracellular total cholesterol concentration was respectively97.5±29.6μg/ml,101.3±36.4μg/ml,110.6±35.μg/ml,121.5±26.4μg/ml, no significant difference was found between intracellular total cholesterol concentration of each group(P>0.05)compared with control group(90.3±30.1μg/ml). Podocytes incubated with different concentrations of oxLDL for48h, no obvious changes were found in groups with low concentration oxLDL (20,40μg/ml) compared with control group (102.6±33.5μg/ml), intracellular total cholesterol concentration was respectively98.4±31.2μg/ml,103.6±31.9μg/ml (P>0.05); whereas significant difference was found in cells treated with oxLDL at80and160μg/ml for48hours and respectively was201.0±20.3μg/ml,278.0±35.1μg/ml (P<0.05, P<0.01). In addition, cells incubated with oxLDL at80and160μg/ml for48hours had significantly higher level of total cholesterol level compared with cells treated with oxLDL at corresponding concentration for24hours, respectively (P<0.05vs24hours for80μg/ml, P<0.01vs24hours for160μg/ml).
     Conclusions:Lipid accumulation had dose-dependent and time-dependent in certain range in mouse glomerulus podocytes induced by oxLDL. Oil red staining showed significant lipid accumulation meanwhile intracellular cholesterol content was increased significantly in vitro cultured conditional immortal mouse podocytes induced by80μg/ml oxLDL for48h,so successfully established the model of lipid accumulation.
     Part2The effect of different stimulating factors on lipid accumulation and CXCL16expression in mouse podocytes induced by oxLDL
     Objectives:Observed the effect of cytokine recombinant mouse IFN-y, CXCL16monoclonal antibody and ADAM10inhibitor on oxLDL uptake and CXCL16expression level in vitro cultured mouse glomerulus podocytes, aiming to determine the influencing factors and regulatory mechanism of lipid accumulation in mouse podocytes,
     Methods:Cultured conditional immortal podocytes of mouse glomerulus (MPC5) in1640complete medium without interferon-y at37℃for10-14d until differentiated and matured, respectively added different treatment factors:1. Podocytes were incubated with oxLDL (80ug/ml) and interferon-y at different concentration (5.0,10.0,20.0U/ml) for48h, and cells were incubated with oxLDL (80ug/ml) and interferon-y (20.0U/ml) for24h and48h;2. Podocytes were incubated with oxLDL (80ug/ml)and CXCL16monoclonal antibody at concentration gradient(0.625,1.25,2.5,5.0,10.0ug/ml) for48h, and cells were treated with oxLDL (80ug/ml) and CXCL16antibody (5.0ug/ml) for24h and48h;3. Podocytes were incubated with oxLDL (80ug/ml) and ADMA10inhibitor at concentration gradient (0.125,0.25,0.5,1.0,2.0ug/ml) for48h. At the same time set group Ctrl (No drugs), group oxLDL (only added80ug/ml oxLDL). Lipid accumulation in podocytes were assessed by oil red O staining, and measured quantitatively by Colorimetric cholesterol detection kit. CXCL16expression were detected by Western blot.
     Results:1. The effect of interferon-y on lipid accumulation and CXCL16expression in podocytes induced by oxLDL.
     1.1The effect of interferon-y on lipid accumulation in oxLDL-induced podocytes: The result of oil red staining showed that no visible lipid drop was observed in control cells, whereas large amount of lipid drops were found in cells treated with only oxLDL (80μg/ml) and both oxLDL and IFN-y at different concentrations. Total cholesterol assay showed significantly increased level of total cholesterol when cells were treated with both oxLDL and IFN-y at different concentrations and respectively was209.2±44.7ug/ml,211.9±22.6ug/ml,273.8±27.1ug/ml (P<0.05;P<0.01); Moreover, cells treated with oxLDL and IFN-y(20U/ml) presented with a significantly elevated total cholesterol level compared to cells treated with only oxLDL (P<0.05). Podocytes incubated with oxLDL (80μg/ml) and IFN-y(20U/ml) for24h, intracellular total cholesterol level was174.9±22.1ug/ml; After incubated for48h, intracellular total cholesterol level was275.3±12.4ug/ml, the result showed intracellular total cholesterol level was increased significantly (P<0.01) as time extended. Compared with cells treated with oxLDL only, cells treated with both oxLDL and IFN-y for24and48hours showed significantly higher level of total cholesterol (P<0.05).
     1.2The effect of interferon-γ on CXCL16expression in podocytes:the effect of IFN-y on oxLDL-induced CXCL16expression was determined by western blot. Compared with control group, CXCL16expression in oxLDL treated podocytes was significantly increased (P<0.05). When cells incubated with oxLDL and IFN-y, CXCL16expression was increased in an IFN-y concentration-dependent manner. Compared with group oxLDL, CXCL16expression was slightly increased in group oxLDL+IFN-y(5U/ml) and oxLDL+IFN-y(10U/ml), whereas CXCL16expression was significantly increased (P<0.05) in group oxLDL+IFN-y(20U/ml). In addition, the effect of incubation time of IFN-y on CXCL16expression was also investigated. Compared with24-hour incubation, CXCL16expression was increased in all cells. However, significant difference between24-hour incubation and48-hour incubation was only found in IFN-y and oxLDL+IFN-y treated cells (P<0.05), indicating that CXCL16expression was increased with incubation time when treated with IFN-y or oxLDL+IFN-γ.
     2. The effect of CXCL16monoclonal antibody on lipid accumulation and CXCL16expression in oxLDL-induced podocytes.
     2.1The effect of CXCL16antibody on lipid accumulation:The result of oil red staining showed no marked change was found in cells treated with oxLDL+anti-CXCL16monoclonal antibodies (0.625, and1.25p,g/ml) compared with cells treated with only oxLDL. However, lipid accumulation was remarkably decreased in cells treated with oxLDL+anti-CXCL16monoclonal antibodies at2.5,5.0and10μg/ml. Meanwhile, compared with cells treated with only oxLDL, cells treated with oxLDL+anti-CXCL16monoclonal antibodies at2.5,5.0and10μg/ml showed significantly reduced level of total cholesterol. Total cholesterol content was respectively203.8±24.8μg/ml and174.1±15.1μg/ml in cells incubated with oxLDL and anti-CXCL16at low concentration (0.625μg/ml,1.25μg/ml); Intracellular total cholesterol content was reduced as dose-dependent in groups incubated with both oxLDL and CXCL16antibody at high concentration(2.5,5.0and10.0μg/ml)for48h, and total cholesterol level was147.2±15.lug/ml,100.7±11.4ug/ml and84.1±9.1ug/ml (P<0.05; P<0.01) respectively.
     2.2The effect of CXCL16antibody on CXCL16expression in podocytes:The result of quantitative test from Western blot showed CXCL16expression in podocytes was reduced as dose-dependent as concentration of CXCL16antibody increased. No significant changes were found in CXCL16expression in cells incubated with both oxLDL and CXCL16antibody at low concentration (0.625μg/ml,1.25μg/ml) for48h compared with cells treated with only oxLDL; But CXCL16expression in podocytes was significantly reduced (P<0.05; P<0.01)in groups that incubated with both oxLDL and CXCL16antibody at high concentration (2.5,5.0and10.0μg/ml) for48h. Podocytes were treated with oxLDL (80μg/ml) and CXCL16antibody (5.0μg/ml) for24h and48h, compared with cells treated with only oxLDL, CXCL16expression was significantly decreased (P<0.05).
     3. The effect of ADAM10inhibitor on lipid accumulation and CXCL16expression in podocytes induced by oxLDL.
     3.1The effect of ADAM10inhibitor on intracellular lipid accumulation:Results of oil red O staining showed that no visible lipid drop was observed in control cells and cells treated with DMSO, whereas large amount of lipid drops were found in cells treated with only oxLDL (80μg/ml). Compared with cells treated with only oxLDL, no marked change was found in cells treated with oxLDL+ADAM10inhibitor (0.125and0.25μg/ml). However, lipid accumulation was remarkably increased in cells treated with oxLDL+ADAM10inhibitor at0.5,1.0and2μg/ml. The result of colorimetry showed total cholesterol level was slightly increased in groups incubated with oxLDL and ADAM10inhibitor at0.125μg/ml or0.25μg/ml compared with group oxLDL and total cholesterol level was respective211.6±12.6μg/ml and227.8±18.3μg/ml, No statistically significant difference was found (P>0.05); whereas significant difference in total cholesterol level was only found when cells incubated with oxLDL+ADAM10inhibitor at0.5,1.0and2.0μg/ml (P<0.05; P<0.01), total cholesterol level was258.5±26.8μg/ml,284.0±23.2μg/ml,338.3±26.6μg/ml respectively.
     3.2The effect of ADAM10inhibitor on CXCL16expression in podocytes:Western blot analysis showed CXCL16expression was slight increased in cells incubated with oxLDL+ADAM10inhibitor at0.125and0.25μg/ml, compared with cells treated with only oxLDL (P>0.05), while CXCL16expression was significant increased in cell incubated with oxLDL+ADAM10inhibitor at0.5,1.0and2.0μg/ml (P<0.05; P<0.01).
     Conclusions:1CXCL16expression was increased in vitro cultured mouse glomerulus podocytes induced by oxLDL, meanwhile intracellular cholesterol level was increased. Which hinted oxLDL could up-regulate the quantities of CXCL16, then was excessively ingested, therefore resulted in cholesterol accumulation in mouse podocytes
     2In vitro podocytes incubated with both oxLDL and IFN-y of certain concentration showed enhanced in intracellular lipid droplets and totle cholesterol content, meanwhile CXCL16expression was up-regulated significantly. Which indicated that interferon-γ could raise CXCL16expression and then facilitate oxLDL uptake in mouse glomerulus podocytes.
     3After treated cells with oxLDL and CXCL16antibody of certain concentration in vitro, intracellular lipid droplets showed obviously reduced accompanied with cholesterol content was significantly decreased, meanwhile CXCL16expression was also significantly dropped. That indicated blocking expression of CXCL16by CXCL16antibody could significantly suppress oxLDL uptake and lipid accumulation in mouse podocytes.
     4Podocytes incubated with oxLDL and ADAM10inhibitor of certain concentration showed both intracellular lipid droplets and cholesterol content were enhanced significantly, accompanied with CXCL16expression was markedly increased. That indicated ADAM10inhibitor could significantly increased CXCL16expression and then facilitated oxLDL uptake in mouse podocytes.
     Part3The interventional mechanism of simvastatin for lipid accumulation in mouse podocytes induced by oxLDL
     Objectives:Investigate the effect of simvastatin on lipid accumulation and the expression of CXCL16and nephrin in murine podocytes induced by oxLDL in order to explore its protection mechanism.
     Methods:Murine podocytes (MPC5) were incubated with oxLDL (80μg/ml) with/without different concentrations of simvastatin (1.0or2.0μg/ml) for48hours. Lipid accumulation in podocytes were assessed by oil red O staining, and measured quantitatively by Colorimetric cholesterol detection kit. CXCL16and nephrin expression were detected by Western blot.
     Results:OxLDL treated MPC5cells exhibited obvious higher intracellular lipid accumulations compared with untreated group. Colorimetric detection found that total cholesterol was90.3±30.1μg/ml in untreated cells and226.5±21.6μg/ml in oxLDL treated cells. The difference was statistically significant (P<0.01). While cells were treated with both oxLDL and simvastatin, we observed much less lipid accumulation. Total cholesterol in oxLDL+simvastatin cells were151.8±6.8μg/ml and135.5±26.9μg/ml under1.0μg/ml or2.0μg/ml of simvastatin treatment respectively. Both were statistically significantly lower than the oxLDL treated cells (P<0.05). Western blot analysis showed that CXCL16expression was significantly increased (P<0.05) in oxLDL treated cells comparing with the untreated cells, and was significantly inhibited by application of simvastatin (P<0.05). The analysis of nephrin expression showed that there were no changes in group simvastatin compared with that of control group(P>0.05). Nephrin expression was significantly reduced by treatment with oxLDL(P<0.01); and was significantly increased by application of simvastatin (P<0.05).
     Conclusions:1.The lipid droplets and total cholesterol concentration were obviously reduced in mouse podocytes since incubated with simvastatin and oxLDL, and the level of CXCL16expression was also markedly lower. All these indicated simvastatin treatment could significantly decrease lipid accumulation in mouse podocytes and this protective effect was realized through inhibition of the expression of CXCL16.
     2. The expression of nephrin was significantly decreased in mouse podocytes induced by oxLDL. After treatment with simvastatin its expression was obviously increased. That hinted nephrin loss might contribute to podocytes lipids damage at the early stage of oxLDL exposure. Simvastatin could prevent podocytes from lipids oxidative damage, which might be due to increasement the expression of nephrin.
     Innovations and meanings:
     1.This study intuitively observed lipid accumulation in mouse glomerular podocytes using oil red O staining for the first time.The method is simple, practical, economic and better repeatability. Establishment of the model of lipid accumulation in mouse podocytes induced by oxLDL may provide credible experiment basis for studying the mechanism and the factors of regulating of oxLDL uptake
     2. This study in vitro cell culture confirmed that CXCL16is the key molecules to mediate the lipid injury in podocytes exposed to oxLDL. Its expression could be regulated by IFN-γ and ADAM10inhibitors, thus affected podocytes to ingest lipids.All those may provide important clues for the early prevention and treatment from lipids oxidative damage in podocytes.
     3. This study firstly in vitro comfirmed that simvastatin could down-regulate CXCL16expression and decrease lipid accumulation in oxLDL-induced mouse podocytes, meanwhile it could enhance the expression of nephrin. All those indicated that simvastatin could alleviate lipids oxidative damage in podocytes, which may also provide some references for the clinical application of statins to protect the kidney.
引文
[1]Moorhead JF, Chan MK, El-Nahas M, et al. Lipid nephrotoxicity in chronic progressive glomerular and tubulo-interstitial disease[J]. Lancet,1982, 2(8):1309-1311.
    [2]Moorhead JF, Brunton C, Varghese Z. Glomerular atherosclerosis[J]. Miner Electrolyte Metab,1997,23(3):287-290.
    [3]Vaziri ND, Navab M, Fogelman AM. HDL metabolism and activity in chronic kidney disease[J]. Nat Rev Nephrol,2010,6(5):287-296.
    [4]王欣,孙洞箫.高脂血症在肾小球损伤中的作用机制研究进展.实用医学杂志,2008,24(7):1260-1261.
    [5]Lee HS. Oxidized LDL, glomerular mesangial cells and collagen[J]. Diabetes Res Clin Pract,1999,45(2):117-122.
    [6]Kasiske BL, O'Donnell MP, Schmitz PG, et al. Renal injury of dietinduced hypercholesterolemia in rats[J]. Kidney Int,1990,37 (3):880-891.
    [7]Lee HS, Jeonq JY, Kim BC, et al. Dietary antioxidant inhibits lipoprotein oxidation and renal injury in experimental focal segmental glomerulosclerosis[J]. Kidney Int,1997,51 (4):1151-1159.
    [8]Witztum JL, Steinberg D. The oxidative modification hypothesis of atherosclerosis:does it hold for humans[J]? Trends Cardiovasc Med,2001,11: 93-102.
    [9]Akiba S, Chiba M, Mukaida Y, et al. Involvement of reactive oxygen species and SP-1 in fibronectin production by oxidized LDL[J]. Biochem Biophys Res Commun, 2003,310:491-497.
    [10]Brune B, Zhou J, von Knethen A. Nitric oxide, oxidative stress, and apoptosis[J]. Kidney Int Suppl,2003,2:22-24.
    [11]Chade AR, Mushin OP, Zhu X, et al. Pathways of renal fibrosis and modulation of matrix turnover in experimental hypercholesterolemia[J]. Hypertension,2005, 46:772-779.
    [12]Reddy S, Santanam N, Reddy PP, et al. Interaction of Interceed oxidized regenerated cellulose with macrophages:a potential mechanism by which Interceed may prevent adhesions[J]. Am J Obstet Gynecol,1997,177:1315-1320.
    [13]梁栋,杨洪涛.氧化低密度脂蛋白与肾小球硬化.中国中西医结合肾病杂志,2012,13(12):1117-1118
    [14]Cheng H, Harris RC. The glomerulus-a view from the outside-the podocyte[J]. Int J Biochem Cell Biol,2010,42(9):1380-1387.
    [15]Gipson DS, Gibson K, Gipson PE, et al. Therapeutic approach to FSGS in children[J]. Pediatr Nephrol,2007,22(1):28-36.
    [16]Camici M. Urinary detection of podocyte injury[J]. Biomed Pharmacother, 2007,61(5):245-249
    [17]Shimaoka T, Kume N, Minami M, et al. Molecular cloning of a novel scavenger receptor for oxidized low density lipoprotein, SR-PSOX, on macrophages [J]. J Biol Chem,2000,275(52):40663-40666.
    [18]Matloubian M, David A, Engel S, et al. A transmembrane CXC chemokine is a ligand for HIV-coreceptor Bonzo [J]. Nat Immunol,2000,1(4):298-304.
    [19]Wilbanks A, Zondlo SC, Murphy K, et al. Expression cloning of the STRL33/ BONZO/ TYMSTR ligand reveals elements of CC, CXC, and CX3C chemokines[J]. J Immunol,2001,166(8):5145-5154.
    [20]徐焕宾,熊思东.新发现的CXCL16趋化因子及其受体[J].生命的化学,2003,23(1):8-10.
    [21]Minami M, Kume N, Shimaoka T, et al. Expression of SR-PSOX,a novel cell surface scavenger receptor for phosphatidyl serine and oxidized LDL in human atherosclerotic lesions [J]. Arterioscler Thromb Vasc Biol,2001,21(11):1796-1800.
    [22]Gough PJ, Garton KJ, Wille PT, et al. A disintegrin and metalloproteinase 10-mediated cleavage and shedding regulates the cell surface expression of CXC chemokine ligand 16[J]. J Immunol,2004,172:3678-3685
    [23]Ludwig A, Hundhausen C, Lambert MH, et al. Metalloproteinase inhibitors for the disintegrin-like metalloproteinases ADAM 10 and ADAM 17 that differentially block constitutive and phorbol ester-inducible shedding of cell surface molecules[J]. Comb Chem High Throughput Screen,2005,8:161-171
    [24]Garcia GE, TruongLD,Li P, et al. Inhibition of CXCL16 attenuates inflammatory and progressive phases of anti-glomerular basement membrane antibody-associated glomerulonephritis [J]. Am J Pathol,2007,170 (5):1485-1496
    [25]Gutwein P, Abdel-Bakky MS, Schramme A, et al. CXCL16 is expressed in podocytes and acts as a scavenger receptor for oxidized low-density lipoprotein[J]. AmJPathol,2009,174:2061-2072.
    [26]Wagsater D, Olofsson PS, Norgren L, et al. The chemokine andscavenger recep tor CXCL16 /SR2PSOX is expressed in human vascular smooth muscle cells and is induced by interferon gamma[J]. Biochem Biophys Res Commun,2004,325(4): 1187-1193.
    [27]Wuttge DM, Zhou X, Sheikine Y, et al. CXCL16 /SR2PSOX is aninterferon-gamma regulated chemokine and scavenger receptor expressed in atheroscleroticlesions [J]. Arterioscler Thromb Vasc Biol,2004,24 (4):750-755
    [28]Raja SG, Dreyfus GD. Statins:much more than just a lipid-lowering therapy[J]. Indian Heart J,2004,56(3):204-209.
    [29]Menzel S, Moeller MJ. Role of the podocyte in proteinuria [J]. Pediatr Nephrol,2011,26(10):1775-1780.
    [30]Nosadini R, Tonolo G. Role of oxidized low density lipoproteins and free fatty acids in the pathogenesis of glomerulopathy and tubulointerstitial lesions in type 2 diabetes[J]. Nutrition, Metabolism & Cardiovascular Diseases,2011,21:79-85
    [31]Stitt-Cavanagh E, Macleod L, Kennedy C. The podocyte in diabetic kidney disease [J]. Scientific World Journal,2009,9 (10):1127-1139
    [32]邹琦,郑祥雄.肾小球足细胞功能认识的进展.医学综述,2011,17(19):2910-2913
    [33]Wiggins RC. The spectrum of podocytopathies:a unifying view of glomerular diseases[J]. Kidney Int,2007,71:1205-1214
    [34]Gutwein P, Abdel-Bakky MS, Doberstein K, et al. CXCL16 and oxLDL are induced in the onset of diabetic nephropathy[J]. Cell Mol Med,2009,13 (9): 3809-3825
    [35]Vaziri ND, Navab M, Fogelman AM. HDL metabolism and activity in chronic kidney disease [J]. Nat Rev Nephrol,2010,6 (5):287-296.
    [36]Joles JA. Early mechanisms of renal injury in hypercholesterolemic or hypertriglyceridemic rats[J]. J Am Soc Nephrol,2000,11(4):669-683
    [37]Bussolati B, Deregibus MC, Fonsato V, et al. Statins prevent oxidized LDL-induced injury of glomerular podocytes by activating the phosphatidylinositol3-kinase/AKT signaling pathway [J]. J Am Soc Nephrol 2005, 16:1936-1947.
    [38]王玉洁,曹灵,孙兴旺.足细胞与蛋白尿发病机制的研究进展.山东医药,2012,52(9):90-92
    [39]Mundel P, Kriz W. Structure and function of podocytes:An update [J]. Anat Embryol,1995,192:385-397
    [40]Pavenstadt H, Kriz W, Kretzler M. Cell biology of glomerular podocyte [J]. Physiol Rev,2003,83:253-307
    [41]Mundel P, Reiser J, Zuniga M, et al. Rearrangements of the cytoskeleton and cell contacts induce process formation during differentiation of conditionally immortalized mouse podocyte lines [J]. Exp Cell Res,1997,236(1):248-258.
    [42]陈洪宇,覃志成,王永钧.脂质对小鼠肾足细胞增生影响.医学研究杂志,2006,35(8):25-28.
    [43]屈长青,李东伟,朱茂英.一种改进的培养脂肪细胞染色法.中国组织化学与细胞化学杂志,2008,17(3):329-330.
    [1]孙颖,常志文.趋化因子CXCL16与动脉粥样硬化的研究现状[J].中国动脉硬化,2008,16(4):329-331.
    [2]Kugiyama K, Kerns SA, Morrisett JD, et al. Impairment of endotheliumdependent arterial relaxation by lysolecithin in modified low-density lipoproteins[J]. Nature,1990,344:160-162.
    [3]Stiko-Rahm A, Hultgardh-Nilsson A, Regnstrom J, et al. Nativeand oxidized LDL enhances production of PDGF AA and the surface expression of PDGF receptors in cultured human smoothmuscle cells[J]. Arterioscler Thromb,1992,12(9):1099-1109.
    [4]Yui S, Sasaki T, Miyazaki A, et al. Induction of murine macrophage growth by modified LDLs[J]. ArteriosclerThromb,1993,13(3):331-337.
    [5]Steinberg D, Parthasarathy S, Carew TE, et al. Beyond cholesterol. Modifications of low-density lipoprotein that increase its atherogenicity[J]. N Engl J Med,1989, 320(14):915-924.
    [6]Schramme A, Abdel-Bakky MS, Gutwein P, et al.Characterization of CXCL16 and ADAM10 in the normal and transplanted kidney[J]. Kidney Int,2008, 74(3):328-338.
    [7]Wu T, Xie C, Wang HW, et al.Elevated urinary VCAM-1, P-selectin, soluble TNF receptor-1, and CXC chemokine ligand16 in multiple murine lupuss trains and human lupusnephritis[J]. J Imm,2007,179(10):7166-7175.
    [8]Okamura DM, Lpez-Guisa JM, Koelsch K, et al.Atherogenic scavenger receptor modulation in the tubulointerstitium in response to chronic renal injury [J]. Am J Physiol Renal Physiol,2007,293(2):575-585.
    [9]Yang SH, Kim SJ, Kim N, et al.NKT cells inhibit the development of experimental crescentic glomerulonephritis [J]. J Am Soc Nephrol,2008, 19(9):1663-1671.
    [10]Garcia GE, Truong LD,Li P, et al. Inhibition of CXCL16 attenuates inflammatory and progressive phases of anti-glomerular basement membrane antibody-associated glomerulonephritis [J]. Am J Pathol,2007,170 (5):1485-1496
    [11]Sercan O, Hammerling GJ, Arnold B, et al. Innate immune cells contribute to the IFN-dependent regulation of antigen Specific CD8+ T cell homeostAsis [J]. J Immunol,2006,176:735-739.
    [12]Wuttge DM, Zhou X, Sheikine Y, et al. CXCL16/SRPSOX is an interferon gamma regulated chemokine and scavenger receptor expressed in atherosclerotic lesions [J]. Arterioscler Thromb Vase Biol,2004,24:750-755.
    [13]杨婷,全智华.干扰素对鼠源性平滑肌细胞内源性CXC配体16表达及细胞内脂质蓄积的影响.中国动脉硬化杂志,2008,16(3):185-188
    [14]Wagsater D, Olofsson PS, Norgren L,et al. The chemokine and scavenger receptor CXCL16/SR-PSOX is expressed in human vascular smooth muscle cells and is induced by interferon [J]. Biochemical and Biophysical Research Communications, 2004,325 (4):1187-1193
    [15]Gutwein P, Abdel-Bakky MS, Schramme A, et al. CXCL16 is expressed in podocytes and acts as a scavenger receptor for oxidized low-density lipoprotein[J]. Am J Pathol,2009,174:2061-2072.
    [16]Gough PJ, Garton KJ, Wille PT, et al. A disintegrin and metalloproteinase 10-mediated cleavage and shedding regulates the cell surface expression of CXC chemokine ligand 16[J]. J Immunol,2004,172:3678-3685
    [17]Matloubian M, David A, Engel S, et al. A transmembrane CXC chemokine is a ligand for HIV-coreceptor Bonzo [J]. Nat Immunol,2000,1:298-304.
    [18]Wilbanks A, Zondlo S C, Murphy K, et al. Expression cloning of the STRL33 /BONZO/TYMSTR ligand reveals elements of CC, CXC, and CX3C chemokines [J]. Immunol,2001,166:5145-5154.
    [19]Abel S, Hundhausen C, Mentlein R, et al. The transmembrane CXC chemokine ligand 16 is induced by IFN-gamma and TNF-alpha and shed by the activityof the disintegrin like metalloproteinAse ADAM10 [J]. J Immunol,2004,172 (6):362-372.
    [20]徐焕宾,熊思东.新发现的CXCL16趋化因子及其受体[J].生命的化学, 2003,23(1):8-10.
    [21]Bussolati B, Deregibus MC, Fonsato V, et al. Statins prevent oxidized LDL-induced injury of glomerular podocytes by activating the phosphatidylinositol 3-kinase/AKT signaling pathway [J].J Am Soc Nephrol,2005,16:1936-1947.
    [22]Dai C, Stolz DB, Bastacky SI, et al. Essential role of integrin linked kinase in podocyte biology:Bridging the integrin and slit diaphragm signaling. J Am Soc Nephrol,2006,17(8):2164-2175.
    [23]林青,宋艳芳,祝先进等.IFN-γ诱导的肾小管上皮细胞CXCL9、CXCL10和CXCL11的表达.细胞与分子免疫学杂志,2013,29(2):137-140
    [24]Leon ML, Zuckerman SH. Gamma interferon:a central mediator in atherosclerosis[J]. Inflamm Res,2005,54:395-411.
    [25]Shimaoka T, Nakayama T, Fukumoto N, et al. Cell surface an chored SR-PSOX/ CXC chemokine ligand 16 mediates firm adhesion of CXC chemokine receptor 6 expressing cells [J]. J Leukoc Biol,2004,75(2):267-274.
    [1]Menzel S, Moeller MJ. Role of the podocyte in proteinuria [J]. Pediatr Nephrol,2011,26(10):1775-1780.
    [2]李卫国,李宇宁.足细胞对损伤的反应.临床儿科杂,2012,30(11):1091-1094
    [3]Mundel P, Shankland SJ. Podocyte biology and response to injury[J]. J Am Soc Nephrol,2002,13:3005-3015
    [4]Kerjaschki D, Caught FF. Podocyte damage and the molecular bases of focal glomerulo sclerosis [J]. Clin Invest,2001,108:1583.
    [5]Doublier S, Salvidio G, Lupia E, et al. Nephrin expression is reduced in human diabetic nephropathy:evidence for a distinct role for glycated albumin and angiotensin. Diabetes,2003,52:1023-1030.
    [6]Raja SG, Dreyfus GD. Statins:much more than just a lipid-lowering therapy[J]. Indian Heart J,2004,56(3):204-209.
    [7]Smith C, Halvorsen B, Otterdal K, et al. High levels and inflammatory effects of soluble CXC ligand 16 (CXCL16) in coronary artery disease:down-regulatory effects of statins [J]. Cardiovasc Res,2008,79(1):195-203.
    [8]Bruni F, Pasqui AL, Pastorelli M, et al.Different effect of statins on platelet oxidized-LDL receptor (CD36 and LOX-1)expression in hypercholesterolemic subjects [J]. Clinical and Applied Thrombosis Hemostasis,2005,11:417-428.
    [9]孙颖,王翠英,李敏,et al.可托伐他汀钙、非诺贝特下调ApoE基因敲除小鼠动脉组织CXCL16的表达.临床和实验医学杂志,2012,11(21):1677-1680
    [10]于冬青,邓华聪,刘金波.辛伐他汀对糖尿病大鼠肾脏氧化应激的抑制作用[J].国际病理生理杂志,2006,22(4):824-825
    [11]姚新明,叶山东,陈燕,等.辛伐他汀对糖尿病大鼠MMP-9表达的影响及对肾脏保护的研究[J].中国老年学杂志,2009,29(12):1472-1475
    [12]Bussolati B, Deregibus MC, Fonsato V, et al. Statins prevent oxidized LDL-induced injury of glomerular podocytes by activating the phosphatidylinositol3-kinase/AKT signaling pathway [J]. J Am Soc Nephrol,2005, 74 16:1936-1947
    [13]Tonolo G, Velussi M, Brocco E, et al. Simvastatin maintains steady patterns of GFR and improves AER and expression of slit diaphragm proteins in type II diabetes [J]. Kidney Int,2006,70:177-186.
    [14]陈姝君,陈海平,刘奇.辛伐他汀对糖尿病大鼠足细胞损伤保护作用的实验研究.中国糖尿病杂志,2009,17(11):861-863
    [15]石琦屏,冯烈,卢筱华,等.辛伐他汀对糖尿病鼠肾nephrin基因表达的影响.广东医学,2006,27:1799-1801.
    [16]Planavila A,Laguna JC,Vazquez-Carrera M. Atorvastatin improves peroxisome proliferators-activated receptor signaling in cardiac hypertrophyby preventing nuclear factor-kappa B activation. Biochim Biophys Acta,2005,1687(1):76-83.
    [17]Huber TB, Hartleben B, Kim J, et al. Nephrin and CD2AP associate with phosphoinositide 3-OH kinase and stimulate AKT-dependent signaling. Mol Cell Biol. 2003,4917-4928
    [1]Pavenstadt H, Kriz W, Kretzler M. Cell biology of glomerular podocyte [J]. Physiol Rev,2003,83:253-307
    [2]Salmon AH, Neal CR, Harper SJ. New aspects of glomerular filtration barrier structure and function:five layers (at least) not three [J]. Curr Opin Nephrol Hypertens,2009,18(3):197-205
    [3]Shankland SJ. The podocyte's response to injury:role in proteinuria and glomerulosclerosis [J]. Kidney Int,2006,69(12):2131-2147.
    [4]Cheng H, Harris RC.The glomerulus-a view from the outside-the podocyte[J].Int J Biochem Cell Biol,2010,42(9):1380-1387.
    [5]Nosadini R, Tonolo G. Role of oxidized low density lipoproteins and free fatty acids in the pathogenesis of glomerulopathy and tubulointerstitial lesions in type 2 diabetes[J].Nutrition, Metabolism & Cardiovascular Diseases,2011,21:79-85
    [6]Stitt-Cavanagh E, Macleod L, Kennedy C. The podocyte in diabetic kidney disease [J]. Scientific World Journal,2009,9 (10):1127-1139
    [7]Wiggins RC. The spectrum of podocytopathies:a unifying view of glomerular diseases[J]. Kidney Int,2007,71:1205-1214
    [8]Machado JR, Rocha LP, Neves PD,et al. An overview of molecular mechanism of nephrotic syndrome[J]. Int J Nephrol,2012,93(11):7623-7628.
    [9]Ronco P, Debiec H. Membranous glomerulopathy:the evolving story[J].Curr Opin Nephrol Hypertens,2010,19(3):254-259.
    [10]Ronco P, Debiec H. Review Podocyte antigens and glomerular disease[J]. Nephron Exp Nephrol.2007,107(2):e41-46.
    [11]Menzel S, Moeller MJ. Role of the podocyte in proteinuria [J]. Pediatr Nephrol,2011,26(10):1775-1780.
    [12]Gipson DS, Gibson K, Gipson PE, et al. Therapeutic approach to FSGS in children [J]. Pediatr Nephrol,2007,22(1):28-36.
    [13]Camici M. Urinary detection of podocyte injury [J]. Biomed Pharmacother, 2007,61(5):245-249.
    [14]Tryggvason K, Patrakka J, Wartiovaara J. Hereditary proteinuria syndromes and mechanisms of proteinuria [J]. N Engl J Med,2006,354(13):1387-1401.
    [15]Dai C, Stolz DB, Bastacky SI, et al. Essential role of integrin linked kinase in podocyte biology:Bridging the integrin and slit diaphragm signaling [J]. J Am Soc Nephrol,2006,17(8):2164-2175.
    [16]Patrakka J, Tryggvason K. Nephrin:a unique structural and signaling protein of the kidney filter [J]. Trends MolMed,2007,13(9):396-403.
    [17]Arias LF, Vieco BE, Arteta AA, et al. Expression of nephrin, Podocin and α-actinin-4 in renal tissue of patients with proteinuria[J]. Nefrologia,2009,29(6): 569-575.
    [18]Ronco P, Debiec H. Molecular pathomechanisms of membranous nephropathy: from Heymann nephritis to alloimmunization[J]. J Am Soc Nephrol,2005, 16(5):1205-1213
    [19]Qian Y, Feldman E, Pennathur S, et al. From fibros is to sclerosis mechanisms of glomerulo scleros is in diabetic nephropathy [J]. Diabetes,2008,57(6):1439.
    [20]Vaziri ND, Navab M, Fogelman AM. HDL metabolism and activity in chronic kidney disease [J]. Nat Rev Nephrol,2010,6 (5):287-296.
    [21]王欣,孙洞箫.高脂血症在肾小球损伤中的作用机制研究进展.实用医学杂志,2008,24(7):1260-1261.
    [22]Hong HK, Song CY, Kim BC, et al. ERK contributes to the effects of Smad signaling on oxidized LDL-induced PAI-1 expression in human mesangial cells [J]. Transl Res,2006,148 (4):171-179.
    [23]Song CY, Kim BC, Hong HK, et al. Oxidized LDL activates PAI-1 transcription through autocrine activation of TGF-β signaling in mesangial cells [J]. Kidney Int,2005,67 (5):1743-1752.
    [24]Derynck R, Zhang YE. Smad-dependent and Smadindependent pathways in TGF-beta family signalling [J]. Nature,2003,425 (6958):577-584.
    [25]Lee HS, Song CY. Oxidized low-density lipoprotein and oxidative stress in the development of glomerulosclerosis [J]. Am J Nephrol,2009,29 (1):62-70.
    [26]Song CY, Kim BC, Lee HS. Lovastatin inhibits oxidized low density lipoprotein-induced plasminogen activator inhibitor and transforming growth factor-betal expression via a decrease in Ras / extracellular signal-regulated kinase activity in mesangial cells [J]. Transl Res,2008,151 (1):27-35.
    [27]Bro S, Binder CJ, Witztum JL, et al. Inhibition of the reninangiotensin system abolishes the proatherogenic effect of uremia in apolipoprotein E-deficient mice [J]. Arterioscler Thromb Vasc Biol,2007,27 (5):1080-1086.
    [28]Whaley-Connell AT, Morris EM, RehmerN, et al. Albumin activation of NADPH oxidase activity is mediated via Racl in proximal tubule cells [J]. Am J Nephrol,2007,27 (1):15-23.
    [29]Vaziri ND, Navab M, Fogelman AM. HDL metabolism and activity in chronic kidney disease [J]. Nat Rev Nephrol,2010,6 (5):287-296.
    [30]Joles JA. Early mechanisms of renal injury in hypercholesterolemic or hypertriglyceridemic rats[J]. J Am Soc Nephrol,2000,11(4):669-683
    [31]Mundel P, Kriz W. Structure and function of podocytes:An update [J]. Anat Embryol(Berl),1995,192(5):385-387.
    [32]Kerjaschki D, Caught FF. Podocyte damage and the molecular bases of focal glomerulo sclerosis [J]. Clin Invest,2001,108:1583.
    [33]Doublier S, Salvidio G, Lupia E, et al. Nephrin expression is reduced in human diabetic nephropathy:evidence for a distinct role for glycated albumin and angiotensin. Diabetes,2003,52:1023-1030.
    [34]徐兰,杨海春,马骥,等.新生小鼠足细胞损伤对肾小球发育的影响.中华肾脏病杂志,2006,22(10):617-622.
    [35]梁丹丹,曾彩虹.局灶节段性肾小球硬化的发病机制.肾脏病与透析肾移植杂志,2009,18(1):70-75.
    [36]陈洪宇,覃志成,王永钧.脂质对小鼠肾足细胞增生影响.医学研究杂志,2006,35(8):25-28.
    [37]Bussolati B, Deregibus MC, Fonsato V, et al. Statins prevent oxidized LDL-induced injury of glomerular podocytes by activating the phosphatidylinositol3-kinase/AKT signaling pathway [J]. J Am Soc Nephrol,2005, 16:1936-1947.
    [38]Norlander AE, Saleh MA, Madhur MS. CXCL16:a chemokine-causing chronic kidney disease [J]. Hypertension,2013,62(6):1008-1010.
    [39]Matloubian M, David A, Engel S, et al. A transmembrane CXC chemokine is a ligand for HIV-coreceptor Bonzo [J].Nat Immunol,2000,1(4):298-304.
    [40]Wilbanks A, Zondlo SC, Murphy K, et al. Expression cloning of the STRL33/ BONZO/TYMSTR ligand reveals elements of CC, CXC, and CX3C chemokines[J]. J Immunol,2001,166(8):5145-5154.
    [41]Chandrasekar B, Bysani S, Mummidi S. CXCL16 signals via Gi, phosphatidylinositol 3-kinase, Akt, I kappa B kinase, and nuclear factor-kappa B and induces cell-cell adhesion and aortic smooth muscle cell proliferation [J]. J Biol Chem,2004,279 (5):3188-3196.
    [42]徐焕宾,熊思东.新发现的CXCL16趋化因子及其受体[J].生命的化学,2003,23(1):8-10.
    [43]Shimaoka T, Nakayama T, Fukumoto N, et al. Cell surface-anchored SR-PSOX/CXC chemokine ligand 16 mediates firm adhesion of CXC chemokine receptor 6-expressing cells[J]. J Leukoc Biol,2004,75(2):261-274.
    [44]Shimaoka T, Nakayama T, Kume N, et al. Cutting edge:SR-PSCX/CXC chemokine ligand 16 mediates bacterial phagocytosis by APCs through its chemokine domain[J]. J Immunol,2003,17(4):1647-1651.
    [45]Abel S, Hundhausen C, Mentlein R, et al. The transmembrane CXC-chemokine ligand 16 is induced by IFN-gamma and TNF-alpha and shed by the activity of the disintegrin-like metalloproteinase ADAM10 [J].JImmunol,2004,172 (10):6362-6372.
    [46]Gough PJ, Garton KJ, Wille PT, et al. A disintegrin and metalloproteinase 10-mediated cleavage and shedding regulates the cell surface expression of CXC chemokine ligand 16 [J], J Immunol,2004,172(6):3678-685.
    [47]Ludwig A, Hundhausen C, Lambert MH, et al. Metalloproteinase inhibitors for the disintegrin-like metalloproteinases ADAM10 and ADAM 17 that differentially block constitutive and phorbol ester-inducible shedding of cell surface molecules[J]. Comb Chem High Throughput Screen,2005,8(2):161-171.
    [48]Shimaoka T, Kume N, Minami M, et al. Molecular cloning of a novel scavenger receptor for oxidized low density lipoprotein, SR-PSOX, on macrophages[J]. J Biol Chem,2000,275 (52):40663-40666.
    [49]Day C, Patel R, Guillen C, et al. The chemokine CXCL16 is highly and constitutively expressed by human bronchial epithelial cells [J]. Experimental Lung Research,2009,35(4):272-283.
    [50]Garcia GE, Truong LD, Li P, et al. Inhibition of CXCL16 Attenuates Inflammatory and Progressive Phases of Anti-Glomerular Basement Membrane Antibody-Associated Glomerulonephritis[J]. Am J Pathol,2007,170(5):1485-1496.
    [51]Gutwein P, Abdel-Bakky MS, Doberstein K, et al. CXCL16 and oxLDL are induced in the onset of diabetic nephropathy[J]. J Cell Mol Med,2009,13(9B): 3809-3825.
    [52]Okamura DM, Lopez-Guisa JM, Koelsch K, et al. Atherogenic scavenger receptor modulation in the tubulointerstitium in response to chronic renal injury [J]. Am J Physiol Renal Physiol,2007,293(2):575-585.
    [53]Wagsater D, Olofsson PS, Norgren L, et al. The chemokine and scavenger receptor CXCL16/SR-PSOX is expressed in human vascular smooth muscle cells and is induced by interferon gamma[J]. Biochem Biophys Res Commun,2004,325 (4): 1187-1193.
    [54]Wuttge DM, Zhou X, Sheikine Y, et al. CXCL16/SR-PSOX is an interferon-gamma regulated chemokine and scavenger receptor expressed in atherosclerotic lesions [J]. Arterioscler Thromb Vasc Biol,2004,24 (4):750-755
    [55]Xu H, XuW, Chu Y, et al. Involvement of up-regulated CXC chemokine ligand 16/scavenger receptor that binds phosphatidylserine and oxidized lipoprotein in endotoxin induced lethal liver injury via regulation of T-cell recruitment and adhesion [J]. Infect Immun,2005,73 (7):4007-4016.
    [56]Matsumura S,WangB, Kawashima N, et al. Radiation2induced CXCL16 release by breast cancer cells attracts effector T cells[J].Immunol,2008,181 (5):3099-3107.
    [57]Smith C, Halvorsen B, Otterdal K, et al. High levels and inflammatory effects of soluble CXC ligand 16 (CXCL16) in coronary artery disease:down-regulatory effects of statins [J]. Cardiovasc Res,2008,79(1):195-203.
    [58]Garcia GE, TruongLD,Li P, et al. Inhibition of CXCL16 attenuates inflammatory and progressive phases of anti-glomerular basement membrane antibody-associated glomerulonephritis [J]. Am J Pathol,2007,170 (5):1485-1496
    [59]Ponticelli C:Membranous nephropathy[J]. J Nephrol,2007,20:268-287.
    [60]Gutwein P, Abdel-Bakky MS, Schramme A,et al. CXCL16 Is Expressed in Podocytes and Acts as a Scavenger Receptor for Oxidized Low-Density Lipoprotein[J]. Am J Pathol,2009,174(6):2061-2072.
    [61]Wu T, Xie C, Wang HW, et al. Elevated Urinary VCAM-1, P-Selectin, Soluble TNF Receptor-1, and CXC Chemokine Ligand 16 in Multiple Murine Lupus Strains and Human Lupus Nephritis[J]. J Immunol,2007,179(10):7166-7175.
    [62]Singh S, Wu T, Xie C, et al. Urine VCAM-1 as a marker of renal pathology activity index in lupus nephritis[J]. Arthritis Res Ther,2012,14(4):R164.
    [63]Reyes-Thomas J, Blanco I, Putterman C. Urinary biomarkers in lupus nephritis[J]. Clin Rev Allergy Immunol,2011,40(3):138-150.
    [64]Teramoto K, Negoro N, Kitamoto K, et al. Microarray analysis of glomerular gene expression in murine lupus nephritis[J]. J Pharmacol Sci,2008,106(1):56-67.
    [65]Schramme A, Abdel-Bakky MS, Gutwein P, et al. Characterization of CXCL16 and ADAM10 in the normal and transplanted kidney[J], Kidney Int,2008, 74(3):328-338.

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700