首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到18条相似文献,搜索用时 78 毫秒
1.
正三磷酸腺苷结合盒转运体A1(ATP-binding cassette transporter A1,ABCA1)作为介导细胞内脂质流出,维持细胞脂质代谢平衡的重要跨膜蛋白,对动脉粥样硬化(atherosclerosis,AS)的防治具有重要意义[1].近日,清华大学结构生物学高精尖创新中心的颜宁教授与龚欣博士组成的研究团队(Cell,2017,169:1228-1239)采用冷冻电子显微镜技术,经过重组人全长ABCA1蛋白制备、透射电子显微  相似文献   

2.
作为一种有效的降脂药物,普罗布考能够降低血浆高密度脂蛋白胆固醇(HDL-C)水平并抑制动脉粥样硬化,但其机制尚未完全阐明.本研究的目的旨在进一步阐明普罗布考降脂及抗动脉粥样硬化的机理.将新西兰白兔随机分为4组:正常饮食组、正常饮食+普罗布考组、高脂饮食组(HFD组)、高脂饮食+普罗布考组(HFD+P组).结果显示,处理7周后,与HFD组比较,HFD+P组动脉粥样硬化病变程度、肝脏脂质蓄积明显减轻,血浆甘油三脂、总胆固醇、低密度脂蛋白胆固醇及HDL-C 水平降低,肝脏中清道夫受体-BⅠ(SR-BⅠ)以及肝脏与小肠中三磷酸腺苷结合盒转运体(ABC)G5(ABCG5)、ABCG8表达上调,肝脏中ABCA1表达下调,主动脉弓与血浆肿瘤坏死因子α、白介素1、白介素6、单核趋化蛋白1水平降低.这些结果表明普罗布考的抗动脉粥样硬化作用可能与其调控ABCA1、SR-BⅠ、ABCG5、ABCG8表达及抑制促炎介质的分泌有关.  相似文献   

3.
唐小二  唐尚书  唐朝克 《生命的化学》2021,41(10):2215-2221
三磷酸腺苷结合盒转运体Al(ATP binding cassette transporter Al,ABCAl)和三磷酸腺苷结合盒转运体G1 (ATP binding cassette transporter G1,ABCGl)促进细胞内胆固醇流出.微小RNAs(microRNAs,miRNAs)包括miR-33、mi...  相似文献   

4.
三磷酸腺苷结合盒转运体A1(ABCA1)是体内胆固醇逆向转运的关键环节.对氧磷是广泛使用的有机磷农药的活性代谢产物.研究发现,对氧磷能增加巨噬细胞中胆固醇的堆积,但具体机制还不清楚.以RAW264.7巨噬细胞源性泡沫细胞为研究对象,观察对氧磷对RAW264.7巨噬细胞源性泡沫细胞ABCA1表达和胆固醇流出的影响并探讨其机制.结果显示,对氧磷以时间和剂量依赖的方式增加RAW264.7巨噬细胞源性泡沫细胞中总胆固醇、游离胆固醇和胆固醇酯水平,降低ABCA1表达和胆固醇流出,同时对氧磷降低细胞中环磷酸腺苷(cAMP)的水平及腺苷酸环化酶(AC)的活性和增加磷酸二酯酶(PDE)的活性,而cAMP的类似物双丁酰环腺苷酸(dBcAMP)能够阻断对氧磷降低ABCA1表达和部分阻断对氧磷降低胆固醇流出,对氧磷导致的cAMP水平的降低也可被AC激动剂福斯高林(Forskolin)和PDE抑制剂3-异丁基-1-甲基黄嘌呤(IBMX)所阻断.以上结果表明,对氧磷通过cAMP信号通路下调RAW264.7巨噬细胞源性泡沫细胞ABCA1的表达,降低细胞内胆固醇流出和增加细胞内胆固醇堆积.  相似文献   

5.
以THP-1巨噬细胞源性泡沫细胞为研究对象,观察干扰素-γ(IFN-γ)对THP-1巨噬细胞源性泡沫细胞胆固醇流出和三磷酸腺苷结合盒转运体A1(ABCA1)表达的影响.以便探讨IFN-γ在动脉粥样硬化发生发展中的作用.采用液体闪烁计数器检测细胞内胆固醇流出, 高效液相色谱分析细胞内总胆固醇、游离胆固醇和胆固醇酯含量.运用逆转录-多聚酶链反应和蛋白质印迹分别检测ABCA1 mRNA与ABCA1蛋白质的表达, 采用流式细胞术检测细胞平均ABCA1荧光强度.发现IFN-γ引起THP-1巨噬细胞源性泡沫细胞总胆固醇、游离胆固醇与胆固醇酯呈时间依赖性增加, 而ABCA1 mRNA和蛋白质表达、细胞平均ABCA1荧光强度以及apoA-1介导的胆固醇流出呈时间依赖性减少, 细胞内胆固醇增多.结果表明IFN-γ抑制THP-1巨噬细胞源性泡沫细胞ABCA1表达及细胞内胆固醇流出,同时增加细胞内胆固醇聚积.  相似文献   

6.
用贵州小香猪建立动脉粥样硬化动物模型,探讨动脉粥样硬化小型猪三磷酸腺苷结合盒转运体 A1(ABCA1) 表达的变化 . 采用血管内膜损伤法加高脂高胆固醇饲料喂养贵州小香猪,建立动脉粥样硬化动物模型 . 血浆总胆固醇、甘油三酯和高密度脂蛋白胆固醇的浓度均用氧化酶法测定,采用逆转录聚合酶链反应检测 ABCA1mRNA 水平,蛋白质印迹和免疫组织化学检测 ABCA1 蛋白质的表达 . 喂养 12 个月后,实验组与正常对照组比较,空腹血浆总胆固醇、甘油三酯和高密度脂蛋白胆固醇水平升高;实验组小型猪主动脉、髂动脉、颈总动脉和冠状动脉可见动脉粥样硬化斑块和脂质条纹;实验组小型猪肝组织、主动脉、小肠组织 ABCA1 表达上调 . 结果提示,采用血管内膜损伤法加高脂高胆固醇饲料喂养小型猪可建立动脉粥样硬化动物模型 . 动脉粥样硬化小型猪肝组织、主动脉和小肠组织 ABCA1 表达上调 .  相似文献   

7.
ABCA1抗动脉粥样硬化的作用主要通过以下两种途径:介导细胞内胆固醇流出和抑制炎症。载脂蛋白与ABCA1的相互作用可激活多个信号通路,包括JAK2/STAT3、蛋白激酶A(PKA)、Rho家族G蛋白CDC42和蛋白激酶C(PKC)等信号通路。ABCA1通过修饰细胞膜脂筏或直接激活信号通路而介导脂质流出和发挥抗炎功能。对这些信号通路的认识,能为动脉粥样硬化相关疾病提供新的治疗靶点。  相似文献   

8.
为探讨mi R-33s在核因子κB(NF-κB)抑制三磷酸腺苷结合盒转运体A1(ABCA1)表达及胆固醇流出中的作用,THP-1巨噬细胞源性泡沫细胞经不同浓度脂多糖(LPS)处理,活化NF-κB,或以PDTC(NF-κB抑制剂)预处理细胞后再加入LPS,实时荧光定量PCR检测细胞mi R-33s及其宿主基因胆固醇调节元件结合蛋白(SREBPs)的表达,蛋白质印迹法检测SREBPs的蛋白质表达,染色体免疫共沉淀检测NF-κB p65与SREBPs启动子区结合情况;LPS处理基础上,转染mi R-33s抑制物或mi R-33s模拟物,RT-PCR检测ABCA1 m RNA表达水平,蛋白质印迹法检测ABCA1蛋白水平,液体闪烁计数仪检测细胞内的胆固醇流出.结果显示,NF-κB活化促进mi R-33s及SREBPs的表达,使用PDTC抑制NF-κB,细胞内mi R-33s和SREBPs的表达下降;NF-κB p65可与SREBPs启动子区直接结合;转染mi R-33s抑制剂后,NF-κB活化对ABCA1的抑制作用减弱,胆固醇流出增强;相反,转染mi R-33s抑制物,NF-κB活化对ABCA1的抑制作用增强,胆固醇流出减弱.结果提示,NF-κB活化可促进mi R-33s表达,抑制ABCA1及胆固醇流出.  相似文献   

9.
三磷酸腺苷结合盒转运体A1(ABCA1)具有介导细胞内脂质流出,维持细胞脂质稳态的功能.新生的ABCA1必须经过胞内运输和各种化学修饰等过程,最终成为具有功能的成熟转运体,才能行使其转运脂质的功能,因此,ABCA1在胞内的运输过程和正确质膜定位对其介导胆固醇流出的功能至关重要.目前ABCA1相关研究主要集中于脂质转运方面,并提出各种胆固醇流出机制的模型,如通道转运模型、蘑菇状突起模型和胞吞-胞吐转运模型等.最近研究显示,ABCA1还具有调节质膜脂筏结构、参与免疫和炎症调节等新功能.本文主要针对ABCA1的胞内运输过程以及各种功能做一综述,以期为动脉粥样硬化相关疾病提供新的治疗靶点和途径.  相似文献   

10.
ABCA1与NPC1在细胞内胆固醇转运中的作用   总被引:1,自引:0,他引:1  
腺苷三磷酸结合盒转运蛋白A1(ATP-binding cassette transporter A1,ABCA1)是血浆高密度脂蛋白(high-density lipoprotein,HDL)颗粒形成之初的限速步骤。ABCA1通过膜泡运输脂质至细胞表面的HDL载脂蛋白的作用机制尚未完全阐明。C型尼曼-匹克病(Niemann-Pick disease type C,NPC)主要由NPC1基因突变引起,NPC1蛋白能促进胆固醇和其他脂质从晚期胞内体/溶酶体流入其他细胞结构。ABCA1和NPC1相互作用保持细胞内脂质平衡,与Tangier病和N C P病等病理过程密切相关。  相似文献   

11.
Mutations in the ATP-binding cassette transporter A1 (ABCA1) are a major cause of decreased HDL cholesterol (HDL-C), which infers an increased risk of cardiovascular disease (CVD). Many ABCA1 mutants show impaired localization to the plasma membrane. The aim of this study was to investigate whether the chemical chaperone, sodium 4-phenylbutyrate (4-PBA) could improve cellular localization and function of ABCA1 mutants. Nine different ABCA1 mutants (p.A594T, p.I659V, p.R1068H, p.T1512M, p.Y1767D, p.N1800H, p.R2004K, p.A2028V, p.Q2239N) expressed in HEK293 cells, displaying different degrees of mislocalization to the plasma membrane and discrete impacts on cholesterol efflux, were subject to treatment with 4-PBA. Treatment restored localization to the plasma membrane and increased cholesterol efflux function for the majority of mutants. Treatment with 4-PBA also increased ABCA1 protein expression in all transfected cell lines. In fibroblast cells obtained from low HDL-C subjects expressing two of the ABCA1 mutants (p.R1068H and p.N1800H), 4-PBA increased cholesterol efflux without any increase in ABCA1 expression. Our study is the first to investigate the effect of the chemical chaperone, 4-PBA on ABCA1 and shows that it is capable of restoring plasma membrane localization and enhancing the cholesterol efflux function of mutant ABCA1s both in vitro and ex vivo. These results suggest 4-PBA may warrant further investigation as a potential therapy for increasing cholesterol efflux and HDL-C levels.  相似文献   

12.
13.
ABCA1 mediates the transport of cellular cholesterol and phospholipids to HDL apolipoproteins. Apolipoprotein A-I (apoA-I) interactions with ABCA1-expressing cells elicit several responses, including removing cellular lipids, stabilizing ABCA1 protein, and activating Janus kinase 2 (JAK2). Here, we used synthetic apolipoprotein-mimetic peptides to characterize the relationship between these responses. Peptides containing one amphipathic helix of L- or D-amino acids (2F, D-2F, or 4F) and a peptide containing two helices (37pA) all promoted ABCA1-dependent cholesterol efflux, competed for apoA-I binding to ABCA1-expressing cells, blocked covalent cross-linking of apoA-I to ABCA1, and inhibited ABCA1 degradation. 37pA was cross-linked to ABCA1, confirming the direct binding of amphipathic helices to ABCA1. 2F, 4F, 37pA, and D-37pA all stimulated JAK2 autophosphorylation. Inhibition of JAK2 greatly reduced peptide-mediated cholesterol efflux, peptide binding to ABCA1-expressing cells, and peptide cross-linking to ABCA1, indicating that these processes require an active JAK2. In contrast, apoA-I and peptides stabilized ABCA1 protein even in the absence of an active JAK2, implying that this process is independent of JAK2 and lipid efflux-promoting binding of amphipathic helices to ABCA1. These findings show that amphipathic helices coordinate the activity of ABCA1 by several distinct mechanisms that are likely to involve different cell surface binding sites.  相似文献   

14.
Heterogeneity of high density lipoprotein generated by ABCA1 and ABCA7   总被引:2,自引:0,他引:2  
The assembly of HDL by helical apolipoprotein and cellular lipid was studied using HEK293 cells to which ecdysone-inducible human ABCA1 or human ABCA7 was transfected. Expression of both ABCA1 and ABCA7 was induced linearly proportional to ponasterone A concentration in the medium. In the experimental conditions used, the ABC protein expression levels limited the rate of lipid release when the apolipoprotein concentration was high, and the apolipoprotein concentration was rate-limiting when the ABC protein expression levels were high. When ABCA1 expression increased in conditions in which it was rate-limiting, relative cholesterol content to phospholipid increased in the HDL produced. In contrast, it was constant when ABCA7 expression increased. To investigate the background mechanism, the HDL particles were analyzed by density gradient ultracentrifugation and high performance lipid chromatography. The ABCA1-mediated reaction produced two distinct HDLs, large cholesterol-rich and small cholesterol-poor particles, and the ABCA7-mediated reaction generated mostly small cholesterol-poor particles. The increase of HDL assembly with the increase of ABCA1 expression was predominant in large cholesterol-rich particles, whereas only small cholesterol-poor HDL increased as ABCA7 expression increased. We conclude that ABCA1 generates cholesterol-rich and cholesterol-poor HDL and that the former is more prominently dependent on the increase of ABCA1 expression. ABCA7 produces this HDL subfraction only as a very minor component.  相似文献   

15.
胆固醇是细胞质膜的重要组成成分。然而,过多的胆固醇累积可导致细胞中毒。异常的胆固醇胞内迁移与蓄积是造成许多心血管疾病如动脉粥样硬化的分子基础。细胞内胆固醇稳态由胆固醇的吸收、合成及外排等一系列过程调控。在哺乳动物细胞中,调节胆固醇合成、吸收和外排是维持体内胆固醇平衡的必要生理过程。本综述着重概述了三磷酸腺苷结合盒转运体(ABC)家族,如ABCA1、ABCG1、ABCG5和ABCG8的细胞功能及生理作用,以及这些转运体在调控胆固醇胞外转运中的分子机制。  相似文献   

16.
ATP结合盒转运体A1(ABCA1)在细胞内胆固醇流出中起着重要作用,增加它的表达可促进细胞内胆固醇流出,降低动脉粥样硬化风险.ABCA1的表达受到多种因素的调控,包括肝X受体、过氧化物酶体增殖物激活受体、钙蛋白酶抑制剂和miRNA等.本文主要综述ABCA1的表达与调控,以及它对动脉粥样硬化性心血管疾病发生发展的影响,...  相似文献   

17.
It has been suggested that the signal transduction initiated by apolipoprotein A-I (apoA-I) activates key proteins involved in cholesterol efflux. ABCA1 serves as a binding partner for apoA-I, but its participation in apoA-I-induced signaling remains uncertain. We show that the exposure of human fibroblasts to ABCA1 ligands (apolipoproteins and amphipathic helical peptides) results in the generation of intracellular signals, including activation of the small G-protein Cdc42, protein kinases (PAK-1 and p54JNK), and actin polymerization. ApoA-I-induced signaling was abrogated by glyburide, an inhibitor of the ABC transporter family, and in fibroblasts from patients with Tangier disease, which do not express ABCA1. Conversely, induction of ABCA1 expression with the liver X receptor agonist, T0901317, and the retinoid X receptor agonist, R0264456, potentiated apoA-I-induced signaling. Similar effects were observed in HEK293 cells overexpressing ABCA1-green fluorescent protein (GFP) fusion protein, but not ABCA1-GFP (K939M), which fails to hydrolyze ATP, or a nonfunctional ABCA1-GFP with a truncated C terminus. We further found that Cdc42 coimmunoprecipitates with ABCA1 in ABCA1-GFP-expressing HEK293 cells exposed to apoA-I but not in cells expressing ABCA1 mutants. We conclude that ABCA1 transduces signals from apoA-I by complexing and activating Cdc42 and downstream kinases and, therefore, acts as a full apoA-I receptor.  相似文献   

18.
The ability of HDL to support macrophage cholesterol efflux is an integral part of its atheroprotective action. Augmenting this ability, especially when HDL cholesterol efflux capacity from macrophages is poor, represents a promising therapeutic strategy. One approach to enhancing macrophage cholesterol efflux is infusing blood with HDL mimics. Previously, we reported the synthesis of a functional mimic of HDL (fmHDL) that consists of a gold nanoparticle template, a phospholipid bilayer, and apo A-I. In this work, we characterize the ability of fmHDL to support the well-established pathways of cellular cholesterol efflux from model cell lines and primary macrophages. fmHDL received cell cholesterol by unmediated (aqueous) and ABCG1- and scavenger receptor class B type I (SR-BI)-mediated diffusion. Furthermore, the fmHDL holoparticle accepted cholesterol and phospholipid by the ABCA1 pathway. These results demonstrate that fmHDL supports all the cholesterol efflux pathways available to native HDL and thus, represents a promising infusible therapeutic for enhancing macrophage cholesterol efflux. fmHDL accepts cholesterol from cells by all known pathways of cholesterol efflux: unmediated, ABCG1- and SR-BI-mediated diffusion, and through ABCA1.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号