首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 187 毫秒
1.
二酮酸类化合物(DKAs)是目前最有前景的HIV-1整合酶(integrase, IN)抑制剂.为了解DKAs引起的多种耐药株共有的耐药性机理,选择3种S-1360引起的IN耐药突变体,用分子对接和分子动力学模拟,研究了野生型和突变型IN与S-1360的结合模式,基于该结合模式探讨了3种耐药突变体所共有的耐药性机理.结果表明:在突变体中,S-1360结合到耐药突变IN核心区中的位置靠近功能loop 3区却远离与 DNA结合的关键残基,结合位置不同导致S-1360的抑制作用部分丧失;残基138到166区域的柔性对IN发挥生物学功能很重要,S-1360能与DNA结合的关键残基N155及K159形成氢键,这2个氢键作用降低了该区域的柔性,突变体中无类似氢键,因而该区域柔性增高;在突变体中,S-1360的苯环远离病毒DNA结合区,不能阻止病毒DNA末端暴露给宿主DNA;T66I突变导致残基Ⅰ的长侧链占据IN的活性口袋,阻止抑制剂以与野生型中相同的方式结合到活性中心,这均是产生抗药性的重要原因.这些模拟结果与实验结果吻合,可为抗IN的抑制剂设计和改造提供帮助.  相似文献   

2.
HIV-1整合酶催化病毒DNA与宿主细胞基因组整合,是病毒复制所需的关键酶之一,也是抗病毒药物研发的重要靶点.IN及其核心结构域均能在体外催化去整合反应.本研究表达纯化了IN和IN-CCD蛋白,建立了一种检测IN和IN-CCD去整合活性的微孔板式高通量方法.设计了生物素和地高辛修饰的去整合DNA底物,运用链亲和素标记的珠子捕获反应产物,再通过酶标地高辛抗体及随后的酶联免疫吸附实验方法对地高辛定量以检测去整合.结果显示,IN和IN-CCD催化的去整合反应信号(A405)分别达到1.6和1.2,而背景信号值低于0.05;IN去整合反应更倾向于使用Mn2+而不是Mg2+作为金属辅助离子;研究还发现,已知的IN抑制剂baicalein是IN-CCD抑制剂.以上结果表明,本工作建立的检测方法能高通量、高灵敏度和高特异性地研究去整合反应,并能够应用于以IN为靶点,特别是以IN-CCD为靶点的HIV抑制剂的筛选.  相似文献   

3.
病毒DNA整合到被感染的宿主细胞的染色体上需要逆转录病毒基因组的有效复制,而这一反应是由病毒编码的酶——整合酶(Inte-grase,IN)调节的。由于IN在逆转录病毒生命周期的早期阶段起着关键的作用,所以它已经成了对HIV-1基因治疗的一个非常吸引人的靶蛋白。由于对这个酶缺少有效的抑制剂,人们开始探讨新的抑制其活性的方法,包括抗体的使  相似文献   

4.
HIV-1复制需要HIV-1整合酶将其环状DNA整合进宿主DNA中,这其中包括2个重要反应,即“3′-加工”和“链转移”,两者均由HIV-1整合酶催化完成.阻断其中的任一反应,都能达到抑制HIV-1复制的目的.因此,了解HIV-1整合酶的完整结构和聚合状态,对深入探讨其作用机理及设计新型抑制剂具有重要的指导作用.然而,迄今为止仅有HIV-1整合酶单独结构域的晶体结构可供参考,而其全酶晶体结构尚未获得解析.本研究利用分子模拟技术,通过蛋白质 蛋白质/DNA分子对接、动力学模拟等方法,构建了全长整合酶四聚体的结构模型、HIV-1 DNA与整合酶复合物的结构模型,进一步从理论上证实HIV-1整合酶是以四聚体形态发挥催化作用,明确“3′-加工”和“链转移”在HIV-1整合酶上的催化位点.同时,通过与作用机理相似的细菌转座子Tn5转座酶等的结构比对,推测HIV-1整合酶的核心结构域中应有第2个Mg2+存在,其位置螯合于Asp64与Glu152之间.在HIV-1整合酶结构研究的基础上,有望进一步设计出新的抗艾滋病药物.  相似文献   

5.
HIV-1整合酶催化病毒cDNA与宿主细胞基因组DNA的整合,是病毒在宿主细胞中增殖的一个关键酶。 3'加工是整合酶催化整合过程的第一步反应,3'加工反应动力学的研究对整合酶催化机理研究和以整合酶为靶点的药物研发都具有重要意义。构建了野生型HIV-1整合酶重组质粒,在大肠杆菌BL21中诱导表达,通过对包涵体变性、复性,纯化得到了整合酶蛋白。 基于分子信标原理,设计了荧光和淬灭基团标记的DNA底物,通过荧光信号实时监测3' 加工反应,对酶反应的动力学性质进行研究。 结果表明,纯化的整合酶蛋白具有较高的活性,酶反应表现出显著的Mg2+偏好性。 酶动力学研究 (Km = 131.79 nmol/L,Kcat = 0.0042 min -1) 表明,该分子信标方法和设计的DNA底物可用于整合酶3'加工反应动力学研究以及酶反应性质的研究。  相似文献   

6.
HIV-1整合酶催化病毒cDNA与宿主细胞基因组DNA的整合,是病毒在宿主细胞中增殖的一个关键酶.3'加工是整合酶催化整合过程的第一步反应,3'加工反应动力学的研究对整合酶催化机理研究和以整合酶为靶点的药物研发都具有重要意义.构建了野生型HIV-1整合酶重组质粒,在大肠杆菌BL21中诱导表达,通过对包涵体变性、复性,纯化得到了整合酶蛋白.基于分子信标原理,设计了荧光和淬灭基团标记的DNA底物,通过荧光信号实时监测3'加工反应,对酶反应的动力学性质进行研究.结果表明,纯化的整合酶蛋白具有较高的活性,酶反应表现出显著的Mg2+偏好性.酶动力学研究(Km=131.79 nmol/L,Kcat=0.0042 min-1)表明,该分子信标方法和设计的DNA底物可用于整合酶3'加工反应动力学研究以及酶反应性质的研究.  相似文献   

7.
HIV-1整合酶是由HIV病毒pol基因编码的分子量为32KD的蛋白质,是HIV病毒复制的必需酶之一,它催化病毒DNA整合入宿主染色体DNA。人类细胞中没有HIV 整合酶的类似物[1],理论上抑制整合酶对人体副作用很小。因此HIV-1整合酶成为继HIV-1蛋白酶,逆转录酶后治疗艾滋病的富有吸引力和合理的靶标。本文综述了HIV整合酶结构,抑制剂的研究以及以HIV-1 整和酶为靶点治疗AIDS方法的最新研究进展。  相似文献   

8.
人类免疫缺陷病毒(HIV-1)整合酶抑制剂筛选及其活性测定   总被引:1,自引:0,他引:1  
整合酶作用的病毒DNA整合进宿主DNA的过程是反转录病毒在宿主细胞中增殖的关键步骤.由于在正常人类细胞中不存在相似的功能蛋白,其抑制剂对人体的副作用可能很小,相对于经典AIDS治疗药物的毒副作用,整合酶抑制剂理论上要具有优势.在线性七肽库中筛选与整合酶有特异结合作用的噬菌体展示肽,选取TPSHSSR和HPERATL 2条肽,它们可以竞争性地抑制展示相应肽段的噬菌体与整合酶的结合,同时它们对整合酶的整合活性也有一定程度的抑制,半数抑制率分别为IC50=(54.56±5.18)μmol/L,IC50=(28.29±1.32)μmol/L.这些多肽可用于治疗艾滋病新药的开发应用及整合酶结构及作用机制的研究.  相似文献   

9.
前期工作已用分子对接方法获得了HIV-1整合酶与L708,906抑制剂分子的复合物模型(IN_L708,906),现从距离、能量和氢键三个方面详细地分析了IN_L708,906模型中的关键残基.结果表明,复合物模型与蛋白质晶体库中整合酶(IN)与5CITEP的结合模式相近.用主成分分析和动力学交叉相关图方法分别研究了IN_L708,906复合物模型和IN单体的运动模式及相关性差异.计算结果显示,L708,906抑制剂的结合使得IN功能loop区残基柔性下降、分子规律性运动的丧失及集团运动相关性的无序增加,这些可能是酶活性下降的主要因素.模拟结果将有利于基于芳香二酮酸类的抗HIV药物设计.  相似文献   

10.
马传贫病毒是反转录病毒科慢病毒属的成员之一,它在体内要通过反转录酶的作用合成DNA,整合到宿主的基因组中进行复制,由于反转录酶没有校正功能,使病毒在体内复制过程中错误拷贝RNA基因组,导致高频率的遗传变异。近年国内外学者对EIAV的研究发现变异的基因主要集中在env、gag、LTR和S2等区域,这些区域基因的变异与病毒的毒力、病毒在体内的复制水平及免疫原性密切相关。由于EIAV是慢病毒中最简单的病毒,它具有独特的发病进程和明显的病程分界,使其成为研究包括HIV内其它慢病毒基因变异与临床症状之间相互关系的理想动物模型。因此对EIAV基因变异情况的研究具有重要意义。  相似文献   

11.
The HIV-1 Integrase protein (IN) mediates the integration of the viral cDNA into the host genome. IN is an emerging target for anti-HIV drug design, and the first IN-inhibitor was recently approved by the FDA. We have developed a new approach for inhibiting IN by "shiftides": peptides derived from its cellular binding protein LEDGF/p75 that inhibit IN by shifting its oligomerization equilibrium from the active dimer to an inactive tetramer. In addition, we described two peptides derived from the HIV-1 Rev protein that interact with IN and inhibit its activity in vitro and in cells. In the current study, we show that the Rev-derived peptides also act as shiftides. Analytical gel filtration and cross-linking experiments showed that IN was dimeric when bound to the viral DNA, but tetrameric in the presence of the Rev-derived peptides. Fluorescence anisotropy studies revealed that the Rev-derived peptides inhibited the DNA binding of IN. The Rev-derived peptides inhibited IN catalytic activity in vitro in a concentration-dependent manner. Inhibition was much more significant when the peptides were added to free IN before it bound the viral DNA than when the peptides were added to a preformed IN-DNA complex. This confirms that the inhibition is due to the ability of the peptides to shift the oligomerization equilibrium of the free IN toward a tetramer that binds much weaker to the viral DNA. We conclude that protein-protein interactions of IN may serve as a general valuable source for shiftide design.  相似文献   

12.
A tetramer of HIV-1 integrase (IN) stably associates with the viral DNA ends to form a fully functional concerted integration intermediate. LEDGF/p75, a key cellular binding partner of the lentiviral enzyme, also stabilizes a tetrameric form of IN. However, functional assays have indicated the importance of the order of viral DNA and LEDGF/p75 addition to IN for productive concerted integration. Here, we employed Förster Resonance Energy Transfer (FRET) to monitor assembly of individual IN subunits into tetramers in the presence of viral DNA and LEDGF/p75. The IN–viral DNA and IN–LEDGF/p75 complexes yielded significantly different FRET values suggesting two distinct IN conformations in these complexes. Furthermore, the order of addition experiments indicated that FRET for the preformed IN–viral DNA complex remained unchanged upon its subsequent binding to LEDGF/p75, whereas pre-incubation of LEDGF/p75 and IN followed by addition of viral DNA yielded FRET very similar to the IN–LEDGF/p75 complex. These findings provide new insights into the structural organization of IN subunits in functional concerted integration intermediates and suggest that differential multimerization of IN in the presence of various ligands could be exploited as a plausible therapeutic target for development of allosteric inhibitors.  相似文献   

13.
Integration of the retrovirus linear DNA genome into the host chromosome is an essential step in the viral replication cycle, and is catalyzed by the viral integrase (IN). Evidence suggests that IN functions as a dimer that cleaves a dinucleotide from the 3′ DNA blunt ends while a dimer of dimers (tetramer) promotes concerted integration of the two processed ends into opposite strands of a target DNA. However, it remains unclear why a dimer rather than a monomer of IN is required for the insertion of each recessed DNA end. To help address this question, we have analyzed crystal structures of the Rous sarcoma virus (RSV) IN mutants complete with all three structural domains as well as its two-domain fragment in a new crystal form at an improved resolution. Combined with earlier structural studies, our results suggest that the RSV IN dimer consists of highly flexible N-terminal domains and a rigid entity formed by the catalytic and C-terminal domains stabilized by the well-conserved catalytic domain dimerization interaction. Biochemical and mutational analyses confirm earlier observations that the catalytic and the C-terminal domains of an RSV IN dimer efficiently integrates one viral DNA end into target DNA. We also show that the asymmetric dimeric interaction between the two C-terminal domains is important for viral DNA binding and subsequent catalysis, including concerted integration. We propose that the asymmetric C-terminal domain dimer serves as a viral DNA binding surface for RSV IN.  相似文献   

14.
A tetramer model for human immunodeficiency virus type 1 (HIV-1) integrase (IN) with DNA representing long terminal repeat (LTR) termini was previously assembled to predict the IN residues that interact with the LTR termini; these predictions were experimentally verified for nine amino acid residues [Chen, A., Weber, I. T., Harrison, R. W. & Leis, J. (2006). Identification of amino acids in HIV-1 and avian sarcoma virus integrase subsites required for specific recognition of the long terminal repeat ends. J. Biol. Chem., 281, 4173-4182]. In a similar strategy, the unique amino acids found in avian sarcoma virus IN, rather than HIV-1 or Mason-Pfizer monkey virus IN, were substituted into the structurally related positions of HIV-1 IN. Substitutions of six additional residues (Q44, L68, E69, D229, S230, and D253) showed changes in the 3′ processing specificity of the enzyme, verifying their predicted interaction with the LTR DNA. The newly identified residues extend interactions along a 16-bp length of the LTR termini and are consistent with known LTR DNA/HIV-1 IN cross-links. The tetramer model for HIV-1 IN with LTR termini was modified to include two IN binding domains for lens-epithelium-derived growth factor/p75. The target DNA was predicted to bind in a surface trench perpendicular to the plane of the LTR DNA binding sites of HIV-1 IN and extending alongside lens-epithelium-derived growth factor. This hypothesis is supported by the in vitro activity phenotype of HIV-1 IN mutant, with a K219S substitution showing loss in strand transfer activity while maintaining 3′ processing on an HIV-1 substrate. Mutations at seven other residues reported in the literature have the same phenotype, and all eight residues align along the length of the putative target DNA binding trench.  相似文献   

15.
The HIV-1 Rev and integrase (IN) proteins control important functions in the viral life cycle. We have recently discovered that the interaction between these proteins results in inhibition of IN enzymatic activity. Peptides derived from the Rev and IN binding interfaces have a profound effect on IN catalytic activity: Peptides derived from Rev inhibit IN, while peptides derived from IN stimulate IN activity by inhibiting the Rev-IN interaction. This inhibition leads to multi integration, genomic instability and specific death of virus-infected cells. Here we used protein docking combined with refinement and energy function ranking to suggest a structural model for the Rev-IN complex. Our results indicate that a Rev monomer binds IN at two sites that match our experimental binding data: (1) IN residues 66-80 and 118-128; (2) IN residues 174-188. According to our model, IN binds Rev and its cellular cofactor, lens epithelium derived growth factor (LEDGF), through overlapping interfaces. This supports previous observations that IN is regulated by a tight interplay between Rev and LEDGF. Rev may bind either the IN dimer or tetramer. Accordingly, Rev is suggested to inhibit IN by two possible mechanisms: (i) shifting the oligomerization equilibrium of IN from an active dimer to an inactive tetramer; (ii) displacing LEDGF from IN, resulting in inhibition of IN binding to the viral DNA. Our model is expected to contribute to the development of lead compounds that inhibit the Rev-IN interaction and thus lead to multi-integration of viral cDNA and consequently to apoptosis of HIV-1 infected cells.  相似文献   

16.
17.
The HIV-1 integrase enzyme (IN) catalyzes integration of viral DNA into the host genome. We previously developed peptides that inhibit IN in vitro and HIV-1 replication in cells. Here we present the design, synthesis and evaluation of several derivatives of one of these inhibitory peptides, the 20-mer IN1. The peptide corresponding to the N-terminal half of IN1 (IN1 1–10) was easier to synthesize and much more soluble than the 20-mer IN1. IN1 1–10 bound IN with improved affinity and inhibited IN activity as well as HIV replication and integration in infected cells. While IN1 bound the IN tetramer, its shorter derivatives bound dimeric IN. Mapping the peptide binding sites in IN provided a model that explains this difference. We conclude that IN1 1–10 is an improved lead compound for further development of IN inhibitors.  相似文献   

18.
Pandey KK  Bera S  Grandgenett DP 《Biochemistry》2011,50(45):9788-9796
The assembly mechanism for the human immunodeficiency virus type 1 (HIV) synaptic complex (SC) capable of concerted integration is unknown. Molecular and structural studies have established that the HIV SC and prototype foamy virus (PFV) intasome contain a tetramer of integrase (IN) that catalyzes concerted integration. HIV IN purified in the presence of 1 mM EDTA and 10 mM MgSO(4) was predominately a monomer. IN efficiently promoted concerted integration of micromolar concentrations of 3'-OH recessed and blunt-ended U5 long terminal repeat (LTR) oligonucleotide (ODN) substrates (19-42 bp) into circular target DNA. Varying HIV IN to U5 DNA showed that an IN dimer:DNA end molar ratio of 1 was optimal for concerted integration. Integration activities decreased with an increasing length of the ODN, starting from the recessed 18/20 or 19/21 bp set to the 31/33 and 40/42 bp set. Under these conditions, the average fidelity for the HIV 5 bp host site duplication with recessed and blunt-ended substrates was 56%. Modifications of U5 LTR sequences beyond 21 bp from the terminus on longer DNA (1.6 kb) did not alter the ~32 bp DNaseI protective footprint, suggesting viral sequences beyond 21 bp were not essential for IN binding. The results suggest IN binds differentially to an 18/20 bp than to a 40/42 bp ODN substrate for concerted integration. The HIV IN monomer may be a suitable candidate for attempting crystallization of an IN-DNA complex in the absence or presence of strand transfer inhibitors.  相似文献   

19.
Integration of the HIV-1 cDNA into the human genome is catalyzed by the viral integrase (IN) protein. Several studies have shown the importance of cellular cofactors that interact with integrase and affect viral integration and infectivity. In this study, we produced a stable complex between HIV-1 integrase, viral U5 DNA, the cellular cofactor LEDGF/p75 and the integrase binding domain of INI1 (INI1-IBD), a subunit of the SWI/SNF chromatin remodeling factor. The stoichiometry of the IN/LEDGF/INI1-IBD/DNA complex components was found to be 4/2/2/2 by mass spectrometry and Fluorescence Correlation Spectroscopy. Functional assays showed that INI1-IBD inhibits the 3′ processing reaction but does not interfere with specific viral DNA binding. Integration assays demonstrate that INI1-IBD decreases the amount of integration events but inhibits by-product formation such as donor/donor or linear full site integration molecules. Cryo-electron microscopy locates INI1-IBD within the cellular DNA binding site of the IN/LEDGF complex, constraining the highly flexible integrase in a stable conformation. Taken together, our results suggest that INI1 could stabilize the PIC in the host cell, by maintaining integrase in a stable constrained conformation which prevents non-specific interactions and auto integration on the route to its integration site within nucleosomes, while LEDGF organizes and stabilizes an active integrase tetramer suitable for specific vDNA integration. Moreover, our results provide the basis for a novel type of integrase inhibitor (conformational inhibitor) representing a potential new strategy for use in human therapy.  相似文献   

20.
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