首页> 外文学位 >Lipoxygenase investigations lead to the discovery of potent inhibitors and their mechanisim of action.
【24h】

Lipoxygenase investigations lead to the discovery of potent inhibitors and their mechanisim of action.

机译:脂氧合酶的研究导致了有效抑制剂及其作用机理的发现。

获取原文
获取原文并翻译 | 示例

摘要

The research in this dissertation describes the discovery of potent and selective inhibitors of various human lipoxygenases (LOXs). The binding modes of specific inhibitors and substrates in the active site of human 15-lipoxygenase-1 (h15-LOX-1) were also investigated. Oxo-lipids, a large family of oxidized human lipoxygenase products, are involved in different inflammatory responses in the cell. Oxo-lipids contain electrophilic sites that can potentially form covalent bonds through a Michael addition mechanism with nucleophilic residues in protein active sites and increase inhibitor potency. Due to the resemblance of oxo-lipids to LOX substrates, the inhibitor potency of 4 different oxo-lipids; 5-oxo-6,8,11,14-(E,Z,Z,Z)-eicosatetraenoic acid (5-oxo-ETE), 15-oxo-5,8,11,13-(Z,Z,Z,E)-eicosatetraenoic acid (15-oxo-ETE), 12-oxo-5,8,10,14-(Z,Z,E,Z)-eicosatetraenoic acid (12-oxo-ETE), and 13-oxo-9,11-(Z,E)-octadecadienoic acid (13-oxo-ODE) were determined against a library of LOX isozymes; leukocyte 5-lipoxygenase (h5-LOX), human reticulocyte 15-lipoxygenase-1 (h15-LOX-1), human platelet 12-lipoxygenase (h12-LOX), human epithelial 15-lipoxygenase-2 (h15-LOX-2), soybean 15-lipoxygenase-1 (s15-LOX-1), and rabbit reticulocyte 15-LOX (r15-LOX). 15-oxo-ETE exhibited the highest potency against h12-LOX of all the oxo-lipids studied, with an IC50 = 1 +/- 0.1 microM and was highly selective. Time-dependent studies did not demonstrate irreversible inhibition with 15-oxo-ETE, however, this does not preclude the role of its Michael acceptor in increasing its potency. Steady-state inhibition kinetic experiments determined 15-oxo-ETE to be a mixed inhibitor against h12-LOX, with a Kic value of 0.087 +/- 0.008 microM. These data are the first observations that oxo-lipids can inhibit LOX isozymes and may be another mechanism in which LOX products regulate LOX activity.;Epithelial h15-LOX-2 is of clinical interest due to its link in the progression of macrophages to foam cells, which are present in atherosclerotic plaques. Coronary artery disease is the primary cause of deaths in men and women in America. The discovery of novel inhibitors that are selective and potent against h15-LOX-2 may aid in identifying this protein's role in heart disease. Two novel molecular scaffolds, (MLS000545091 and MLS000536924), were identified using High Throughput Screening (HTS) that are selective and potent against h15-LOX-2.;Human reticulocyte 15-Lipoxygenase-1 (h15-LOX-1 or h12/15-LOX) is a lipid-oxidizing enzyme that can directly oxidize lipid membranes in the absence of a phospholipase, leading to a direct attack on organelles, such as the mitochondria. This cytotoxic activity of h15-LOX-1 is up-regulated in neurons and endothelial cells after a stroke and thought to contribute to both neuronal cell death and blood-brain barrier leakage. Stroke is the fifth leading cause of death and the first leading cause of disability in America. The discovery of inhibitors that selectively target recombinant h15-LOX-1 in vitro, as well as possessing activity against the murine ortholog ex vivo, could potentially support a novel therapeutic strategy for the treatment of stroke. A new family of inhibitors was identified in a High Throughput Screen that are selective and potent against recombinant h15-LOX-1 and cellular mouse 15-LOX (m15-LOX). MLS000099089 (compound 99089), the parent molecule, exhibits an IC50 potency of 3.4 +/- 0.5 microM against h15-LOX-1 in vitro and an ex vivo IC50 potency of approximately 10 microM in a mouse neuronal cell line, HT-22. These data indicate that 99089 and related derivatives may serve as a starting point for the development of anti-stroke therapeutics due to their ability to selectively target h15-LOX-1 in vitro and m15-LOX ex vivo..;The discovery of h15-LOX-1 inhibitors could potentially be novel therapeutics in the treatment of stroke, however, little is known about the inhibitor/active site interaction due to the lack of a protein crystal structure. Site-directed mutagenesis and molecular modeling were utilized to gain a better structural understanding of inhibitor interactions with the active site of h15-LOX-1. Eight mutants (R402L, R404L, F414I, F414W, E356Q, Q547L, L407A, I417A) of h15-LOX-1 were generated to determine whether these active site residues interact with two structurally similar h15-LOX-1 inhibitors, a ML094 derivative and ML351. IC50 values and steady-state inhibition kinetics were determined with the eight mutants and four of the mutants affected inhibitor potency relative to wild type h15-LOX-1 (F414I, F414W, E356Q and L407A). The data indicate that the ML094 derivative and ML351 bind to similar sites in the active site but have subtle differences in their binding modes.;Lipoxygenases are capable of catalyzing dioxygenation and dehydration reactions when reacting with conjugated mono-hydroperoxy fatty acids. Dioxygenation of mono-hydroperoxide fatty acids lead to di-hydroperoxide products while dehydration of mono-hydroperoxides followed by hydrolysis lead to di-hydroxide products. The fatty acid kinetics, product profile and mechanism of product formation was investigated between WT human 15-lipoxygenase-1 and 15S-HpETE in comparison to the reaction between an active site mutant, F414I, and 15S-HpETE. The substrate affinity and kcat of 15S-HpETE turnover were comparable for both WT h15-LOX-1 and F414I. Alternatively, the substrate affinity and kcat of AA turnover varied for WT h15-LOX-1 and F414I leading to a decreased catalytic efficiency of AA turnover by F414I relative to WT h15-LOX-1. The kinetic data suggests that the active site mutant, F414I, is less efficient at turning over AA relative to WT but comparable at turning over 15S-HpETE. 5S,15S-DiHpETE, 8S,15S-DiHETE and 14R,15S-DiHpETE were identified as the major products in the reaction for both WT 15-LOX-1 and F414I, possibly indicating that this active site mutation does not significantly affect the binding mode of 15S-HpETE in the active site. Comparison of the mass spectra of the reduced and unreduced reactions of 15S-HpETE and both enzymes reveal that 14R,15S-DiHpETE and 5S,15S-DiHETE are dioxygenation products while 8S,15S-DiHETE is the hydrolyzed dehydration product. In contrast to rabbit 15-LOX, h15-LOX-1 does not react with 15S-DiHETE with or without 13S-HpODE.
机译:本文的研究描述了各种人脂氧合酶(LOXs)有效和选择性抑制剂的发现。还研究了人类15-脂氧合酶-1(h15-LOX-1)的活性位点中特定抑制剂和底物的结合模式。氧脂是氧化型人脂氧合酶产物的一大家族,参与细胞中不同的炎症反应。含氧脂质含有亲电性位点,该位点可通过迈克尔加成机理与蛋白质活性位点中的亲核残基潜在地形成共价键,并增加抑制剂的效力。由于含氧脂质与LOX底物相似,因此有4种不同含氧脂质的抑制剂效力。 5-氧代6,8,11,14-(E,Z,Z,Z)-二十碳四烯酸(5-氧代-ETE),15-氧代5,8,11,13-(Z,Z,Z ,E)-二十碳四烯酸(15-oxo-ETE),12-oxo-5,8,10,14-(Z,Z,E,Z)-二十碳四烯酸(12-oxo-ETE)和13-oxo针对LOX同工酶文库确定了-9,11-(Z,E)-十八碳二烯酸(13-氧代-ODE);白细胞5-脂氧合酶(h5-LOX),人网状细胞15-脂氧合酶-1(h15-LOX-1),人血小板12-脂氧合酶(h12-LOX),人上皮15-脂氧合酶-2(h15-LOX-2) ,大豆15-脂氧合酶-1(s15-LOX-1)和兔网织红细胞15-LOX(r15-LOX)。 15-oxo-ETE在所有研究的脂质脂质中均表现出最高的针对h12-LOX的效力,IC50 = 1 +/- 0.1 microM,具有高度选择性。时间依赖性研究并未显示15-oxo-ETE具有不可逆的抑制作用,但是,这并不排除其Michael受体在增强其效力方面的作用。稳态抑制动力学实验确定15-oxo-ETE是针对h12-LOX的混合抑制剂,Kic值为0.087 +/- 0.008 microM。这些数据是氧代脂质可以抑制LOX同工酶的第一个观察结果,并且可能是LOX产物调节LOX活性的另一种机制。上皮h15-LOX-2由于其与巨噬细胞向泡沫细胞的进展有关而具有临床意义。 ,它们存在于动脉粥样硬化斑块中。在美国,冠状动脉疾病是导致男性和女性死亡的主要原因。对h15-LOX-2具有选择性和效力的新型抑制剂的发现可能有助于鉴定该蛋白在心脏病中的作用。使用高通量筛选(HTS)鉴定了两个新颖的分子支架(MLS000545091和MLS000536924),它们对h15-LOX-2具有选择性和效力;人网状细胞15-Lipoxygenase-1(h15-LOX-1或h12 / 15 -LOX)是一种脂质氧化酶,可以在不存在磷脂酶的情况下直接氧化脂质膜,从而直接攻击细胞器,例如线粒体。中风后,h15-LOX-1的这种细胞毒性活性在神经元和内皮细胞中被上调,并被认为有助于神经元细胞死亡和血脑屏障渗漏。中风是美国的第五大死亡原因和第一大残疾原因。在体外选择性靶向重组h15-LOX-1的抑制剂的发现以及对小鼠直系同源物具有体外活性的发现可能潜在地支持了一种新的中风治疗策略。在高通量筛选中发现了一个新的抑制剂家族,这些抑制剂具有选择性,并且对重组h15-LOX-1和细胞小鼠15-LOX(m15-LOX)具有有效作用。 MLS000099089(化合物99089)是母体分子,在小鼠神经元细胞系HT-22中,在体外对h15-LOX-1的IC50效能为3.4 +/- 0.5 microM,在体外的IC50效能约为10 microM。这些数据表明,由于99089及其相关衍生物在体外和体外选择性靶向h15-LOX-1的能力,它们可以作为抗中风治疗药物开发的起点。 LOX-1抑制剂可能是治疗中风的新型疗法,但是由于缺乏蛋白质晶体结构,对抑制剂/活性部位相互作用的了解甚少。利用定点诱变和分子建模可以更好地理解抑制剂与h15-LOX-1活性位点的相互作用。产生了h15-LOX-1的八个突变体(R402L,R404L,F414I,F414W,E356Q,Q547L,L407A,I417A),以确定这些活性位点残基是否与两种结构相似的h15-LOX-1抑制剂,ML094衍生物和ML351。相对于野生型h15-LOX-1(F414I,F414W),用八个突变体和四个受影响的突变体测定抑制剂效能来确定IC50值和稳态抑制动力学,E356Q和L407A)。数据表明,ML094衍生物和ML351与活性位点的相似位点结合,但是它们的结合方式有细微的差别。脂氧化酶与共轭单氢过氧脂肪酸反应时能够催化双加氧和脱水反应。一氢过氧化物脂肪酸的双氧合导致二氢过氧化物产物,而一氢过氧化物的脱水然后水解导致二氢过氧化物产物。与活性位点突变体F414I和15S-HpETE之间的反应相比,研究了WT人15-脂加氧酶-1和15S-HpETE之间的脂肪酸动力学,产物特征和产物形成的机理。 WT h15-LOX-1和F414I的15S-HpETE转换的底物亲和力和kcat相当。或者,对于WT h15-LOX-1和F414I,AA转换的底物亲和力和kcat变化,导致相对于WT h15-LOX-1,F414I导致AA转换的催化效率降低。动力学数据表明,活性位点突变体F414I相对于WT在翻转AA方面效率较低,但在翻转15S-HpETE方面却相当。 5S,15S-DiHpETE,8S,15S-DiHETE和14R,15S-DiHpETE被确定为WT 15-LOX-1和F414I反应的主要产物,可能表明该活性位点突变不会显着影响结合活动站点中的15S-HpETE模式。比较15S-HpETE和两种酶的还原反应和未还原反应的质谱,发现14R,15S-DiHpETE和5S,15S-DiHETE是双加氧产物,而8S,15S-DiHETE是水解的脱水产物。与兔子15-LOX相反,在有或没有13S-HpODE的情况下,h15-LOX-1都不与15S-DiHETE反应。

著录项

  • 作者

    Armstrong, Michelle Marie.;

  • 作者单位

    University of California, Santa Cruz.;

  • 授予单位 University of California, Santa Cruz.;
  • 学科 Biochemistry.;Chemistry.
  • 学位 Ph.D.
  • 年度 2016
  • 页码 180 p.
  • 总页数 180
  • 原文格式 PDF
  • 正文语种 eng
  • 中图分类
  • 关键词

相似文献

  • 外文文献
  • 中文文献
  • 专利
获取原文

客服邮箱:kefu@zhangqiaokeyan.com

京公网安备:11010802029741号 ICP备案号:京ICP备15016152号-6 六维联合信息科技 (北京) 有限公司©版权所有
  • 客服微信

  • 服务号