首页> 外文学位 >Using novel small molecule modulators as a tool to elucidate the role of the Myocyte Enhancer Factor 2 (MEF2) family of transcription factors in leukemia.
【24h】

Using novel small molecule modulators as a tool to elucidate the role of the Myocyte Enhancer Factor 2 (MEF2) family of transcription factors in leukemia.

机译:使用新型小分子调节剂作为阐明白血病中转录因子肌细胞增强因子2(MEF2)家族作用的工具。

获取原文
获取原文并翻译 | 示例

摘要

The Myocyte Enhancer Factor 2 (MEF2) family of transcription factors are DNA-bound proteins that regulate gene expression via their interaction with co-regulatory proteins. This interaction between MEF2 and its co-factors is necessary for the development and proper function of many mammalian cell types, and is a major factor in the onset and progression of many cancers, specifically leukemia. Understanding the MEF2:co-factor interaction in leukemia, which is often times an epigenetic event, is of utmost importance to those searching for therapies that have high specificity and low chance of side effects.;In the first part of this paper, the structural characteristics of the MEF2 family that allow its interaction with co-regulators is examined. The study revealed that the structure of MEF2 proteins are not majorly altered by the binding of a co-regulator, such as class IIa histone deacetylases (HDACs), Cabin1, or p300, at the protein-protein interface, and are inherently able to be bound while interacting with DNA. This overruled previous notions that the hydrophobic groove, or "binding pocket", of MEF2 was not present unless it was interacting with a co-regulator.;The second, and most involved, study of this paper examines the interaction of MEF2 and class IIa HDACs in leukemia. This study was aided by the availability of a small molecule benzamide compound NKL-30, which we call a MEF2 modulator (MEF2m), that was used as a tool to observe the consequences of inhibiting the protein-protein interaction both in vitro and in vivo. Our MEF2m were able to preferentially decrease the viability of leukemia cells over non-cancerous control B cells. We were also able to show that our compound was able to ablate the MEF2:class IIa HDAC interaction and result in the cytoplasmic shuttling of these co-repressors. Additionally, we were able to show that the expression of the protein NR4A1/Nur77, a MEF2 target gene associated with apoptosis, increased after treatment with NKL-30, likely because of the derepression of MEF2 afforded by the exodus of class IIa HDACs from the nucleus. Finally, we were able to show in a mammalian leukemia model utilizing Nalm6 cells, that mice treated with NKL-30 enjoy an increased lifespan compared to those receiving vehicle control injections.;The third and final part of this work involves the gene expression analysis of B cells that have been treated with our MEF2m. Both healthy and leukemic B cells were treated with our small molecule compound or a solvent control and then data was collected and analyzed using mRNA-seq. We have shown that our MEF2m often preferentially activates gene expression, possibly due to the derepression seen on MEF2 and additional off-target effects. While these differentially regulated genes seen after drug treatment do not fit a single profile or pathway, we do know that the treatment of leukemia cells with our MEF2m usually results in death. Therefore, this data is crucial in our quest for understanding this phenomenon. This work is still ongoing and can be massaged for more data and relationships with the help of future replication and analyses.;In summation, the work presented in this dissertation addresses the possibility of a small molecule compound to specifically target leukemia cells for death over healthy cells due to its ability to bind to MEF2 proteins, disallowing the binding of co-repressors, and resulting in an epigenetic alteration event. The mechanism and consequence of this event, while not yet completely understood, is detailed within the following chapters and the data suggest that we are on the right track toward understanding a way to develop targeted therapies for previously untreatable or high mortality-rate diseases.
机译:肌细胞增强因子2(MEF2)家族的转录因子是与DNA结合的蛋白,它们通过与共同调节蛋白的相互作用来调节基因表达。 MEF2及其辅助因子之间的这种相互作用对于许多哺乳动物细胞类型的发育和正常功能是必需的,并且是许多癌症(尤其是白血病)的发作和发展的主要因素。了解白血病中经常发生的表观遗传事件MEF2:辅助因子相互作用,对于那些寻找具有高特异性和低副作用可能性的疗法而言,是至关重要的。在本文的第一部分,结构研究了MEF2家族允许其与共调节剂相互作用的特性。研究表明,MEF2蛋白质的结构在蛋白质-蛋白质界面上不会受到共同调节剂(例如IIa类组蛋白脱乙酰基酶(HDACs),Cabin1或p300)的结合的主要改变,并且固有地能够被与DNA相互作用时结合。这推翻了先前的观点,即除非MEF2与共调节剂相互作用,否则不存在MEF2的疏水沟或“结合口袋”。;第二,也是最涉及的研究是本文研究了MEF2和IIa类的相互作用。白血病中的HDAC。这项研究得益于小分子苯甲酰胺化合物NKL-30(我们称为MEF2调节剂(MEF2m))的可用性,该化合物用作观察抑制体内外蛋白质相互作用的后果的工具。我们的MEF2m能够比非癌性对照B细胞优先降低白血病细胞的活力。我们还能够证明我们的化合物能够消除MEF2:IIa类HDAC相互作用,并导致这些共阻遏物的胞质穿梭。此外,我们能够显示与细胞凋亡相关的MEF2靶基因蛋白NR4A1 / Nur77的表达在用NKL-30处理后增加了,这可能是由于IIa类HDAC的外流提供了对MEF2的抑制作用。核。最后,我们能够在利用Nalm6细胞的哺乳动物白血病模型中证明,与接受媒介物对照注射的小鼠相比,经NKL-30治疗的小鼠的寿命更长。;这项工作的第三部分也是最后一部分涉及用我们的MEF2m处理过的B细胞。健康和白血病B细胞均用我们的小分子化合物或溶剂对照处理,然后收集数据并使用mRNA-seq分析。我们已经表明,我们的MEF2m通常会优先激活基因表达,这可能是由于在MEF2上出现的抑制和其他脱靶作用所致。尽管在药物治疗后看到的这些差异调节基因不符合单个特征或途径,但我们确实知道用我们的MEF2m治疗白血病细胞通常会导致死亡。因此,这些数据对于我们寻求了解这种现象至关重要。这项工作仍在进行中,可以在将来的复制和分析的帮助下获得更多的数据和关系。总而言之,本文提出的工作解决了小分子化合物特异性靶向白血病细胞导致健康死亡的可能性。细胞由于其结合MEF2蛋白的能力,不允许共抑制因子的结合,并导致表观遗传学改变。在下一章中将详细介绍该事件的机制和后果,尽管尚未完全了解,但数据表明我们在正确的方向上了解开发针对先前无法治愈或高死亡率的疾病的靶向疗法的方法。

著录项

  • 作者

    Philips, Michael A.;

  • 作者单位

    University of Southern California.;

  • 授予单位 University of Southern California.;
  • 学科 Biology Molecular.
  • 学位 Ph.D.
  • 年度 2013
  • 页码 148 p.
  • 总页数 148
  • 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