WDR5

WDR5
  • 文章类型: Journal Article
    核小体重塑和脱乙酰酶(NuRD)复合物在染色质调节和转录抑制中起关键作用。在老鼠身上,甲基-CpG结合结构域3同工型C(MBD3C)与组蛋白H3结合蛋白WD含重复序列的蛋白5(WDR5)特异性相互作用,并形成WDR5-MBD3C/NuRD复合物。尽管这种相互作用对胚胎干细胞基因调控具有功能意义,WDR5识别MBD3C的分子机制仍然难以捉摸。这里,我们确定了WDR5与MBD3C蛋白衍生的肽(残基40-51)复合的晶体结构,分辨率为1.9µ。结构分析显示,MBD3C利用独特的结合模式与WDR5相互作用,其中MBD3CArg43和Phe47分别参与识别WDR5小表面上的WDR5相互作用(WIN)位点和Tyr191相关B位点。值得注意的是,该结合诱导WDR5Tyr191~91°旋转,产生疏水B位点。此外,突变实验结合等温滴定量热法(ITC)测定证实了Arg43和Phe47在介导WDR5结合亲和力中的重要性.通过确定与WDR5结合的各种肽的结构,我们证明了WDR5WIN位点和B位点可以同时被含有\'\'Arg-Cys/Ser-Arg-Val-Phe\'共有序列的WIN基序肽识别。总的来说,这项研究揭示了WDR5-MBD3C亚复合物形成的结构基础,并为WDR5对WIN基序的识别模式提供了新的见解。此外,这些发现揭示了WDR5靶向抗癌小分子抑制剂或肽模拟药物的基于结构的设计.
    The nucleosome remodeling and deacetylase (NuRD) complex plays a pivotal role in chromatin regulation and transcriptional repression. In mice, methyl-CpG binding domain 3 isoform C (MBD3C) interacts specifically with the histone H3 binding protein WD repeat-containing protein 5 (WDR5) and forms the WDR5-MBD3C/Norde complex. Despite the functional significance of this interaction on embryonic stem cell gene regulation, the molecular mechanism underlying MBD3C recognition by WDR5 remains elusive. Here, we determined the crystal structure of WDR5 in complex with the peptide (residues 40-51) derived from the MBD3C protein at a resolution of 1.9 Å. Structural analysis revealed that MBD3C utilizes a unique binding mode to interact with WDR5, wherein MBD3C Arg43 and Phe47 are involved in recognizing the WDR5-interacting (WIN) site and Tyr191-related B site on the small surface of WDR5, respectively. Notably, the binding induces a ∼91° rotation of WDR5 Tyr191, generating the hydrophobic B site. Furthermore, mutation experiments combined with isothermal titration calorimetry (ITC) assays confirmed the importance of both Arg43 and Phe47 in mediating WDR5 binding affinity. By determining structures of various peptides bound to WDR5, we demonstrated that the WDR5 WIN site and B site can be concurrently recognized by WIN motif peptides containing \'\'Arg-Cies/Ser-Arg-Val-Phe\'\' consensus sequence. Overall, this study reveals the structural basis for the formation of the WDR5-MBD3C subcomplex and provides new insights into the recognition mode of WDR5 for the WIN motif. Moreover, these findings shed light on structural-based designs of WDR5-targeted anti-cancer small molecule inhibitors or peptide-mimic drugs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    聚(ADP-核糖)聚合酶-1(PARP1)是将带负电荷的聚(ADP-核糖)(PAR)附着到自身和其他靶蛋白上的核蛋白。虽然它在DNA损伤修复中的功能已经确立,其在靶染色质识别和基因表达调控中的作用还有待进一步了解。该研究表明PARP1通过直接与WDR5结合而与SET1/MLL复合物相互作用。值得注意的是,虽然PARP1不调节WDR5PARA化或H3K4甲基化的整体水平,它对WDR5结合和H3K4甲基化发挥基因座特异性作用。有趣的是,PARP1和WDR5在染色质上显示出广泛的共定位,WDR5促进PARP1调控的靶基因的识别和表达。此外,我们证明了WDR5Win位点的抑制阻碍了PARP1和WDR5之间的相互作用,从而抑制了PARP1与靶基因的结合.最后,WDR5Win位点和PARP的联合抑制显示出对癌细胞增殖的显著抑制作用。这些发现阐明了染色质识别的复杂机制,基因转录,和肿瘤发生,阐明PARP1和WDR5在这些过程中以前无法识别的角色。
    Poly (ADP-ribose) polymerase-1 (PARP1) is a nuclear protein that attaches negatively charged poly (ADP-ribose) (PAR) to itself and other target proteins. While its function in DNA damage repair is well established, its role in target chromatin recognition and regulation of gene expression remains to be better understood. This study showed that PARP1 interacts with SET1/MLL complexes by binding directly to WDR5. Notably, although PARP1 does not modulate WDR5 PARylation or the global level of H3K4 methylation, it exerts locus-specific effects on WDR5 binding and H3K4 methylation. Interestingly, PARP1 and WDR5 show extensive co-localization on chromatin, with WDR5 facilitating the recognition and expression of target genes regulated by PARP1. Furthermore, we demonstrated that inhibition of the WDR5 Win site impedes the interaction between PARP1 and WDR5, thereby inhibiting PARP1 from binding to target genes. Finally, the combined inhibition of the WDR5 Win site and PARP shows a profound inhibitory effect on the proliferation of cancer cells. These findings illuminate intricate mechanisms underlying chromatin recognition, gene transcription, and tumorigenesis, shedding light on previously unrecognized roles of PARP1 and WDR5 in these processes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    染色质相关蛋白WD重复结构域5(WDR5)是癌症药物发现的有希望的靶标,大多数努力阻断蛋白质上的精氨酸结合腔,称为“胜利”位点,将WDR5连接到染色质。WIN位点抑制剂(WINi)在体外对多种癌细胞类型具有活性,其中最值得注意的是源自MLL重排(MLLr)白血病的那些。肽模拟物WINi最初被提议通过与造血干细胞扩增相关的基因的失调来抑制MLLr细胞。我们对小分子WINi的发现和审讯,然而,揭示了它们在MLLr细胞系中起作用以抑制核糖体蛋白基因(RPG)转录,诱导核仁应力,激活p53。因为没有专门针对RPG表达的抗癌策略的先例,我们采用综合的多组学方法来进一步研究WINi在人类MLLr癌细胞中的作用机制。我们证明WINi诱导核糖体存量的消耗,伴随着广泛而适度的翻译窒息和可变mRNA剪接的变化,使p53拮抗剂MDM4失活。我们还表明,WINi与包括venetoclax和BET-bromodomain抑制剂在内的药物具有协同作用。一起,这些研究强化了这样一个概念,即WINi是一种新型的核糖体导向抗癌疗法,并为支持其在MLLr白血病和其他恶性肿瘤中的临床应用提供了资源.
    The chromatin-associated protein WD Repeat Domain 5 (WDR5) is a promising target for cancer drug discovery, with most efforts blocking an arginine-binding cavity on the protein called the \'WIN\' site that tethers WDR5 to chromatin. WIN site inhibitors (WINi) are active against multiple cancer cell types in vitro, the most notable of which are those derived from MLL-rearranged (MLLr) leukemias. Peptidomimetic WINi were originally proposed to inhibit MLLr cells via dysregulation of genes connected to hematopoietic stem cell expansion. Our discovery and interrogation of small-molecule WINi, however, revealed that they act in MLLr cell lines to suppress ribosome protein gene (RPG) transcription, induce nucleolar stress, and activate p53. Because there is no precedent for an anticancer strategy that specifically targets RPG expression, we took an integrated multi-omics approach to further interrogate the mechanism of action of WINi in human MLLr cancer cells. We show that WINi induce depletion of the stock of ribosomes, accompanied by a broad yet modest translational choke and changes in alternative mRNA splicing that inactivate the p53 antagonist MDM4. We also show that WINi are synergistic with agents including venetoclax and BET-bromodomain inhibitors. Together, these studies reinforce the concept that WINi are a novel type of ribosome-directed anticancer therapy and provide a resource to support their clinical implementation in MLLr leukemias and other malignancies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:在有丝分裂期间,细胞依赖于复制染色体的适当附着和分离来产生两个相同的后代。在由MYC癌基因的过度表达或失调定义的癌症中,该过程变得受损,导致基因组不稳定和肿瘤进化。最近发现染色质调节剂WDR5-一种关键的MYC辅因子-通过与主激酶PDPK1的直接相互作用来调节有丝分裂中所需基因的表达。然而,PDPK1和WDR5在过表达MYC的癌症中是否有助于类似的有丝分裂基因调控仍不清楚.因此,表征WDR5和PDPK1对MYC水平高的细胞中有丝分裂基因表达的影响,我们在MYCN扩增定义的神经母细胞瘤细胞系中进行了比较转录组学分析,这导致高细胞水平的N-MYC蛋白。
    结果:使用RNA-seq分析,我们在多种工程化CHP-134神经母细胞瘤细胞系中鉴定了N-MYC和PDPK1调控的基因,并将它们与之前发表的在抑制WDR5后CHP-134细胞中收集的基因表达数据进行了比较.我们发现,正如预期的那样,N-MYC调节多种基因,包括与有丝分裂有关的,但是PDPK1调节参与发育的特定基因集,信令,和有丝分裂。对N-MYC-和PDPK1调节基因的分析揭示了一小组与纺锤体极形成和染色体分离相关的共同控制基因。与也受WDR5调控的基因重叠。我们还发现N-MYC通过WDR5-PDPK1相互作用与PDPK1物理相互作用,表明有丝分裂基因表达的调节可以通过N-MYC-WDR5-PDPK1连接来实现。
    结论:总体而言,我们确定了一小群基因高度富集在与通常由N-MYC调控的有丝分裂过程相关的功能基因类别中,WDR5和PDPK1,并表明三个调节因子之间的三方相互作用可能负责设定N-MYC扩增细胞系中有丝分裂基因调节的水平。本研究为今后研究确定N-MYC,WDR5和PDPK1收敛于细胞周期相关过程。
    BACKGROUND: During mitosis the cell depends on proper attachment and segregation of replicated chromosomes to generate two identical progeny. In cancers defined by overexpression or dysregulation of the MYC oncogene this process becomes impaired, leading to genomic instability and tumor evolution. Recently it was discovered that the chromatin regulator WDR5-a critical MYC cofactor-regulates expression of genes needed in mitosis through a direct interaction with the master kinase PDPK1. However, whether PDPK1 and WDR5 contribute to similar mitotic gene regulation in MYC-overexpressing cancers remains unclear. Therefore, to characterize the influence of WDR5 and PDPK1 on mitotic gene expression in cells with high MYC levels, we performed a comparative transcriptomic analysis in neuroblastoma cell lines defined by MYCN-amplification, which results in high cellular levels of the N-MYC protein.
    RESULTS: Using RNA-seq analysis, we identify the genes regulated by N-MYC and PDPK1 in multiple engineered CHP-134 neuroblastoma cell lines and compare them to previously published gene expression data collected in CHP-134 cells following inhibition of WDR5. We find that as expected N-MYC regulates a multitude of genes, including those related to mitosis, but that PDPK1 regulates specific sets of genes involved in development, signaling, and mitosis. Analysis of N-MYC- and PDPK1-regulated genes reveals a small group of commonly controlled genes associated with spindle pole formation and chromosome segregation, which overlap with genes that are also regulated by WDR5. We also find that N-MYC physically interacts with PDPK1 through the WDR5-PDPK1 interaction suggesting regulation of mitotic gene expression may be achieved through a N-MYC-WDR5-PDPK1 nexus.
    CONCLUSIONS: Overall, we identify a small group of genes highly enriched within functional gene categories related to mitotic processes that are commonly regulated by N-MYC, WDR5, and PDPK1 and suggest that a tripartite interaction between the three regulators may be responsible for setting the level of mitotic gene regulation in N-MYC amplified cell lines. This study provides a foundation for future studies to determine the exact mechanism by which N-MYC, WDR5, and PDPK1 converge on cell cycle related processes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    前信使RNA的转录和剪接紧密协调,但是这种功能耦合是如何在人类疾病中被破坏的,还有待探索。使用等基因细胞系,患者样本,和突变小鼠模型,我们调查了SF3B1中癌症相关突变如何改变转录.我们发现这些突变降低了RNA聚合酶II(RNAPII)沿基因体的伸长率及其在启动子处的密度。延伸缺陷是由于突变体SF3B1的蛋白质-蛋白质相互作用受损而导致的剪接体组装前的破坏。降低的启动子近端RNAPII密度降低了染色质可及性和启动子处的H3K4me3标记。通过一个不偏不倚的屏幕,我们确定了Sin3/HDAC/H3K4me途径中的表观遗传因素,which,当调制时,逆转录和染色质变化。我们的发现揭示了剪接因子突变状态如何通过受损的染色质景观转录相关变化在功能上表现为表观遗传疾病。我们还提出了靶向Sin3/HDAC复合物作为治疗策略的基本原理。
    Transcription and splicing of pre-messenger RNA are closely coordinated, but how this functional coupling is disrupted in human diseases remains unexplored. Using isogenic cell lines, patient samples, and a mutant mouse model, we investigated how cancer-associated mutations in SF3B1 alter transcription. We found that these mutations reduce the elongation rate of RNA polymerase II (RNAPII) along gene bodies and its density at promoters. The elongation defect results from disrupted pre-spliceosome assembly due to impaired protein-protein interactions of mutant SF3B1. The decreased promoter-proximal RNAPII density reduces both chromatin accessibility and H3K4me3 marks at promoters. Through an unbiased screen, we identified epigenetic factors in the Sin3/HDAC/H3K4me pathway, which, when modulated, reverse both transcription and chromatin changes. Our findings reveal how splicing factor mutant states behave functionally as epigenetic disorders through impaired transcription-related changes to the chromatin landscape. We also present a rationale for targeting the Sin3/HDAC complex as a therapeutic strategy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    我们提出了一种用户友好的分子生成管道,称为PocketCrafter,专门设计用于促进药物发现过程中的命中发现活动。此工作流程利用了三维(3D)生成建模方法Pocket2Mol,对于从空间角度对分子进行从头设计,用于靶向蛋白质结构,然后是化学物理特性和药物相似性的过滤器,结构-活性关系分析,和群集以生成顶级虚拟命中脚手架。在我们的WDR5案例研究中,在基于虚拟支架的诺华存档库中进行有针对性的搜索后,我们获得了一组集中的2029种化合物.随后,我们通过实验分析了这些化合物,产生了一种新型的化学支架系列,该系列在生化和生物物理测定中表现出活性。PocketCrafter成功地制作了一个有效的端到端3D生成化学工作流程的原型,用于探索新的化学支架,这代表了早期药物发现中用于命中识别的有希望的方法。
    We present a user-friendly molecular generative pipeline called Pocket Crafter, specifically designed to facilitate hit finding activity in the drug discovery process. This workflow utilized a three-dimensional (3D) generative modeling method Pocket2Mol, for the de novo design of molecules in spatial perspective for the targeted protein structures, followed by filters for chemical-physical properties and drug-likeness, structure-activity relationship analysis, and clustering to generate top virtual hit scaffolds. In our WDR5 case study, we acquired a focused set of 2029 compounds after a targeted searching within Novartis archived library based on the virtual scaffolds. Subsequently, we experimentally profiled these compounds, resulting in a novel chemical scaffold series that demonstrated activity in biochemical and biophysical assays. Pocket Crafter successfully prototyped an effective end-to-end 3D generative chemistry-based workflow for the exploration of new chemical scaffolds, which represents a promising approach in early drug discovery for hit identification.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    WDR5是一种保守的核蛋白,可支持表观遗传调节复合物和月光的组装,其功能范围从招募MYC癌蛋白到染色质,以促进有丝分裂的完整性。它也是抗癌治疗的高价值目标,与小分子WDR5抑制剂和降解物进行广泛的临床前评估。WDR5抑制剂最初被认为是表观遗传调节剂,提出通过逆转组蛋白H3赖氨酸4甲基化的致癌模式来抑制癌细胞-这一观点一直持续到今天。这个前提,然而,无法承受当代检查,并建立了可能永远无法满足的WDR5抑制剂的机制和效用的期望。这里,我们强调了关于WDR5抑制剂作为表观遗传调节剂的显著误解,并为它们作为核糖体导向的抗癌治疗提供了一个统一的模型,这有助于集中了解何时以及如何最好地理解和利用这些药物的肿瘤抑制特性.
    WDR5 is a conserved nuclear protein that scaffolds the assembly of epigenetic regulatory complexes and moonlights in functions ranging from recruiting MYC oncoproteins to chromatin to facilitating the integrity of mitosis. It is also a high-value target for anti-cancer therapies, with small molecule WDR5 inhibitors and degraders undergoing extensive preclinical assessment. WDR5 inhibitors were originally conceived as epigenetic modulators, proposed to inhibit cancer cells by reversing oncogenic patterns of histone H3 lysine 4 methylation-a notion that persists to this day. This premise, however, does not withstand contemporary inspection and establishes expectations for the mechanisms and utility of WDR5 inhibitors that can likely never be met. Here, we highlight salient misconceptions regarding WDR5 inhibitors as epigenetic modulators and provide a unified model for their action as a ribosome-directed anti-cancer therapy that helps focus understanding of when and how the tumor-inhibiting properties of these agents can best be understood and exploited.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:癌基因MYCN与神经母细胞瘤(NB)的恶性进展和不良预后密切相关。最近,长链非编码RNA(lncRNAs)已被认为是各种癌症的关键调节因子。然而,lncRNAs是否有助于NB中MYCN的过表达尚不清楚.
    方法:微阵列分析用于分析MYCN扩增和MYCN非扩增NB细胞系之间差异表达的lncRNAs。生物信息学分析用于鉴定MYCN基因座附近的lncRNA。qRT-PCR检测lncRNAAC142119.1在NB细胞系和组织中的表达水平。进行了功能增益和功能丧失试验以研究AC142119.1在NB中的生物学效应。荧光原位杂交,RNA下拉,RNA免疫沉淀,质谱,RNA电泳迁移率变化,通过RNA纯化测定进行染色质免疫沉淀和染色质分离以验证AC142119.1和WDR5蛋白以及MYCN启动子之间的相互作用。
    结果:在MYCN扩增的NB组织中AC142119.1显著升高,高级INSS阶段和高风险,并与NB患者的低生存率相关。此外,AC142119.1的强制表达增强了NB细胞的体外和体内增殖。此外,AC142119.1专门招募WDR5蛋白与MYCN启动子相互作用,进一步启动MYCN的转录并加速NB的进展。
    结论:我们鉴定了一种新的lncRNAAC142119.1,它通过与WDR5蛋白和MYCN启动子相互作用,通过表观遗传学启动MYCN的转录促进NB的进展,表明AC142119.1可能是NB的潜在诊断生物标志物和治疗靶标。
    Oncogene MYCN is closely related with malignant progression and poor prognosis of neuroblastoma (NB). Recently, long non-coding RNAs (lncRNAs) have been recognized as crucial regulators in various cancers. However, whether lncRNAs contribute to the overexpression of MYCN in NB is unclear.
    Microarray analysis were applied to analyze the differentially expressed lncRNAs between MYCN-amplified and MYCN-non-amplified NB cell lines. Bioinformatic analyses were utilized to identify lncRNAs nearby MYCN locus. qRT-PCR was used to detect the expression level of lncRNA AC142119.1 in NB cell lines and tissues. Gain- and loss-of-function assays were conducted to investigate the biological effect of AC142119.1 in NB. Fluorescence in situ hybridization, RNA pull-down, RNA immunoprecipitation, mass spectrometry, RNA electrophoretic mobility shift, chromatin immunoprecipitation and chromatin isolation by RNA purification assays were performed to validate the interaction between AC142119.1 and WDR5 protein as well as MYCN promoter.
    AC142119.1 was significantly elevated in NB tissues with MYCN amplification, advanced INSS stage and high risk, and associated with poor survival of NB patients. Moreover, enforced expression of AC142119.1 reinforced the proliferation of NB cells in vitro and in vivo. Additionally, AC142119.1 specifically recruited WDR5 protein to interact with MYCN promoter, further initiating the transcription of MYCN and accelerating NB progression.
    We identified a novel lncRNA AC142119.1, which promoted the progression of NB through epigenetically initiating the transcription of MYCN via interacting with both WDR5 protein and the promoter of MYCN, indicating that AC142119.1 might be a potential diagnostic biomarker and therapeutic target for NB.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    保守的WD40重复蛋白WDR5与细胞核内外的多种蛋白相互作用。然而,目前尚不清楚WDR5在复合物之间的分布是否以及如何受到调控.这里,我们表明,在人SCRIB基因中双重编码的未注释的微蛋白EMBOW(WDR5的内源性微蛋白结合剂)与WDR5相互作用,并调节其与多个相互作用伙伴的结合,包括KMT2A和KIF2A。脑膜受细胞周期调节,在G1期晚期和G2/M期有两个表达最大值。EMBOW的丢失减少了WDR5与KIF2A的相互作用,异常缩短有丝分裂纺锤体长度,延长G2/M期,并延迟细胞增殖。相比之下,丧失EMBOW增加了WDR5与KMT2A的相互作用,导致WDR5与脱靶基因结合,错误地增加H3K4me3水平,并激活这些基因的转录。一起,这些结果提示EMBOW是WDR5的调节因子,调节其相互作用并防止其在多种情况下的脱靶结合.
    The conserved WD40-repeat protein WDR5 interacts with multiple proteins both inside and outside the nucleus. However, it is currently unclear whether and how the distribution of WDR5 between complexes is regulated. Here, we show that an unannotated microprotein EMBOW (endogenous microprotein binder of WDR5) dually encoded in the human SCRIB gene interacts with WDR5 and regulates its binding to multiple interaction partners, including KMT2A and KIF2A. EMBOW is cell cycle regulated, with two expression maxima at late G1 phase and G2/M phase. Loss of EMBOW decreases WDR5 interaction with KIF2A, aberrantly shortens mitotic spindle length, prolongs G2/M phase, and delays cell proliferation. In contrast, loss of EMBOW increases WDR5 interaction with KMT2A, leading to WDR5 binding to off-target genes, erroneously increasing H3K4me3 levels, and activating transcription of these genes. Together, these results implicate EMBOW as a regulator of WDR5 that regulates its interactions and prevents its off-target binding in multiple contexts.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    多步降解过程的ProteesolutionTogeting嵌合体(PROTACs)对其合理发展提出了挑战,因为决定PROTACs效率的限速步骤在很大程度上仍然未知。此外,当前使用的终点测定的通量缓慢,不允许对更大系列的PROTACs进行全面分析.这里,我们使用NanoLuciferase和HaloTag开发了基于细胞的测定法,该测定法允许测量PROTAC诱导的降解和三元复合物形成动力学以及细胞中的稳定性。使用开发用于降解WD40重复结构域蛋白5(WDR5)的PROTACs,这些PROTACs在早期降解级联中的作用模式的表征揭示了三元复合物形成和稳定性的关键作用。比较一系列三元络合物晶体结构强调了有效的E3-靶界面对于三元络合物稳定性的重要性。开发的测定法概述了使用一系列活细胞测定法监测早期PROTAC诱导的降解途径的关键步骤的合理优化PROTAC的策略。
    The multi-step degradation process of PROteolysis TArgeting Chimeras (PROTACs) poses a challenge for their rational development, as the rate-limiting steps that determine PROTACs efficiency remain largely unknown. Moreover, the slow throughput of currently used endpoint assays does not allow the comprehensive analysis of larger series of PROTACs. Here, we developed cell-based assays using the NanoLuciferase and HaloTag that allow measuring PROTAC-induced degradation and ternary complex formation kinetics and stability in cells. Using PROTACs developed for the degradation of WD40 repeat domain protein 5 (WDR5), the characterization of the mode of action of these PROTACs in the early degradation cascade revealed a key role of ternary complex formation and stability. Comparing a series of ternary complex crystal structures highlighted the importance of an efficient E3-target interface for ternary complex stability. The developed assays outline a strategy for the rational optimization of PROTACs using a series of live cell assays monitoring key steps of the early PROTAC-induced degradation pathway.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号