proteolysis-targeting chimera (PROTAC)

  • 文章类型: Journal Article
    SMARCA2和SMARCA4是SWI/SNF复合物的亚基,SWI/SNF复合物是染色质重塑复合物,是促进基因表达的关键表观遗传调节因子。SMARCA4功能缺失突变的肿瘤依赖于SMARCA2细胞存活,这种合成致死性是治疗癌症的潜在治疗策略。
    当前的评论集中在声称与SMARCA2的溴结构域位点结合的蛋白水解靶向嵌合体(PROTAC)降解物的专利申请中,并在2019年1月至2023年6月之间发布。共评估了9个不同申请人的29个申请。
    SMARCA2/4溴结构域抑制剂不会对癌症增殖产生预期效果;然而,公司已经将溴结构域结合剂转化为PROTACs来降解蛋白质,与SMARCA4相比,更喜欢SMARCA2。在缺乏SMARCA4的情况下,SMARCA2的选择性降解很可能是有效的,同时允许正常组织有足够的安全裕度。随着最近披露的几项专利申请,瞄准SMARCA2的兴趣应该继续,特别是现在在临床上使用PreludeTherapeutics的选择性SMARCA2PROTAC。临床试验的结果将影响选择性SMARCA2PROTACs开发的演变。
    UNASSIGNED: SMARCA2 and SMARCA4 are subunits of the SWI/SNF complex which is a chromatin remodeling complex and a key epigenetic regulator that facilitates gene expression. Tumors with loss of function mutations in SMARCA4 rely on SMARCA2 for cell survival and this synthetic lethality is a potential therapeutic strategy to treat cancer.
    UNASSIGNED: The current review focuses on patent applications that claim proteolysis-targeting chimeras (PROTAC) degraders that bind the bromodomain site of SMARCA2 and are published between January 2019-June 2023. A total of 29 applications from 9 different applicants were evaluated.
    UNASSIGNED: SMARCA2/4 bromodomain inhibitors do not lead to desired effects on cancer proliferation; however, companies have converted bromodomain binders into PROTACs to degrade the protein, with a preference for SMARCA2 over SMARCA4. Selective degradation of SMARCA2 is most likely required to be efficacious in the SMARCA4-deficient setting, while allowing for sufficient safety margin in normal tissues. With several patent applications disclosed recently, interest in targeting SMARCA2 should continue, especially with a selective SMARCA2 PROTAC now in the clinic from Prelude Therapeutics. The outcome of the clinical trials will influence the evolution of selective SMARCA2 PROTACs development.
    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
    哺乳动物开关/蔗糖不可发酵(mSWI/SNF)ATPase降解剂已被证明在增强子驱动的癌症中有效,可以阻止致癌转录因子染色质的可及性。这里,我们开发了AU-24118,一种口服生物可利用的蛋白水解靶向嵌合体(PROTAC)降解剂,用于mSWI/SNFATPases(SMARCA2和SMARCA4)和PBRM1。AU-24118在去势抵抗性前列腺癌(CRPC)模型中显示出肿瘤消退,通过联合恩杂鲁胺治疗进一步增强。用于CRPC患者的标准治疗雄激素受体(AR)拮抗剂。重要的是,AU-24118在小鼠和大鼠的临床前分析中表现出良好的药代动力学特征,在小鼠中进一步的毒性测试显示了良好的安全性。由于获得性耐药在靶向癌症治疗中是常见的,实验旨在探索长期mSWI/SNFATPasePROTAC治疗可能产生的潜在耐药机制。暴露于高剂量mSWI/SNFATPase降解剂长期治疗的前列腺癌细胞系产生SMARCA4溴结构域突变和ABCB1(ATP结合盒亚家族B成员1)过表达作为获得性耐药机制。有趣的是,虽然SMARCA4突变提供了对mSWI/SNF降解剂的特异性抗性,ABCB1过表达对靶向含溴结构域蛋白4和AR的其他有效PROTAC降解物提供了更广泛的抗性。ABCB1抑制剂,zosuquidar,测试的所有三个PROTAC降级器的反向电阻。合并,这些发现为增强子驱动型癌症患者的临床翻译定位了mSWI/SNF降解产物,并确定了克服可能出现的耐药机制的策略.
    Mammalian switch/sucrose nonfermentable (mSWI/SNF) ATPase degraders have been shown to be effective in enhancer-driven cancers by functioning to impede oncogenic transcription factor chromatin accessibility. Here, we developed AU-24118, an orally bioavailable proteolysis-targeting chimera (PROTAC) degrader of mSWI/SNF ATPases (SMARCA2 and SMARCA4) and PBRM1. AU-24118 demonstrated tumor regression in a model of castration-resistant prostate cancer (CRPC) which was further enhanced with combination enzalutamide treatment, a standard of care androgen receptor (AR) antagonist used in CRPC patients. Importantly, AU-24118 exhibited favorable pharmacokinetic profiles in preclinical analyses in mice and rats, and further toxicity testing in mice showed a favorable safety profile. As acquired resistance is common with targeted cancer therapeutics, experiments were designed to explore potential mechanisms of resistance that may arise with long-term mSWI/SNF ATPase PROTAC treatment. Prostate cancer cell lines exposed to long-term treatment with high doses of a mSWI/SNF ATPase degrader developed SMARCA4 bromodomain mutations and ABCB1 (ATP binding cassette subfamily B member 1) overexpression as acquired mechanisms of resistance. Intriguingly, while SMARCA4 mutations provided specific resistance to mSWI/SNF degraders, ABCB1 overexpression provided broader resistance to other potent PROTAC degraders targeting bromodomain-containing protein 4 and AR. The ABCB1 inhibitor, zosuquidar, reversed resistance to all three PROTAC degraders tested. Combined, these findings position mSWI/SNF degraders for clinical translation for patients with enhancer-driven cancers and define strategies to overcome resistance mechanisms that may arise.
    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
    间接反应(IDR)和失活转化(TI)包括两个基于机制的药效学(PD)模型阵列,广泛用于描述延迟的药物作用。IDR模型-IV(刺激反应损失)和TI(不可逆损失)已被描述为具有明显的“特征”曲线;典型的IDR-IV反应时间曲线显示缓慢下降,其中最小反应随剂量增加而变化,而TI配置文件的特点是响应急剧下降与较早移动的最低点。在这里,我们证明了IDR-IV和TI模型在使用相同的线性和非线性药理作用项实施时的数学收敛性.实际上,根据PK或PD参数,IDR-IV中的峰值响应时间可以更早或更晚移动(例如,Kel,Smax)和效果类型。提出了连接mRNA和蛋白质周转的广义动态模型。IDR-IV和TI的适用性,线性或非线性项作用于降解/分解代谢/响应损失,通过模型拟合三种蛋白水解靶向嵌合体(PROTACs)和两种配体缀合的小干扰RNA(siRNA)的PK/PD效应来证明。这项工作阐明了数学性质,收敛,以及IDR-IV和TI的预期反应,证明了它们对靶向基因沉默和蛋白质降解剂的适用性,并说明了设计良好的体内研究如何覆盖广泛的剂量范围和丰富的采样时间点可以影响PK-PD模型的结构和参数分辨率。
    Indirect response (IDR) and turnover with inactivation (TI) comprise two arrays of mechanism-based pharmacodynamic (PD) models widely used to describe delayed drug effects. IDR Model-IV (stimulation of response loss) and TI (irreversible loss) have been described with discerning \"signature\" profiles; classical IDR-IV response-time profiles display slow declines where peak response shifts later with increasing dose, whereas TI profiles feature steep response declines with earlier-shifting nadirs. Herein, we demonstrate mathematical convergence of IDR-IV and TI models upon implementation with identical linear versus nonlinear pharmacologic effect terms. Time of peak response in IDR-IV can in fact shift earlier or later depending on PK or PD parameters (e.g., kel, Smax) and effect type. A generalized dynamic model linking mRNA and protein turnover is proposed. Applicability of IDR-IV and TI, with either linear or nonlinear terms acting on degradation/catabolism/loss of response, is demonstrated through model-fitting PK-PD effects of three proteolysis-targeting chimeras (PROTACs) and two ligand-conjugated small interfering RNAs (siRNA). This work clarifies mathematical properties, convergence, and expected responses of IDR-IV and TI, demonstrates their applicability for targeted gene-silencing and protein-degrading agents, and illustrates how well-designed in vivo studies covering broad dose ranges with richly sampled time-points can influence PK-PD model structure and parameter resolution.
    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
    由于活性氧(ROS)的异常积累而引起的氧化应激是人类大量疾病的引发剂,因此,消除和预防过量ROS是预防此类疾病发展的重要方面。核因子红系2相关因子2(NRF2)是防御氧化应激的必需转录因子,其功能受Kelch样ECH相关蛋白1(KEAP1)的负控制。因此,通过抑制KEAP1激活NRF2被视为对抗氧化应激相关疾病的策略。这里,我们产生了一个基于cereblon(CRBN)的蛋白水解靶向嵌合体(PROTAC),我们将其命名为SD2267,它诱导KEAP1的蛋白酶体降解并导致NRF2激活。按照预期,SD2267绑定到KEAP1,招募CRBN,并诱导KEAP1的降解。此外,MG132(一种蛋白酶体降解抑制剂)降低了SD2267的KEAP1降解功效,但氯喹(一种自噬抑制剂)没有降低,这表明SD2267对KEAP1的降解是蛋白酶体降解依赖性和自噬依赖性。KEAP1降解后,SD2267诱导NRF2的核易位,导致NRF2靶基因的表达,并减弱对乙酰氨基酚(APAP)在肝细胞中诱导的ROS积累。基于体内药代动力学研究,在APAP诱导的肝损伤小鼠模型中,以1或3mg/kg腹膜内注射SD2267。我们观察到SD2267降解肝KEAP1并减轻APAP诱导的肝损伤。总结,我们描述了KEAP1靶向PROTAC(SD2267)的合成及其在体外和体内的功效和作用方式.结果表明,SD2267可用于治疗与氧化应激相关的肝脏疾病。
    Oxidative stress due to abnormal accumulation of reactive oxygen species (ROS) is an initiator of a large number of human diseases, and thus, the elimination and prevention of excessive ROS are important aspects of preventing the development of such diseases. Nuclear factor erythroid 2-related factor 2 (NRF2) is an essential transcription factor that defends against oxidative stress, and its function is negatively controlled by Kelch-like ECH-associated protein 1 (KEAP1). Therefore, activating NRF2 by inhibiting KEAP1 is viewed as a strategy for combating oxidative stress-related diseases. Here, we generated a cereblon (CRBN)-based proteolysis-targeting chimera (PROTAC), which we named SD2267, that induces the proteasomal degradation of KEAP1 and leads to NRF2 activation. As was intended, SD2267 bound to KEAP1, recruited CRBN, and induced the degradation of KEAP1. Furthermore, the KEAP1 degradation efficacy of SD2267 was diminished by MG132 (a proteasomal degradation inhibitor) but not by chloroquine (an autophagy inhibitor), which suggested that KEAP1 degradation by SD2267 was proteasomal degradation-dependent and autophagy-independent. Following KEAP1 degradation, SD2267 induced the nuclear translocation of NRF2, which led to the expression of NRF2 target genes and attenuated ROS accumulation induced by acetaminophen (APAP) in hepatocytes. Based on in vivo pharmacokinetic study, SD2267 was injected intraperitoneally at 1 or 3 mg/kg in APAP-induced liver injury mouse model. We observed that SD2267 degraded hepatic KEAP1 and attenuated APAP-induced liver damage. Summarizing, we described the synthesis of a KEAP1-targeting PROTAC (SD2267) and its efficacy and mode of action in vitro and in vivo. The results obtained suggest that SD2267 could be used to treat hepatic diseases related to oxidative stress.
    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
    泛素化是一种引人入胜的翻译后修饰,自发现以来一直受到关注。在这次审查中,我们首先提供E3泛素连接酶的简要概述,深入研究分类,泛素化的特点和机制。然后,我们专门检查由N/C-degrons介导的泛素化途径,讨论它们的独特特征和底物识别机制。最后,我们提供有关针对N/C-Degron途径的抑制剂的发展现状的见解,以及PROTAC(PROteesolutionTogetingChimeras)领域的有希望的进展。总的来说,这篇综述提供了对迅速发展的泛素生物学领域的全面了解。
    Ubiquitination is a fascinating post-translational modification that has received continuous attention since its discovery. In this review, we first provide a concise overview of the E3 ubiquitin ligases, delving into classification, characteristics and mechanisms of ubiquitination. We then specifically examine the ubiquitination pathways mediated by the N/C-degrons, discussing their unique features and substrate recognition mechanisms. Finally, we offer insights into the current state of development pertaining to inhibitors that target the N/C-degron pathways, as well as the promising advances in the field of PROTAC (PROteolysis TArgeting Chimeras). Overall, this review offers a comprehensive understanding of the rapidly-evolving field of ubiquitin biology.
    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
    动脉粥样硬化是心血管疾病死亡和残疾的主要原因。与脂质积累和慢性炎症相关的动脉粥样硬化导致动脉壁中的斑块形成和颈动脉的管腔狭窄。目前的方法如手术或他汀类药物治疗在治疗动脉粥样硬化斑块方面面临巨大挑战。促炎性M1巨噬细胞的浸润在动脉粥样硬化斑块的发生和发展中起着至关重要的作用。最近的一项研究表明,TRIM24是Trim家族蛋白的E3泛素连接酶,充当抑制抗炎M2巨噬细胞极化的瓣膜,TRIM24的消除为实现M2极化开辟了一条途径。蛋白水解靶向嵌合体(PROTAC)技术已成为靶向蛋白质选择性降解的新型工具。但是PROTAC试剂的低生物利用度和细胞特异性阻碍了其在动脉粥样硬化斑块治疗中的应用。在这项研究中,我们通过用M2巨噬细胞膜(MELT)包被负载PROTAC降解剂(dTRIM24)的PLGA纳米颗粒来构建一种生物启发的PROTAC,用于动脉粥样硬化治疗。熔体的特征在于形态,尺寸,和稳定性。MELT显示对M1巨噬细胞的特异性增强以及dTRIM24的酸性响应性释放。静脉给药后,MELT显示通过与M1巨噬细胞结合并诱导有效和精确的TRIM24降解,在高脂肪和高胆固醇饮食喂养的动脉粥样硬化(ApoE-/-)小鼠的动脉粥样硬化斑块中的积累显着改善,从而导致M2巨噬细胞的极化,导致斑块形成大大减少。这些结果表明,MELT可以被认为是靶向动脉粥样硬化斑块和缓解动脉粥样硬化进展的潜在治疗剂。为动脉粥样硬化的靶向治疗提供了有效的策略。
    Atherosclerosis is a major cause of death and disability in cardiovascular disease. Atherosclerosis associated with lipid accumulation and chronic inflammation leads to plaques formation in arterial walls and luminal stenosis in carotid arteries. Current approaches such as surgery or treatment with statins encounter big challenges in curing atherosclerosis plaque. The infiltration of proinflammatory M1 macrophages plays an essential role in the occurrence and development of atherosclerosis plaque. A recent study shows that TRIM24, an E3 ubiquitin ligase of a Trim family protein, acts as a valve to inhibit the polarization of anti-inflammatory M2 macrophages, and elimination of TRIM24 opens an avenue to achieve the M2 polarization. Proteolysis-targeting chimera (PROTAC) technology has emerged as a novel tool for the selective degradation of targeting proteins. But the low bioavailability and cell specificity of PROTAC reagents hinder their applications in treating atherosclerosis plaque. In this study we constructed a type of bioinspired PROTAC by coating the PROTAC degrader (dTRIM24)-loaded PLGA nanoparticles with M2 macrophage membrane (MELT) for atherosclerosis treatment. MELT was characterized by morphology, size, and stability. MELT displayed enhanced specificity to M1 macrophages as well as acidic-responsive release of dTRIM24. After intravenous administration, MELT showed significantly improved accumulation in atherosclerotic plaque of high fat and high cholesterol diet-fed atherosclerotic (ApoE-/-) mice through binding to M1 macrophages and inducing effective and precise TRIM24 degradation, thus resulting in the polarization of M2 macrophages, which led to great reduction of plaque formation. These results suggest that MELT can be considered a potential therapeutic agent for targeting atherosclerotic plaque and alleviating atherosclerosis progression, providing an effective strategy for targeted atherosclerosis therapy.
    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
    蛋白水解靶向嵌合体(PROTACs)通过诱导肿瘤过度表达的致癌蛋白的降解而迅速成为癌症治疗的潜在治疗策略。它们可以通过募集E3连接酶并利用泛素-蛋白酶体途径特异性催化目标致癌蛋白的降解。由于他们的行动方式是普遍的,不可逆转的,可回收,持久的,适用于“不可食用的”蛋白质,PROTACs正在逐渐取代常规小分子抑制剂的作用。此外,它们的应用领域正在扩展到癌症免疫疗法,作为参与免疫抑制肿瘤微环境的各种类型的致癌蛋白。然而,不良的水溶性和低的细胞渗透性极大地限制了药代动力学(PK)性质,这就需要使用适当的递送系统进行癌症免疫疗法。在这次审查中,一般特征,发展状况,首先简要介绍PROTACs的PK。接下来,介绍了最近对各种类型的被动或主动靶向递送系统在PROTACs中的应用的研究,描述了它们对PROTACs的PK和肿瘤靶向能力的影响。最后,综述了近年来用于肿瘤免疫治疗的PROTACs给药系统。采用适当的PROTAC递送系统有望加速PROTAC的临床翻译,以及提高其对癌症治疗的疗效。
    Proteolysis-targeting chimeras (PROTACs) are rapidly emerging as a potential therapeutic strategy for cancer therapy by inducing the degradation of tumor-overexpressing oncogenic proteins. They can specifically catalyze the degradation of target oncogenic proteins by recruiting E3 ligases and utilizing the ubiquitin-proteasome pathway. Since their mode of action is universal, irreversible, recyclable, long-lasting, and applicable to \'undruggable\' proteins, PROTACs are gradually replacing the role of conventional small molecular inhibitors. Moreover, their application areas are being expanded to cancer immunotherapy as various types of oncogenic proteins that are involved in immunosuppressive tumor microenvironments. However, poor water solubility and low cell permeability considerably restrict the pharmacokinetic (PK) property, which necessitates the use of appropriate delivery systems for cancer immunotherapy. In this review, the general characteristics, developmental status, and PK of PROTACs are first briefly covered. Next, recent studies on the application of various types of passive or active targeting delivery systems for PROTACs are introduced, and their effects on the PK and tumor-targeting ability of PROTACs are described. Finally, recent drug delivery systems of PROTACs for cancer immunotherapy are summarized. The adoption of an adequate delivery system for PROTAC is expected to accelerate the clinical translation of PROTACs, as well as improve its efficacy for cancer therapy.
    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
    TYK2,近端膜结合酪氨酸激酶JAK家族的成员,已成为治疗自身免疫性疾病的有吸引力的靶标。在这里,我们报告发现了一流的有效和亚型选择性TYK2降解剂。通过各种长度的烷基接头将来自已知变构TYK2抑制剂的TYK2配体与作为E3连接酶配体的VHL配体缀合,我们快速鉴定出具有中等TYK2降解活性的TYK2降解剂5。降解剂5在Jurkat细胞测定中诱导TYK2降解而不影响亚型激酶(JAK1、JAK2和JAK3)的蛋白质水平。此外,修饰降解剂5的TYK2配体部分产生更有效的TYK2降解剂37,并保留对JAK的选择性。我们的亚型选择性TYK2降解剂代表了研究TYK2降解生物学的有价值的化学探针。
    TYK2, a member of the JAK family of proximal membrane-bound tyrosine kinases, has emerged as an attractive target for the treatment of autoimmune diseases. Herein, we report the discovery of first-in-class potent and subtype-selective TYK2 degraders. By conjugating a TYK2 ligand from a known allosteric TYK2 inhibitor with a VHL ligand as the E3 ligase ligand via alkyl linkers of various lengths, we rapidly identified TYK2 degrader 5 with moderate TYK2 degradation activity. Degrader 5 induced TYK2 degradation without affecting the protein level of subtype kinases (JAK1, JAK2, and JAK3) in Jurkat cellular assays. Furthermore, modifying the TYK2 ligand moiety of degrader 5 yielded the more potent TYK2 degrader 37 with retained selectivity for JAKs. Our subtype-selective TYK2 degraders represent valuable chemical probes for investigating the biology of TYK2 degradation.
    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
    蛋白水解靶向嵌合体(PROTAC)是选择性降解感兴趣的蛋白质(POI)的分子。将招募小鼠双分钟2(MDM2)的配体掺入到PROTACs中,形成所谓的基于MDM2的协议,由于其双重作用机制,已在癌症治疗中显示出希望:招募MDM2的PROTAC阻止其与p53的结合,不仅导致POI的降解,而且导致p53抑制剂的细胞内水平增加,随着一整套生物过程的激活,如细胞周期停滞或凋亡。此外,这些PROTACs,在某些情况下,允许目标的退化,具有纳摩尔效力,随着时间的推移,以快速和持续的方式,对抗性和耐受性发展的敏感性较低,而不会引起蛋白质表达的变化,并且对目标有选择性,包括各自的同工型或突变,和细胞类型,克服了与针对相同治疗靶标使用抑制剂相关的一些限制。因此,这篇综述的目的是分析和讨论为降解致癌蛋白而开发的基于MDM2的PROTACs的特征,并了解它们作为未来抗癌药物的潜力。
    Proteolysis-targeting chimeras (PROTACs) are molecules that selectively degrade a protein of interest (POI). The incorporation of ligands that recruit mouse double minute 2 (MDM2) into PROTACs, forming the so-called MDM2-based PROTACs, has shown promise in cancer treatment due to its dual mechanism of action: a PROTAC that recruits MDM2 prevents its binding to p53, resulting not only in the degradation of POI but also in the increase of intracellular levels of the p53 suppressor, with the activation of a whole set of biological processes, such as cell cycle arrest or apoptosis. In addition, these PROTACs, in certain cases, allow for the degradation of the target, with nanomolar potency, in a rapid and sustained manner over time, with less susceptibility to the development of resistance and tolerance, without causing changes in protein expression, and with selectivity to the target, including the respective isoforms or mutations, and to the cell type, overcoming some limitations associated with the use of inhibitors for the same therapeutic target. Therefore, the aim of this review is to analyze and discuss the characteristics of MDM2-based PROTACs developed for the degradation of oncogenic proteins and to understand what potential they have as future anticancer 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
    蛋白水解靶向嵌合体(PROTACs)将感兴趣的蛋白质(POI)带入E3泛素连接酶的空间附近,促进POI泛素化和蛋白酶体降解。PROTACs依赖于内源性细胞机制来介导POI降解,因此,POI的亚细胞定位和对E3连接酶的获取可能会影响PROTAC的疗效.为了询问POI的亚细胞环境是否影响PROTAC介导的降解,我们表达了由不同定位信号组成的Halo或FKBP12F36V(dTAG)构建体,并测试了它们通过靶向Halo或dTAG的vonHippel-Lindau(VHL)-或cereblon(CRBN)-招募PROTACs降解的功效.POI定位于细胞核,细胞质,线粒体外膜,内质网,高尔基,过氧化物酶体或溶酶体。差异定位的Halo或FKBP12F36V蛋白使用相同的各自PROTACs表现出不同的降解水平,因此,表明POI的亚细胞环境可以影响PROTAC介导的POI降解的功效。
    Proteolysis-targeting chimeras (PROTACs) bring a protein of interest (POI) into spatial proximity of an E3 ubiquitin ligase, promoting POI ubiquitylation and proteasomal degradation. PROTACs rely on endogenous cellular machinery to mediate POI degradation, therefore the subcellular location of the POI and access to the E3 ligase being recruited potentially impacts PROTAC efficacy. To interrogate whether the subcellular context of the POI influences PROTAC-mediated degradation, we expressed either Halo or FKBP12F36V (dTAG) constructs consisting of varying localization signals and tested the efficacy of their degradation by von Hippel-Lindau (VHL)- or cereblon (CRBN)-recruiting PROTACs targeting either Halo or dTAG. POIs were localized to the nucleus, cytoplasm, outer mitochondrial membrane, endoplasmic reticulum, Golgi, peroxisome or lysosome. Differentially localized Halo or FKBP12F36V proteins displayed varying levels of degradation using the same respective PROTACs, suggesting therefore that the subcellular context of the POI can influence the efficacy of PROTAC-mediated POI degradation.
    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