NEDD8 Protein

NEDD8 蛋白
  • 文章类型: Journal Article
    (1)背景:neddylation通路在癌症的发生和发展中起着关键作用。MLN4924,NEDD8激活酶(NAE)的有效小分子抑制剂,有效干预neddylation途径的早期阶段。通过激发不同的细胞反应,如癌细胞的衰老和凋亡,MLN4924还对肿瘤微环境(TME)和肿瘤病毒感染的细胞内的非恶性细胞发挥调节作用,从而阻碍肿瘤的发作。因此,MLN4924已被广泛认为是一种有效的抗癌药物。(2)最近的发现:尽管如此,最近的发现阐明了neddylation途径的其他方面,揭示了其积极参与各种不利于癌细胞生存的生物过程。这种新发现的理解强调了MLN4924在肿瘤治疗中的双重作用,具有抗癌和抗癌作用。这种二分法在本文中被称为MLN4924的“双刃效应”。本文深入研究了Neddylation通路与癌症之间的复杂关系,对MLN4924双刃效应的潜在原因进行了机理探索和分析,促癌症Neddylation底物的积累。(3)观点:在这里,目的是提供理论支持和新见解,以指导针对neddylation途径的下一代抗癌药物的开发。
    (1) Background: The neddylation pathway assumes a pivotal role in the initiation and progression of cancer. MLN4924, a potent small-molecule inhibitor of the NEDD8-activating enzyme (NAE), effectively intervenes in the early stages of the neddylation pathway. By instigating diverse cellular responses, such as senescence and apoptosis in cancer cells, MLN4924 also exerts regulatory effects on non-malignant cells within the tumor microenvironment (TME) and tumor virus-infected cells, thereby impeding the onset of tumors. Consequently, MLN4924 has been widely acknowledged as a potent anti-cancer drug. (2) Recent findings: Nevertheless, recent findings have illuminated additional facets of the neddylation pathway, revealing its active involvement in various biological processes detrimental to the survival of cancer cells. This newfound understanding underscores the dual role of MLN4924 in tumor therapy, characterized by both anti-cancer and pro-cancer effects. This dichotomy is herein referred to as the \"double-edged effects\" of MLN4924. This paper delves into the intricate relationship between the neddylation pathway and cancer, offering a mechanistic exploration and analysis of the causes underlying the double-edged effects of MLN4924-specifically, the accumulation of pro-cancer neddylation substrates. (3) Perspectives: Here, the objective is to furnish theoretical support and novel insights that can guide the development of next-generation anti-cancer drugs targeting the neddylation pathway.
    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
    在西方世界,药物性肝损伤(DILI)是急性肝衰竭(ALF)和肝移植的重要原因。对乙酰氨基酚(APAP)过量是DILI的主要原因,通过坏死导致肝细胞死亡。这里,我们确定了neddylation,涉及线粒体功能的重要翻译后修饰,在APAP诱导的肝损伤(AILI)患者和APAP过量治疗的小鼠的肝活检中上调。MLN4924,神经元前体细胞表达的发育下调蛋白8(NEDD8)激活酶(NAE-1)的抑制剂,在AILI中改善坏死和增强肝再生。要了解Neddylation如何干扰AILI,在APAP过量使用和不使用MLN4924的情况下,研究了全身生物素化NEDD8(bioNEDD8)和泛素(bioUB)转基因小鼠。胞苷二磷酸二酰甘油(CDP-DAG)合酶TAM41,负责产生线粒体活性必需的心磷脂,被发现在AILI下调节,并通过抑制Neddylation恢复其水平。了解AILI中的这种泛素样串扰对于开发用于DILI治疗的有前途的靶向抑制剂至关重要。
    Drug-induced liver injury (DILI) is a significant cause of acute liver failure (ALF) and liver transplantation in the Western world. Acetaminophen (APAP) overdose is a main contributor of DILI, leading to hepatocyte cell death through necrosis. Here, we identified that neddylation, an essential post-translational modification involved in the mitochondria function, was upregulated in liver biopsies from patients with APAP-induced liver injury (AILI) and in mice treated with an APAP overdose. MLN4924, an inhibitor of the neuronal precursor cell-expressed developmentally downregulated protein 8 (NEDD8)-activating enzyme (NAE-1), ameliorated necrosis and boosted liver regeneration in AILI. To understand how neddylation interferes in AILI, whole-body biotinylated NEDD8 (bioNEDD8) and ubiquitin (bioUB) transgenic mice were investigated under APAP overdose with and without MLN4924. The cytidine diphosphate diacylglycerol (CDP-DAG) synthase TAM41, responsible for producing cardiolipin essential for mitochondrial activity, was found modulated under AILI and restored its levels by inhibiting neddylation. Understanding this ubiquitin-like crosstalk in AILI is essential for developing promising targeted inhibitors for DILI treatment.
    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:紫杉醇(PTX)治疗耐药是导致三阴性乳腺癌(TNBC)预后不良的重要因素,因此,迫切需要确定联合治疗的新靶点。Neddylation是一种翻译后过程,它引入了一种称为神经前体细胞的泛素样蛋白,该蛋白在发育中表达下调蛋白8(NEDD8)。以前的研究已经发现,在多种肿瘤中,neddylation被激活,但其与PTX化疗敏感性的关系尚未见报道。
    方法:使用公共数据库和免疫组织化学验证了PTX敏感和PTX不敏感的TNBC组织之间UBC12和NEDD8表达水平的差异。通过体外和体内功能实验观察Neddylation抑制联合PTX治疗对肿瘤进展的影响。共同IP,采用westernblot和PCR方法研究其分子机制。分子对接用于模拟UBC12和TRIM25的蛋白质结合。分子动力学模拟观察TRIM25蛋白构象的变化。
    结果:我们发现在对PTX不敏感的TNBC中,NEDD8和NEDD8接合酶UBC12高度表达。用NEDD8激活酶(NAE)抑制剂mln4924或UBC12敲低治疗显着增加了肿瘤对PTX的敏感性,这种敏感性的增加与UBC12介导的自噬激活有关。机械上,UBC12可以在K117处将NEDD8转移到含有25(TRIM25)的E3泛素连接酶三方基序。分子动力学模拟表明,TRIM25的neddylation修饰降低了其RING域的空间位阻,促进TRIM25和泛素化底物的结合。随后,TRIM25通过增加转录因子EB(TFEB)的K63-聚泛素化促进自噬相关基因的核易位和转录,从而降低肿瘤对PTX的敏感性。
    结论:Neddylation在PTX不敏感的TNBC中被激活。具体来说,UBC12/TRIM25/TFEB轴介导的自噬基因转录激活降低了TNBC对PTX的敏感性。Neddylation抑制与PTX治疗的组合显示出协同抗肿瘤作用。
    BACKGROUND: Paclitaxel (PTX) treatment resistance is an important factor leading to poor prognosis in triple-negative breast cancer (TNBC), therefore there is an urgent need to identify new target for combination therapy. Neddylation is a post-translational process that introduces a ubiquitin-like protein called neural precursor cell expressed developmentally downregulated protein 8 (NEDD8). Previous studies have found that neddylation is activated in multiple tumors, but its relationship with PTX chemotherapy sensitivity has not been reported.
    METHODS: Differences in UBC12 and NEDD8 expression levels between PTX-sensitive and PTX-insensitive TNBC tissues were validated using public databases and immunohistochemistry. The in vitro and in vivo functional experiments were used to observe the effect of neddylation inhibition combined with PTX therapy on tumor progression. Co-IP, western blot and PCR assays were used to investigate the molecular mechanisms. Molecular docking was used to simulate the protein binding of UBC12 and TRIM25. Molecular dynamics simulation was used to observe the changes in TRIM25 protein conformation.
    RESULTS: We found that in TNBC that is insensitive to PTX, NEDD8 and NEDD8 conjugating enzyme UBC12 are highly expressed. Treatment with the NEDD8-activating enzyme (NAE) inhibitor mln4924 or knockdown of UBC12 significantly increased the sensitivity of the tumor to PTX, and this increase in sensitivity is related to UBC12-mediated autophagy activation. Mechanistically, UBC12 can transfer NEDD8 to E3 ubiquitin ligase tripartite motif containing 25 (TRIM25) at K117. Molecular dynamics simulations indicate that the neddylation modification of TRIM25 reduces steric hindrance in its RING domain, facilitating the binding of TRIM25 and ubiquitylated substrates. Subsequently, TRIM25 promotes the nuclear translocation of transcription factor EB (TFEB) and transcription of autophagy related genes by increasing K63-polyubiquitination of TFEB, thereby reducing tumor sensitivity to PTX.
    CONCLUSIONS: Neddylation is activated in PTX-insensitive TNBC. Specifically, autophagy gene transcriptional activation mediated by the UBC12/TRIM25/TFEB axis reduces TNBC sensitivity to PTX. Neddylation suppression combination with PTX treatment shows a synergistic anti-tumor effect.
    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
    费城染色体阳性(Ph)白血病是一种致命的血液恶性肿瘤。尽管使用酪氨酸激酶抑制剂(TKIs)的标准治疗在延长患者生存期方面取得了显著成功,不容忍,复发,对于Ph+白血病患者,TKI耐药仍然是严重的问题。这里,我们报道了一个新的白血病发生过程,其中RAPSYN和BCR-ABL共同出现在Ph+白血病中,和RAPSKYN介导BCR-ABL的neddylation。因此,NeddylatedBCR-ABL通过竞争其c-CBL介导的降解来增强稳定性。此外,SRC磷酸化RAPSYN以激活其NEDD8E3连接酶活性,促进BCR-ABL稳定和疾病进展。此外,与基于PROTAC的降解剂的体内无效相反,RAPPYN表达的缺失,或其连接酶活性降低BCR-ABL稳定性,反过来,抑制肿瘤的形成和生长。总的来说,这些发现代表了癌蛋白和白血病细胞的酪氨酸激酶活性的替代方法,并为靶向RAPSYN介导的BCR-ABLneddylation治疗Ph+白血病提供了理论基础.
    慢性粒细胞白血病(简称CML)约占美国成年人诊断的所有血癌的15%。该病症的特征在于不成熟免疫细胞的过度产生,其干扰适当的血液功能。它与基因重组(一种突变)有关,该基因重组导致白细胞产生异常的“BCR-ABL”酶,该酶始终处于打开状态。反过来,这种过度活跃的蛋白质会导致细胞活得更长,分裂不受控制。目前可用于控制疾病的一些最有效的药物通过阻断BCR-ABL的活性起作用。然而,随着时间的推移,某些患者可能会对这些治疗产生抗药性,导致他们复发。因此,需要其他方法来控制这种疾病;特别是,一个有希望的研究途径在于探索是否有可能减少患病细胞中存在的酶的量。作为这项努力的一部分,赵,戴,Li,张等人。专注于RAPPYN,CML细胞中以前未知的支架蛋白。在其他组织中,它最近被证明参与neddylation-一种过程,蛋白质接受某些化学“标签”,从而改变它们的行为方式。实验表明,与健康志愿者相比,RAPSYN在CML患者的白细胞中以高得多的水平存在。通过实验降低CML细胞中RAPSYN的含量导致这些细胞的分裂速度降低-无论是在培养皿中还是在小鼠体内注射时,同时也与BCR-ABL水平降低有关。其他生化实验表明,RAPSYN坚持与BCR-ABL添加化学“标签”,保护异常蛋白质免受降解,提高其总体水平。最后,团队展示了SRC,一种在新出现的癌症中经常失调的酶,可以激活RAPSYN进行Neddylation的能力;这种机制可以促进BCR-ABL稳定,反过来,疾病进展。一起来看,这些实验表明了一种控制BCR-ABL水平的新方法。未来的研究应该调查RAPSYN是否也能稳定白血病对现有药物耐药的患者的BCR-ABL。最终,RAPSYN可能为克服CML患者的耐药性提供新的靶点。
    Philadelphia chromosome-positive (Ph+) leukemia is a fatal hematological malignancy. Although standard treatments with tyrosine kinase inhibitors (TKIs) have achieved remarkable success in prolonging patient survival, intolerance, relapse, and TKI resistance remain serious issues for patients with Ph+ leukemia. Here, we report a new leukemogenic process in which RAPSYN and BCR-ABL co-occur in Ph+ leukemia, and RAPSYN mediates the neddylation of BCR-ABL. Consequently, neddylated BCR-ABL enhances the stability by competing its c-CBL-mediated degradation. Furthermore, SRC phosphorylates RAPSYN to activate its NEDD8 E3 ligase activity, promoting BCR-ABL stabilization and disease progression. Moreover, in contrast to in vivo ineffectiveness of PROTAC-based degraders, depletion of RAPSYN expression, or its ligase activity decreased BCR-ABL stability and, in turn, inhibited tumor formation and growth. Collectively, these findings represent an alternative to tyrosine kinase activity for the oncoprotein and leukemogenic cells and generate a rationale of targeting RAPSYN-mediated BCR-ABL neddylation for the treatment of Ph+ leukemia.
    Chronic myeloid leukemia (CML for short) accounts for about 15% of all blood cancers diagnosed in adults in the United States. The condition is characterized by the overproduction of immature immune cells that interfere with proper blood function. It is linked to a gene recombination (a type of mutation) that leads to white blood cells producing an abnormal ‘BCR-ABL’ enzyme which is always switched on. In turn, this overactive protein causes the cells to live longer and divide uncontrollably. Some of the most effective drugs available to control the disease today work by blocking the activity of BCR-ABL. Yet certain patients can become resistant to these treatments over time, causing them to relapse. Other approaches are therefore needed to manage this disease; in particular, a promising avenue of research consists in exploring whether it is possible to reduce the amount of the enzyme present in diseased cells. As part of this effort, Zhao, Dai, Li, Zhang et al. focused on RAPSYN, a scaffolding protein previously unknown in CML cells. In other tissues, it has recently been shown to participate in neddylation – a process by which proteins receive certain chemical ‘tags’ that change the way they behave. The experiments revealed that, compared to healthy volunteers, RAPSYN was present at much higher levels in the white blood cells of CML patients. Experimentally lowering the amount of RAPSYN in CML cells led these to divide less quickly – both in a dish and when injected in mice, while also being linked to decreased levels of BCR-ABL. Additional biochemical experiments indicated that RAPSYN sticks with BCR-ABL to add chemical ‘tags’ that protect the abnormal protein against degradation, therefore increasing its overall levels. Finally, the team showed that SRC, an enzyme often dysregulated in emerging cancers, can activate RAPSYN’s ability to conduct neddylation; such mechanism could promote BCR-ABL stabilization and, in turn, disease progression. Taken together, these experiments indicate a new way by which BCR-ABL levels are controlled. Future studies should investigate whether RAPSYN also stabilizes BCR-ABL in patients whose leukemias have become resistant to existing drugs. Eventually, RAPSYN may offer a new target for overcoming drug-resistance in CML patients.
    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
    酪氨酸激酶抑制剂已成为费城染色体阳性(Ph)白血病患者的标准治疗方法。然而,一系列的问题,包括耐药性,复发和不容忍,仍然是未满足的医疗需求。这里,我们报道了Ph白血病细胞系中基于靶向siRNA的脂质纳米颗粒用于Ph白血病的基因治疗,特异性靶向Ph白血病细胞中最近鉴定的NEDD8E3连接酶RAPSYN,以破坏致癌BCR-ABL的neddylation。为了实现Ph+白血病治疗的特异性,将抗CD79B单克隆抗体的单链片段可变区(scFv)共价缀合在OA2-siRAPSYN脂质纳米颗粒的表面,以生成靶向脂质纳米颗粒(scFv-OA2-siRAPSYN).通过纳米颗粒有效沉默白血病细胞系中的RAPSYN基因,BCR-ABL明显降解,同时抑制增殖和促进凋亡。具体的目标,在细胞系来源的小鼠模型中进一步评估和证明了治疗效果和系统安全性.本研究不仅解决了Ph+白血病的临床需要,但也使基因疗法对一个不太容易用药的目标。
    Tyrosine kinase inhibitors have been the standard treatment for patients with Philadelphia chromosome-positive (Ph+) leukemia. However, a series of issues, including drug resistance, relapse and intolerance, are still an unmet medical need. Here, we report the targeted siRNA-based lipid nanoparticles in Ph+ leukemic cell lines for gene therapy of Ph+ leukemia, which specifically targets a recently identified NEDD8 E3 ligase RAPSYN in Ph+ leukemic cells to disrupt the neddylation of oncogenic BCR-ABL. To achieve the specificity for Ph+ leukemia therapy, a single-chain fragment variable region (scFv) of anti-CD79B monoclonal antibody was covalently conjugated on the surface of OA2-siRAPSYN lipid nanoparticles to generate the targeted lipid nanoparticles (scFv-OA2-siRAPSYN). Through effectively silencing RAPSYN gene in leukemic cell lines by the nanoparticles, BCR-ABL was remarkably degraded accompanied by the inhibition of proliferation and the promotion of apoptosis. The specific targeting, therapeutic effects and systemic safety were further evaluated and demonstrated in cell line-derived mouse models. The present study has not only addressed the clinical need of Ph+ leukemia, but also enabled gene therapy against a less druggable target.
    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
    免疫检查点阻断疗法旨在激活免疫系统以消除癌细胞。然而,临床获益仅记录在一部分患者中.这里,我们在肿瘤免疫共培养系统中利用全基因组CRISPR/Cas9筛选,重点关注三阴性乳腺癌(TNBC)。我们发现NEDD8在癌细胞中的丢失导致nivolumab(抗PD-1)的脆弱性。NEDD8的遗传缺失最初仅延迟细胞分裂,但恢复后细胞增殖不受影响。由于NEDD8基因通常是必需的,我们通过额外的CRISPR筛选验证了这一观察结果,并使用蛋白质组学揭示了NEDD8缺陷细胞中增强的免疫原性.在雌性免疫活性小鼠中,PD-1阻断缺乏对已建立的EO771乳腺癌肿瘤的功效。相比之下,我们观察到PD-1阻断后CD8+T细胞介导的针对Nedd8缺陷型EO771肿瘤的肿瘤消退。实质上,我们提供的证据表明NEDD8在TNBC中是有条件必需的,并且是PD-1/L1阻断治疗的协同药物靶点.
    Immune checkpoint blockade therapy aims to activate the immune system to eliminate cancer cells. However, clinical benefits are only recorded in a subset of patients. Here, we leverage genome-wide CRISPR/Cas9 screens in a Tumor-Immune co-Culture System focusing on triple-negative breast cancer (TNBC). We reveal that NEDD8 loss in cancer cells causes a vulnerability to nivolumab (anti-PD-1). Genetic deletion of NEDD8 only delays cell division initially but cell proliferation is unaffected after recovery. Since the NEDD8 gene is commonly essential, we validate this observation with additional CRISPR screens and uncover enhanced immunogenicity in NEDD8 deficient cells using proteomics. In female immunocompetent mice, PD-1 blockade lacks efficacy against established EO771 breast cancer tumors. In contrast, we observe tumor regression mediated by CD8+ T cells against Nedd8 deficient EO771 tumors after PD-1 blockade. In essence, we provide evidence that NEDD8 is conditionally essential in TNBC and presents as a synergistic drug target for PD-1/L1 blockade 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
    E3连接酶-degron相互作用决定了泛素-蛋白酶体系统的特异性。我们最近发现FEM1B,Cullin2-RING连接酶(CRL2)的底物受体,识别包含C末端脯氨酸的C-degrons。通过求解与不同C-degrons结合的CRL2FEM1B的几种低温-EM结构,我们阐明了复杂的二聚体组装。此外,我们揭示了未修饰和内化的CRL2FEM1B的不同二聚化状态,以揭示NEDD8介导的CRL2FEM1B激活机制。我们的研究还表明,FEM1B利用两部分机制来识别底物内的C末端脯氨酸和上游芳族残基。这些结构性发现,补充体外泛素化和体内基于细胞的测定,证明CRL2FEM1B介导的多泛素化和随后的蛋白质周转取决于FEM1B-degron相互作用和E3连接酶复合物的二聚化状态。总的来说,这项研究加深了我们对Cullin-RINGE3连接酶底物选择如何介导蛋白质周转的分子理解。
    The E3 ligase-degron interaction determines the specificity of the ubiquitin‒proteasome system. We recently discovered that FEM1B, a substrate receptor of Cullin 2-RING ligase (CRL2), recognizes C-degrons containing a C-terminal proline. By solving several cryo-EM structures of CRL2FEM1B bound to different C-degrons, we elucidate the dimeric assembly of the complex. Furthermore, we reveal distinct dimerization states of unmodified and neddylated CRL2FEM1B to uncover the NEDD8-mediated activation mechanism of CRL2FEM1B. Our research also indicates that, FEM1B utilizes a bipartite mechanism to recognize both the C-terminal proline and an upstream aromatic residue within the substrate. These structural findings, complemented by in vitro ubiquitination and in vivo cell-based assays, demonstrate that CRL2FEM1B-mediated polyubiquitination and subsequent protein turnover depend on both FEM1B-degron interactions and the dimerization state of the E3 ligase complex. Overall, this study deepens our molecular understanding of how Cullin-RING E3 ligase substrate selection mediates protein turnover.
    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
    泛素连接通常由标志E3催化结构域执行。两个这样的领域,\'cullin-RING\'和\'RBR\',分别存在于数百种人类E3连接酶中,并与E2酶合作催化泛素化。然而,具有RBX1的脊椎动物特异性CUL9复合物(也称为ROC1),由于其与TP53的肿瘤抑制性相互作用而感兴趣,独特地包括cullin-RING和RBR结构域。这里,cryo-EM,生物化学和细胞测定阐明了1.8-MDa六聚体人CUL9-RBX1组装。在一个二聚体亚复合体中,E2结合的RBR结构域通过其自身的cullin结构域的neddylation并以反式从相邻的CUL9-RBX1定位而被激活。我们的数据显示CUL9在RBX1结合的cullins中独特,依赖于后生动物特异性UBE2F内丁化酶,而RBR结构域保护它免受去乙酰化。基质被招募到各个上游领域,而泛素化依赖于CUL9的内化cullin和RBR结构域实现自组装和嵌合cullin-RING/RBRE3连接酶活性。
    Ubiquitin ligation is typically executed by hallmark E3 catalytic domains. Two such domains, \'cullin-RING\' and \'RBR\', are individually found in several hundred human E3 ligases, and collaborate with E2 enzymes to catalyze ubiquitylation. However, the vertebrate-specific CUL9 complex with RBX1 (also called ROC1), of interest due to its tumor suppressive interaction with TP53, uniquely encompasses both cullin-RING and RBR domains. Here, cryo-EM, biochemistry and cellular assays elucidate a 1.8-MDa hexameric human CUL9-RBX1 assembly. Within one dimeric subcomplex, an E2-bound RBR domain is activated by neddylation of its own cullin domain and positioning from the adjacent CUL9-RBX1 in trans. Our data show CUL9 as unique among RBX1-bound cullins in dependence on the metazoan-specific UBE2F neddylation enzyme, while the RBR domain protects it from deneddylation. Substrates are recruited to various upstream domains, while ubiquitylation relies on both CUL9\'s neddylated cullin and RBR domains achieving self-assembled and chimeric cullin-RING/RBR E3 ligase activity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Review
    Neddylation,类似于泛素化,代表蛋白质的翻译后修饰,其中神经前体细胞表达的发育下调蛋白8(NEDD8)通过一系列反应在底物蛋白上进行修饰。Neddylation在动物细胞的生长和增殖中起着关键作用。在结直肠癌(CRC)中,它主要有助于扩散,肿瘤细胞的转移和存活,降低患者总体生存率。NEDD8介导的Neddylation通路的战略操纵在通过调节癌细胞内多种生物反应来调节肿瘤生长的潜力方面具有巨大的治疗前景。如DNA损伤反应和细胞凋亡,在其他人中。MLN4924是NEDD8的抑制剂,它与铂类药物和伊立替康联合使用,以及通过药物再利用筛选的循环抑制剂和NEDD激活酶抑制剂,已发现发挥有希望的抗肿瘤作用。本综述总结了在理解NEDD8在促进CRC方面的作用方面取得的最新进展,表明NEDD8是一个有前途的抗CRC目标。
    Neddylation, akin to ubiquitination, represents a post‑translational modification of proteins wherein neural precursor cell‑expressed developmentally downregulated protein 8 (NEDD8) is modified on the substrate protein through a series of reactions. Neddylation plays a pivotal role in the growth and proliferation of animal cells. In colorectal cancer (CRC), it predominantly contributes to the proliferation, metastasis and survival of tumor cells, decreasing overall patient survival. The strategic manipulation of the NEDD8‑mediated neddylation pathway holds immense therapeutic promise in terms of the potential to modulate the growth of tumors by regulating diverse biological responses within cancer cells, such as DNA damage response and apoptosis, among others. MLN4924 is an inhibitor of NEDD8, and its combined use with platinum drugs and irinotecan, as well as cycle inhibitors and NEDD activating enzyme inhibitors screened by drug repurposing, has been found to exert promising antitumor effects. The present review summarizes the recent progress made in the understanding of the role of NEDD8 in the advancement of CRC, suggesting that NEDD8 is a promising anti‑CRC target.
    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
    Cullin(CUL)-RING(真正有趣的新基因)E3泛素(Ub)连接酶(CRLs)是最大的E3家族。E3CRL核心连接酶是由与ROC1/RBX1RING指蛋白结合的CULC末端结构域形成的亚复合物,它作为一个枢纽,调解和组织与E2、Ub、Nedd8和ARIH家族蛋白,从而导致Ub转移到E3结合的底物。本报告描述了小分子化合物对CRL依赖性泛素化的调节,包括KH-4-43,#33和苏拉明,以CRL核心连接酶为目标。我们还显示KH-4-43和#33均抑制CRL4CRBN对CK1α的泛素化。然而,当使用Neddylated形式的CRL4CRBN时,任一化合物对该反应的抑制作用显著降低。另一方面,#33和KH-4-43几乎相等地抑制CRL1β-TrCP和Nedd8-CRL1β-TrCP对β-联蛋白的泛素化。因此,CRL1β-TrCP的neddylation化不会负面影响对#33和KH-4-43的抑制作用的敏感性。这些发现表明neddylation改变KH-4-43/#33对CRL抑制的敏感性的作用取决于具体的CRL类型。苏拉明,一种针对CUL基本峡谷的化合物,能有效抑制CRL1/4依赖的泛素化,无论Neddylation状态如何,与用KH-4-43/#33观察到的结果相反。观察到的KH-4-43/#33的差异药物敏感性似乎反映了CUL特异性Nedd8对CRL的影响,如最近的高分辨率结构生物学研究所揭示的。高度多样化的CRL核心连接酶结构可以为小分子调节剂的特异性靶向提供机会。
    Cullin (CUL)-RING (Really Interesting New Gene) E3 ubiquitin (Ub) ligases (CRLs) are the largest E3 family. The E3 CRL core ligase is a subcomplex formed by the CUL C-terminal domain bound with the ROC1/RBX1 RING finger protein, which acts as a hub that mediates and organizes multiple interactions with E2, Ub, Nedd8, and the ARIH family protein, thereby resulting in Ub transfer to the E3-bound substrate. This report describes the modulation of CRL-dependent ubiquitination by small molecule compounds including KH-4-43, #33, and suramin, which target the CRL core ligases. We show that both KH-4-43 and #33 inhibit the ubiquitination of CK1α by CRL4CRBN. However, either compound\'s inhibitory effect on this reaction is significantly reduced when a neddylated form of CRL4CRBN is used. On the other hand, both #33 and KH-4-43 inhibit the ubiquitination of β-catenin by CRL1β-TrCP and Nedd8-CRL1β-TrCP almost equally. Thus, neddylation of CRL1β-TrCP does not negatively impact the sensitivity to inhibition by #33 and KH-4-43. These findings suggest that the effects of neddylation to alter the sensitivity of CRL inhibition by KH-4-43/#33 is dependent upon the specific CRL type. Suramin, a compound that targets CUL\'s basic canyon, can effectively inhibit CRL1/4-dependent ubiquitination regardless of neddylation status, in contrast to the results observed with KH-4-43/#33. This observed differential drug sensitivity of KH-4-43/#33 appears to echo CUL-specific Nedd8 effects on CRLs as revealed by recent high-resolution structural biology efforts. The highly diversified CRL core ligase structures may provide opportunities for specific targeting by small molecule modulators.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号