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
    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
    翻译后修饰和微调的蛋白质周转在哺乳动物早期胚胎发育中非常重要。除了经典的蛋白质降解促进泛素化,泛素化修饰的新形式尚未完全理解。这里,我们证明了一种泛素化修饰的功能和潜在机制,Neddylation,在小鼠植入前胚胎发育中。用特异性抑制剂治疗,受精卵显示出显着降低的卵裂率,几乎所有都未能进入4细胞阶段。转录谱分析显示基因在涉及细胞命运决定和细胞分化的途径中差异表达,包括几个下调合子基因组激活(ZGA)标记基因。检测到磷酸化RNA聚合酶II的水平降低,表明胚胎细胞核内的基因转录受损。蛋白质组数据显示差异表达的蛋白质富含组蛋白修饰。我们证实了甲基转移酶(KMT2D)表达的降低和组蛋白H3K4me3的降低。同时,乙酰转移酶(CBP/p300)减少,而去乙酰化酶(HDAC6)增加,导致组蛋白H3K27ac的衰减。此外,我们观察到YAP1和RPL13活性的上调,提示Hippo信号通路下游反应的潜在异常。总之,我们发现抑制neddylation可引起早期胚胎的表观遗传变化,并导致相关下游信号通路异常。这项研究揭示了调节哺乳动物胚胎发育的新形式的泛素化,并可能有助于进一步研究女性不孕症的病理学。
    Post-translational modification and fine-tuned protein turnover are of great importance in mammalian early embryo development. Apart from the classic protein degradation promoting ubiquitination, new forms of ubiquitination-like modification are yet to be fully understood. Here, we demonstrate the function and potential mechanisms of one ubiquitination-like modification, neddylation, in mouse preimplantation embryo development. Treated with specific inhibitors, zygotes showed a dramatically decreased cleavage rate and almost all failed to enter the 4-cell stage. Transcriptional profiling showed genes were differentially expressed in pathways involving cell fate determination and cell differentiation, including several down-regulated zygotic genome activation (ZGA) marker genes. A decreased level of phosphorylated RNA polymerase II was detected, indicating impaired gene transcription inside the embryo cell nucleus. Proteomic data showed that differentially expressed proteins were enriched in histone modifications. We confirmed the lowered in methyltransferase (KMT2D) expression and a decrease in histone H3K4me3. At the same time, acetyltransferase (CBP/p300) reduced, while deacetylase (HDAC6) increased, resulting in an attenuation in histone H3K27ac. Additionally, we observed the up-regulation in YAP1 and RPL13 activities, indicating potential abnormalities in the downstream response of Hippo signaling pathway. In summary, we found that inhibition of neddylation induced epigenetic changes in early embryos and led to abnormalities in related downstream signaling pathways. This study sheds light upon new forms of ubiquitination regulating mammalian embryonic development and may contribute to further investigation of female infertility pathology.
    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
    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
    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
    阿霉素(Dox)是临床肿瘤学中广泛使用的化学治疗剂,用于治疗各种癌症。然而,其临床应用受到其显著副作用的限制。其中,心肌病的发展,以心脏重塑和最终的心力衰竭为特征,是Dox化疗后的主要问题。在我们目前的调查中,我们展示了MLN4924通过直接抑制NEDD8激活酶减轻阿霉素诱导的心脏毒性的功效,NAE。MLN4924证明了多柔比星治疗后稳定线粒体功能的能力,减少心肌细胞凋亡,减轻氧化应激诱导的心肌损伤,增强心脏收缩功能,减轻心脏纤维化,并阻碍与心力衰竭相关的心脏重塑。在机械层面,MLN4924通过抑制NEDD8激活酶干预了Neddylation过程,NAE,在阿霉素治疗后的鼠心脏组织内。这种干预导致NEDD8蛋白表达的抑制,Neddylation活性降低,以及心脏保护作用的相应表现。总的来说,我们的研究结果认为MLN4924是一种潜在的治疗途径,通过NAE抑制减弱增强的neddylation活性来减轻阿霉素诱导的心脏毒性,从而为患病的患者提供了一种可行且有希望的治疗方式。
    Doxorubicin (DOX) is a widely utilized chemotherapeutic agent in clinical oncology for treating various cancers. However, its clinical use is constrained by its significant side effects. Among these, the development of cardiomyopathy, characterized by cardiac remodeling and eventual heart failure, stands as a major concern following DOX chemotherapy. In our current investigation, we have showcased the efficacy of MLN4924 in mitigating doxorubicin-induced cardiotoxicity through direct inhibition of the NEDD8-activating enzyme, NAE. MLN4924 demonstrated the ability to stabilize mitochondrial function post-doxorubicin treatment, diminish cardiomyocyte apoptosis, alleviate oxidative stress-induced damage in the myocardium, enhance cardiac contractile function, mitigate cardiac fibrosis, and impede cardiac remodeling associated with heart failure. At the mechanistic level, MLN4924 intervened in the neddylation process by inhibiting the NEDD8 activating enzyme, NAE, within the murine cardiac tissue subsequent to doxorubicin treatment. This intervention resulted in the suppression of NEDD8 protein expression, reduction in neddylation activity, and consequential manifestation of cardioprotective effects. Collectively, our findings posit MLN4924 as a potential therapeutic avenue for mitigating doxorubicin-induced cardiotoxicity by attenuating heightened neddylation activity through NAE inhibition, thereby offering a viable and promising treatment modality for afflicted patients.
    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
    糖尿病视网膜病变(DR)是糖尿病(DM)的高度危险和广泛的并发症。积累的活性氧(ROS)在DR的发展中起着核心作用。这项研究的目的是研究间充质干细胞(MSC)衍生的小细胞外囊泡(sEV)对调节DR中ROS和视网膜损伤的影响和机制。玻璃体内注射sEV抑制Cullin3neddylation,稳定的Nrf2,减少的ROS,减少视网膜炎症,抑制了Müller胶质增生,并减轻了DR。基于MSC-sEVmiRNA测序,生物信息学软件,和双荧光素酶报告分析,miR-143-3p被鉴定为MSC-sEV在调节神经前体细胞发育下调8(NEDD8)介导的Neddylation中的作用的关键效应。sEV能够被Müller细胞内化。与晚期糖基化终产物(AGEs)诱导的Müller细胞相比,sEV共培养减少Cullin3neddylation,激活Nrf2信号通路以对抗ROS诱导的炎症。当用AGEs诱导的Müller细胞的上清液处理内皮细胞时,内皮细胞的屏障功能受损,但用AGEs诱导的Müller细胞与sEV共培养的上清液处理后恢复。sEV的保护作用是,然而,当miR-143-3p在sEV中被抑制时受损。此外,当NEDD8在Müller细胞中过表达时,sEV的保护功效减弱。这些发现显示MSC-sEV递送miR-143-3p以抑制Cullin3neddylation,稳定Nrf2以抵消ROS诱导的炎症并减少血管渗漏。我们的研究结果表明,MSC-sEV可能是DR的潜在纳米治疗剂,Cullin3neddylation可能是DR治疗的新靶点。
    Diabetic retinopathy (DR) is a highly hazardous and widespread complication of diabetes mellitus (DM). The accumulated reactive oxygen species (ROS) play a central role in DR development. The aim of this research was to examine the impact and mechanisms of mesenchymal stem cell (MSC)-derived small extracellular vesicles (sEV) on regulating ROS and retinal damage in DR. Intravitreal injection of sEV inhibited Cullin3 neddylation, stabilized Nrf2, decreased ROS, reduced retinal inflammation, suppressed Müller gliosis, and mitigated DR. Based on MSC-sEV miRNA sequencing, bioinformatics software, and dual-luciferase reporter assay, miR-143-3p was identified to be the key effector for MSC-sEV\'s role in regulating neural precursor cell expressed developmentally down-regulated 8 (NEDD8)-mediated neddylation. sEV were able to be internalized by Müller cells. Compared to advanced glycation end-products (AGEs)-induced Müller cells, sEV coculture decreased Cullin3 neddylation, activated Nrf2 signal pathway to combat ROS-induced inflammation. The barrier function of endothelial cells was impaired when endothelial cells were treated with the supernatant of AGEs-induced Müller cells, but was restored when treated with supernatant of AGEs-induced Müller cells cocultured with sEV. The protective effect of sEV was, however, compromised when miR-143-3p was inhibited in sEV. Moreover, the protective efficacy of sEV was diminished when NEDD8 was overexpressed in Müller cells. These findings showed MSC-sEV delivered miR-143-3p to inhibit Cullin3 neddylation, stabilizing Nrf2 to counteract ROS-induced inflammation and reducing vascular leakage. Our findings suggest that MSC-sEV may be a potential nanotherapeutic agent for DR, and that Cullin3 neddylation could be a new target for DR 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: 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: Review
    人类的衰老与腹部分布和身体脂肪的重塑以及肥胖等代谢性疾病的患病率逐渐增加有关。2型糖尿病和脂肪肝,以及发生代谢并发症的风险。通过了解与年龄相关的代谢紊乱发作的详细机制,可以改善当前的治疗方法。Neddylation,将泛素样蛋白NEDD8添加到底物蛋白的翻译后修饰,最近与年龄相关的代谢性疾病有关,开辟新的调查途径,并提出治疗这些疾病的潜在目标。在这次审查中,我们将关注NEDD8介导的Neddylation在年龄相关的代谢失调中的潜在作用,胰岛素抵抗,肥胖,2型糖尿病和脂肪肝。我们认为NEDD8介导的neddylation的改变有助于引发胰岛素抵抗和年龄相关的代谢失调的发展。因此强调NEDD8是预防年龄相关代谢性疾病的有希望的治疗靶标。
    Aging in humans is associated with abdominal distribution and remodeling of body fat and a parallel gradual increase in the prevalence of metabolic diseases such as obesity, type 2 diabetes mellitus and fatty liver disease, as well as the risk of developing metabolic complications. Current treatments might be improved by understanding the detailed mechanisms underlying the onset of age-related metabolic disorders. Neddylation, a post-translational modification that adds the ubiquitin-like protein NEDD8 to substrate proteins, has recently been linked to age-related metabolic diseases, opening new avenues of investigation and raising a potential target for treatment of these diseases. In this review, we will focus on the potential role of NEDD8-mediated neddylation in age-related metabolic dysregulation, insulin resistance, obesity, type 2 diabetes mellitus and fatty liver. We propose that alterations in NEDD8-mediated neddylation contribute to triggering insulin resistance and the development of age-related metabolic dysregulation, thus highlighting NEDD8 as a promising therapeutic target for preventing age-related metabolic diseases.
    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