Proteasome inhibitors

蛋白酶体抑制剂
  • 文章类型: Journal Article
    计算药物基因组学可以潜在地确定已经批准的药物的新适应症,并确定具有相似作用机制的化合物。这里,我们使用基于转录组学数据和基于结构的虚拟筛选的综合药物重定位方法来鉴定具有与三种已知蛋白酶体抑制剂(PIs;硼替佐米,MG-132和MLN-2238)。然后进行候选化合物的体外验证以评估蛋白酶体蛋白水解活性,泛素化蛋白质的积累,细胞活力,和药物在A375黑色素瘤和MCF7乳腺癌细胞中的诱导表达。使用这种方法,我们确定了六个具有PI性质的化合物((-)-激动素-核苷,Manumycin-A,盐酸嘌呤霉素,抗霉素,马来酸替加色罗德,和thapsigargin)。尽管对接评分确定了它们与β5亚基结合的能力,我们的体外研究表明,这些化合物在一定程度上抑制了β1,β2和β5催化位点。如硼替佐米所示,只有Manumycin-A,盐酸嘌呤霉素,和马来酸替加色罗导致泛素化蛋白的过度积累和HMOX1表达升高。一起来看,我们的综合药物重定位方法和随后的体外验证研究鉴定出6种化合物,这些化合物具有与蛋白酶体抑制剂相似的性质.
    Computational pharmacogenomics can potentially identify new indications for already approved drugs and pinpoint compounds with similar mechanism-of-action. Here, we used an integrated drug repositioning approach based on transcriptomics data and structure-based virtual screening to identify compounds with gene signatures similar to three known proteasome inhibitors (PIs; bortezomib, MG-132, and MLN-2238). In vitro validation of candidate compounds was then performed to assess proteasomal proteolytic activity, accumulation of ubiquitinated proteins, cell viability, and drug-induced expression in A375 melanoma and MCF7 breast cancer cells. Using this approach, we identified six compounds with PI properties ((-)-kinetin-riboside, manumycin-A, puromycin dihydrochloride, resistomycin, tegaserod maleate, and thapsigargin). Although the docking scores pinpointed their ability to bind to the β5 subunit, our in vitro study revealed that these compounds inhibited the β1, β2, and β5 catalytic sites to some extent. As shown with bortezomib, only manumycin-A, puromycin dihydrochloride, and tegaserod maleate resulted in excessive accumulation of ubiquitinated proteins and elevated HMOX1 expression. Taken together, our integrated drug repositioning approach and subsequent in vitro validation studies identified six compounds demonstrating properties similar to proteasome inhibitors.
    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
    蛋白酶体抑制剂(PIs),如硼替佐米和卡菲佐米,是治疗多发性骨髓瘤(MM)的骨干药物。在这项研究中,我们研究了硼替佐米在MM细胞中的相互作用物,并确定了二氢硫辛酸酰胺脱氢酶(DLD)是硼替佐米的分子靶标。DLD催化二氢硫胺素氧化形成硫胺素,也产生NADH的反应。我们的数据表明硼替佐米与DLD结合并抑制MM细胞中DLD的酶促功能。DLD敲低MM细胞(DLD-KD)的NADH水平降低。减少的NADH抑制了细胞中蛋白酶体复合物的组装。因此,DLD-KDMM细胞的基础水平蛋白酶体活性降低,对硼替佐米更敏感。由于PI在诊所的许多抗MM方案中使用,我们发现DLD的高表达与MM的不良预后相关。考虑到DLD在蛋白酶体组装中的调节作用,我们评估了MM细胞中的DLD靶向治疗。DLD抑制剂CPI-613在体外和体内显示出与硼替佐米的协同抗MM作用。总的来说,我们的发现阐明了DLD是硼替佐米在MM中的替代分子靶标。DLD靶向可能会增加MM对PI的敏感性。
    Proteasome inhibitors (PIs), such as bortezomib and calfizomib, were backbone agents in the treatment of multiple myeloma (MM). In this study, we investigated bortezomib interactors in MM cells and identified dihydrolipoamide dehydrogenase (DLD) as a molecular target of bortezomib. DLD catalyzes the oxidation of dihydrolipoamide to form lipoamide, a reaction that also generates NADH. Our data showed that bortezomib bound to DLD and inhibited DLD\'s enzymatic function in MM cells. DLD knocked down MM cells (DLD-KD) had decreased levels of NADH. Reduced NADH suppressed assembly of proteasome complex in cells. As a result, DLD-KD MM cells had decreased basal-level proteasome activity and were more sensitive to bortezomib. Since PIs were used in many anti-MM regimens in clinics, we found that high expression of DLD correlated with inferior prognosis of MM. Considering the regulatory role of DLD in proteasome assembly, we evaluated DLD targeting therapy in MM cells. DLD inhibitor CPI-613 showed a synergistic anti-MM effect with bortezomib in vitro and in vivo. Overall, our findings elucidated DLD as an alternative molecular target of bortezomib in MM. DLD-targeting might increase MM sensitivity to PIs.
    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
    多发性骨髓瘤(MM)是一种普遍但无法治愈的血液系统恶性肿瘤。尽管蛋白酶体抑制剂在治疗MM方面的疗效已得到证实,部分患者对硼替佐米治疗的耐药性持续存在。这项病例对照研究探讨了循环内皮祖细胞(EPCs)作为预测MM患者对基于蛋白酶体抑制剂的治疗联合地塞米松的反应的生物标志物的潜力。这项研究是对105例接受硼替佐米加地塞米松治疗的MM患者和90例健康个体作为对照组进行的。利用8色多参数流式细胞术,我们评估了循环EPC的水平,在诊断时和一个治疗周期(4周)后,通过CD34FITC和CD309PE标志物鉴定。我们的发现表明,对治疗反应较差的患者显示出明显高于反应良好的患者的CD34/CD309值(p<0.001)。建立基于CD34/CD309表达的反应描述,其中对于百分比(产生100%灵敏度和94.1%特异性)的截止值≤0.9,对于绝对值(也具有100%灵敏度和94.1%特异性)的截止值≤12.5。这些结果强调了EPC人口水平的潜力,通过CD34/CD309定量,作为接受蛋白酶体抑制剂和地塞米松治疗的MM患者免疫调节治疗的预测生物标志物。
    Multiple myeloma (MM) is a prevalent yet incurable hematologic malignancy. Despite the proven efficacy of proteasome inhibitors in treating MM, resistance to Bortezomib-based treatments persists in a subset of patients. This case control study explores the potential of circulating endothelial progenitor cells (EPCs) as biomarkers for predicting response to Proteasome Inhibitor based therapy combined with Dexamethasone in MM patients. This study was conducted on 105 MM patients receiving bortezomib plus dexamethasone therapy and 90 healthy individuals as a control group. Utilizing 8-color multi-parameter flow cytometry, we assessed the levels of circulating EPCs, identified through CD34 FITC and CD309 PE markers at diagnosis and after one treatment cycle (4 weeks). Our findings revealed that patients exhibiting poor response to therapy showed significantly higher CD34/CD309 values than those with a good response (p < 0.001). The delineation of response based on CD34/CD309 expression was established with a cutoff ≤ 0.9 for percentage (yielding 100% sensitivity and 94.1% specificity) and ≤ 12.5 for absolute value (also with 100% sensitivity and 94.1% specificity). These results underscore the potential of EPC population levels, as quantified by CD34/CD309, to serve as a predictive biomarker for immunomodulatory treatment in MM patients undergoing Proteasome Inhibitor and Dexamethasone 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
    癌细胞的免疫原性受多种因素的影响,包括主要组织相容性复合体I类(MHC-I)的表达,抗原表达,和蛋白酶体产生的表位肽的库。恶性胸膜间皮瘤细胞系ACC-MEOS-4(MESO-4)表达高水平的MHC-I和Wilms肿瘤1(WT1)肿瘤抗原。使用对HLA-A*24:02限制性WT1表位(WT1235,CMTWNQMNL)具有特异性的功能性T细胞报告基因测定,我们寻找增强MESO-4免疫原性的因素,重点是蛋白酶体,在抗原加工机制中起着核心作用。ONX-0914,免疫蛋白酶体亚基β5i的选择性抑制剂,在低浓度下剂量依赖性地增强免疫原性而没有细胞毒性。此外,识别WT1的CD8+T淋巴细胞对用ONX-0914预处理的MESO-4显示更大的细胞毒性。MESO-4表达标准蛋白酶体(SP)和免疫蛋白酶体(IP)。值得注意的是,IP具有与SP不同的催化活性,有利于在抗原呈递细胞和癌细胞中产生对MHC-I具有高亲和力的抗原肽。体外,免疫蛋白酶体消化试验和质谱分析表明,IP在疏水残基后内部切割WT1235。重要的是,ONX-0914减轻了WT1235表位的这种内部切割。这些结果表明,ONX-0914可以防止IP对WT1235的内部破坏性裂解,从而促进MESO-4对WT1表位的特异性呈递。总之,选择性IP抑制剂可能通过指导特定肿瘤表位的呈递提供调节癌细胞免疫原性的手段。
    The immunogenicity of cancer cells is influenced by several factors, including the expression of the major histocompatibility complex class I (MHC-I), antigen expression, and the repertoire of proteasome-produced epitope peptides. The malignant pleural mesothelioma cell line ACC-MEOS-4 (MESO-4) expresses high levels of MHC-I and Wilms tumor 1 (WT1) tumor antigens. Using a functional T cell reporter assay specific for the HLA-A*24:02 restricted WT1 epitope (WT1235, CMTWNQMNL), we searched for factors that augmented the immunogenicity of MESO-4, focusing on proteasomes, which have a central role in the antigen processing machinery. ONX-0914, a selective inhibitor of the immunoproteasome subunit β5i, enhanced immunogenicity dose-dependently at low concentrations without cytotoxicity. In addition, CD8+ T lymphocytes recognizing WT1 showed greater cytotoxicity against MESO-4 pre-treated with ONX-0914. MESO-4 expresses a standard proteasome (SP) and immunoproteasome (IP). Notably, IP has distinct catalytic activity from SP, favoring the generation of antigenic peptides with high affinity for MHC-I in antigen-presenting cells and cancer cells. In vitro, immunoproteasome digestion assay and mass spectrometry analysis showed that IP cleaved WT1235 internally after the hydrophobic residues. Importantly, this internal cleavage of the WT1235 epitope was mitigated by ONX-0914. These results suggest that ONX-0914 prevents the internal destructive cleavage of WT1235 by IP, thereby promoting the specific presentation of the WT1 epitope by MESO-4. In conclusion, selective IP inhibitors might offer a means to modulate cancer cell immunogenicity by directing the presentation of particular tumor epitopes.
    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
    目的:蛋白酶体是在真核生物中对蛋白质稳定至关重要的保守蛋白酶,并且是原生动物寄生虫的有希望的药物靶标。然而,溶组织内阿米巴的蛋白酶体仍未得到充分研究。该研究的目的是分析变形虫蛋白酶体与宿主的底物结合袋的差异,和β5催化亚基的计算模型,目的是寻找选择性抑制剂。
    结果:比较序列分析揭示了溶组织大肠杆菌蛋白酶体的底物结合位点的差异,特别是在催化β亚基的S1和S3袋中,暗示底物偏好和对宿主蛋白酶体抑制剂的敏感性存在差异。与人类蛋白酶体相比,对MG132介导的阿米巴蛋白酶体β5亚基糜蛋白酶活性抑制的敏感性明显较低,这一点得到了强有力的支持。也反映在溶组织大肠杆菌对MG132抑制增殖的敏感性较低。β4和β5亚基的计算模型,对接的β4-β5模型揭示了β4-β5之间的结合袋,与利什曼原虫相似。内脏利什曼病的选择性抑制剂,LXE408和化合物8,与这个口袋对接。这种基于功能和序列的分析预测了阿米巴和宿主蛋白酶体之间的差异,可用于开发合理设计的蛋白酶体,针对溶组织大肠杆菌的选择性抑制剂。
    OBJECTIVE: Proteasomes are conserved proteases crucial for proteostasis in eukaryotes and are promising drug targets for protozoan parasites. Yet, the proteasomes of Entamoeba histolytica remain understudied. The study\'s objective was to analyse the differences in the substrate binding pockets of amoeba proteasomes from those of host, and computational modelling of β5 catalytic subunit, with the goal of finding selective inhibitors.
    RESULTS: Comparative sequence analysis revealed differences in substrate binding sites of E. histolytica proteasomes, especially in the S1 and S3 pockets of the catalytic beta subunits, implying differences in substrate preference and susceptibility to inhibitors from host proteasomes. This was strongly supported by significantly lower sensitivity to MG132 mediated inhibition of amoebic proteasome β5 subunit\'s chymotryptic activity compared to human proteasomes, also reflected in lower sensitivity of E. histolytica to MG132 for inhibition of proliferation. Computational models of β4 and β5 subunits, and a docked β4-β5 model revealed a binding pocket between β4-β5, similar to that of Leishmania tarentolae. Selective inhibitors for visceral leishmaniasis, LXE408 and compound 8, docked well to this pocket. This functional and sequence-based analysis predicts differences between amoebic and host proteasomes that can be utilized to develop rationally designed, selective inhibitors against E. histolytica.
    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
    疟疾仍然是全球健康问题,因为耐药性威胁着治疗计划。我们通过表型筛选鉴定了具有抗疟疾活性的哌啶甲酰胺(SW042)。对SW042抗性恶性疟原虫(Pf)寄生虫的选择揭示了Pf_蛋白酶体β5活性位点(Pfβ5)的点突变。口服给药后,有效的类似物(SW584)在人疟疾小鼠模型中显示出功效。SW584具有低的产生抗性的倾向(抗性的最小接种物[MIR]>109)并且与双氢青蒿素协同。通过将His8标签引入到β7上促进了Pf_蛋白酶体的纯化。Pfβ5的抑制与寄生虫杀伤有关,不抑制人蛋白酶体同工型或显示细胞毒性。Pf_proteasome_SW584低温电子显微镜(cryo-EM)结构显示SW584与催化苏氨酸非共价结合,在β5/β6/β3亚基界面的未探索口袋中,Pf和人蛋白酶体之间存在物种差异。识别一个可逆的,物种选择性,具有低耐药倾向的口服活性系列为这一基本目标的药物治疗提供了途径。
    Malaria remains a global health concern as drug resistance threatens treatment programs. We identified a piperidine carboxamide (SW042) with anti-malarial activity by phenotypic screening. Selection of SW042-resistant Plasmodium falciparum (Pf) parasites revealed point mutations in the Pf_proteasome β5 active-site (Pfβ5). A potent analog (SW584) showed efficacy in a mouse model of human malaria after oral dosing. SW584 had a low propensity to generate resistance (minimum inoculum for resistance [MIR] >109) and was synergistic with dihydroartemisinin. Pf_proteasome purification was facilitated by His8-tag introduction onto β7. Inhibition of Pfβ5 correlated with parasite killing, without inhibiting human proteasome isoforms or showing cytotoxicity. The Pf_proteasome_SW584 cryoelectron microscopy (cryo-EM) structure showed that SW584 bound non-covalently distal from the catalytic threonine, in an unexplored pocket at the β5/β6/β3 subunit interface that has species differences between Pf and human proteasomes. Identification of a reversible, species selective, orally active series with low resistance propensity provides a path for drugging this essential target.
    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
    丙酮酸脱氢酶激酶(PDK),磷酸化丙酮酸脱氢酶复合物,调节骨骼肌中的葡萄糖代谢。PDK1,一种同工酶,其表达受缺氧诱导因子-1α(HIF-1α)控制,被认为在肌肉对缺氧的适应中起作用。虽然HIF-1α对PDK1的转录上调有很好的表征,控制骨骼肌中PDK1蛋白水解的机制尚未得到彻底研究。蛋白酶体抑制剂MG132矛盾地降低了人类癌细胞和大鼠L6肌管中PDK1的丰度,这表明MG132可能指导PDK1进行自噬降解。我们当前研究的目标是确定(1)MG132是否抑制原发性人类肌管中的PDK1水平,(2)是否氯喹,自噬的抑制剂,防止MG132诱导的L6肌管中PDK1的抑制,和(3)PYR-41,泛素化的抑制剂,抑制L6肌管中的PDK1。使用qPCR和/或免疫印迹,我们发现,尽管HIF-1α蛋白显著上调,MG132没有改变PDK1在培养的原代人肌管中的表达,同时抑制L6肌管中的PDK1mRNA和蛋白。在与氯喹和MG132共同处理期间,L6肌管中的PDK1水平也被抑制。PYR-41显著增加原发性人类肌管和L6肌管中HIF-1α的丰度,同时降低了PDK1的丰度。在用PYR-41处理的L6肌管中,氯喹增加了表皮生长因子受体的丰度,但不能阻止PDK1的抑制。总的来说,我们的结果表明,培养的肌管通过MG132,PYR-41和/或氯喹无法抑制的途径降解PDK1.
    Pyruvate dehydrogenase kinase (PDK), which phosphorylates the pyruvate dehydrogenase complex, regulates glucose metabolism in skeletal muscle. PDK1, an isozyme whose expression is controlled by hypoxia-inducible factor-1α (HIF-1α), is thought to play a role in muscle adaptation to hypoxia. While transcriptional upregulation of PDK1 by HIF-1α is well characterised, mechanisms controlling proteolysis of PDK1 in skeletal muscle have not been thoroughly investigated. Proteasome inhibitor MG132 paradoxically reduced the abundance of PDK1 in human cancer cells and rat L6 myotubes, suggesting that MG132 might direct PDK1 towards autophagic degradation. The objectives of our current study were to determine (1) whether MG132 suppresses PDK1 levels in primary human myotubes, (2) whether chloroquine, an inhibitor of autophagy, prevents MG132-induced suppression of PDK1 in L6 myotubes, and (3) whether PYR-41, an inhibitor of ubiquitination, suppresses PDK1 in L6 myotubes. Using qPCR and/or immunoblotting, we found that despite markedly upregulating HIF-1α protein, MG132 did not alter the PDK1 expression in cultured primary human myotubes, while it suppressed both PDK1 mRNA and protein in L6 myotubes. The PDK1 levels in L6 myotubes were suppressed also during co-treatment with chloroquine and MG132. PYR-41 markedly increased the abundance of HIF-1α in primary human and L6 myotubes, while reducing the abundance of PDK1. In L6 myotubes treated with PYR-41, chloroquine increased the abundance of the epidermal growth factor receptor, but did not prevent the suppression of PDK1. Collectively, our results suggest that cultured myotubes degrade PDK1 via a pathway that cannot be inhibited by MG132, PYR-41, and/or chloroquine.
    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
    雷公藤甲素(TP)是中药雷公藤的主要活性和毒性成份。F.(TWHF),表现出各种治疗生物活性。在毒性作用中,TP的肝毒性值得重视。以前,我们的研究小组提出了TP相关肝毒性的新观点:脂多糖(LPS)刺激下的肝超敏反应.然而,TP/LPS诱导肝超敏反应的机制尚不清楚.在这项研究中,我们从抑制蛋白酶体活性的角度研究了TP/LPS诱导的超敏反应的机制,激活的内质网应激(ERS)相关的凋亡,和活性氧(ROS)的积累。我们的结果表明,N-乙酰半胱氨酸(NAC),一种常见的ROS抑制剂,降低裂解的caspase-3和裂解的PARP的表达,与FLIP增强相关联。此外,4-苯基丁酸(4-PBA),一种ERS抑制剂,能够通过减少ERS相关凋亡蛋白的表达来减轻TP/LPS诱导的肝毒性(GRP78,p-eIF2α/eIF2α,ATF4,CHOP,裂解的caspase-3和裂解的PARP)和ROS水平,ATF4是不可或缺的调解人。此外,蛋白酶体活性抑制剂MG-132进一步加重了ERS相关的细胞凋亡,这表明蛋白酶体活性的抑制在TP/LPS相关的肝损伤中也起着重要作用。总之,我们认为TP/LPS可能通过抑制蛋白酶体活性和通过ATF4增强ROS产生来上调ERS相关凋亡的激活。
    Triptolide (TP) is a major active and toxic composition of the Chinese medicine Tripterygium wilfordii Hook. F. (TWHF), exhibiting various therapeutic bioactivities. Among the toxic effects, the hepatotoxicity of TP deserves serious attention. Previously, our research group proposed a new view of TP-related hepatotoxicity: hepatic hypersensitivity under lipopolysaccharide (LPS) stimulation. However, the mechanism of TP/LPS-induced hepatic hypersensitivity remains unclear. In this study, we investigated the mechanism underlying TP/LPS-induced hypersensitivity from the perspective of the inhibition of proteasome activity, activated endoplasmic reticulum stress (ERS)-related apoptosis, and the accumulation of reactive oxygen species (ROS). Our results showed that N-acetylcysteine (NAC), a common ROS inhibitor, decreased the expression of cleaved caspase-3 and cleaved PARP, which are associated with FLIP enhancement. Moreover, 4-phenylbutyric acid (4-PBA), an ERS inhibitor, was able to alleviate TP/LPS-induced hepatotoxicity by reducing ERS-related apoptosis protein expression (GRP78, p-eIF2α/eIF2α, ATF4, CHOP, cleaved caspase-3 and cleaved PARP) and ROS levels, with ATF4 being an indispensable mediator. In addition, the proteasome activity inhibitor MG-132 further aggravated ERS-related apoptosis, which indicated that the inhibition of proteasome activity also plays an important role in TP/LPS-related liver injuries. In summary, we propose that TP/LPS may upregulate the activation of ERS-associated apoptosis by inhibiting proteasome activity and enhancing ROS production through ATF4.
    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
    蛋白质稳态是一个紧密保守的过程,通过泛素蛋白酶体系统(UPS)以不依赖泛素或依赖泛素的方式进行调节。在过去的二十年里,蛋白酶体已经成为一个极好的治疗靶点通过抑制催化核心颗粒,抑制负责识别和结合泛素化蛋白的亚基,最近,通过使用蛋白水解靶向嵌合体(PROTACs)的靶向蛋白质降解。大多数已开发的蛋白酶体核心颗粒抑制剂依赖于通过未引发底物通道内的结合相互作用获得选择性。尽管这允许针对不同的蛋白酶体同工型产生选择性抑制剂和化学探针,关于可以在引发的底物通道内利用以增加效力或选择性的相互作用,仍然未知。在这里,我们讨论了与引发的底物袋相互作用的小分子,以及它们的差异如何引起活性的改变。利用与蛋白酶体的引发的底物袋的额外相互作用可以允许产生用于扰乱或监测蛋白酶体活性的改进的化学工具。
    Protein homeostasis is a tightly conserved process that is regulated through the ubiquitin proteasome system (UPS) in a ubiquitin-independent or ubiquitin-dependent manner. Over the past two decades, the proteasome has become an excellent therapeutic target through inhibition of the catalytic core particle, inhibition of subunits responsible for recognizing and binding ubiquitinated proteins, and more recently, through targeted protein degradation using proteolysis targeting chimeras (PROTACs). The majority of the developed inhibitors of the proteasome\'s core particle rely on gaining selectivity through binding interactions within the unprimed substrate channel. Although this has allowed for selective inhibitors and chemical probes to be generated for the different proteasome isoforms, much remains unknown about the interactions that could be harnessed within the primed substrate channel to increase potency or selectivity. Herein, we discuss small molecules that interact with the primed substrate pocket and how their differences may give rise to altered activity. Taking advantage of additional interactions with the primed substrate pocket of the proteasome could allow for the generation of improved chemical tools for perturbing or monitoring proteasome 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: Journal Article
    癌细胞破坏了正常细胞的多种特性,包括逃避严格的细胞周期调控,获得对细胞死亡的抵抗力,重塑肿瘤微环境。最近对癌症的标志进行了更新和总结。核因子红系2相关因子1(NFE2L1,也称为NRF1)属于帽领(CNC)碱性区域亮氨酸拉链(bZIP)家族。它作为转录因子,在发育和生长过程中对于维持细胞的同质性和器官完整性是必不可少的。以及对病理生理应激源的适应性反应。此外,NFE2L1在神经母细胞瘤的临床蛋白酶体抑制剂治疗中介导蛋白酶体反弹效应,多发性骨髓瘤,和三阴性乳腺癌,快速诱导蛋白酶体抑制剂抗性。最近的研究表明,NFE2L1在各种癌细胞系中介导细胞增殖和代谢重编程。我们将“癌症标志”提供的框架与最近对NFE2L1的研究相结合,总结了NFE2L1在癌症中的作用和机制。这些正在进行的努力旨在促进开发靶向NFE2L1途径及其活性的潜在新型癌症疗法。
    Cancer cells subvert multiple properties of normal cells, including escaping strict cell cycle regulation, gaining resistance to cell death, and remodeling the tumor microenvironment. The hallmarks of cancer have recently been updated and summarized. Nuclear factor erythroid 2-related factor 1 (NFE2L1, also named NRF1) belongs to the cap\'n\'collar (CNC) basic-region leucine zipper (bZIP) family. It acts as a transcription factor and is indispensable for maintaining both cellular homoeostasis and organ integrity during development and growth, as well as adaptive responses to pathophysiological stressors. In addition, NFE2L1 mediates the proteasome bounce-back effect in the clinical proteasome inhibitor therapy of neuroblastoma, multiple myeloma, and triple-negative breast cancer, which quickly induces proteasome inhibitor resistance. Recent studies have shown that NFE2L1 mediates cell proliferation and metabolic reprogramming in various cancer cell lines. We combined the framework provided by \"hallmarks of cancer\" with recent research on NFE2L1 to summarize the role and mechanism of NFE2L1 in cancer. These ongoing efforts aim to contribute to the development of potential novel cancer therapies that target the NFE2L1 pathway and its activity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号