NEK7

NEK7
  • 文章类型: Journal Article
    镰刀病毒是一种嗜神经病原体,需要逃避外周宿主免疫进入中枢神经系统来完成感染。NLRP3炎性体激活对于宿主防御病原体入侵至关重要。这项研究表明,裂解病毒的基质蛋白(M)可以抑制NLRP3炎性体激活的引发步骤和激活步骤。具体来说,M的裂解病毒可以与NEK7竞争结合NLRP3,这限制了下游凋亡相关的斑点样蛋白,其中含有CARD(ASC)寡聚化。Lyssavirus中M的第158位的丝氨酸氨基酸对于限制ASC寡聚化至关重要。此外,在M处具有G158S突变的重组实验室减毒狂犬病病毒(狂犬病狂犬病病毒[RABV])可降低骨髓来源的树突状细胞(BMDCs)中白介素1β(IL-1β)的产生,以促进狂犬病病毒侵入大脑,从而提高小鼠的致病性。一起来看,这项研究揭示了一种常见的机制,通过该机制,lyssavirus抑制NLRP3炎性体激活,以逃避宿主防御。
    Lyssavirus is a kind of neurotropic pathogen that needs to evade peripheral host immunity to enter the central nervous system to accomplish infection. NLRP3 inflammasome activation is essential for the host to defend against pathogen invasion. This study demonstrates that the matrix protein (M) of lyssavirus can inhibit both the priming step and the activation step of NLRP3 inflammasome activation. Specifically, M of lyssavirus can compete with NEK7 for binding to NLRP3, which restricts downstream apoptosis-associated speck-like protein containing a CARD (ASC) oligomerization. The serine amino acid at the 158th site of M among lyssavirus is critical for restricting ASC oligomerization. Moreover, recombinant lab-attenuated lyssavirus rabies (rabies lyssavirus [RABV]) with G158S mutation at M decreases interleukin-1β (IL-1β) production in bone-marrow-derived dendritic cells (BMDCs) to facilitate lyssavirus invasion into the brain thereby elevating pathogenicity in mice. Taken together, this study reveals a common mechanism by which lyssavirus inhibits NLRP3 inflammasome activation to evade host defenses.
    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
    NLRP3炎性体,先天免疫的关键组成部分,与各种炎症性疾病有关。众所周知,泛素编辑酶A20可以调节炎症并维持稳态。然而,A20调节NLRP3炎性体的确切分子机制仍然知之甚少。这里,我们的研究显示,缺乏A20的巨噬细胞表现出蛋白丰度增加和NIMA相关激酶7(NEK7)mRNA水平升高.重要的是,A20直接与NEK7结合,介导其K48连接的泛素化,从而靶向NEK7用于蛋白酶体降解。我们的结果表明,A20增强了NEK7在K189和K293泛素化位点的泛素化,K189在NEK7与A20的结合中起关键作用,尽管没有显着影响NEK7与NLRP3之间的相互作用。此外,A20破坏了NEK7与NLRP3复合体的联系,可能通过OTU结构域和/或ZnF4和ZnF7基序的协同作用。重要的是,在A20缺陷条件下,NEK7缺失显著减弱NLRP3炎性体的激活,在体外和体内。本研究揭示了A20抑制NLRP3炎性体的机制。
    The NLRP3 inflammasome, a pivotal component of innate immunity, has been implicated in various inflammatory disorders. The ubiquitin-editing enzyme A20 is well known to regulate inflammation and maintain homeostasis. However, the precise molecular mechanisms by which A20 modulates the NLRP3 inflammasome remain poorly understood. Here, our study revealed that macrophages deficient in A20 exhibit increased protein abundance and elevated mRNA level of NIMA-related kinase 7 (NEK7). Importantly, A20 directly binds with NEK7, mediating its K48-linked ubiquitination, thereby targeting NEK7 for proteasomal degradation. Our results demonstrate that A20 enhances the ubiquitination of NEK7 at K189 and K293 ubiquitinated sites, with K189 playing a crucial role in the binding of NEK7 to A20, albeit not significantly influencing the interaction between NEK7 and NLRP3. Furthermore, A20 disrupts the association of NEK7 with the NLRP3 complex, potentially through the OTU domain and/or synergistic effect of ZnF4 and ZnF7 motifs. Significantly, NEK7 deletion markedly attenuates the activation of the NLRP3 inflammasome in A20-deficient conditions, both in vitro and in vivo. This study uncovers a mechanism by which A20 inhibits the NLRP3 inflammasome.
    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
    癌症构成了重大的全球健康挑战,并显著增加了死亡率。NEK7,与NIMA蛋白激酶家族相关,在纺锤体组装和细胞分裂中起着至关重要的作用。NEK7的失调与各种癌症的发生和进展密切相关,尤其是结肠癌和乳腺癌,使其成为癌症治疗的有希望的目标。然而,高质量NEK7抑制剂的短缺凸显了对新治疗策略的需求.在这项研究中,我们采用了多学科的方法,包括虚拟筛查,分子对接,药代动力学,分子动力学模拟(MD),和MM/PBSA计算,综合评价天然化合物作为NEK7抑制剂。通过各种对接策略,我们确定了三种天然化合物:(-)-巴拉诺,双金属酸,还有Scutellarin.分子对接揭示了在诸如GLU112和ALA114的残基上的显着相互作用,对接评分为-15.054,-13.059和-11.547kcal/mol,分别,突出了它们作为NEK7抑制剂的潜力。MD证实了这些化合物在NEK7结合位点的稳定性。模拟过程中的氢键分析揭示了一致的相互作用,支持他们强大的约束力。MM/PBSA分析确定了其他有助于结合亲和力的关键氨基酸,包括ILE20,VAL28,ILE75,LEU93,ALA94,LYS143,PHE148,LEU160和THR161,对稳定复合物至关重要。这项研究表明,这些化合物的结合能超过了dabrafenib,根据MM/PBSA计算,强调它们作为NEK7抑制剂的有效性。ADME/T预测显示这些化合物的口服毒性较低,表明了他们进一步发展的潜力。这项研究强调了这些天然化合物作为创造具有显著生物活性的更有效衍生物的基础的前景。为未来的实验验证铺平了道路。
    Cancer poses a significant global health challenge and significantly contributes to mortality. NEK7, related to the NIMA protein kinase family, plays a crucial role in spindle assembly and cell division. The dysregulation of NEK7 is closely linked to the onset and progression of various cancers, especially colon and breast cancer, making it a promising target for cancer therapy. Nevertheless, the shortage of high-quality NEK7 inhibitors highlights the need for new therapeutic strategies. In this study, we utilized a multidisciplinary approach, including virtual screening, molecular docking, pharmacokinetics, molecular dynamics simulations (MDs), and MM/PBSA calculations, to evaluate natural compounds as NEK7 inhibitors comprehensively. Through various docking strategies, we identified three natural compounds: (-)-balanol, digallic acid, and scutellarin. Molecular docking revealed significant interactions at residues such as GLU112 and ALA114, with docking scores of -15.054, -13.059, and -11.547 kcal/mol, respectively, highlighting their potential as NEK7 inhibitors. MDs confirmed the stability of these compounds at the NEK7-binding site. Hydrogen bond analysis during simulations revealed consistent interactions, supporting their strong binding capacity. MM/PBSA analysis identified other crucial amino acids contributing to binding affinity, including ILE20, VAL28, ILE75, LEU93, ALA94, LYS143, PHE148, LEU160, and THR161, crucial for stabilizing the complex. This research demonstrated that these compounds exceeded dabrafenib in binding energy, according to MM/PBSA calculations, underscoring their effectiveness as NEK7 inhibitors. ADME/T predictions showed lower oral toxicity for these compounds, suggesting their potential for further development. This study highlights the promise of these natural compounds as bases for creating more potent derivatives with significant biological activities, paving the way for future experimental validation.
    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
    焦亡,gasdermin介导的裂解细胞死亡,是癌症研究的新热点,诱导肿瘤焦亡已成为癌症治疗的新目标。槲皮素(Que),一种天然物质,展示了有希望的抗癌作用。然而,需要更多的信息来充分了解Que在结肠癌中的功能和机制。这项研究揭示了Que诱导的结肠癌体内外焦凋亡的潜在机制。Que通过GasderminD(GSDMD)介导的焦亡抑制结肠癌细胞的生长。耗尽GSDMD,而不是gasderminE(GSDME),逆转Que对结肠癌细胞的细胞毒性作用。Que治疗上调NIMA相关激酶7(NEK7)蛋白表达,从而促进NLRP3炎性体的组装和GSDMD的裂解。NEK7沉默导致结肠癌细胞在体外和体内生长。机械上,NEK7抑郁症克制NLRP3炎性小体-GSDMD通路的激活,从而减弱由Que在结肠癌细胞中引发的焦亡。此外,较低的NEK7和NLRP3表达水平表明结肠癌进展.我们的研究结果揭示了Que抗结肠癌活性的新模式,激活NEK7介导的焦亡可能是结肠癌的一个有希望的治疗靶点,这为Que的临床应用提供了新的实验依据。
    Pyroptosis, a gasdermin-mediated lytic cell death, is a new hotspot topic in cancer research, and induction of tumor pyroptosis has emerged as a new target in cancer management. Quercetin (Que), a natural substance, demonstrates promising anticancer action. However, further information is required to fully comprehend the function and mechanism of Que in pyroptosis in colon cancer. This study revealed the underlying mechanism of Que-induced pyroptosis in colon cancer in vitro and in vivo. Que inhibited colon cancer cell growth through gasdermin D (GSDMD)-mediated pyroptosis. Depletion of GSDMD, rather than gasdermin E (GSDME), reversed the cytotoxic effects of Que on colon cancer cells. Que treatment upregulated NIMA-related kinase 7 (NEK7) protein expression, thus facilitating the assembly of the NLRP3 inflammasome and cleavage of GSDMD. NEK7 silencing resulted in colon cancer cell growth in vitro and in vivo. Mechanistically, NEK7 depression restrained the activation of the NLRP3 inflammasome-GSDMD pathway, thus attenuating pyroptosis triggered by Que in colon cancer cells. Furthermore, lower NEK7 and NLRP3 expression levels indicated colon cancer progression. Our results unveiled a novel pattern of anti-colon cancer activity of Que, and activation of NEK7-mediated pyroptosis is potentially a promising therapeutic target for colon cancer, which provides novel experimental proof for the clinical application of Que.
    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
    NIMA相关激酶7(NEK7)和磷蛋白磷酸酶-1催化亚基α(PPP1CA)是胰腺导管腺癌中最常见的过表达蛋白,这是最常见的胰腺癌。本研究的目的是确定一种可能的NEK7和PPP1CA治疗性抑制剂。为了这次调查,从IMPPAT植物化学物质库中检索到5000种化合物,与我们各自的目标蛋白对接。此外,参考化合物,吉西他滨,这是美国食品和药物管理局(FDA)批准的药物,与靶蛋白对接。两种靶向蛋白质的参考化合物的结合能为-6.5kcal/mol。对于两个靶标具有最低结合能的常见配体是boeravinoneB(PubChemID:14018348),NEK7为-9.2kcal/mol,PPP1CA为-7.6kcal/mol。通过密度泛函理论(DFT)和分子动力学模拟分析进一步研究了该化合物。均方根偏差(RMSD),均方根波动(RMSF),回转半径(Rg),和氢键分析表明boeravinoneB与靶蛋白(NEK7和PPP1CA)的稳定性。由RamaswamyH.Sarma沟通。
    NIMA-related kinase 7 (NEK7) and phosphoprotein phosphatase-1 catalytic subunit alpha (PPP1CA) are the most common proteins overexpressed in pancreatic ductal adenocarcinoma, which is the most common type of pancreatic cancer. The goal of the current study was to identify a possible NEK7 and PPP1CA therapeutic inhibitor. For this investigation, 5000 compounds were retrieved from the IMPPAT library of phytochemicals, which were docked with our respective target proteins. Also, a reference compound, gemcitabine, which is a Food and Drug Administration (FDA) approved drug, was docked with the target proteins. The binding energy of the reference compound for both the targeted proteins was -6.5 kcal/mol. The common ligand with the lowest binding energy for both targets is boeravinone B (PubChem ID: 14018348) with -9.2 kcal/mol of NEK7 and -7.6 kcal/mol for PPP1CA. The compound was further investigated through density function theory (DFT) and molecular dynamic simulation analysis. The root mean square deviation (RMSD), root mean square fluctuation (RMSF), radius of gyration (Rg), and hydrogen bonding analysis indicated the stability of the boeravinone B with the target proteins (NEK7 and PPP1CA).Communicated by Ramaswamy H. Sarma.
    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
    在大约5%的非小细胞肺癌患者中发现EML4-ALK致癌融合蛋白。存在不同的EML4-ALK融合变体,变体3(V3)与明显高于其他常见变体的风险相关。如V1。V3患者对靶向ALK抑制剂的反应较差,有加速的转移率和较差的总体生存率。已经描述了EML4-ALKV3下游的途径,其不依赖于ALK催化活性,但依赖于NEK9和NEK7激酶。已经提出EML4-ALKV3:NEK9:NEK7复合物在微管上的组装导致细胞形成间充质样形态并表现出增强的迀移。然而,该复合体的下游目标仍有待确定。这里,我们证明了基于微管的驱动蛋白运动蛋白,响应于EML4-ALKV3的表达,将Eg5募集至相间微管,而Eg5的化学抑制逆转细胞的间充质形态。此外,我们显示,NEK7的耗竭干扰Eg5募集到微管中表达EML4-ALKV3的细胞,并导致细胞长度减少,但这是通过在一个位点共表达Eg5的磷模拟突变体而逆转的,S1033,被NEK7磷酸化。有趣的是,我们还发现Eg5-S1033D的表达导致表达EML4-ALKV1的细胞采用更多的间充质样形态。一起来看,我们提出Eg5在表达EML4-ALKV3的细胞中充当NEK7的底物,并且Eg5磷酸化促进这些细胞典型的形态改变。
    Echinoderm microtubule-associated protein-like 4 (EML4)-anaplastic lymphoma kinase (ALK) oncogenic fusion proteins are found in approximately 5% of non-small cell lung cancers. Different EML4-ALK fusion variants exist with variant 3 (V3) being associated with a significantly higher risk than other common variants, such as variant 1 (V1). Patients with V3 respond less well to targeted ALK inhibitors, have accelerated rates of metastasis, and have poorer overall survival. A pathway has been described downstream of EML4-ALK V3 that is independent of ALK catalytic activity but dependent on the NEK9 and NEK7 kinases. It has been proposed that assembly of an EML4-ALK V3-NEK9-NEK7 complex on microtubules leads to cells developing a mesenchymal-like morphology and exhibiting enhanced migration. However, downstream targets of this complex remain unknown. Here, we show that the microtubule-based kinesin, Eg5, is recruited to interphase microtubules in cells expressing EML4-ALK V3, whereas chemical inhibition of Eg5 reverses the mesenchymal morphology of cells. Furthermore, we show that depletion of NEK7 interferes with Eg5 recruitment to microtubules in cells expressing EML4-ALK V3 and cell length is reduced, but this is reversed by coexpression of a phosphomimetic mutant of Eg5, in a site, S1033, phosphorylated by NEK7. Intriguingly, we also found that expression of Eg5-S1033D led to cells expressing EML4-ALK V1 adopting a more mesenchymal-like morphology. Together, we propose that Eg5 acts as a substrate of NEK7 in cells expressing EML4-ALK V3 and Eg5 phosphorylation promotes the mesenchymal morphology typical of these cells.
    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
    脑出血(ICH)导致严重的神经损伤,它的发展是由METTL3驱动的。本研究旨在通过体外实验研究METTL3在ICH中的作用。为此,用血红素处理HT-22细胞以体外模拟ICH,然后用流式细胞术评估细胞焦亡,乳酸脱氢酶释放分析,酶联免疫吸附测定,和西方印迹。此外,使用甲基化RNA免疫沉淀评估NEK7的N6-甲基腺苷(m6A)甲基化,RNA免疫沉淀,双荧光素酶报告分析,和定量实时聚合酶链反应。结果表明,METTL3的敲低抑制了血红素诱导的焦凋亡,并抑制了METTL3下调导致的NEK7的m6A甲基化,降低NEK7mRNA稳定性。通过过表达NEK7消除了对METTL3诱导的细胞焦亡的影响,而IGF2BP2增加了NEK7的表达。同样,IGF2BP2沉默下调METTL3介导的NEK7表达。总之,METTL3的沉默通过抑制被IGF2BP2识别的NEK7的m6A甲基化来抑制血红素诱导的HT-22细胞的焦亡。这些发现旨在确定ICH的新治疗策略。
    Intracerebral hemorrhage (ICH) induces severe neurological damage, and its progression is driven by METTL3. This study aimed to investigate the role of METTL3 in ICH via in vitro experiments. For this purpose, HT-22 cells were treated with hemin to mimic ICH in vitro, followed by evaluating cell pyroptosis using flow cytometry, lactic dehydrogenase release analysis, enzyme-linked immunosorbent assay, and western blotting. Moreover, N6-methyl adenosine (m6A) methylation of NEK7 was assessed using methylated RNA immunoprecipitation, RNA immunoprecipitation, dual-luciferase reporter assay, and quantitative real-time polymerase chain reaction. Results indicated that knockdown of METTL3 inhibited hemin-induced pyroptosis and suppressed m6A methylation of NEK7 due to METTL3 downregulation, reducing NEK7 mRNA stability. The effects on METTL3-induced cell pyroptosis were abrogated by overexpressing NEK7, while IGF2BP2 increased NEK7 expression. Similarly, IGF2BP2 silence downregulated NEK7 expression mediated by METTL3. In conclusion, silencing of METTL3 inhibited hemin-induced HT-22 cell pyroptosis by suppressing m6A methylation of NEK7, which was recognized by IGF2BP2. These findings are envisaged to identify a novel therapeutic strategy for ICH.
    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
    已经报道了O-GlcNAc转移酶(OGT)诱导的O-连接的N-乙酰葡糖胺化(O-GlcNAc酰化)在多种人类疾病中的作用。然而,其在骨关节炎(OA)进展中的具体功能仍未确定。
    本研究集中于OA中OGT诱导的O-GlcNAcylation的靶蛋白及其特定的功能机制。
    使用蛋白质印迹在体外和体内OA模型中测量总O-GlcNAc和OGT的水平。通过SafraninO染色检测OGT基因敲除对OA进展的影响,免疫组化染色和OARSI评分评价。OGT沉默对LPS诱导的软骨细胞损伤的影响通过进行功能丧失测定来评估。进行免疫共沉淀(co-IP)以验证OGT诱导的O-GlcNAcylation对NEK7和NLRP3之间的相互作用的影响。使用蛋白质印迹分析OGT在调节NEK7的O-GlcNAcylation和磷酸化水平中的作用。
    OGT引入的O-GlcNAcylation水平在体外和体内OA模型中均增加。OGT的基因敲除减轻了模型小鼠中的OA进展。此外,OGT的沉默抑制了LPS诱导的软骨细胞焦亡。此外,OGT的沉默抑制了O-GlcNAcylation并增强了NEK7在S260位点的磷酸化,从而阻断NEK7与NLRP3的结合。
    OGT诱导的NEK7O-GlcNAcylation通过抑制NEK7和NLRP3之间的相互作用促进软骨细胞的焦亡促进OA进展。
    UNASSIGNED: The role of O-GlcNAc transferase (OGT)-induced O-linked N-acetylglucosaminylation (O-GlcNAcylation) has been reported in multiple human diseases. However, its specific functions in osteoarthritis (OA) progression remain undetermined.
    UNASSIGNED: This study focused on the target proteins of OGT-induced O-GlcNAcylation in OA and the specific functional mechanism.
    UNASSIGNED: The levels of total O-GlcNAc and OGT were measured in both in vitro and in vivo OA models using western blot. The effects of OGT knockout on OA progression were detected through Safranin O staining, immunohistochemical staining and OARSI score evaluation. The effects of OGT silencing on LPS-induced chondrocyte injury were assessed by performing loss-of function assays. Co-immunoprecipitation (co-IP) was conducted to verify the effect of OGT-induced O-GlcNAcylation on the interaction between NEK7 and NLRP3. The role of OGT in modulating the O-GlcNAcylation and phosphorylation levels of NEK7 was analysed using western blot.
    UNASSIGNED: The OGT-indued O-GlcNAcylation level was increased in both in vitro and in vivo OA models. Knockout of OGT mitigated OA progression in model mice. Additionally, silencing of OGT suppressed LPS-induced chondrocyte pyroptosis. Moreover, silencing of OGT inhibited the O-GlcNAcylation and enhanced the phosphorylation of NEK7 at S260 site, thereby blocking the binding of NEK7 with NLRP3.
    UNASSIGNED: OGT-induced NEK7 O-GlcNAcylation promotes OA progression by promoting chondrocyte pyroptosis via the suppressing interaction between NEK7 and NLRP3.
    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NEK7对NLRP3炎性体激活至关重要,NEK7抑制剂可用作痛风的治疗剂,由NLRP3炎性体引起的代表性疾病。
    我们基于2,7-取代的噻吩并[3,2-d]嘧啶衍生物(SLC3031〜3035和SLC3037)的生化动力学谱分析设计了NEK7抑制剂。在用LPS+尿酸单钠(MSU)处理骨髓来源的巨噬细胞后,通过IL-1b的ELISA和IL-1b成熟的免疫印迹评估炎性体活化。使用免疫沉淀和蓝色天然凝胶电泳检查NLPR3与NEK7的结合和寡聚化,分别。通过研究MSU注射小鼠的食物垫组织的总体和组织病理学变化来研究体内作用,连同组织中IL-1b和ASC斑点的成熟测定。
    SLC3037通过阻断NLRP3与NEK7的结合或寡聚化而抑制MSU和其他炎症小体激活剂的炎症小体,和随后的ASC寡聚化/磷酸化。SLC3037显著降低了MSU的脚垫厚度和炎症,其效果优于秋水仙碱。SLC3037显着降低了食物垫中IL-1b和ASC斑点的含量或成熟。秋水仙素降低了肠绒毛的数量和高度,但SLC3037没有降低。
    SLC3037是一种阻断NEK7与NLRP3结合的NLRP3抑制剂,可能是一种新的药物,可以对抗与NLRP3炎性体激活相关的疾病,如痛风,心血管疾病,代谢综合征或神经退行性疾病。
    Since NEK7 is critical for NLRP3 inflammasome activation, NEK7 inhibitors could be employed as therapeutic agents against gout, a representative disease caused by NLRP3 inflammasome.
    We designed NEK7 inhibitors based on biochemical kinome profiling of 2,7-substituted thieno[3,2-d]pyrimidine derivatives (SLC3031~3035 and SLC3037). Inflammasome activation was assessed by ELISA of IL-1b and immunoblotting of IL-1b maturation after treatment of bone marrow-derived macrophages with LPS+monosodium urate (MSU). NLPR3 binding to NEK7 and oligomerization were examined using immunoprecipitation and Blue Native gel electrophoresis, respectively. In vivo effect was investigated by studying gross and histopathological changes of food pad tissue of MSU-injected mice, together with assays of maturation of IL-1b and ASC speck in the tissue.
    SLC3037 inhibited inflammasome by MSU and other inflammasome activators through blockade of NLRP3 binding to NEK7 or oligomerization, and subsequent ASC oligomerization/phosphorylation. SLC3037 significantly reduced foot pad thickness and inflammation by MSU, which was superior to the effects of colchicine. SLC3037 significantly reduced content or maturation of IL-1b and ASC speck in the food pad. The number and height of intestinal villi were decreased by colchicine but not by SLC3037.
    SLC3037, a NLRP3 inhibitor blocking NEK7 binding to NLRP3, could be a novel agent against diseases associated with NLRP3 inflammasome activation such as gout, cardiovascular diseases, metabolic syndrome or neurodegenerative diseases.
    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
    由NLRP3炎性体异常激活引起的过度炎症有助于多种人类疾病的发病机理,但针对NLRP3炎性体的临床药物仍不可用。在这项研究中,我们确定了恩替尼(ENB),美国食品和药物管理局(FDA)批准的抗癌剂,作为NLRP3炎性体的靶向抑制剂来治疗相关疾病。ENB特异性阻断NLRP3而不影响其他炎性体的激活。此外,我们证明ENB直接与NEK7的精氨酸121(R121)结合,并阻断NEK7和NLRP3之间的相互作用,从而抑制炎症小体的组装和激活。体内研究表明,ENB对NLRP3炎症相关疾病的小鼠模型有显著的改善作用,包括脂多糖(LPS)诱导的全身性炎症,尿酸单钠(MSU)诱导的腹膜炎,高脂饮食(HFD)诱导的2型糖尿病(T2D)。这些数据表明,ENB是NEK7的靶向抑制剂,具有强抗NLRP3炎性体活性,使其成为治疗炎症相关疾病的潜在候选药物。
    Excessive inflammation caused by abnormal activation of the NLRP3 inflammasome contributes to the pathogenesis of multiple human diseases, but clinical drugs targeting the NLRP3 inflammasome are still not available. In this study, we identify entrectinib (ENB), a US Food and Drug Administration (FDA)-approved anti-cancer agent, as a target inhibitor of the NLRP3 inflammasome to treat related diseases. ENB specifically blocks NLRP3 without affecting activation of other inflammasomes. Furthermore, we demonstrate that ENB directly binds to arginine 121 (R121) of NEK7 and blocks the interaction between NEK7 and NLRP3, thereby inhibiting inflammasome assembly and activation. In vivo studies show that ENB has a significant ameliorative effect on mouse models of NLRP3 inflammasome-related diseases, including lipopolysaccharide (LPS)-induced systemic inflammation, monosodium urate (MSU)-induced peritonitis, and high-fat diet (HFD)-induced type 2 diabetes (T2D). These data show that ENB is a targeted inhibitor of NEK7 with strong anti-NLRP3 inflammasome activity, making it a potential candidate drug for the treatment of inflammasome-related diseases.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号