TNF

TNF
  • 文章类型: Journal Article
    肿瘤坏死因子(TNF)是一种促炎细胞因子,其功能性同三聚体形式与TNF受体(TNFR)相互作用以激活下游凋亡,坏死,和炎症信号通路。这些途径的过度激活导致各种炎性疾病,这使得TNF成为一个有希望的治疗靶点。这里,基于它们的相对结合能从TNF-TNFR的界面中选择12聚体肽,并将其命名为“TNF抑制诱饵”(TID)。这些诱饵肽抑制TNF介导的细胞因子分泌和细胞死亡,以及下游信号效应的激活。有效的TIDs通过破坏TNF的功能性同三聚体形式的形成来抑制TNF信号传导。在TID的衍生物中,通过破坏TNF三聚体形成,TID3c在基于细胞的测定中显示稍微更好的功效。此外,TID3c将TNF寡聚化为高分子量构型。计算机建模和模拟表明,TID3c及其亲本肽,TID3,通过氢键和静电相互作用与TNF形成稳定的复合物,这使他们成为开发基于肽的抗TNF治疗剂的有希望的领导者。
    Tumor necrosis factor (TNF) is a pro-inflammatory cytokine and its functional homotrimeric form interacts with the TNF receptor (TNFR) to activate downstream apoptotic, necroptotic, and inflammatory signaling pathways. Excessive activation of these pathways leads to various inflammatory diseases, which makes TNF a promising therapeutic target. Here, 12-mer peptides were selected from the interface of TNF-TNFR based upon their relative binding energies and were named \'TNF-inhibiting decoys\' (TIDs). These decoy peptides inhibited TNF-mediated secretion of cytokines and cell death, as well as activation of downstream signaling effectors. Effective TIDs inhibited TNF signaling by disrupting the formation of TNF\'s functional homotrimeric form. Among derivatives of TIDs, TID3c showed slightly better efficacy in cell-based assays by disrupting TNF trimer formation. Moreover, TID3c oligomerized TNF to a high molecular weight configuration. In silico modeling and simulations revealed that TID3c and its parent peptide, TID3, form a stable complex with TNF through hydrogen bonds and electrostatic interactions, which makes them the promising lead to develop peptide-based anti-TNF therapeutics.
    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
    抗逆转录病毒治疗(ART)已将HIV从致命疾病转变为慢性病,然而,合并症仍然存在。免疫恢复不完全和慢性免疫激活,尤其是在肠粘膜中,有助于这些并发症。炎性体,由先天免疫受体激活的多蛋白复合物,似乎在这些炎症反应中发挥作用。特别是,初步数据表明IFI16和NLRP3炎性体参与慢性HIV感染.这项研究探讨了HIV(PWH)患者单核细胞中的炎性体功能;将22例接受抑制病毒血症的ART治疗和17例未经治疗的PWH与33例HIV阴性供体进行了比较。在体外用LPS和ATP活化的炎性小体引发单核细胞。在供体的子集中检查IFI16和NLRP3mRNA表达。未刺激的单核细胞中的IFI16和NLRP3表达与未处理的PWH中的CD4T细胞计数呈负相关。对于IFI16,与病毒载量也呈正相关。来自未经处理的PWH的单核细胞显示IL-1α的释放增加,IL-1β,和TNF与治疗的PWH和HIV阴性供体相比。然而,PWH中的循环单核细胞未预先引发体内炎症小体激活。研究结果表明IFI16,NLRP3和HIV进展之间存在联系,强调它们在心血管疾病等合并症中的潜在作用。该研究提供了对HIV发病机制中炎症小体调节及其对治疗干预的影响的见解。
    Antiretroviral treatment (ART) has converted HIV from a lethal disease to a chronic condition, yet co-morbidities persist. Incomplete immune recovery and chronic immune activation, especially in the gut mucosa, contribute to these complications. Inflammasomes, multi-protein complexes activated by innate immune receptors, appear to play a role in these inflammatory responses. In particular, preliminary data indicate the involvement of IFI16 and NLRP3 inflammasomes in chronic HIV infection. This study explores inflammasome function in monocytes from people with HIV (PWH); 22 ART-treated with suppressed viremia and 17 untreated PWH were compared to 33 HIV-negative donors. Monocytes were primed with LPS and inflammasomes activated with ATP in vitro. IFI16 and NLRP3 mRNA expression were examined in a subset of donors. IFI16 and NLRP3 expression in unstimulated monocytes correlated negatively with CD4 T cell counts in untreated PWH. For IFI16, there was also a positive correlation with viral load. Monocytes from untreated PWH exhibit increased release of IL-1α, IL-1β, and TNF compared to treated PWH and HIV-negative donors. However, circulating monocytes in PWH are not pre-primed for inflammasome activation in vivo. The findings suggest a link between IFI16, NLRP3, and HIV progression, emphasizing their potential role in comorbidities such as cardiovascular disease. The study provides insights into inflammasome regulation in HIV pathogenesis and its implications for therapeutic interventions.
    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
    细胞间相互作用涉及许多因素,这些因素导致通过细胞竞争消除细胞。基于细胞适应性比较的体细胞组织中细胞选择的上下文相关过程。在这里,我们使用果蝇的一系列遗传测试来探索多效细胞因子肿瘤坏死因子(TNF)在Myc介导的细胞竞争(也称为Myc超竞争或Myc细胞竞争)中的相对贡献。我们发现唯一的果蝇TNF,Eiger(Egr),其受体Grindelwald(Grnd/TNFR),在Myc细胞竞争过程中,需要衔接蛋白Traf4和Traf6来消除野生型“失败者”细胞。尽管Egr和Grnd之间的相互作用通常通过激活细胞内JunN末端激酶(JNK)应激信号通路导致细胞死亡,我们的实验表明,在Myc细胞竞争中,规范JNK信号的许多组成部分对于细胞死亡是可有可无的,包括JNKKKTak1,JNKK半翅目(Hep)和JNK篮子(Bsk)。我们的结果表明,Egr/Grnd信号参与Myc细胞竞争,但其作用在很大程度上独立于JNK信号通路。
    Numerous factors have been implicated in the cell-cell interactions that lead to elimination of cells via cell competition, a context-dependent process of cell selection in somatic tissues that is based on comparisons of cellular fitness. Here we use a series of genetic tests in Drosophila to explore the relative contribution of the pleiotropic cytokine Tumor Necrosis Factor ⍺ (TNF⍺) in Myc-mediated cell competition (also known as Myc super-competition or Myc cell competition). We find that the sole Drosophila TNF, Eiger (Egr), its receptor Grindelwald (Grnd/TNFR), and the adaptor proteins Traf4 and Traf6 are required to eliminate wild-type \"loser\" cells during Myc cell competition. Although typically the interaction between Egr and Grnd leads to cell death by activating the intracellular Jun N-terminal Kinase (JNK) stress signaling pathway, our experiments reveal that many components of canonical JNK signaling are dispensable for cell death in Myc cell competition, including the JNKKK Tak1, the JNKK Hemipterous (Hep) and the JNK Basket (Bsk). Our results suggest that Egr/Grnd signaling participates in Myc cell competition, but functions in a role that is largely independent of the JNK signaling pathway.
    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
    背景:银屑病的全球患病率为1-3%,在不同的种族和地理区域观察到的差异。疾病易感性和对抗肿瘤坏死因子-α(TNFα)药物的反应提示了不同的遗传调节机制,其中可能包括NLR家族pyrin结构域包含3(NLRP3)多态性。NLRP3基因多态性的评价,银屑病患者血清CRP和TNFα水平及NLRP3(rs10754558)基因多态性,CRP和TNFα与疾病严重程度的关系及其作为银屑病对甲氨蝶呤和阿达木单抗反应的生物标志物的作用。该研究共有75名被诊断为寻常型银屑病的患者,将他们与75名健康个体的对照组进行比较。
    结果:NLRP3基因型和等位基因分布在银屑病患者和对照组之间有非常显著的差异(P=0.002,0.004)。杂合子基因型GC(OR=3.67,95CI:1.75-7.68,P=0.0006),与牛皮癣的风险增加有关。此外,GC基因型与银屑病治疗无反应显著相关(OR=11.7,95CI:3.24-42.28,P=0.0002)。关于血清CRP和TNFα水平,银屑病患者和对照组之间存在高度统计学差异(P<0.0001),银屑病患者PASI50的应答者和非应答者之间也存在高度统计学差异(P<0.0001)。
    结论:NLRP3(rs10754558)基因型GC与银屑病的严重形式和对银屑病药物的无应答有关。因此,NLRP3(rs10754558)基因多态性是银屑病患者预后的重要生物标志物。血清TNFα可用作银屑病患者对治疗反应的预测因子。仍然需要更多的研究来评估NLRP3基因多态性在与银屑病相关的遗传风险和治疗结果中的作用。
    BACKGROUND: Psoriasis has a global prevalence of 1-3%, with variations observed across different ethnic groups and geographical areas. Disease susceptibility and response to anti-tumor necrosis factor-α (TNFα) drugs suggest different genetic regulatory mechanisms which may include NLR family pyrin domain containing 3 (NLRP3) polymorphism. Evaluation of the NLRP3 gene polymorphism, the serum level of CRP and TNFα in psoriasis patients and assessment of the NLRP3 (rs10754558) gene polymorphism, CRP and TNFα with disease severity and their role as biomarkers for response to Methotrexate and Adalimumab in psoriasis. The study had a total of 75 patients diagnosed with psoriasis vulgaris, who were compared to a control group of 75 healthy individuals.
    RESULTS: There was a highly significant difference in NLRP3 genotypes and alleles distribution between psoriasis patients and controls (P = 0.002,0.004). The heterozygote genotype GC (OR = 3.67,95%CI:1.75-7.68, P = 0.0006), was linked with increased risk of psoriasis. Additionally, The GC genotype was significantly associated with nonresponse to psoriasis therapy (OR = 11.7,95%CI:3.24-42.28, P = 0.0002). Regarding serum CRP and TNFα levels, there was a highly statistically significant difference between psoriasis patients and controls (P < 0.0001), and there was also a highly statistically significant difference between responders and non-responders in psoriasis patients regarding PASI 50 (P < 0.0001).
    CONCLUSIONS: The NLRP3 (rs10754558) genotypes GC was associated with the severe form of psoriasis and with nonresponse to psoriasis medication. Therefore, NLRP3 (rs10754558) gene polymorphism is an important prognostic biomarker in psoriasis patients. The serum TNFα can be used as a predictor for response to therapy in psoriasis patients. More research for evaluation of role of the NLRP3 gene polymorphism in the genetic risks and treatment outcomes associated with psoriasis is still required.
    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:LLT-1是众所周知的天然杀伤(NK)细胞抑制性受体NKRP1A的配体。这里,我们检测了胶质母细胞瘤(GBM)组织中NLRC4炎性体成分和LLT-1的表达,以阐明这些因子之间的潜在关联和相互作用.
    方法:收集GBM组织用于RNA测序(RNA-seq)和免疫荧光实验。通过免疫荧光评估LLT-1和其他蛋白质的共定位。计算分析利用来自中国胶质瘤基因组图谱和CHA医疗记录的296至52名患者的RNA-seq数据,分别。这些数据进行了生存,非负矩阵分解聚类,基因本体富集,和蛋白质-蛋白质相互作用分析。通过单细胞RNA-seq分析证实肿瘤和免疫细胞之间的受体-配体相互作用。
    结果:在GBM组织中,LLT-1主要与表达胶质纤维酸性蛋白(GFAP)的星形胶质细胞共定位,但没有像Iba-1这样的小胶质细胞标记。此外,LLT-1和活化的NLRC4炎性体主要在肿瘤内星形胶质细胞中共表达,提示LLT-1,NLRC4和神经胶质瘤恶性肿瘤之间的关联。LLT-1高表达与不良预后相关,特别是在间充质亚型中,并与TNF和NOD样受体信号通路富集有关,表明在肿瘤炎症和进展中的潜在作用。在单细胞层面,间充质样恶性细胞显示高NF,NLR,与其他恶性细胞类型相比,IL-1信号通路富集。
    结论:我们揭示了NLRC4炎性体活性与LLT-1表达之间的关联,提示涉及TNF的新调节途径,炎性体,和IL-1,可能提供新的NK细胞介导的抗神经胶质瘤方法。
    OBJECTIVE: LLT-1 is a well-known ligand for the natural killer (NK) cell inhibitory receptor NKRP1A. Here, we examined NLRC4 inflammasome components and LLT-1 expression in glioblastoma (GBM) tissues to elucidate potential associations and interactions between these factors.
    METHODS: GBM tissues were collected for RNA sequencing (RNA-seq) and Immunofluorescent experiments. Colocalization of LLT-1 and other proteins was assessed by immunofluorescence. Computational analyses utilized RNA-seq data from 296 to 52 patients from the Chinese Glioma Genome Atlas and CHA medical records, respectively. These data were subjected to survival, non-negative matrix factorization clustering, Gene Ontology enrichment, and protein-protein interaction analyses. Receptor-ligand interactions between tumor and immune cells were confirmed by single-cell RNA-seq analysis.
    RESULTS: In GBM tissues, LLT-1 was predominantly colocalized with glial fibrillary acidic protein (GFAP)-expressing astrocytes, but not with microglial markers like Iba-1. Additionally, LLT-1 and activated NLRC4 inflammasomes were mainly co-expressed in intratumoral astrocytes, suggesting an association between LLT-1, NLRC4, and glioma malignancy. High LLT-1 expression correlates with poor prognosis, particularly in the mesenchymal subtype, and is associated with TNF and NOD-like receptor signaling pathway enrichment, indicating a potential role in tumor inflammation and progression. At the single-cell level, mesenchymal-like malignant cells showed high NF, NLR, and IL-1 signaling pathway enrichment compared to other malignant cell types.
    CONCLUSIONS: We revealed an association between NLRC4 inflammasome activity and LLT-1 expression, suggesting a novel regulatory pathway involving TNF, inflammasomes, and IL-1, potentially offering new NK-cell-mediated anti-glioma approaches.
    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
    CD49d,由基因整合素α4编码,是细胞粘附受体的重要成员,它在各种免疫细胞中广泛表达,以触发针对入侵病原体的免疫反应。在本研究中,在太平洋牡蛎Crassostreagigas中研究了CgCD49d的表达及其对TNF表达的调节作用。有五个Int-alpha域,在CgCD49d中鉴定的整合素_α2区和插入的独特FG-GAP重复区。CgCD49d转录本在血细胞中特异性表达,LPS和脾弧菌刺激后,其在血细胞中的mRNA表达水平增加。CgCD49d用其抗体阻断后,脾静脉刺激后,MAPK信号通路中CgJNK的磷酸化水平和CgTNF转录本显著降低。在使用其抑制剂抑制CgJNK的磷酸化水平后,CgRel的核易位受到抑制,CgTNF转录本在脾V.刺激后也显着降低。此外,发现CgCD49d主要在粒细胞亚群中表达,和AlexaFluor488缀合的CgCD49d抗体标记的粒细胞,在共聚焦显微镜下在CgCD49d+粒细胞表面上有一圈绿色荧光信号,占血细胞总数的24.9±4.53%。总的来说,这些结果表明,CgCD49d通过介导MAPK通路促进牡蛎血细胞中TNF的表达抵抗细菌侵袭,它可以用作表面标记,以对血细胞中的粒细胞亚群进行分型和分类。
    CD49d, encoded by the gene Integrin α4, is a significant member of cell adhesion receptors, which is widely expressed in various immune cells to trigger immune responses against invading pathogens. In the present study, the expression of CgCD49d and its regulatory role in TNF expression were investigated in the Pacific oyster Crassostrea gigas. There were five Int-alpha domains, an Integrin_alpha2 region and a unique FG-GAP repeat region inserted identified in CgCD49d. CgCD49d transcript was specifically expressed in haemocytes, and its mRNA expression level in haemocytes increased after LPS and Vibrio splendidus stimulation. After CgCD49d was blocked by using its antibody, the phosphorylation level of CgJNK in the MAPK signaling pathway and CgTNF transcripts decreased significantly post V. splendidus stimulation. After phosphorylation level of CgJNK was inhibited by using its inhibitor, the nuclear translocation of CgRel was restrained and CgTNF transcripts also decreased significantly post V. splendidus stimulation. Furthermore, CgCD49d was found to be mainly expressed in the agranulocyte subpopulation, and Alexa Fluor 488-conjugated CgCD49d antibody labeled agranulocytes with a circle of green fluorescence signals on CgCD49d+ agranulocyte surface under Confocal microscopy, which accounted for 24.9 ± 4.53% of total haemocytes. Collectively, these results suggested that CgCD49d promoted TNF expression in oyster haemocytes against bacterial invasion by mediating MAPK pathway, and it could be used as a surface marker to type and sort a subset of agranulocyte subpopulation among haemocytes.
    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
    肿瘤坏死因子(TNF)是许多细胞因子之一-负责免疫系统细胞之间通讯的蛋白质分子。TNF因其在实验系统中具有惊人的抗肿瘤作用而被发现并被命名。但是它在整个生物体中的主要生理功能与肿瘤的保护完全无关。这篇简短的评论讨论了早期基因组编辑技术产生的“人造”小鼠模型,这使我们能够确定TNF在健康和某些疾病中的真正功能,并揭示改善TNF依赖性疾病治疗的潜在策略。
    Tumor necrosis factor (TNF) is one of many cytokines - protein molecules responsible for communication between the cells of immune system. TNF was discovered and given its grand name because of its striking antitumor effects in experimental systems, but its main physiological functions in the context of whole organism turned out to be completely unrelated to protection against tumors. This short review discusses \"man-made\" mouse models generated by early genome-editing technologies, which enabled us to establish true functions of TNF in health and certain diseases as well as to unravel potential strategies for improving therapy of TNF-dependent diseases.
    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
    NLR构成了一个很大的,对健康和疾病至关重要的高度保守的胞浆模式识别受体家族,使它们成为关键的治疗靶点。NLRC5是一种神秘的NLR,具有与炎症和传染病相关的突变,但对其作为先天免疫传感器和细胞死亡调节因子的功能知之甚少。因此,我们筛查了NLRC5在感染反应中的作用,PAMPs,DAMPs,和细胞因子。我们发现NLRC5作为驱动炎性细胞死亡的先天免疫传感器,全角下垂,响应特定的配体,包括PAMP/血红素和血红素/细胞因子组合。NLRC5与NLRP12和PANphosome组件相互作用形成细胞死亡复合物,这表明NLR网络的形式与植物中的类似。机械上,TLR信号和NAD+水平调节NLRC5表达和ROS产生以控制细胞死亡。此外,NLRC5缺陷小鼠在溶血和炎症模型中受到保护,表明NLRC5可能是一个潜在的治疗靶点。
    NLRs constitute a large, highly conserved family of cytosolic pattern recognition receptors that are central to health and disease, making them key therapeutic targets. NLRC5 is an enigmatic NLR with mutations associated with inflammatory and infectious diseases, but little is known about its function as an innate immune sensor and cell death regulator. Therefore, we screened for NLRC5\'s role in response to infections, PAMPs, DAMPs, and cytokines. We identified that NLRC5 acts as an innate immune sensor to drive inflammatory cell death, PANoptosis, in response to specific ligands, including PAMP/heme and heme/cytokine combinations. NLRC5 interacted with NLRP12 and PANoptosome components to form a cell death complex, suggesting an NLR network forms similar to those in plants. Mechanistically, TLR signaling and NAD+ levels regulated NLRC5 expression and ROS production to control cell death. Furthermore, NLRC5-deficient mice were protected in hemolytic and inflammatory models, suggesting that NLRC5 could be a potential therapeutic 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
    间充质干细胞(MSCs)用于细胞治疗;尽管如此,它们的应用受到移植后在促炎微环境中的低存活率的限制。骨髓间充质干细胞的巨自噬/自噬激活构成了应激适应通路,促进细胞稳态。我们的蛋白质组学数据表明RUBCNL/PACER(RUN和富含半胱氨酸的结构域含有beclin1相互作用蛋白样),自噬的正调节因子,也参与了细胞死亡。因此,我们筛选了在RUBCNL功能丧失或获得的情况下各种细胞死亡刺激下MSC的存活。当RUBCNL表达时,保护MSC免受TNF(肿瘤坏死因子)诱导的细胞死亡的影响。TNF通过诱导RIPK1激酶依赖性细胞凋亡或坏死促进炎症。我们确定,当半胱天冬酶失活时,MSC在TNF感应和坏死后屈服于RIPK1激酶依赖性凋亡。我们表明RUBCNL是RIPK1依赖性凋亡和坏死的负调节因子。此外,RUBCNL突变体失去调节自噬的能力,保留它们在负调节细胞死亡中的功能。我们还发现RUBCNL与RIPK1形成复合物,RIPK1响应TNF而分解。根据这一发现,RUBCNL表达限制RIPK1-TNFRSF1A/TNFR1复合物I的组装,表明RUBCNL和RIPK1之间的复合物形成抑制TNF信号传导。这些结果为RIPK1介导的细胞死亡和自噬机制之间的串扰提供了新的见解,并表明RUBCNL,由于其在自噬和凋亡/坏死中的功能二重性,可以靶向提高MSCs的治疗效果。
    Mesenchymal stem cells (MSCs) are used in cell therapy; nonetheless, their application is limited by their poor survival after transplantation in a proinflammatory microenvironment. Macroautophagy/autophagy activation in MSCs constitutes a stress adaptation pathway, promoting cellular homeostasis. Our proteomics data indicate that RUBCNL/PACER (RUN and cysteine rich domain containing beclin 1 interacting protein like), a positive regulator of autophagy, is also involved in cell death. Hence, we screened MSC survival upon various cell death stimuli under loss or gain of function of RUBCNL. MSCs were protected from TNF (tumor necrosis factor)-induced regulated cell death when RUBCNL was expressed. TNF promotes inflammation by inducing RIPK1 kinase-dependent apoptosis or necroptosis. We determine that MSCs succumb to RIPK1 kinase-dependent apoptosis upon TNF sensing and necroptosis when caspases are inactivated. We show that RUBCNL is a negative regulator of both RIPK1-dependent apoptosis and necroptosis. Furthermore, RUBCNL mutants that lose the ability to regulate autophagy, retain their function in negatively regulating cell death. We also found that RUBCNL forms a complex with RIPK1, which disassembles in response to TNF. In line with this finding, RUBCNL expression limits assembly of RIPK1-TNFRSF1A/TNFR1 complex I, suggesting that complex formation between RUBCNL and RIPK1 represses TNF signaling. These results provide new insights into the crosstalk between the RIPK1-mediated cell death and autophagy machineries and suggest that RUBCNL, due to its functional duality in autophagy and apoptosis/necroptosis, could be targeted to improve the therapeutic efficacy of MSCs. Abbreviations: BAF: bafilomycin A1; CASP3: caspase 3; Caspases: cysteine-aspartic proteases; cCASP3: cleaved CASP3; CQ: chloroquine; CHX: cycloheximide; cPARP: cleaved poly (ADP-ribose) polymerase; DEPs: differential expressed proteins; ETO: etoposide; MEF: mouse embryonic fibroblast; MLKL: mixed lineage kinase domain-like; MSC: mesenchymal stem cell; MTORC1: mechanistic target of rapamycin kinase complex 1; Nec1s: necrostatin 1s; NFKB/NF-kB: nuclear factor of kappa light polypeptide gene enhancer in B cells; PLA: proximity ligation assay; RCD: regulated cell death; RIPK1: receptor (TNFRSF)-interacting serine-threonine kinase 1; RIPK3: receptor-interacting serine-threonine kinase 3; RUBCNL/PACER: RUN and cysteine rich domain containing beclin 1 interacting protein like; siCtrl: small interfering RNA nonsense; siRNA: small interfering RNA; TdT: terminal deoxynucleotidyl transferase; Tm: tunicamycin; TNF: tumor necrosis factor; TNFRSF1A/TNFR1: tumor necrosis factor receptor superfamily, member 1a.
    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
    受体相互作用丝氨酸/苏氨酸蛋白激酶1(RIPK1)作为一个关键的应激前哨,协调细胞存活,炎症,和免疫原性细胞死亡(ICD)。尽管RIPK1的催化功能是触发细胞死亡所必需的,其非催化支架功能介导强大的促生存信号。因此,癌细胞可以劫持RIPK1来阻止坏死和逃避免疫检测。我们产生了选择性降解人和鼠RIPK1的小分子蛋白水解靶向嵌合体(PROTAC)。PROTAC介导的RIPK1耗尽下调TNFR1和TLR3/4信号中枢,增强NF-κB的输出,MAPK,和IFN信号。此外,RIPK1降解同时促进RIPK3活化和凋亡诱导。我们进一步证明,RIPK1降解通过使癌细胞对治疗诱导的TNF和干扰素敏感来增强放射疗法和免疫疗法的免疫刺激作用。这促进了ICD,抗肿瘤免疫,和持久的治疗反应。因此,通过PROTACs靶向RIPK1是克服放射疗法或免疫疗法耐药性并增强抗癌疗法的一种有前景的方法。
    Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) functions as a critical stress sentinel that coordinates cell survival, inflammation, and immunogenic cell death (ICD). Although the catalytic function of RIPK1 is required to trigger cell death, its non-catalytic scaffold function mediates strong pro-survival signaling. Accordingly, cancer cells can hijack RIPK1 to block necroptosis and evade immune detection. We generated a small-molecule proteolysis-targeting chimera (PROTAC) that selectively degraded human and murine RIPK1. PROTAC-mediated depletion of RIPK1 deregulated TNFR1 and TLR3/4 signaling hubs, accentuating the output of NF-κB, MAPK, and IFN signaling. Additionally, RIPK1 degradation simultaneously promoted RIPK3 activation and necroptosis induction. We further demonstrated that RIPK1 degradation enhanced the immunostimulatory effects of radio- and immunotherapy by sensitizing cancer cells to treatment-induced TNF and interferons. This promoted ICD, antitumor immunity, and durable treatment responses. Consequently, targeting RIPK1 by PROTACs emerges as a promising approach to overcome radio- or immunotherapy resistance and enhance anticancer therapies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号