Receptors, Prostaglandin E, EP4 Subtype

受体,前列腺素 E,EP4 亚型
  • 文章类型: Journal Article
    前列腺素E2(PGE2)是炎性痛觉过敏的主要贡献者,然而,它调节伤害性轴突活动的程度尚不完全清楚。我们开发并表征了微流体细胞培养模型,以研究背根神经节神经元轴突的敏化。我们表明,将PGE2应用于流体分离的轴突会导致其对去极化刺激的反应敏感。有趣的是,将PGE2应用于DRG轴突会引起直接和持续的尖峰活动,传播到体细胞。EP4受体抑制剂和cAMP合成阻断剂消除了轴突的持续活性和膜去极化。对加标活性机制的进一步研究表明,Nav1.8钠通道阻滞剂抑制了PGE2诱发的去极化,但对TTX或扎特拉定的应用是难以反应的。有趣的是,通过用T16Ainh-A01阻断ANO1通道来阻断轴突的去极化。我们进一步表明,用Na-K-2Cl协同转运蛋白NKCC1抑制剂布美他尼治疗后,由于轴突内氯化物梯度的变化,PGE2引起的轴突反应发生了变化,但不是VU01240551氯化钾转运蛋白KCC2的抑制剂。我们的数据证明了PGE2/EP4/cAMP途径的新作用,该途径最终导致了通过ANO1通道的氯化物电流介导的感觉轴突的持续去极化。因此,使用微流体培养模型,我们为PGE2在炎性疼痛中的潜在双重功能提供了证据:它使痛觉轴突的去极化诱发反应敏感,并通过激活ANO1和Nav1.8通道直接触发动作电位.
    Prostaglandin E2 (PGE2) is a major contributor to inflammatory pain hyperalgesia, however, the extent to which it modulates the activity of nociceptive axons is incompletely understood. We developed and characterized a microfluidic cell culture model to investigate sensitisation of the axons of dorsal root ganglia neurons. We show that application of PGE2 to fluidically isolated axons leads to sensitisation of their responses to depolarising stimuli. Interestingly the application of PGE2 to the DRG axons elicited a direct and persistent spiking activity propagated to the soma. Both the persistent activity and the membrane depolarisation in the axons are abolished by the EP4 receptor inhibitor and a blocker of cAMP synthesis. Further investigated into the mechanisms of the spiking activity showed that the PGE2 evoked depolarisation was inhibited by Nav1.8 sodium channel blockers but was refractory to the application of TTX or zatebradine. Interestingly, the depolarisation of axons was blocked by blocking ANO1 channels with T16Ainh-A01. We further show that PGE2-elicited axonal responses are altered by the changes in chloride gradient within the axons following treatment with bumetanide a Na-K-2Cl cotransporter NKCC1 inhibitor, but not by VU01240551 an inhibitor of potassium-chloride transporter KCC2. Our data demonstrate a novel role for PGE2/EP4/cAMP pathway which culminates in a sustained depolarisation of sensory axons mediated by a chloride current through ANO1 channels. Therefore, using a microfluidic culture model, we provide evidence for a potential dual function of PGE2 in inflammatory pain: it sensitises depolarisation-evoked responses in nociceptive axons and directly triggers action potentials by activating ANO1 and Nav1.8 channels.
    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
    SonicHedgehog(SHH)是胚胎模式的驱动力,当损坏时,引发发育障碍和癌症。SHH效应反应是通过初级纤毛(PC)组织的,该纤毛随着细胞周期的生长和缩回并响应细胞外提示。PC稳态的破坏会破坏SHH调节,对维持纤毛健康的途径施加显著压力。在SHH刺激的细胞中确保纤毛鲁棒性的机制尚不清楚。在这里,我们揭示了由SHH激活磷脂酶A2α引起的串扰电路,该电路驱动纤毛E型前列腺素受体4(EP4)信号传导以确保PC功能并稳定纤毛长度。我们证明了SHH-EP4串扰的阻断会使PC循环AMP(cAMP)平衡不稳定,减慢纤毛运输,减少纤毛长度,并减弱SHH途径的诱导。因此,Ep4-/-小鼠表现出缩短的神经上皮PC和改变的SHH依赖性神经元细胞命运规范。因此,SHH启动不同纤毛受体之间的协调,以维持PC功能和长度稳态,以实现强大的下游信号传导。
    Sonic Hedgehog (SHH) is a driver of embryonic patterning that, when corrupted, triggers developmental disorders and cancers. SHH effector responses are organized through primary cilia (PC) that grow and retract with the cell cycle and in response to extracellular cues. Disruption of PC homeostasis corrupts SHH regulation, placing significant pressure on the pathway to maintain ciliary fitness. Mechanisms by which ciliary robustness is ensured in SHH-stimulated cells are not yet known. Herein, we reveal a crosstalk circuit induced by SHH activation of Phospholipase A2α that drives ciliary E-type prostanoid receptor 4 (EP4) signaling to ensure PC function and stabilize ciliary length. We demonstrate that blockade of SHH-EP4 crosstalk destabilizes PC cyclic AMP (cAMP) equilibrium, slows ciliary transport, reduces ciliary length, and attenuates SHH pathway induction. Accordingly, Ep4-/- mice display shortened neuroepithelial PC and altered SHH-dependent neuronal cell fate specification. Thus, SHH initiates coordination between distinct ciliary receptors to maintain PC function and length homeostasis for robust downstream signaling.
    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
    常规,长期使用阿司匹林可能与遗传变异协同作用,特别是那些机械相关的途径,赋予对结直肠癌(CRC)风险的保护作用。我们利用了52项临床试验的汇总数据,队列,病例对照研究包括30,806例CRC病例和41,861例欧洲血统对照,在常规阿司匹林/非甾体抗炎药(NSAID)使用和估算的遗传变异之间进行全基因组相互作用扫描。调整多次比较后,我们在6q24.1中发现了常规阿司匹林/NSAID使用与变体之间的统计学显着相互作用(排名最高的rs72833769),有影响TBC1D7(TSC1-TSC2复合物的亚基,MTOR活动的关键调节器),和5p13.1中的变体(最高命中rs350047),它与PTGER4(编码直接参与阿司匹林作用模式的细胞表面受体)的表达有关。具有功能影响的遗传变异可能会调节常规使用阿司匹林的化学预防作用,我们的研究确定了推定的先前未识别的目标,以进行额外的机械询问。
    Regular, long-term aspirin use may act synergistically with genetic variants, particularly those in mechanistically relevant pathways, to confer a protective effect on colorectal cancer (CRC) risk. We leveraged pooled data from 52 clinical trial, cohort, and case-control studies that included 30,806 CRC cases and 41,861 controls of European ancestry to conduct a genome-wide interaction scan between regular aspirin/nonsteroidal anti-inflammatory drug (NSAID) use and imputed genetic variants. After adjusting for multiple comparisons, we identified statistically significant interactions between regular aspirin/NSAID use and variants in 6q24.1 (top hit rs72833769), which has evidence of influencing expression of TBC1D7 (a subunit of the TSC1-TSC2 complex, a key regulator of MTOR activity), and variants in 5p13.1 (top hit rs350047), which is associated with expression of PTGER4 (codes a cell surface receptor directly involved in the mode of action of aspirin). Genetic variants with functional impact may modulate the chemopreventive effect of regular aspirin use, and our study identifies putative previously unidentified targets for additional mechanistic interrogation.
    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
    少突胶质细胞谱系细胞,包括NG2胶质细胞,经历各种神经退行性疾病的显著变化。这里,我们确定了NG2神经胶质对朊病毒毒性的神经保护作用。朊病毒感染后,小脑器官型培养切片(COCS)和朊病毒接种小鼠的大脑中的NG2胶质细胞被激活。在这两个模型系统中,NG2神经胶质的耗竭加剧了朊病毒诱导的神经变性并加速了朊病毒病理。NG2胶质细胞的缺失增强了小胶质细胞对前列腺素E2(PGE2)的生物合成,通过与EP4受体结合增强朊病毒的神经毒性。PGE2生物合成的药理学或遗传学抑制减弱了COCS和小鼠的朊病毒诱导的神经变性,减少NG2-神经胶质细胞耗竭COCS增强的神经变性感染后,并抑制了NG2-胶质细胞耗竭小鼠中朊病毒病的加速。这些数据揭示了朊病毒疾病中NG2神经胶质和小胶质细胞之间的非细胞自主相互作用,并表明PGE2信号传导可能代表了针对朊病毒疾病的可行靶标。
    Oligodendrocyte-lineage cells, including NG2 glia, undergo prominent changes in various neurodegenerative disorders. Here, we identify a neuroprotective role for NG2 glia against prion toxicity. NG2 glia were activated after prion infection in cerebellar organotypic cultured slices (COCS) and in brains of prion-inoculated mice. In both model systems, depletion of NG2 glia exacerbated prion-induced neurodegeneration and accelerated prion pathology. Loss of NG2 glia enhanced the biosynthesis of prostaglandin E2 (PGE2) by microglia, which augmented prion neurotoxicity through binding to the EP4 receptor. Pharmacological or genetic inhibition of PGE2 biosynthesis attenuated prion-induced neurodegeneration in COCS and mice, reduced the enhanced neurodegeneration in NG2-glia-depleted COCS after prion infection, and dampened the acceleration of prion disease in NG2-glia-depleted mice. These data unveil a non-cell-autonomous interaction between NG2 glia and microglia in prion disease and suggest that PGE2 signaling may represent an actionable target against prion 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
    目的:了解前列腺素E2(PGE2)介导的树突状细胞(DC)免疫抑制机制。
    方法:对4T1荷瘤小鼠(TBM)进行体内实验。在骨髓来源的DC(BMDCs)中进行了体外实验,或脾细胞。通过ELISA/ELIspot监测细胞因子。通过RT-PCR/流式细胞术监测基因表达。
    结果:在计算机上,在体外,4T1TBM体内实验显示PGE2通过EP4受体在DCs中诱导IL-6/pSTAT3信号传导,导致他们的功能障碍。这些效应被EP4抗体中和逆转,EP4拮抗剂,和STAT3抑制肽。PGE2诱导的IL-6可以被miR-365调节,因为其模拟物抑制PGE2诱导的IL-6,并且抑制剂增加DC中IL-6的水平。人类乳腺癌的生物信息学分析也揭示了PGE2和IL-6之间的强比较相关性(相关分析R)(R=0.94)。携带PTGS-2KD4T1肿瘤的小鼠肿瘤负荷降低,PGE2,EP4,IL-6和pSTAT3信号,以及改善的DC和T细胞功能。用环氧合酶-2(COX-2)抑制剂或EP4拮抗剂治疗小鼠可降低肿瘤负荷,EP4拮抗剂的这种作用在体内清除CD11c细胞后被消除,表明PGE2信号在DCs中在肿瘤进展中的关键作用。
    结论:总之,我们的数据强调了树突状细胞在介导PGE2介导的免疫抑制和EP4或STAT3抑制剂中的重要性,或者使用miR365模拟物可以恢复癌症中的免疫原性.
    OBJECTIVE: To understand the mechanism of prostaglandin E2 (PGE2)-mediated immunosuppression in dendritic cells (DCs).
    METHODS: In vivo experiments were conducted on 4T1 tumor bearing mice (TBM). In vitro experiments were performed in bone marrow-derived DCs (BMDCs), or spleen cells. Cytokines were monitored by ELISA/ELIspot. Gene expression was monitored by RT-PCR/flow cytometry.
    RESULTS: In silico, in vitro, and in vivo experiments in 4T1 TBM revealed that PGE2 induced IL-6/pSTAT3 signaling through EP4 receptors in DCs, resulting in their dysfunction. These effects were reversed by EP4 antibody neutralization, EP4 antagonist, and STAT3 inhibitory peptides. PGE2 induced IL-6 was regulated by miR-365, as its mimic inhibited PGE2 induced IL-6 and the inhibitor increased lL-6 levels in DC. Bio-informatic analysis in human mammary cancers also revealed a strong compared co-relation between PGE2 and IL-6 (Correlation AnalyzeR) (R = 0.94). Mice bearing PTGS-2 KD 4T1 tumors had decreased tumor burden, PGE2, EP4, IL-6, and pSTAT3 signaling, along with improved DCs and T cell functions. Treatment of mice with a cyclooxygenase-2 (COX-2) inhibitor or EP4 antagonist decreased tumor burden, and this effect of EP4 antagonist was abrogated upon in vivo depletion of CD11c cells, indicating the crucial role of PGE2 signaling in DCs in tumor progression.
    CONCLUSIONS: In summary, our data highlights the importance of dendritic cells in mediating PGE2-mediated immunosuppression and the use of EP4 or STAT3 inhibitors or miR365 mimics can restore immunogenicity in cancer.
    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: English Abstract
    BACKGROUND: Tumor microenvironment (TME) is one of the important factors in tumorigenesis and progression, in which tumor-associated macrophages (TAMs) play an important role in non-small cell lung cancer (NSCLC) progression. However, the mechanism of TAMs in NSCLC progression remains unclear, so this study aimed to investigate the role of TAMs in NSCLC progression and to find potential therapeutic targets.
    METHODS: Gene Expression Profiling Interactive Analysis (GEPIA) database was used to analyze the expression of prostaglandin E2 receptor 4 (EP4) mRNA in NSCLC and normal lung tissues; the protein expression levels of cyclooxygenase-2 (COX-2), EP4, cluster of differentiation 86 (CD86), CD163 and CD31 were detected by immunohistochemistry (IHC) in 120 NSCLC tissues and 24 paracancerous tissues specimens. The nude mouse lung adenocarcinoma cell A549 and macrophage RAW264.7 co-transplanted tumor model was established. And the samples were collected by gavage with EP4 inhibitor E7046, and then stained with hematoxylin-eosin (HE), IHC, and immunofluorescence (IF), and then detected by Western blot for the epithelial mesenchymal transformation (EMT) of the tumor tissues of the nude mice in each group. Western blot was used to detect the expressions of EMT related protiens in each group of nude mice; full-length transcriptome sequencing was used to screen the key genes causing liver metastasis and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis was performed.
    RESULTS: EP4 mRNA expression level in NSCLC tissues was generally lower than that in normal lung tissues (P<0.05); COX-2, EP4, CD163, CD31 proteins were differentially expressed in NSCLC tissues and adjacent tissues, and differences were observed in many clinicopathological parameters of NSCLC patients; RAW264.7 shortened the latency period of tumorigenesis of A549 and promoted the proliferation of tumors and liver metastasis of tumors, and E7046 could reduce tumor cell proliferation activity, tumor tissue vascular density and M2-type macrophage infiltration in nude mice; IF staining showed that macrophages were mainly distributed around the metastatic foci of tumors; Western blot results showed that compared with A549 alone transplantation group, the relative expression of E-cadherin protein in tumor tissues of mice in A549 and RAW264.7 co-transplantation group was significantly decreased, and the difference was statistically significant (P<0.05), while the relative expression of N-cadherin protein was up-regulated, but the difference was not statistically significant (P>0.05); the main pathways enriched in the differential genes of the full-length transcriptome were the PI3K-AKT and MAPK signaling pathways.
    CONCLUSIONS: During NSCLC development, the COX-2/PGE2/EP4 axis may promote tumor progression by inducing macrophage functional activation, and EP4 may be a potential new target for tumor immunotherapy. This study provides new perspectives and ideas for in-depth exploration of the mechanisms of NSCLC development, as well as a theoretical basis for the development of new therapeutic strategies for NSCLC.
    【中文题目:COX-2/PGE2/EP4轴诱导巨噬细胞功能活化在NSCLC发展过程中的作用】 【中文摘要:背景与目的 肿瘤微环境(tumor microenvironment, TME)是肿瘤发生、发展的重要因素之一,其中肿瘤相关巨噬细胞(tumor-associated macrophages, TAMs)在非小细胞肺癌(non-small cell lung cancer, NSCLC)进展中起着重要作用。然而,TAMs在NSCLC发展过程中的作用机制仍不清楚,因此本研究旨在探讨TAMs在NSCLC发展过程中的作用,并寻找潜在的治疗靶点。方法 使用GEPIA(Gene Expression Profiling Interactive Analysis)数据库分析前列腺素E2受体4(prostaglandin E2 receptor 4, EP4)mRNA在NSCLC和正常肺组织中的表达情况;利用免疫组化(immunohistochemistry, IHC)方法检测120例NSCLC组织及24例癌旁组织标本中环氧合酶-2(cyclooxygenase-2, COX-2)、EP4、分化簇86(cluster of differentiation 86, CD86)、CD163、CD31的蛋白表达水平;建立裸鼠肺腺癌细胞A549与巨噬细胞RAW264.7共移植瘤模型,使用EP4抑制剂E7046灌胃,收集样本行苏木素-伊红(hematoxylin-eosin, HE)、IHC和免疫荧光(immunofluorescence, IF)染色,免疫印迹(Western blot)检测各组裸鼠肿瘤组织上皮间充质转化(epithelial-mesenchymal transformation, EMT)相关蛋白表达情况;全长转录组测序技术筛选引起肿瘤肝转移的关键基因并进行KEGG(Kyoto Encyclopedia of Genes and Genomes)富集分析。结果 EP4 mRNA在NSCLC组织中表达水平普遍低于正常肺组织(P<0.05) ;COX-2、EP4、CD163、CD31蛋白在NSCLC组织和癌旁组织中差异性表达且在NSCLC患者多项临床病理参数中存在差异;RAW264.7缩短了A549的成瘤潜伏期,促进肿瘤增殖及肿瘤肝转移,E7046可降低裸鼠肿瘤细胞增殖活性、肿瘤组织血管密度及M2型巨噬细胞浸润;IF染色显示巨噬细胞主要分布在肿瘤转移灶周围;Western blot结果显示与A549单独移植组相比,A549与RAW264.7共移植组小鼠肿瘤组织中E-钙黏蛋白(E-cadherin)相对表达量明显降低,差异具有统计学意义(P<0.05),N-钙黏蛋白(N-cadherin)相对表达量上调,但差异无统计学意义(P>0.05);全长转录组差异基因主要富集的通路为PI3K-AKT、MAPK信号通路。结论 在NSCLC发生发展过程中,COX-2/PGE2/EP4轴可能通过诱导巨噬细胞功能活化来促进肿瘤的进展,EP4可能是具有潜力的肿瘤免疫治疗新靶点。本研究为深入探讨NSCLC的发生发展机制提供了新的视角和思路,同时为开发新的NSCLC治疗策略提供了理论依据。
】 【中文关键词:肺肿瘤;PGE2;EP4;巨噬细胞;肿瘤免疫】.
    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
    淋巴结转移,主要由口腔鳞状细胞癌(OSCC)细胞的迁移引起,作为一个关键的预后标志。我们以前已经证明,EP4,前列腺素E2(PGE2)受体的亚型,通过Ca2+信号协调OSCC细胞迁移。EP4通过Ca2+信号传导影响细胞迁移的确切机制,然而,不清楚。我们的研究旨在阐明EP4如何通过该途径控制OSCC细胞迁移。我们发现用激动剂(ONO-AE1-473)激活EP4可增加细胞内Ca2水平和人口腔癌细胞(HSC-3)的迁移,但不是人牙龈成纤维细胞(HGnF)。进一步的RNA测序将EP4与钙调蛋白样蛋白6(CALML6)连接,其角色在OSCC中仍未定义。通过蛋白质-蛋白质相互作用网络分析,CALML6和钙/钙调蛋白依赖性蛋白激酶激酶2(CaMKK2)之间有很强的联系,EP4激活也促进线粒体功能。在HSC-3细胞中过度表达EP4会增加小鼠的实验性肺转移,而用STO-609抑制CaMKK2可显着降低这些转移。这将CaMKK2定位为治疗OSCC转移的潜在新靶标。我们的发现强调CALML6是EP4驱动的线粒体呼吸的关键调节因子,通过CaMKK2途径影响细胞迁移和转移。
    Lymph node metastasis, primarily caused by the migration of oral squamous cell carcinoma (OSCC) cells, stands as a crucial prognostic marker. We have previously demonstrated that EP4, a subtype of the prostaglandin E2 (PGE2) receptor, orchestrates OSCC cell migration via Ca2+ signaling. The exact mechanisms by which EP4 influences cell migration through Ca2+ signaling, however, is unclear. Our study aims to clarify how EP4 controls OSCC cell migration through this pathway. We find that activating EP4 with an agonist (ONO-AE1-473) increased intracellular Ca2+ levels and the migration of human oral cancer cells (HSC-3), but not human gingival fibroblasts (HGnF). Further RNA sequencing linked EP4 to calmodulin-like protein 6 (CALML6), whose role remains undefined in OSCC. Through protein-protein interaction network analysis, a strong connection is identified between CALML6 and calcium/calmodulin-dependent protein kinase kinase 2 (CaMKK2), with EP4 activation also boosting mitochondrial function. Overexpressing EP4 in HSC-3 cells increases experimental lung metastasis in mice, whereas inhibiting CaMKK2 with STO-609 markedly lowers these metastases. This positions CaMKK2 as a potential new target for treating OSCC metastasis. Our findings highlight CALML6 as a pivotal regulator in EP4-driven mitochondrial respiration, affecting cell migration and metastasis via the CaMKK2 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
    前列腺素E2(PGE2)系统在癌症进展中的参与早已被认识到。PGE2作为自分泌和旁分泌信号分子在人体内具有多效性作用。已经观察到高水平的肿瘤内PGE2和PGE2的关键代谢酶的过表达,并且表明有助于肿瘤进展。这已被声称适用于不同类型的实体瘤,包括,但不限于,肺,乳房,还有结肠癌.PGE2对已知促进肿瘤发展的肿瘤细胞和血管生成具有直接作用。然而,PGE2驱动癌症发生的主要机制之一目前被认为是锚定在抑制的抗肿瘤免疫,从而为癌症免疫疗法提供了可能的治疗靶点。EP2和EP4(PGE2的两种受体)正在成为与此目的最相关的受体。本文旨在总结PGE2在免疫系统中的已知作用及其在肿瘤微环境中的功能。意义陈述:前列腺素E2(PGE2)长期以来被认为是癌症中的信号分子。它在肿瘤中的存在与疾病进展反复相关。阐明其对肿瘤微环境免疫成分的影响,突出了PGE2受体拮抗剂在癌症治疗中的潜力,特别是与免疫检查点抑制剂疗法组合。辅助治疗可以提高应答率和基于免疫的疗法的功效。
    The involvement of the prostaglandin E2 (PGE2) system in cancer progression has long been recognized. PGE2 functions as an autocrine and paracrine signaling molecule with pleiotropic effects in the human body. High levels of intratumoral PGE2 and overexpression of the key metabolic enzymes of PGE2 have been observed and suggested to contribute to tumor progression. This has been claimed for different types of solid tumors, including, but not limited to, lung, breast, and colon cancer. PGE2 has direct effects on tumor cells and angiogenesis that are known to promote tumor development. However, one of the main mechanisms behind PGE2 driving cancerogenesis is currently thought to be anchored in suppressed antitumor immunity, thus providing possible therapeutic targets to be used in cancer immunotherapies. EP2 and EP4, two receptors for PGE2, are emerging as being the most relevant for this purpose. This review aims to summarize the known roles of PGE2 in the immune system and its functions within the tumor microenvironment. SIGNIFICANCE STATEMENT: Prostaglandin E2 (PGE2) has long been known to be a signaling molecule in cancer. Its presence in tumors has been repeatedly associated with disease progression. Elucidation of its effects on immunological components of the tumor microenvironment has highlighted the potential of PGE2 receptor antagonists in cancer treatment, particularly in combination with immune checkpoint inhibitor therapeutics. Adjuvant treatment could increase the response rates and the efficacy of immune-based therapies.
    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
    具有抗原的CD8T细胞的扩增对于癌症患者的肿瘤浸润淋巴细胞(TIL)过继细胞疗法(ACT)的成功至关重要1。白细胞介素-2(IL-2)通过促进扩增和细胞毒性能力2,3充当CD8+细胞毒性T淋巴细胞功能的关键调节因子。因此,理解肿瘤微环境中IL-2感知的机制障碍对于实施策略以重振IL-2应答和T细胞抗肿瘤应答是至关重要的。在这里,我们报道了前列腺素E2(PGE2),肿瘤微环境4,5中已知的免疫应答的负调节因子以高浓度存在于来自患者的肿瘤组织中,并通过PGE2受体EP2和EP4导致人CD8+TIL中的IL-2感知受损。机械上,PGE2通过下调IL-2Rγc链抑制TIL中的IL-2传感,导致IL-2Rβ-IL2Rγc膜二聚体的组装缺陷。这导致受损的IL-2-mTOR适应和PGC1α转录抑制,在肿瘤反应性TIL中引起氧化应激和铁细胞死亡。在ACT的TIL扩增期间PGE2信号对EP2和EP4的抑制导致IL-2感知增加,导致肿瘤反应性TIL的增殖增强,并在体内转移细胞后增强肿瘤控制。我们的研究揭示了肿瘤微环境中PGE2介导的人类TILs受损的基本特征。这些发现对癌症免疫疗法和细胞疗法具有治疗意义。并能够开发有针对性的策略来增强IL-2感知并放大TIL中的IL-2反应,从而促进具有增强治疗潜力的效应T细胞的扩增。
    Expansion of antigen-experienced CD8+ T cells is critical for the success of tumour-infiltrating lymphocyte (TIL)-adoptive cell therapy (ACT) in patients with cancer1. Interleukin-2 (IL-2) acts as a key regulator of CD8+ cytotoxic T lymphocyte functions by promoting expansion and cytotoxic capability2,3. Therefore, it is essential to comprehend mechanistic barriers to IL-2 sensing in the tumour microenvironment to implement strategies to reinvigorate IL-2 responsiveness and T cell antitumour responses. Here we report that prostaglandin E2 (PGE2), a known negative regulator of immune response in the tumour microenvironment4,5, is present at high concentrations in tumour tissue from patients and leads to impaired IL-2 sensing in human CD8+ TILs via the PGE2 receptors EP2 and EP4. Mechanistically, PGE2 inhibits IL-2 sensing in TILs by downregulating the IL-2Rγc chain, resulting in defective assembly of IL-2Rβ-IL2Rγc membrane dimers. This results in impaired IL-2-mTOR adaptation and PGC1α transcriptional repression, causing oxidative stress and ferroptotic cell death in tumour-reactive TILs. Inhibition of PGE2 signalling to EP2 and EP4 during TIL expansion for ACT resulted in increased IL-2 sensing, leading to enhanced proliferation of tumour-reactive TILs and enhanced tumour control once the cells were transferred in vivo. Our study reveals fundamental features that underlie impairment of human TILs mediated by PGE2 in the tumour microenvironment. These findings have therapeutic implications for cancer immunotherapy and cell therapy, and enable the development of targeted strategies to enhance IL-2 sensing and amplify the IL-2 response in TILs, thereby promoting the expansion of effector T cells with enhanced therapeutic potential.
    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
    癌症特异性TCF1+干细胞样CD8+T细胞可以通过扩增和效应细胞分化驱动保护性抗癌免疫1-4;然而,这种反应在肿瘤中是功能失调的。目前的癌症免疫治疗2,5-9可以通过TCF1+干细胞样CD8+T细胞在一些但不是所有患者中促进抗癌反应。这种变化指向限制TCF1+CD8+T细胞介导的抗癌免疫的目前不明确的机制。在这里,我们证明了肿瘤来源的前列腺素E2(PGE2)限制了肿瘤内TCF1CD8T细胞的增殖扩增和效应子分化,促进癌症免疫逃逸。PGE2不影响引流淋巴结中TCF1+CD8+T细胞的引发。PGE2通过CD8+T细胞中的EP2和EP4(EP2/EP4)受体信号传导起作用,以限制源自TCF1+肿瘤浸润性CD8+T淋巴细胞(TIL)的早期和晚期效应T细胞群的肿瘤内生成。癌症特异性CD8+T细胞中EP2/EP4信号传导的消融挽救了它们在肿瘤内的扩增和效应子分化,并导致多种小鼠癌症模型中的肿瘤消除。机械上,白介素-2(IL-2)信号通路的抑制是PGE2介导的TCF1+TIL应答抑制的基础.总之,我们揭示了限制TCF1+TIL的IL-2反应性并阻止源自这些细胞的抗癌T细胞反应的关键机制。这项研究确定了PGE2-EP2/EP4轴作为分子靶标,以恢复抗癌TIL中的IL-2反应性以实现癌症免疫控制。
    Cancer-specific TCF1+ stem-like CD8+ T cells can drive protective anticancer immunity through expansion and effector cell differentiation1-4; however, this response is dysfunctional in tumours. Current cancer immunotherapies2,5-9 can promote anticancer responses through TCF1+ stem-like CD8+ T cells in some but not all patients. This variation points towards currently ill-defined mechanisms that limit TCF1+CD8+ T cell-mediated anticancer immunity. Here we demonstrate that tumour-derived prostaglandin E2 (PGE2) restricts the proliferative expansion and effector differentiation of TCF1+CD8+ T cells within tumours, which promotes cancer immune escape. PGE2 does not affect the priming of TCF1+CD8+ T cells in draining lymph nodes. PGE2 acts through EP2 and EP4 (EP2/EP4) receptor signalling in CD8+ T cells to limit the intratumoural generation of early and late effector T cell populations that originate from TCF1+ tumour-infiltrating CD8+ T lymphocytes (TILs). Ablation of EP2/EP4 signalling in cancer-specific CD8+ T cells rescues their expansion and effector differentiation within tumours and leads to tumour elimination in multiple mouse cancer models. Mechanistically, suppression of the interleukin-2 (IL-2) signalling pathway underlies the PGE2-mediated inhibition of TCF1+ TIL responses. Altogether, we uncover a key mechanism that restricts the IL-2 responsiveness of TCF1+ TILs and prevents anticancer T cell responses that originate from these cells. This study identifies the PGE2-EP2/EP4 axis as a molecular target to restore IL-2 responsiveness in anticancer TILs to achieve cancer immune control.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号