Indoleamine-Pyrrole 2,3,-Dioxygenase

吲哚胺 - 吡咯 2, 3, - 双加氧酶
  • 文章类型: Journal Article
    自噬靶向嵌合体(AUTAC)已经成为一种强大的方式,可以选择性地降解肿瘤相关的致病蛋白。但其低生物利用度和非特异性分布显著限制了其治疗效果。受AUTAC分子鸟嘌呤结构的启发,我们在这里报道了由AUTAC分子GN[吲哚胺2,3-双加氧酶(IDO)降解剂]和核苷类似物甲氨蝶呤(MTX)通过超分子相互作用进行的超分子人工纳米AUTAC(GMNP)工程,用于肿瘤特异性蛋白降解。它们的纳米结构允许精确定位并递送到癌细胞中,细胞内的酸性环境可以破坏超分子相互作用,释放MTX来根除肿瘤细胞,调节肿瘤相关巨噬细胞,激活树突状细胞,并诱导自噬。具体来说,诱导的自噬促进释放的GN降解免疫抑制性IDO,进一步增强效应T细胞活性,抑制肿瘤生长和转移。这项研究为构建纳米平台提供了独特的策略,以推进AUTAC在肿瘤免疫治疗领域的发展。
    Autophagy-targeting chimera (AUTAC) has emerged as a powerful modality that can selectively degrade tumor-related pathogenic proteins, but its low bioavailability and nonspecific distribution significantly restrict their therapeutic efficacy. Inspired by the guanine structure of AUTAC molecules, we here report supramolecular artificial Nano-AUTACs (GM NPs) engineered by AUTAC molecule GN [an indoleamine 2,3-dioxygenase (IDO) degrader] and nucleoside analog methotrexate (MTX) through supramolecular interactions for tumor-specific protein degradation. Their nanostructures allow for precise localization and delivery into cancer cells, where the intracellular acidic environment can disrupt the supramolecular interactions to release MTX for eradicating tumor cells, modulating tumor-associated macrophages, activating dendritic cells, and inducing autophagy. Specifically, the induced autophagy facilitates the released GN for degrading immunosuppressive IDO to further enhance effector T cell activity and inhibit tumor growth and metastasis. This study offers a unique strategy for building a nanoplatform to advance the field of AUTAC in tumor immunotherapy.
    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
    胰腺癌是恶性肿瘤相关死亡的第四大原因,随着胰腺癌干细胞驱动的耐药性的迅速发展。然而,胰腺导管腺癌(PDAC)的干性和化疗耐药维持机制尚不清楚.这里,我们证明了双尾C同源物1(BICC1),调节大量细胞质mRNA的RNA结合蛋白,促进PDAC中的化学抗性和干性。机械上,BICC1通过上调吲哚胺2,3-双加氧酶-1(IDO1)表达激活PDAC中的色氨酸分解代谢,一种色氨酸分解代谢酶.色氨酸代谢物水平的增加有助于NAD+合成和氧化磷酸化,导致干细胞样表型。阻断BICC1/IDO1/色氨酸代谢信号传导极大地改善了具有高BICC1水平的若干PDAC模型中的吉西他滨(GEM)功效。这些发现表明BICC1是驱动PDAC的干性和化学抗性的关键色氨酸代谢调节剂,因此是针对化学抗性的组合治疗策略的潜在靶标。
    Pancreatic adenocarcinoma is the fourth leading cause of malignancy-related deaths, with rapid development of drug resistance driven by pancreatic cancer stem cells. However, the mechanisms sustaining stemness and chemotherapy resistance in pancreatic ductal adenocarcinoma (PDAC) remain unclear. Here, we demonstrate that Bicaudal C homolog 1 (BICC1), an RNA binding protein regulating numerous cytoplasmic mRNAs, facilitates chemoresistance and stemness in PDAC. Mechanistically, BICC1 activated tryptophan catabolism in PDAC by up-regulating indoleamine 2,3-dioxygenase-1 (IDO1) expression, a tryptophan-catabolizing enzyme. Increased levels of tryptophan metabolites contribute to NAD+ synthesis and oxidative phosphorylation, leading to a stem cell-like phenotype. Blocking BICC1/IDO1/tryptophan metabolism signaling greatly improves the gemcitabine (GEM) efficacy in several PDAC models with high BICC1 level. These findings indicate that BICC1 is a critical tryptophan metabolism regulator that drives the stemness and chemoresistance of PDAC and thus a potential target for combinatorial therapeutic strategy against chemoresistance.
    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
    吲哚胺2,3-双加氧酶(IDO),在肝细胞癌(HCC)中高表达,在创造免疫抑制肿瘤微环境中起着关键作用。抑制IDO活性已成为一种有前途的免疫治疗策略;然而,IDO抑制剂向肿瘤部位的递送受到限制,限制其治疗效果。在这项研究中,我们开发了一种用于靶向递送IDO抑制剂NLG919的磁性涡旋纳米递送系统,该系统与磁性热疗相结合,以逆转肝癌的免疫抑制微环境并抑制肿瘤生长.该系统包括加载有NLG919(NLG919/PI-FVIOs)的热响应性聚乙烯亚胺涂覆的亚铁磁性涡流畴氧化铁纳米环(PI-FVIOs)。在热效应下,NLG919可以精确地从输送系统中释放,对抗IDO介导的免疫抑制,并与NLG919/PI-FVIOs介导的磁热力学(MTD)治疗诱导的免疫原性细胞死亡(ICD)协同作用,导致有效的HCC抑制。体内研究表明,这种联合疗法通过增强细胞毒性T淋巴细胞的积累和抑制肿瘤内的调节性T细胞来显著抑制肿瘤生长和转移。总的来说,我们的研究结果表明,NLG919/PI-FVIOs可以通过破坏IDO途径和激活ICD来诱导有效的抗肿瘤免疫应答,为肝癌治疗提供了一个有希望的治疗途径。
    Indoleamine 2,3-dioxygenase (IDO), highly expressed in hepatocellular carcinoma (HCC), plays a pivotal role in creating an immune-suppressive tumor microenvironment. Inhibiting IDO activity has emerged as a promising immunotherapeutic strategy; however, the delivery of IDO inhibitors to the tumor site is constrained, limiting their therapeutic efficacy. In this study, we developed a magnetic vortex nanodelivery system for the targeted delivery of the IDO inhibitor NLG919, integrated with magnetic hyperthermia therapy to reverse the immune-suppressive microenvironment of liver cancer and inhibit tumor growth. This system comprises thermoresponsive polyethylenimine-coated ferrimagnetic vortex-domain iron oxide nanorings (PI-FVIOs) loaded with NLG919 (NLG919/PI-FVIOs). Under thermal effects, NLG919 can be precisely released from the delivery system, counteracting IDO-mediated immune suppression and synergizing with NLG919/PI-FVIOs-mediated magnetothermodynamic (MTD) therapy-induced immunogenic cell death (ICD), resulting in effective HCC suppression. In vivo studies demonstrate that this combination therapy significantly inhibits tumor growth and metastasis by enhancing the accumulation of cytotoxic T lymphocytes and suppressing regulatory T cells within the tumor. Overall, our findings reveal that NLG919/PI-FVIOs can induce a potent antitumor immune response by disrupting the IDO pathway and activating the ICD, offering a promising therapeutic avenue for HCC treatment.
    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
    髓系来源的抑制细胞(MDSCs)通过表达一些免疫抑制分子在维持母胎耐受中起着至关重要的作用。如吲哚胺2,3-双加氧酶(IDO)。弓形虫(T.gondii)感染可以破坏母胎界面的免疫微环境,导致不良妊娠结局。然而,弓形虫是否影响dMDSCs中IDO的表达及其作用的分子机制尚不清楚。在这里我们展示,在感染的dMDSCs中,IDO的mRNA水平升高,但蛋白水平降低。机械上,dMDSCs中IDO转录水平的上调是通过STAT3/p52-RelB途径调节的,而IDO表达的降低是由于弓形虫感染后SOCS3增加引起的降解。在体内,IDO-/-感染小鼠的不良妊娠结局比广型感染小鼠更为严重,经外源性犬尿氨酸治疗后明显改善。此外,弓形虫感染诱导的dMDSCs中IDO的减少导致通过Kyn/AhR/SP1信号通路调节的dNK细胞中TGF-β和IL-10的表达下调,最终导致dNK细胞功能障碍,并导致不良妊娠结局的发生。这项研究揭示了弓形虫感染引起的不良妊娠结局的新分子机制。
    Myeloid-derived suppressor cells (MDSCs) play a crucial role in maintaining maternal-fetal tolerance by expressing some immune-suppressive molecules, such as indoleamine 2,3-dioxygenase (IDO). Toxoplasma gondii (T. gondii) infection can break the immune microenvironment of maternal-fetal interface, resulting in adverse pregnancy outcomes. However, whether T. gondii affects IDO expression in dMDSCs and the molecular mechanism of its effect are still unclear. Here we show, the mRNA level of IDO is increased but the protein level decreased in infected dMDSCs. Mechanistically, the upregulation of transcriptional levels of IDO in dMDSCs is regulated through STAT3/p52-RelB pathway and the decrease of IDO expression is due to its degradation caused by increased SOCS3 after T. gondii infection. In vivo, the adverse pregnancy outcomes of IDO-/- infected mice are more severe than those of wide-type infected mice and obviously improved after exogenous kynurenine treatment. Also, the reduction of IDO in dMDSCs induced by T. gondii infection results in the downregulation of TGF-β and IL-10 expression in dNK cells regulated through Kyn/AhR/SP1 signal pathway, eventually leading to the dysfunction of dNK cells and contributing the occurrence of adverse pregnancy outcomes. This study reveals a novel molecular mechanism in adverse pregnancy outcome induced by T. gondii infection.
    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
    吲哚胺2,3-双加氧酶1(IDO)是沿着犬尿氨酸(NFK)途径的色氨酸(Trp)代谢酶。在病理条件下,肿瘤细胞过度表达的IDO导致色氨酸的消耗和代谢产物的积累,抑制局部免疫反应并形成免疫逃逸。因此,抑制IDO活性是肿瘤免疫治疗的策略之一,二十多年来,针对这一靶点的药物设计一直是研究的焦点。除了IDO,同一家族的色氨酸双加氧酶(TDO)也可以在人体中催化相同的生化反应,但它具有与IDO不同的组织分布和底物选择性。基于高效力和低交叉反应性的特定目标的药物设计的原则,在这个主题中,从热力学和动力学的角度研究了IDO和TDO对小分子抑制剂的活性和选择性。目的是阐明实现IDO和TDO抑制剂的有利生物活性和选择性的结构要求。具体来说,通过分子对接和分子动力学模拟初步研究了八个家族的抑制剂与IDO和TDO的相互作用,通过分子力学/广义Born表面积(MM/GBSA)方法预测了抑制剂结合的热力学数据。其次,我们探索了JKloops的自由能源景观,IDO/TDO的动力学控制元件,使用温度复制交换分子动力学(T-REMD)模拟,并阐明了IDO/TDO构象变化的规则与抑制剂选择性机理之间的联系。此外,通过自适应转向分子动力学(ASMD)方法模拟了C1抑制剂(NLG919)的结合和解离过程,这不仅解决了可能的稳定,亚稳态,和C1抑制剂-IDO/TDO相互作用的过渡态,而且还准确预测了C1抑制剂结合和解离的动力学数据。总之,我们构建了从酶(IDO/TDO)构象激活到抑制剂结合/解离的完整过程,并以每个环节的热力学和动力学数据为线索,验证了IDO/TDO对抑制剂选择性的控制机制。这对于我们了解肿瘤免疫治疗药物的设计原则,避免免疫治疗药物的耐药性具有重要意义。
    Indoleamine 2,3-dioxygenase 1 (IDO) is a tryptophan (Trp) metabolic enzyme along the kynurenine (NFK) pathway. Under pathological conditions, IDO overexpressed by tumor cells causes depletion of tryptophan and the accumulation of metabolic products, which inhibit the local immune response and form immune escape. Therefore, the suppression of IDO activity is one of the strategies for tumor immunotherapy, and drug design for this target has been the focus of research for more than two decades. Apart from IDO, tryptophan dioxygenase (TDO) of the same family can also catalyze the same biochemical reaction in the human body, but it has different tissue distribution and substrate selectivity from IDO. Based on the principle of drug design with high potency and low cross-reactivity to specific targets, in this subject, the activity and selectivity of IDO and TDO toward small molecular inhibitors were studied from the perspective of thermodynamics and kinetics. The aim was to elucidate the structural requirements for achieving favorable biological activity and selectivity of IDO and TDO inhibitors. Specifically, the interactions of inhibitors from eight families with IDO and TDO were initially investigated through molecular docking and molecular dynamics simulations, and the thermodynamic data for binding of inhibitors were predicted by the molecular mechanics/generalized Born surface area (MM/GBSA) method. Secondly, we explored the free energy landscape of JKloops, the kinetic control element of IDO/TDO, using temperature replica exchange molecular dynamics (T-REMD) simulations and elucidated the connection between the rules of IDO/TDO conformational changes and the inhibitor selectivity mechanism. Furthermore, the binding and dissociation processes of the C1 inhibitor (NLG919) were simulated by the adaptive steering molecular dynamics (ASMD) method, which not only addressed the possible stable, metastable, and transition states for C1 inhibitor-IDO/TDO interactions, but also accurately predicted kinetic data for C1 inhibitor binding and dissociation. In conclusion, we have constructed a complete process from enzyme (IDO/TDO) conformational activation to inhibitor binding/dissociation and used the thermodynamic and kinetic data of each link as clues to verify the control mechanism of IDO/TDO on inhibitor selectivity. This is of great significance for us to understand the design principles of tumor immunotherapy drugs and to avoid drug resistance of immunotherapy drugs.
    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
    肿瘤微环境(TME)中鼻咽癌(NPC)介导的免疫抑制通常导致其他有希望的免疫疗法失败。在这项研究中,我们确定肿瘤固有的FLI1是损害T细胞抗肿瘤免疫的关键介质。机制研究表明,FLI1协调CBP和STAT1的表达,促进染色质可及性和IDO1的转录激活,以响应T细胞释放的IFN-γ。这种调节级联最终导致IDO1表达增强,导致肿瘤细胞中犬尿氨酸(Kyn)的合成增加。这个,反过来,促进CD8+T细胞耗竭和调节性T细胞(Treg)分化。有趣的是,我们发现FLI1的药理学抑制能有效地阻断CBP/STAT1-IDO1-Kyn轴,从而激发自发和检查点治疗诱导的免疫反应,最终导致肿瘤根除增强。总之,我们的发现将FLI1介导的Kyn代谢描述为NPC中的一种免疫逃避机制,为潜在的治疗干预提供有价值的见解。
    Nasopharyngeal carcinoma (NPC)-mediated immunosuppression within the tumor microenvironment (TME) frequently culminates in the failure of otherwise promising immunotherapies. In this study, we identify tumor-intrinsic FLI1 as a critical mediator in impairing T cell anti-tumor immunity. A mechanistic inquiry reveals that FLI1 orchestrates the expression of CBP and STAT1, facilitating chromatin accessibility and transcriptional activation of IDO1 in response to T cell-released IFN-γ. This regulatory cascade ultimately leads to augmented IDO1 expression, resulting in heightened synthesis of kynurenine (Kyn) in tumor cells. This, in turn, fosters CD8+ T cell exhaustion and regulatory T cell (Treg) differentiation. Intriguingly, we find that pharmacological inhibition of FLI1 effectively obstructs the CBP/STAT1-IDO1-Kyn axis, thereby invigorating both spontaneous and checkpoint therapy-induced immune responses, culminating in enhanced tumor eradication. In conclusion, our findings delineate FLI1-mediated Kyn metabolism as an immune evasion mechanism in NPC, furnishing valuable insights into potential therapeutic interventions.
    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
    抑郁症和肥胖症是具有重大公共卫生影响的普遍疾病。在这项研究中,我们使用高脂饮食(HFD)诱导的肥胖小鼠模型来研究HFD诱导的抑郁样行为的潜在机制.HFD诱导的肥胖小鼠表现出抑郁样行为和海马体积的减少,通过吲哚胺2,3-双加氧酶(IDO)抑制剂1-甲基色氨酸(1-MT)治疗可以逆转。有趣的是,1-MT治疗后未观察到IDO水平的变化,提示1-MT的抗抑郁作用可能涉及其他机制。我们进一步进行了RNA测序分析,以阐明1-MT在HFD诱导的抑郁小鼠中的抗抑郁作用的潜在潜在机制,并发现1-MT治疗和未治疗的HFD诱导的抑郁小鼠之间的细胞外基质(ECM)组织途径中共享差异基因的显着富集。因此,我们假设ECM的变化在1-MT的抗抑郁作用中起关键作用。为此,我们调查了神经周网(PNN),它们是ECM组件,优先鞘鞘旁白蛋白(PV)阳性中间神经元,并参与许多异常。我们发现HFD与PV阳性神经元的过度积累和PNN的上调有关,影响PV阳性神经元的突触传递,并导致海马谷氨酸-γ-氨基丁酸失衡。1-MT有效地扭转了这些变化,强调了1-MT发挥其抗抑郁作用的PNN相关机制。
    Depression and obesity are prevalent disorders with significant public health implications. In this study, we used a high-fat diet (HFD)-induced obese mouse model to investigate the mechanism underlying HFD-induced depression-like behaviors. HFD-induced obese mice exhibited depression-like behaviors and a reduction in hippocampus volume, which were reversed by treatment with an indoleamine 2,3-dioxygenase (IDO) inhibitor 1-methyltryptophan (1-MT). Interestingly, no changes in IDO levels were observed post-1-MT treatment, suggesting that other mechanisms may be involved in the anti-depressive effect of 1-MT. We further conducted RNA sequencing analysis to clarify the potential underlying mechanism of the anti-depressive effect of 1-MT in HFD-induced depressive mice and found a significant enrichment of shared differential genes in the extracellular matrix (ECM) organization pathway between the 1-MT-treated and untreated HFD-induced depressive mice. Therefore, we hypothesized that changes in ECM play a crucial role in the anti-depressive effect of 1-MT. To this end, we investigated perineuronal nets (PNNs), which are ECM assemblies that preferentially ensheath parvalbumin (PV)-positive interneurons and are involved in many abnormalities. We found that HFD is associated with excessive accumulation of PV-positive neurons and upregulation of PNNs, affecting synaptic transmission in PV-positive neurons and leading to glutamate-gamma-aminobutyric acid imbalances in the hippocampus. The 1-MT effectively reversed these changes, highlighting a PNN-related mechanism by which 1-MT exerts its anti-depressive 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
    程序性细胞死亡1配体1(PD-L1)阻断的免疫治疗对NK/T细胞淋巴瘤患者有效。除PD-L1外,吲哚胺2,3-双加氧酶-1(IDO1)是最有前途的免疫治疗靶标之一。通过免疫组织化学(IHC)从230名新诊断的NK/T淋巴瘤患者和来自三个癌症中心的组织样本中观察到高比例的PD-L1和IDO1蛋白,并且与NK/T淋巴瘤患者的总体生存率(OS)差相关。重要的是,PD-L1和IDO1的共表达与NK/T淋巴瘤患者的OS差和限制性平均生存时间短有关,并且是训练队列中的独立预后因素,在58例NK/T淋巴瘤患者中也得到了验证(GSE90597)。此外,根据PD-L1和IDO1表达与年龄一起构建的列线图模型可以提供对OS率和中位生存时间的简明和精确的预测.列线图模型中的高危人群与验证队列中的CD4+T细胞浸润呈正相关,免疫抑制因子水平也是如此。因此,在NK/T淋巴瘤患者中,PD-L1和IDO1高表达与OS差相关.PD-L1和IDO1可能是NK/T淋巴瘤未来免疫检查点阻断(ICB)治疗的潜在靶标。
    Immunotherapy with programmed cell death 1 ligand 1 (PD-L1) blockade was effective in patients with NK/T-cell lymphoma. In addition to PD-L1, indoleamine 2,3-dioxygenase-1 (IDO1) is one of the most promising immunotherapeutic targets. High proportions of PD-L1 and IDO1 proteins were observed by immunohistochemistry (IHC) from 230 newly diagnosed patients with NK/T lymphoma with tissue samples from three cancer centers and were associated with poor overall survival (OS) in patients with NK/T lymphoma. Importantly, the coexpression of PD-L1 and IDO1 was related to poor OS and short restricted mean survival time in patients with NK/T lymphoma and was an independent prognostic factor in the training cohorts, and which was also validated in 58 NK/T lymphoma patients (GSE90597). Moreover, a nomogram model constructed with PD-L1 and IDO1 expression together with age could provide concise and precise predictions of OS rates and median survival time. The high-risk group in the nomogram model had a positive correlation with CD4 + T-cell infiltration in the validation cohort, as did the immunosuppressive factor level. Therefore, high PD-L1 and IDO1 expression was associated with poor OS in patients with NK/T lymphoma. PD-L1 and IDO1 might be potential targets for future immune checkpoint blockade (ICB) therapy for NK/T lymphoma.
    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
    对胰腺导管腺癌(PDAC)的临床治疗仍然存在难以察觉的检查和治疗效果不佳的问题。目前,尽管5-氟尿嘧啶(5-FU),作为临床一线FOLFIRINOX化疗药物,取得了显著的治疗效果。然而,这些不可避免的因素,如低溶解度,缺乏生物特异性,容易诱导免疫抑制环境的形成,严重限制了他们在PDAC的治疗。作为许多肿瘤细胞的重要能量来源,色氨酸(Trp),容易被吲哚胺2,3-双加氧酶1(IDO1)降解为犬尿氨酸(Kyn),激活Kyn-AHR轴以形成促进肿瘤生长和转移的特殊抑制性免疫微环境。然而,我们的研究发现,5-FU可以诱导有效的免疫原性细胞死亡(ICD),通过激活免疫系统进一步治疗肿瘤,而干扰素-γ(IFN-γ)的分泌重新诱导Kyn-AHR轴的激活,导致治疗效率低下。因此,金属基质蛋白酶-2(MMP-2)和内源性GSH双响应脂质体基纳米囊泡,与5-FU(抗癌药物)和NLG919(IDO1抑制剂)共负载,已建成(命名为ENP919@5-FU)。多功能ENP919@5-FU可有效重塑肿瘤免疫抑制微环境,增强化疗疗效,从而有效抑制癌症生长。机械上,高表达MMP-2的PDAC将通过在纳米囊泡表面上脱落PEG推动制备的纳米囊泡停留在肿瘤区域,有效增强肿瘤的摄取。随后,通过高内源性GSH切割含有S-S键的纳米囊泡,导致5-FU和NLG919的持续释放,从而使循环化学免疫疗法能够有效地引起肿瘤消融。此外,ENP919@5-FU联合PD-L1抗体(αPD-L1)对腹腔转移的PDAC模型具有协同抗肿瘤作用。总的来说,ENP919@5-FU纳米囊泡,作为PDAC治疗策略,通过重塑肿瘤微环境循环肿瘤化学免疫疗法扩增显示出优异的抗肿瘤疗效,在精准医学方法中具有很好的潜力。
    Imperceptible examination and unideal treatment effect are still intractable difficulties for the clinical treatment of pancreatic ductal adenocarcinoma (PDAC). At present, despite 5-fluorouracil (5-FU), as a clinical first-line FOLFIRINOX chemo-drug, has achieved significant therapeutic effects. Nevertheless, these unavoidable factors such as low solubility, lack of biological specificity and easy to induce immunosuppressive surroundings formation, severely limit their treatment in PDAC. As an important source of energy for many tumor cells, tryptophan (Trp), is easily degraded to kynurenine (Kyn) by indolamine 2,3- dioxygenase 1 (IDO1), which activates the axis of Kyn-AHR to form special suppressive immune microenvironment that promotes tumor growth and metastasis. However, our research findings that 5-FU can induce effectively immunogenic cell death (ICD) to further treat tumor by activating immune systems, while the secretion of interferon-γ (IFN-γ) re-induce the Kyn-AHR axis activation, leading to poor treatment efficiency. Therefore, a metal matrix protease-2 (MMP-2) and endogenous GSH dual-responsive liposomal-based nanovesicle, co-loading with 5-FU (anti-cancer drug) and NLG919 (IDO1 inhibitor), was constructed (named as ENP919@5-FU). The multifunctional ENP919@5-FU can effectively reshape the tumor immunosuppression microenvironment to enhance the effect of chemoimmunotherapy, thereby effectively inhibiting cancer growth. Mechanistically, PDAC with high expression of MMP-2 will propel the as-prepared nanovesicle to dwell in tumor region via shedding PEG on the nanovesicle surface, effectively enhancing tumor uptake. Subsequently, the S-S bond containing nanovesicle was cut via high endogenous GSH, leading to the continued release of 5-FU and NLG919, thereby enabling circulating chemoimmunotherapy to effectively cause tumor ablation. Moreover, the combination of ENP919@5-FU and PD-L1 antibody (αPD-L1) showed a synergistic anti-tumor effect on the PDAC model with abdominal cavity metastasis. Collectively, ENP919@5-FU nanovesicle, as a PDAC treatment strategy, showed excellent antitumor efficacy by remodeling tumor microenvironment to circulate tumor chemoimmunotherapy amplification, which has promising potential in a precision medicine approach.
    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
    利用亚砜和血红素辅因子之间的关键相互作用,我们使用亚砜作为锚定官能团来开发两个系列的吲哚胺2,3-双加氧酶1(IDO1)抑制剂:2-苄基亚磺酰基苯并恶唑(系列1)和2-苯基亚磺酰基苯并恶唑(系列2)。体外酶促筛选表明,这两个系列都可以在低微摩尔(系列1)或纳摩尔(系列2)水平上抑制IDO1的活性。它们还显示IDO1和色氨酸2,3-双加氧酶2之间的抑制选择性。有趣的是,虽然系列1是这两个系列中效力较低的IDO1抑制剂,在干扰素-γ刺激的BxPC-3细胞中,它对犬尿氨酸的产生表现出更强的抑制活性。酶动力学和结合研究表明,2-亚磺酰基苯并恶唑是色氨酸的非竞争性抑制剂,它们与血红素的亚铁形式相互作用。这些结果证明2-亚磺酰基苯并恶唑作为II型IDO1抑制剂。此外,分子对接研究支持亚砜是与血红素辅因子相互作用的关键官能团。化合物22(系列1)在脂多糖(LPS)刺激的RAW264.7细胞中以浓度依赖性方式抑制NO的产生,并能减轻LPS诱导的小鼠急性肺损伤模型的肺水肿和肺损伤。
    Taking advantage of key interactions between sulfoxide and heme cofactor, we used the sulfoxide as the anchor functional group to develop two series of indoleamine 2, 3-dioxygenase 1 (IDO1) inhibitors: 2-benzylsulfinylbenzoxazoles (series 1) and 2-phenylsulfinylbenzoxazoles (series 2). In vitro enzymatic screening shows that both series can inhibit the activity of IDO1 in low micromolar (series 1) or nanomolar (series 2) levels. They also show inhibitory selectivity between IDO1 and tryptophan 2, 3-dioxygenase 2. Interestingly, although series 1 is less potent IDO1 inhibitors of these two series, it exhibited stronger inhibitory activity toward kynurenine production in interferon-γ stimulated BxPC-3 cells. Enzyme kinetics and binding studies demonstrated that 2-sulfinylbenzoxazoles are non-competitive inhibitors of tryptophan, and they interact with the ferrous form of heme. These results demonstrated 2-sulfinylbenzoxazoles as type II IDO1 inhibitors. Furthermore, molecular docking studies supports the sulfoxide being of the key functional group that interacts with the heme cofactor. Compound 22 (series 1) can inhibit NO production in a concentration dependent manner in lipopolysaccharides (LPS) stimulated RAW264.7 cells, and can relieve pulmonary edema and lung injury in LPS induced mouse acute lung injury models.
    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