RELB

RelB
  • 文章类型: Journal Article
    目的:胆管癌(CCA)是由肝内(iCCA)或肝外(eCCA)胆管引起的侵袭性恶性肿瘤,预后差,治疗选择有限。先前的证据强调了非规范NF-κB信号通路在不同肿瘤类型的起始和侵袭性中的重要贡献。淋巴毒素β(LTβ)刺激NF-κB诱导激酶(NIK),导致转录因子RelB的激活。然而,通过LTβ/NIK/RelB轴的非经典NF-κB信号通路在CCA癌变和进展中的功能贡献尚未确定。
    方法:检测人CCA来源的细胞系和类器官,以确定活化或抑制后NF-κB途径组分的表达。使用实时阻抗测量和流式细胞术分析增殖和细胞死亡。免疫印迹,qRT-PCR,RNA测序和原位杂交用于分析基因和蛋白质表达。使用iCCA的四个体内模型来探测非常规NF-κB途径的激活和调节。
    结果:暴露于LTα1/β2会激活LTβ/NIK/RelB轴并促进CCA中的增殖。用小分子抑制剂B022抑制NIK有效地抑制了患者来源的CCA类器官中的RelB表达以及CCA细胞系中由LTα1/β2刺激的RelB和p52的核共转位。在鼠类CCA中,RelB表达显著增加,并且LTβ是非经典NF-κB信号传导途径的主要配体。
    结论:我们的研究证实,非经典NF-κB轴LTβ/NIK/RelB驱动了胆管癌的发生,并代表了一个候选治疗靶点。
    OBJECTIVE: Cholangiocarcinoma (CCA) is an aggressive malignancy arising from the intrahepatic (iCCA) or extrahepatic (eCCA) bile ducts with poor prognosis and limited treatment options. Prior evidence highlighted a significant contribution of the non-canonical NF-κB signalling pathway in initiation and aggressiveness of different tumour types. Lymphotoxin-β (LTβ) stimulates the NF-κB-inducing kinase (NIK), resulting in the activation of the transcription factor RelB. However, the functional contribution of the non-canonical NF-κB signalling pathway via the LTβ/NIK/RelB axis in CCA carcinogenesis and progression has not been established.
    METHODS: Human CCA-derived cell lines and organoids were examined to determine the expression of NF-κB pathway components upon activation or inhibition. Proliferation and cell death were analysed using real-time impedance measurement and flow cytometry. Immunoblot, qRT-PCR, RNA sequencing and in situ hybridization were employed to analyse gene and protein expression. Four in vivo models of iCCA were used to probe the activation and regulation of the non-canonical NF-κB pathway.
    RESULTS: Exposure to LTα1/β2 activates the LTβ/NIK/RelB axis and promotes proliferation in CCA. Inhibition of NIK with the small molecule inhibitor B022 efficiently suppresses RelB expression in patient-derived CCA organoids and nuclear co-translocation of RelB and p52 stimulated by LTα1/β2 in CCA cell lines. In murine CCA, RelB expression is significantly increased and LTβ is the predominant ligand of the non-canonical NF-κB signalling pathway.
    CONCLUSIONS: Our study confirms that the non-canonical NF-κB axis LTβ/NIK/RelB drives cholangiocarcinogenesis and represents a candidate 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
    已知树突状细胞(DC)功能障碍会加剧肠道病变,但在这种情况下损害DC介导的免疫调节的机制仍不清楚.这里,我们表明,来自实验性结肠炎小鼠模型的肠道树突状细胞表现出显著水平的非经典NF-κB信号,其激活RelB:p52异二聚体。DC中该途径的遗传失活减轻了患有结肠炎的小鼠的肠病理学。RelB的缺乏:p52减少了Axin1的转录,Axin1是β-catenin破坏复合物的关键组成部分,增强Raldh2的β-连环蛋白依赖性表达,其通过促进视黄酸合成赋予耐受性DC属性。非典型NF-κB信号传导的DC特异性损伤导致Tregs和IgA+B细胞的结肠数量增加,促进管腔IgA的产生并促进优生。实验引入的β-连环蛋白单倍体不足的DC缺乏的非经典NF-κB信号调节Raldh2活性,恢复小鼠的致过敏敏感性。最后,炎症性肠病患者在肠道DCs中也显示出有害的非规范NF-κB信号特征.总之,我们确定了树突状细胞中的非经典NF-κB信号传导如何破坏视黄酸的合成以促进肠道炎症。
    Dendritic cell (DC) dysfunction is known to exacerbate intestinal pathologies, but the mechanisms compromising DC-mediated immune regulation in this context remain unclear. Here, we show that intestinal dendritic cells from a mouse model of experimental colitis exhibit significant levels of noncanonical NF-κB signaling, which activates the RelB:p52 heterodimer. Genetic inactivation of this pathway in DCs alleviates intestinal pathologies in mice suffering from colitis. Deficiency of RelB:p52 diminishes transcription of Axin1, a critical component of the β-catenin destruction complex, reinforcing β-catenin-dependent expression of Raldh2, which imparts tolerogenic DC attributes by promoting retinoic acid synthesis. DC-specific impairment of noncanonical NF-κB signaling leads to increased colonic numbers of Tregs and IgA+ B cells, which promote luminal IgA production and foster eubiosis. Experimentally introduced β-catenin haploinsufficiency in DCs with deficient noncanonical NF-κB signaling moderates Raldh2 activity, reinstating colitogenic sensitivity in mice. Finally, inflammatory bowel-disease patients also display a deleterious noncanonical NF-κB signaling signature in intestinal DCs. In sum, we establish how noncanonical NF-κB signaling in dendritic cells can subvert retinoic acid synthesis to fuel intestinal inflammation.
    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
    细胞毒性药物引起的肠道损伤后,控制粘膜愈合的复杂免疫机制仍然知之甚少。这项研究的目的是研究淋巴毒素β受体(LTβR)信号在化疗诱导的肠道损伤中的作用。LTβR缺陷小鼠表现出更高的体重减轻,加剧肠道病理,促炎细胞因子表达增加,IL-22表达减少,甲氨蝶呤(MTX)治疗后肠上皮细胞的增殖。此外,LTβR-/-IL-22-/-小鼠死于MTX治疗,提示LTβR-和IL-22-依赖性途径共同促进粘膜修复。尽管LTβR配体LIGHT和LTβ在MTX治疗后早期在肠道中上调,LIGHT-/-小鼠,但不是LTβ-/-小鼠,显示加剧的疾病。Further,我们揭示了T细胞在粘膜修复中的关键作用,因为T细胞缺陷小鼠未能上调肠道LIGHT表达,并表现出体重减轻和肠道病理增加。对LTβR条件性失活的小鼠的分析表明,LTβR在肠上皮细胞中的信号传导,但不是在Lgr5+肠干细胞中,巨噬细胞或树突状细胞对粘膜修复至关重要。此外,肠上皮细胞中非经典NF-kB途径成员RelB的失活促进了MTX诱导的疾病。基于这些结果,我们提出了一个模型,其中T细胞产生的LIGHT激活肠上皮细胞中的LTβR-RelB信号传导,以促进化疗后的粘膜修复.
    The intricate immune mechanisms governing mucosal healing following intestinal damage induced by cytotoxic drugs remain poorly understood. The goal of this study was to investigate the role of lymphotoxin beta receptor (LTβR) signaling in chemotherapy-induced intestinal damage. LTβR deficient mice exhibited heightened body weight loss, exacerbated intestinal pathology, increased proinflammatory cytokine expression, reduced IL-22 expression, and proliferation of intestinal epithelial cells following methotrexate (MTX) treatment. Furthermore, LTβR-/-IL-22-/- mice succumbed to MTX treatment, suggesting that LTβR- and IL-22- dependent pathways jointly promote mucosal repair. Although both LTβR ligands LIGHT and LTβ were upregulated in the intestine early after MTX treatment, LIGHT-/- mice, but not LTβ-/- mice, displayed exacerbated disease. Further, we revealed the critical role of T cells in mucosal repair as T cell-deficient mice failed to upregulate intestinal LIGHT expression and exhibited increased body weight loss and intestinal pathology. Analysis of mice with conditional inactivation of LTβR revealed that LTβR signaling in intestinal epithelial cells, but not in Lgr5+ intestinal stem cells, macrophages or dendritic cells was critical for mucosal repair. Furthermore, inactivation of the non-canonical NF-kB pathway member RelB in intestinal epithelial cells promoted MTX-induced disease. Based on these results, we propose a model wherein LIGHT produced by T cells activates LTβR-RelB signaling in intestinal epithelial cells to facilitate mucosal repair following chemotherapy treatment.
    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
    小胶质细胞介导的神经炎症是脑缺血再灌注损伤(CI/RI)中脑损伤的主要贡献者之一。最近,发现RNA修饰有助于调节小胶质细胞极化和随后的脑I/R神经炎症的发展。在这里,我们研究了m5CRNA修饰在小胶质细胞诱导的CI/RI神经炎症中的作用和机制。我们发现m5CRNA修饰水平在从腔内大脑中动脉阻塞/再灌注(MCAO/R)小鼠模型中分离的原发性小胶质细胞和BV2小胶质细胞中进行氧糖剥夺和复氧(OGD/R),这种变化伴随着M1/M2极化比的增加。此外,m5C脱甲基酶TET1在小胶质细胞中的表达增加,这促进了M1极化,但阻碍了M2极化。机械上,较高的TET1表达降低了RelB的m5C修饰水平并增强了其mRNA稳定性,这随后增加了M1/M2极化比。总之,这项研究提供了m5CRNA修饰在脑I/R神经炎症发病机制中的作用,并可能加深我们对针对TET1-RelB轴的临床治疗的理解.
    Microglia mediated neuroinflammation is one of the major contributors to brain damage in cerebral ischemia reperfusion injury (CI/RI). Recently, RNA modification was found to contribute to the regulation of microglia polarization and the subsequent development of cerebral I/R neuroinflammation. Herein, we investigated the effect and mechanism of m5C RNA modification in the microglia induced CI/RI neuroinflammation. We found that the m5C RNA modification levels decreased in the primary microglia isolated from a mouse model of intraluminal middle cerebral artery occlusion/reperfusion (MCAO/R) and the BV2 microglial cells subjected to oxygen-glucose deprivation and reoxygenation (OGD/R), and this change was accompanied by an increase in the M1/M2 polarization ratio. Furthermore, the expression of m5C demethylase TET1 in microglia increased, which promoted M1 polarization but impeded M2 polarization. Mechanistically, the higher TET1 expression decreased the m5C modification level of RelB and enhanced its mRNA stability, which subsequently increased the M1/M2 polarization ratio. In conclusion, this study provides insight into the role of m5C RNA modification in the pathogenesis of cerebral I/R neuroinflammation and may deepen our understanding on clinical therapy targeting the TET1-RelB axis.
    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
    非经典NFκB信号通路介导多种细胞存活的生物学功能,增长,成熟,以及对造血细胞和免疫器官的发育和维持重要的分化因子。它的失调与许多免疫病理和恶性肿瘤有关。最初描述为控制NFκB家族成员RelB的信号通路,我们现在知道,非规范信号也控制NFκBRelA和cRel。这里,我们的目的是阐明我们对介导非规范NFκB信号传导的分子网络的理解,并回顾由缺陷或过度活跃的非规范NFκB通路引起的人类疾病.事实证明,RelA和cRel的失调,不是RelB,通常与介导所产生的病理有关。本文分为:免疫系统疾病>分子和细胞生理学癌症>分子和细胞生理学免疫系统疾病>干细胞和发育。
    The noncanonical NFκB signaling pathway mediates the biological functions of diverse cell survival, growth, maturation, and differentiation factors that are important for the development and maintenance of hematopoietic cells and immune organs. Its dysregulation is associated with a number of immune pathologies and malignancies. Originally described as the signaling pathway that controls the NFκB family member RelB, we now know that noncanonical signaling also controls NFκB RelA and cRel. Here, we aim to clarify our understanding of the molecular network that mediates noncanonical NFκB signaling and review the human diseases that result from a deficient or hyper-active noncanonical NFκB pathway. It turns out that dysregulation of RelA and cRel, not RelB, is often implicated in mediating the resulting pathology. This article is categorized under: Immune System Diseases > Molecular and Cellular Physiology Cancer > Molecular and Cellular Physiology Immune System Diseases > Stem Cells and Development.
    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
    治疗选择有限,恶病质仍然是癌症患者的主要挑战。表征肿瘤细胞和免疫微环境之间的相互作用可能有助于确定癌症恶病质的潜在治疗靶标。在这里,我们研究了巨噬细胞通过促进肿瘤分泌TWEAK(TNF样弱凋亡诱导剂)在增强胰腺癌诱导的肌肉萎缩中的关键作用。具体来说,巨噬细胞的消耗逆转了肿瘤细胞诱导的肌肉降解。巨噬细胞通过CCL5/TRAF6/NF-κB途径诱导TWEAK的非自主分泌。TWEAK通过激活MuRF1启动的肌肉重塑促进肌肉萎缩。值得注意的是,肿瘤细胞通过CCL2/CCR2轴募集和重新编程巨噬细胞,并破坏巨噬细胞和肿瘤细胞之间的相互作用减轻肌肉消耗。总的来说,这项研究确定了胰腺癌细胞和巨噬细胞之间的前馈环,肿瘤细胞分泌TWEAK的非自主激活,从而为胰腺癌恶病质提供有希望的治疗靶点。
    With limited treatment options, cachexia remains a major challenge for patients with cancer. Characterizing the interplay between tumor cells and the immune microenvironment may help identify potential therapeutic targets for cancer cachexia. Herein, we investigate the critical role of macrophages in potentiating pancreatic cancer induced muscle wasting via promoting TWEAK (TNF-like weak inducer of apoptosis) secretion from the tumor. Specifically, depletion of macrophages reverses muscle degradation induced by tumor cells. Macrophages induce non-autonomous secretion of TWEAK through CCL5/TRAF6/NF-κB pathway. TWEAK promotes muscle atrophy by activating MuRF1 initiated muscle remodeling. Notably, tumor cells recruit and reprogram macrophages via the CCL2/CCR2 axis and disrupting the interplay between macrophages and tumor cells attenuates muscle wasting. Collectively, this study identifies a feedforward loop between pancreatic cancer cells and macrophages, underlying the non-autonomous activation of TWEAK secretion from tumor cells thereby providing promising therapeutic targets for pancreatic cancer cachexia.
    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
    血液透析(HD)患者的血容量过高与炎症相关。高血容量如何引发炎症尚不完全清楚。我们启动了一项横断面研究,招募了40名通过生物阻抗测量分为等容(N;23)和高容(H;17)组的血液透析患者。与特异性caspase-4抑制剂组合的caspase活性测定用于检测分离的外周血单核细胞(PBMC)中的caspase-4活性。通过磷酸流式细胞术分析转录因子RelA(pS529)和RelB(pS552)。血清内毒素通过基于脂肪细胞裂解物的测定法检测,采用ELISA技术检测IL-6(白细胞介素-6)和TNF-α(肿瘤坏死因子-α)基因表达。高容量患者年龄较大,更常见的糖尿病患者,显示CRP和IL-6水平升高.Caspase-4活性,这与细胞内内毒素检测有关,在H患者中显著升高。尽管RelA表达免疫细胞的频率和这些细胞中的表达密度没有差异,在H患者中,RelB(pS552)阳性染色的单核细胞频率显着降低。H患者的caspase-4活性增加可能表明H患者炎症的原因。RelB(pS552)的翻译后修饰与NF-kB活性的下调有关,可能表明炎症的消退。与H患者相比,N患者更明显。因此,较高的炎症负荷和较低的炎症消退能力是H患者的特征.
    Hypervolemia is associated with inflammation in hemodialysis (HD) patients. How hypervolemia triggers inflammation is not entirely known. We initiated a cross-sectional study enrolling 40 hemodialysis patients who were categorized into normovolemic (N; 23) and hypervolemic (H; 17) groups by bioimpedance measurement. A caspase activity assay in combination with a specific caspase-4 inhibitor was used to detect caspase-4 activity in isolated peripheral blood mononuclear cells (PBMCs). Transcription factors RelA (pS529) and RelB (pS552) were analyzed by phospho-flow cytometry. Serum endotoxins were detected by an amebocyte lysate-based assay, and IL-6 (interleukin-6) and TNF-α (Tumor necrosis factor-α) gene expression were detected using the ELISA technique. Hypervolemic patients were older, more frequently had diabetes and showed increased CRP and IL-6 levels. Caspase-4 activity, which is linked to intracellular endotoxin detection, was significantly elevated in H patients. While the frequency of RelA-expressing immune cells and the expression density in these cells did not differ, the monocytic frequency of cells positively stained for RelB (pS552) was significantly decreased in H patients. Increased caspase-4 activity in H patients may indicate a cause of inflammation in H patients. The post-translational modification of RelB (pS552) is linked to downregulation of NF-kB activity and may indicate the resolution of inflammation, which is more distinct in N patients compared to H patients. Therefore, both higher inflammatory loads and lower inflammatory resolution capacities are characteristics of H patients.
    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
    他莫昔芬(TAM)耐药性仍然是晚期乳腺癌(BCa)治疗的主要障碍。除了雌激素受体(ER)信号通路的竞争性抑制,通过增加活性氧(ROS)来抑制线粒体功能对于增强TAM药效学至关重要。这里,我们表明,RelB通过抑制TAM引起的铁凋亡来促进TAM抗性。TAM诱导的ROS水平促进TAM敏感细胞的铁凋亡,但是在具有高RelB组成水平的TAM抗性细胞中,这种作用得到了缓解。机械上,RelB通过转录上调谷胱甘肽过氧化物酶4(GPX4)抑制铁死亡。因此,在敏感细胞中升高RelB和GPX4会增加TAM抗性,反过来,在抗性细胞中剥夺RelB和GPX4降低了TAM抗性。此外,通过在体外和体内增强铁凋亡来抑制RelB转录激活重新致敏的TAM抗性细胞。在TAM抗性细胞中GPX4的失活通过增加铁凋亡介导的细胞死亡而一致地重新激活TAM。一起,这项研究发现,通过RelB上调的GPX4,铁死亡的抑制有助于BCa的TAM抗性。
    Tamoxifen (TAM) resistance remains a major obstacle in the treatment of advanced breast cancer (BCa). In addition to the competitive inhibition of the estrogen receptor (ER) signaling pathway, damping of mitochondrial function by increasing reactive oxygen species (ROS) is critical for enhancing TAM pharmacodynamics. Here, we showed that RelB contributes to TAM resistance by inhibiting TAM-provoked ferroptosis. TAM-induced ROS level promoted ferroptosis in TAM-sensitive cells, but the effect was alleviated in TAM-resistant cells with high constitutive levels of RelB. Mechanistically, RelB inhibited ferroptosis by transcriptional upregulating glutathione peroxidase 4 (GPX4). Consequently, elevating RelB and GPX4 in sensitive cells increased TAM resistance, and conversely, depriving RelB and GPX4 in resistant cells decreased TAM resistance. Furthermore, suppression of RelB transcriptional activation resensitized TAM-resistant cells by enhancing ferroptosis in vitro and in vivo. The inactivation of GPX4 in TAM-resistant cells consistently resensitized TAM by increasing ferroptosis-mediated cell death. Together, this study uncovered that inhibition of ferroptosis contributes to TAM resistance of BCa via RelB-upregulated GPX4.
    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:实验性自身免疫性脑脊髓炎(EAE)是一种广泛使用的多发性硬化症小鼠模型。而不是诱导免疫反应,致耐受性树突状细胞(tDC)具有诱导免疫耐受的能力。在以往的研究中,我们通过1,25-(OH)2D3和1,25-(OH)2D3DCs诱导tDCs可显着缓解EAE症状。作为1,25-(OH)2D3的下游靶标,抑制DCs中RelB和MyD88的表达可能会诱导tDCs并具有MS的治疗作用。
    方法:用shRNA慢病毒敲除RelB和MyD88的表达以诱导tDC,将这些tDCs过继转移到EAE小鼠,并研究其治疗效果。
    结果:降低RelB表达诱导tDC。转移到EAE小鼠后,具有低RelB表达的tDCs可显着缓解其症状,并减少脊髓中的免疫细胞浸润和脱髓鞘。
    结论:RelB在DCs的抗原呈递功能中起关键作用,具有低RelB表达的tDC是EAE和MS的潜在治疗方法。
    Experimental autoimmune encephalomyelitis (EAE) is a widely used mouse model of multiple sclerosis. Rather than inducing immune response, tolerogenic dendritic cells (tDCs) have the ability to induce immune tolerance. In previous studies, we induced tDCs by 1,25-(OH)2D3 and 1,25-(OH)2D3 DCs significantly alleviated EAE symptoms. As downstream targets of 1,25-(OH)2D3, inhibition of RelB and MyD88 expression in DCs might induce tDCs and has therapeutic effect of MS.
    Knockdown the expression of RelB and MyD88 with shRNA lentivirus to induce tDCs, adoptive transfer these tDCs to EAE mice, and investigate their therapeutic effects.
    Reduction of RelB expression induced tDCs. After transferring into EAE mice, tDCs with low RelB expression significantly alleviate their symptoms as well as reduce the immune cell infiltration and demyelination in spinal cord.
    RelB plays a key role in the antigen presenting function of DCs, and tDCs with low RelB expression is a potential treatment for EAE and MS.
    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: Video-Audio Media
    背景:乳腺癌(BC)是全球女性中最常见的癌症。已知BC干细胞(BCSC)参与乳腺的癌变并有助于治疗抗性。BC的程序性死亡配体1(PD-L1)表达与不良预后相关。靶向PD-L1的免疫疗法具有巨大的潜力,并且在应用于癌症治疗时已经成功。然而,PD-L1是否调节BCSC的形成尚不清楚。
    方法:采用无血清悬浮培养法富集BCSCs。BCSCs的性质通过乳腺球形成试验进行检测,CD44+/Cd24-,醛脱氢酶(ALDH)测定,CSC标记分析,和乳腺球生长测定。为了阐明含溴结构域蛋白4(BRD4)的功能,BCSC的核PD-L1和RelB蛋白,使用BRD4抑制剂和降解剂检查乳腺球形成,PD-L1降解器,和RelB抑制剂。3'的抗肿瘤功能,4\',7,8-四羟基黄酮(THF),一种特定的BRD4抑制剂,通过体内肿瘤模型和小鼠研究,和c-Myc的蛋白质水平,在THF处理下在肿瘤模型中检查PD-L1和RelB。
    结果:BRD4在乳腺CSC中上调,调节BCs的干性。使用BRD4PROTAC下调BRD4,ARV-825和BRD4抑制剂,(+)-JQ1,抑制乳腺球形成并降低乳腺CSC标志物(CD44+/CD24-和ALDH1)的水平,干细胞标记基因,和乳腺球生长。BRD4抑制剂(JQ1)和降解剂(ARV825)通过抑制PD-L1转录物水平下调PD-L1的膜和核部分。PD-L1的敲低抑制乳腺球形成。Verteporfin,PD-L1降解器,抑制PD-L1的转录本和蛋白质水平,并下调RelB的转录本和蛋白质水平。骨化三醇,RelB抑制剂,使用si-RelB敲除RelB通过白细胞介素-6(IL-6)的表达调节乳腺球的形成。THF是一种天然产物,是一种有效的选择性BRD4抑制剂,抑制乳腺球的形成,并通过下调c-Myc来降低CD44+/CD24-和乳腺球的生长水平,PD-L1和RelB。3\',4\',在使用4T1和MC38细胞的鼠肿瘤模型中,7,8-THF显示肿瘤活性和肿瘤和肿瘤引流淋巴结(TDLN)中CD3+CD4+和CD3+CD8+T细胞水平增加。
    结论:结果显示BRD4/核PD-L1/RelB轴在乳腺CSC形成中的重要作用。核PD-L1调节RelB,和RelB/p65复合物诱导IL6和乳腺CSC形成。靶向核PD-L1代表了难治性BC免疫治疗的潜在和新颖工具。视频摘要。
    Breast cancer (BC) is the most common cancer diagnosed in women worldwide. BC stem cells (BCSCs) have been known to be involved in the carcinogenesis of the breast and contribute to therapeutic resistance. The programmed death-ligand 1 (PD-L1) expression of BC correlated with a poor prognosis. Immunotherapies that target PD-L1 have great potential and have been successful when applied to cancer treatment. However, whether PD-L1 regulates BCSC formation is unknown.
    BCSCs were enriched by serum-free suspension culture. The properties of BCSCs were examined by mammosphere formation assay, CD44+/Cd24-, aldehyde dehydrogenase (ALDH) assay, CSC marker analysis, and mammosphere growth assay. To elucidate the functions of bromodomain-containing protein 4 (BRD4), nuclear PD-L1, and RelB proteins in the stemness of BCSCs, mammosphere formation was examined using BRD4 inhibitor and degrader, PD-L1 degrader, and RelB inhibitor. The antitumor function of 3\',4\',7,8-tetrahydroxyflavone (THF), a specific BRD4 inhibitor, was studied through in vivo tumor model and mouse studies, and the protein levels of c-Myc, PD-L1, and RelB were examined in tumor model under THF treatment.
    BRD4 was upregulated in breast CSCs and regulates the stemness of BCs. The downregulation of BRD4 using BRD4 PROTAC, ARV-825, and BRD4 inhibitor, (+)-JQ1, inhibits mammosphere formation and reduces the levels of breast CSC markers (CD44+/CD24- and ALDH1), stem cell marker genes, and mammosphere growth. BRD4 inhibitor (JQ1) and degrader (ARV825) downregulate membrane and nuclear fractions of PD-L1 through the inhibition of PD-L1 transcript levels. The knockdown of PD-L1 inhibits mammosphere formation. Verteporfin, a PD-L1 degrader, inhibits the transcripts and protein levels of PD-L1 and downregulates the transcript and protein levels of RelB. Calcitriol, a RelB inhibitor, and the knockdown of RelB using si-RelB regulate mammosphere formation through interleukin-6 (IL-6) expression. THF is a natural product and a potent selective BRD4 inhibitor, inhibits mammosphere formation, and reduces the levels of CD44+/CD24- and mammosphere growth by downregulating c-Myc, PD-L1, and RelB. 3\',4\',7,8-THF shows tumoricidal activity and increased levels of CD3+CD4+ and CD3+CD8+ T-cells in the tumor and tumor-draining lymph nodes (TDLNs) in the murine tumor model using 4T1 and MC38 cells.
    The results show the first evidence of the essential role of the BRD4/nuclear PD-L1/RelB axis in breast CSC formation. The nuclear PD-L1 regulates RelB, and the RelB/p65 complex induces IL6 and breast CSC formation. Targeting nuclear PD-L1 represents a potential and novel tool for immunotherapies of intractable BC. Video Abstract.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号