c-FLIP

c - FLIP
  • 文章类型: Journal Article
    背景:凋亡调节缺陷是癌细胞的经典特征之一,通常与更积极的治疗选择和失败有关。我们研究了天然产物的组合抗肿瘤作用,physachenolideC(PCC)和硼替佐米,在KRASmut/P53mut肺癌细胞和异种移植小鼠模型中。
    方法:使用细胞活力研究了硼替佐米和PCC组合的体外抗癌作用,迁移,和344SQ的侵袭试验,H23和H358细胞系。此外,联合治疗对细胞代谢关键参数的影响,包括基于癌细胞耗氧率的细胞外酸化率(ECAR)和线粒体氧化磷酸化使用海马测定法进行评估。最后,使用异种移植小鼠模型评价硼替佐米(1mg/kg)和PCC(10mg/kg)组合的抗肿瘤作用.
    结果:我们的数据表明,与单独治疗相比,硼替佐米-PCC组合在降低肺癌细胞活力方面更有效。同样,联合治疗对癌细胞的迁移和侵袭有显著的抑制作用。此外,关键的抗凋亡蛋白c-FLIP被显著抑制,同时癌细胞中细胞代谢的关键参数显著降低.值得注意的是,与对照组相比,硼替佐米或PCC抑制肿瘤生长,在异种移植瘤小鼠模型中,硼替佐米联合PCC对肿瘤生长的抑制作用更为有效.
    结论:这些发现表明PCC使癌细胞对硼替佐米敏感,有可能改善对KRASmut/P53mut肺癌细胞的抗肿瘤作用,具有增强的联合治疗的功效而不引起显著的副作用。
    BACKGROUND: Defects in apoptosis regulation are one of the classical features of cancer cells, often associated with more aggressiveness and failure to therapeutic options. We investigated the combinatorial antitumor effects of a natural product, physachenolide C (PCC) and bortezomib, in KRASmut/P53mut lung cancer cells and xenograft mice models.
    METHODS: The in vitro anticancer effects of the bortezomib and PCC combination were investigated using cell viability, migration, and invasion assays in 344SQ, H23, and H358 cell lines. Furthermore, the effects of combination treatment on the critical parameters of cellular metabolism, including extracellular acidification rate (ECAR) and mitochondrial oxidative phosphorylation based on the oxygen consumption rate of cancer cells were assessed using Seahorse assay. Finally, the antitumor effect of the bortezomib (1 mg/kg) and PCC (10 mg/kg) combination was evaluated using xenograft mice models.
    RESULTS: Our data showed that the bortezomib-PCC combination was more effective in reducing the viability of lung cancer cells in comparison with the individual treatments. Similarly, the combination treatment showed a significant inhibition of cell migration and invasion of cancer cells. Additionally, the key anti-apoptotic protein c-FLIP was significantly inhibited along with a substantial reduction in the key parameters of cellular metabolism in cancer cells. Notably, the bortezomib or PCC inhibited the tumor growth compared to the control group, the tumor growth inhibition was much more effective when bortezomib was combined with PCC in tumor xenograft mice models.
    CONCLUSIONS: These findings demonstrate that PCC sensitizes cancer cells to bortezomib, potentially improving the antitumor effects against KRASmut/P53mut lung cancer cells, with an enhanced efficacy of combination treatments without causing significant side effects.
    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
    催化失活的caspase-8同源蛋白,c-FLIP,是一种有效的抗凋亡蛋白,在各种类型的癌症中高度表达。在通过TNF-R家族的配体激活死亡受体(DR)后,c-FLIP与caspase-8竞争结合衔接蛋白FADD(Fas-相关死亡结构域)。因此,涉及DR的外源性凋亡信号通路被抑制.肿瘤细胞中c-FLIP活性的抑制可能会增强DR介导的凋亡并克服免疫和抗癌药物的耐药性。基于计算机模拟方法,这项工作的目的是鉴定能够选择性结合c-FLIP并阻断其抗凋亡活性的新的小抑制分子.使用c-FLIP的同源3D模型,对来自NCI数据库(国家癌症研究所)的1880种化合物进行了计算机筛选。选择了九种分子进行体外测定,基于它们对c-FLIP的结合亲和力和与caspase-8相比的高选择性。这些分子选择性结合c-FLIP的死亡效应物结构域2(DED2)。我们已经使用过表达c-FLIP的人肺癌细胞系H1703在体外测试了这9种分子的抑制作用。我们的结果表明,这些新鉴定的化合物中的六个在分子下拉法中有效地防止了FADD/c-FLIP相互作用,以及在DISC免疫沉淀测定中。c-FLIP在H1703中的过表达阻止TRAIL介导的细胞凋亡;然而,TRAIL与这些选择的分子的组合通过挽救半胱天冬酶裂解和活化显著恢复TRAIL诱导的细胞死亡。总之,我们的发现表明,新的抑制性化学分子有效地防止c-FLIP募集到DISC复合物中,从而恢复caspase-8依赖性凋亡级联反应。这些结果为设计新的c-FLIP抑制分子铺平了道路,这些分子可以在过表达c-FLIP的肿瘤中用作抗癌剂。
    The catalytically inactive caspase-8-homologous protein, c-FLIP, is a potent antiapoptotic protein highly expressed in various types of cancers. c-FLIP competes with caspase-8 for binding to the adaptor protein FADD (Fas-Associated Death Domain) following death receptors\' (DRs) activation via the ligands of the TNF-R family. As a consequence, the extrinsic apoptotic signaling pathway involving DRs is inhibited. The inhibition of c-FLIP activity in tumor cells might enhance DR-mediated apoptosis and overcome immune and anticancer drug resistance. Based on an in silico approach, the aim of this work was to identify new small inhibitory molecules able to bind selectively to c-FLIP and block its anti-apoptotic activity. Using a homology 3D model of c-FLIP, an in silico screening of 1880 compounds from the NCI database (National Cancer Institute) was performed. Nine molecules were selected for in vitro assays, based on their binding affinity to c-FLIP and their high selectivity compared to caspase-8. These molecules selectively bind to the Death Effector Domain 2 (DED2) of c-FLIP. We have tested in vitro the inhibitory effect of these nine molecules using the human lung cancer cell line H1703, overexpressing c-FLIP. Our results showed that six of these newly identified compounds efficiently prevent FADD/c-FLIP interactions in a molecular pull-down assay, as well as in a DISC immunoprecipitation assay. The overexpression of c-FLIP in H1703 prevents TRAIL-mediated apoptosis; however, a combination of TRAIL with these selected molecules significantly restored TRAIL-induced cell death by rescuing caspase cleavage and activation. Altogether, our findings indicate that new inhibitory chemical molecules efficiently prevent c-FLIP recruitment into the DISC complex, thus restoring the caspase-8-dependent apoptotic cascade. These results pave the way to design new c-FLIP inhibitory molecules that may serve as anticancer agents in tumors overexpressing c-FLIP.
    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
    肿瘤坏死因子相关的凋亡诱导配体(TRAIL)是一种有前途的抗癌药物,因为它选择性地诱导转化细胞的凋亡,而不改变健康细胞的细胞机制。不幸的是,TRAIL抗性机制在多种癌症类型中的存在代表了一个主要障碍,因此限制了TRAIL作为单一药物的使用。越来越多的研究表明,TRAIL介导的细胞凋亡可以通过与抗肿瘤药物联合治疗促进耐药肿瘤。从合成分子到天然产物。在后者中,黄酮类化合物,植物中最普遍的多酚,在抗性细胞系以及具有最小副作用的荷瘤小鼠中显示出改善TRAIL驱动的凋亡的显着能力。这里,我们总结了分子机制,例如死亡受体(DR)4和DR5的上调和关键抗凋亡蛋白的下调[例如,细胞FLICE抑制蛋白(c-FLIP),X-连锁凋亡抑制蛋白(XIAP),survivin],不同类别类黄酮的TRAIL敏化特性(例如,黄酮,黄酮醇,异黄酮,查尔酮,异戊二烯类黄酮)。最后,我们讨论局限性,主要与生物利用度问题有关,以及关于黄酮类化合物在TRAIL治疗中作为佐剂的临床应用的未来观点。
    Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) represents a promising anticancer agent, as it selectively induces apoptosis in transformed cells without altering the cellular machinery of healthy cells. Unfortunately, the presence of TRAIL resistance mechanisms in a variety of cancer types represents a major hurdle, thus limiting the use of TRAIL as a single agent. Accumulating studies have shown that TRAIL-mediated apoptosis can be facilitated in resistant tumors by combined treatment with antitumor agents, ranging from synthetic molecules to natural products. Among the latter, flavonoids, the most prevalent polyphenols in plants, have shown remarkable competence in improving TRAIL-driven apoptosis in resistant cell lines as well as tumor-bearing mice with minimal side effects. Here, we summarize the molecular mechanisms, such as the upregulation of death receptor (DR)4 and DR5 and downregulation of key anti-apoptotic proteins [e.g., cellular FLICE-inhibitory protein (c-FLIP), X-linked inhibitor of apoptosis protein (XIAP), survivin], underlying the TRAIL-sensitizing properties of different classes of flavonoids (e.g., flavones, flavonols, isoflavones, chalcones, prenylflavonoids). Finally, we discuss limitations, mainly related to bioavailability issues, and future perspectives regarding the clinical use of flavonoids as adjuvant agents in TRAIL-based therapies.
    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
    背景:转移性三阴性乳腺癌(mTNBC)患者在初次诊断后5年内发生内脏转移的可能性更高。因此,迫切需要对mTNBC的进展和传播有更深入的了解。
    方法:基于相对和绝对定量(iTRAQ)的LC-MS/MS蛋白质组学方法的等量异位标签用于鉴定肺向转移性细胞中的新型膜相关蛋白。公共领域数据集用于评估候选蛋白的临床相关性。基于细胞的模型和小鼠模型用于通过iTRAQ鉴定的蛋白质分子Sciellin(SCEL)的生物化学和功能表征,以阐明其在促进TNBC细胞的肺定植中的作用和潜在机制。
    结果:基于iTRAQ的LC-MS/MS蛋白质组学方法鉴定了一种在嗜肺转移细胞中过表达的膜相关蛋白SCEL,其高表达与晚期TNBC和患者生存期短显著相关。SCEL表达的下调显着损害了3D集落形成能力,但不损害肺定植(LC)细胞的迁移和侵袭能力。SCEL的敲低降低了TNF-α诱导的LC细胞中NF-κB/c-FLIP促存活和Akt/Erk1/2生长信号通路的激活。具体来说,SCEL表达的敲低将TNF-α介导的细胞存活转变为caspase3依赖性细胞凋亡。相反,SCEL的异位表达可促进TNF-α诱导的NF-κB/c-FLIP促存活和Akt/Erk1/2促生长信号通路的激活。免疫共沉淀(Co-IP)和GST下拉实验结果表明,SCEL可以与TNFR1相互作用以提高其蛋白稳定性。异种移植小鼠模型实验表明,SCEL敲低导致caspase-3活性增加,在转移性肺病变中ki67和TNFR1表达减少以及肿瘤相关巨噬细胞增加。临床上,在TNBC组织中发现SCEL表达与TNFR1呈正相关。最后,我们表明,通过阿达木单抗阻断TNF-α介导的细胞存活信号可有效抑制SCEL阳性的肺定植,而不是尾静脉注射模型中SCEL下调的LC细胞。
    结论:我们的研究结果表明,SCEL通过促进TNF-α/TNFR1/NF-κB/c-FLIP的存活和Akt/Erk1/2信号的增殖在TNBC的转移性肺定植中起重要作用。因此,SCEL可作为阿达木单抗治疗TNBC患者的生物标志物。
    BACKGROUND: Patients with metastatic triple-negative breast cancer (mTNBC) have a higher probability of developing visceral metastasis within 5 years after the initial diagnosis. Therefore, a deeper understanding of the progression and spread of mTNBC is urgently needed.
    METHODS: The isobaric tag for relative and absolute quantitation (iTRAQ)-based LC-MS/MS proteomic approach was applied to identify novel membrane-associated proteins in the lung-tropic metastatic cells. Public domain datasets were used to assess the clinical relevance of the candidate proteins. Cell-based and mouse models were used for biochemical and functional characterization of the protein molecule Sciellin (SCEL) identified by iTRAQ to elucidate its role and underlying mechanism in promoting lung colonization of TNBC cells.
    RESULTS: The iTRAQ-based LC-MS/MS proteomic approach identified a membrane-associated protein SCEL that was overexpressed in the lung-tropic metastatic cells, and its high expression was significantly correlated with the late-stage TNBC and the shorter survival of the patients. Downregulation of SCEL expression significantly impaired the 3D colony-forming ability but not the migration and invasion ability of the lung colonization (LC) cells. Knockdown of SCEL reduced TNF-α-induced activation of the NF-κB/c-FLIP pro-survival and Akt/Erk1/2 growth signaling pathways in the LC cells. Specifically, knockdown of SCEL expression switched TNF-α-mediated cell survival to the caspase 3-dependent apoptosis. Conversely, ectopic expression of SCEL promoted TNF-α-induced activation of NF-κB/c-FLIP pro-survival and Akt/Erk1/2 pro-growth signaling pathway. The result of co-immunoprecipitation (Co-IP) and GST pull-down assay showed that SCEL could interact with TNFR1 to promote its protein stability. The xenograft mouse model experiments revealed that knockdown of SCEL resulted in increase of caspase-3 activity, and decrease of ki67 and TNFR1 expression as well as increase of tumor-associated macrophages in the metastatic lung lesions. Clinically, SCEL expression was found to be positively correlated with TNFR1 in TNBC tissues. Lastly, we showed that blocking TNF-α-mediated cell survival signaling by adalimumab effectively suppressed the lung colonization of the SCEL-positive, but not the SCEL-downregulated LC cells in the tail-vein injection model.
    CONCLUSIONS: Our findings indicate that SCEL plays an essential role in the metastatic lung colonization of TNBC by promoting the TNF-α/TNFR1/NF-κB/c-FLIP survival and Akt/Erk1/2 proliferation signaling. Thus, SCEL may serve as a biomarker for adalimumab treatment of TNBC 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
    土贝莫塞德-1(TBMS-1),一种传统的中草药,通常用作抗癌剂。在这项研究中,我们旨在研究其对肿瘤坏死因子相关凋亡诱导配体(TRAIL)致敏作用.我们的结果表明,即使使用亚致死浓度的TBMS-1或TRAIL的单一疗法也不会影响癌细胞死亡,使用TBMS-1和TRAIL的联合治疗增加了凋亡性细胞死亡。机械上,TBMS-1通过下调STAMBPL1(一种去泛素酶(DUB))使c-FLIP表达不稳定。具体来说,当STAMBPL1和c-FLIP绑定在一起时,STAMBPL1去泛素化c-FLIP。此外,STAMBPL1敲低通过使c-FLIP不稳定而显着增加对TRAIL的敏感性。基于用TBMS-1和TRAIL组合治疗减少肿瘤体积并下调STAMBPL1和c-FLIP表达水平的观察,使用异种移植模型在体内进一步证实了这些发现。总的来说,我们的研究表明,STAMBPL1对c-FLIP稳定至关重要,并且STAMBPL1耗竭通过c-FLIP下调增强TRAIL介导的细胞凋亡。
    Tubeimoside-1 (TBMS-1), a traditional Chinese medicinal herb, is commonly used as an anti-cancer agent. In this study, we aimed to investigate its effect on the sensitization of cancer cells to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Our results revealed that even though monotherapy using TBMS-1 or TRAIL at sublethal concentrations did not affect cancer cell death, combination therapy using TBMS-1 and TRAIL increased apoptotic cell death. Mechanistically, TBMS-1 destabilized c-FLIP expression by downregulating STAMBPL1, a deubiquitinase (DUB). Specifically, when STAMBPL1 and c-FLIP bound together, STAMBPL1 deubiquitylated c-FLIP. Moreover, STAMBPL1 knockdown markedly increased sensitivity to TRAIL by destabilizing c-FLIP. These findings were further confirmed in vivo using a xenograft model based on the observation that combined treatment with TBMS-1 and TRAIL decreased tumor volume and downregulated STAMBPL1 and c-FLIP expression levels. Overall, our study revealed that STAMBPL1 is essential for c-FLIP stabilization, and that STAMBPL1 depletion enhances TRAIL-mediated apoptosis via c-FLIP downregulation.
    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
    肠上皮细胞(IEC)过度凋亡是破坏上皮稳态的关键原因,导致溃疡性结肠炎(UC)的发病机制。武田G蛋白偶联受体5(TGR5)在IEC细胞凋亡中的调控作用及其分子机制尚不清楚。而且还缺乏选择性TGR5激动剂治疗UC的直接证据.这里,我们合成了一种在肠道中高度分布的有效和选择性的TGR5激动剂OM8,并研究了其对IEC凋亡和UC治疗的影响。我们发现OM8有效激活hTGR5和mTGR5,EC50值为202±55nM和74±17nM,分别。口服后,大量的OM8维持在肠道中,血液吸收非常低。在DSS诱导的结肠炎小鼠中,口服OM8减轻结肠炎症状,病理变化和紧密连接蛋白表达受损。除了增强肠干细胞(ISC)的增殖和分化,OM8给药显著降低结肠炎小鼠结肠上皮细胞凋亡率。在体外HT-29和Caco-2细胞中进一步证明了OM8对IEC凋亡的直接抑制作用。在HT-29细胞中,我们证明沉默TGR5,抑制腺苷酸环化酶或蛋白激酶A(PKA)均可阻断OM8诱导的JNK磷酸化抑制,从而消除其对TNF-α诱导的凋亡的拮抗作用。提示OM8对IEC凋亡的抑制作用是通过激活TGR5和cAMP/PKA信号通路介导的。进一步的研究表明,OM8在HT-29细胞中以TGR5依赖性方式上调细胞FLICE抑制蛋白(c-FLIP)的表达。敲除c-FLIP阻断OM8对TNF-α诱导的JNK磷酸化和凋亡的抑制作用,提示c-FLIP对于抑制OM8诱导的IEC凋亡是必不可少的。总之,我们的研究证明了TGR5激动剂在体外通过cAMP/PKA/c-FLIP/JNK信号通路抑制IEC凋亡的新机制,并强调了TGR5激动剂作为治疗UC的新治疗策略的价值。
    Excessive apoptosis of intestinal epithelial cell (IEC) is a crucial cause of disrupted epithelium homeostasis, leading to the pathogenesis of ulcerative colitis (UC). The regulation of Takeda G protein-coupled receptor-5 (TGR5) in IEC apoptosis and the underlying molecular mechanisms remained unclear, and the direct evidence from selective TGR5 agonists for the treatment of UC is also lacking. Here, we synthesized a potent and selective TGR5 agonist OM8 with high distribution in intestinal tract and investigated its effect on IEC apoptosis and UC treatment. We showed that OM8 potently activated hTGR5 and mTGR5 with EC50 values of 202 ± 55 nM and 74 ± 17 nM, respectively. After oral administration, a large amount of OM8 was maintained in intestinal tract with very low absorption into the blood. In DSS-induced colitis mice, oral administration of OM8 alleviated colitis symptoms, pathological changes and impaired tight junction proteins expression. In addition to enhancing intestinal stem cell (ISC) proliferation and differentiation, OM8 administration significantly reduced the rate of apoptotic cells in colonic epithelium in colitis mice. The direct inhibition by OM8 on IEC apoptosis was further demonstrated in HT-29 and Caco-2 cells in vitro. In HT-29 cells, we demonstrated that silencing TGR5, inhibition of adenylate cyclase or protein kinase A (PKA) all blocked the suppression of JNK phosphorylation induced by OM8, thus abolished its antagonizing effect against TNF-α induced apoptosis, suggesting that the inhibition by OM8 on IEC apoptosis was mediated via activation of TGR5 and cAMP/PKA signaling pathway. Further studies showed that OM8 upregulated cellular FLICE-inhibitory protein (c-FLIP) expression in a TGR5-dependent manner in HT-29 cells. Knockdown of c-FLIP blocked the inhibition by OM8 on TNF-α induced JNK phosphorylation and apoptosis, suggesting that c-FLIP was indispensable for the suppression of OM8 on IEC apoptosis induced by OM8. In conclusion, our study demonstrated a new mechanism of TGR5 agonist on inhibiting IEC apoptosis via cAMP/PKA/c-FLIP/JNK signaling pathway in vitro, and highlighted the value of TGR5 agonist as a novel therapeutic strategy for the treatment of UC.
    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
    对抗癌剂的抗性和细胞凋亡导致癌症复发并且与癌症死亡率相关。大量数据提供了令人信服的证据,证明人类癌症是由癌症干细胞(CSC)产生的。表现出自我更新并对抗癌药物有抗性,辐射,和细胞凋亡,并表达增强的上皮向间充质的进展。CSC代表异质性肿瘤细胞群,缺乏特异性细胞靶标,这使得瞄准和根除它们成为一个巨大的挑战。同样,它们与肿瘤微环境的密切关系在开发靶向CSC的新型治疗策略时产生了更大的复杂性.多种机制参与各种癌症中CSC的药物和凋亡抵抗表型。这些包括ATP结合盒膜转运蛋白的表达增强,各种细胞保护和存活信号通路的激活,干性信号通路的失调,异常的DNA修复机制,增加的静止,自噬,增加免疫逃避,线粒体介导的细胞凋亡缺乏,上调抗凋亡蛋白,包括c-FLIP[细胞FLICE(FADD样IL-1β转换酶)-抑制蛋白],Bcl-2家族成员,凋亡蛋白抑制剂,和PI3K/AKT信号。研究这些机制不仅提供了对药物无反应的这些细胞的机械见解,但可能导致开发靶向和有效的治疗方法来根除CSC。一些研究已经确定了靶向CSC的有希望的策略。这些新出现的策略可能有助于在临床环境中靶向CSC相关的耐药性和转移。本文将对CSCs的药物和凋亡抵抗机制以及如何靶向CSCs进行综述。
    Resistance to anticancer agents and apoptosis results in cancer relapse and is associated with cancer mortality. Substantial data have provided convincing evidence establishing that human cancers emerge from cancer stem cells (CSCs), which display self-renewal and are resistant to anticancer drugs, radiation, and apoptosis, and express enhanced epithelial to mesenchymal progression. CSCs represent a heterogeneous tumor cell population and lack specific cellular targets, which makes it a great challenge to target and eradicate them. Similarly, their close relationship with the tumor microenvironment creates greater complexity in developing novel treatment strategies targeting CSCs. Several mechanisms participate in the drug and apoptosis resistance phenotype in CSCs in various cancers. These include enhanced expression of ATP-binding cassette membrane transporters, activation of various cytoprotective and survival signaling pathways, dysregulation of stemness signaling pathways, aberrant DNA repair mechanisms, increased quiescence, autophagy, increased immune evasion, deficiency of mitochondrial-mediated apoptosis, upregulation of anti-apoptotic proteins including c-FLIP [cellular FLICE (FADD-like IL-1β-converting enzyme)-inhibitory protein], Bcl-2 family members, inhibitors of apoptosis proteins, and PI3K/AKT signaling. Studying such mechanisms not only provides mechanistic insights into these cells that are unresponsive to drugs, but may lead to the development of targeted and effective therapeutics to eradicate CSCs. Several studies have identified promising strategies to target CSCs. These emerging strategies may help target CSC-associated drug resistance and metastasis in clinical settings. This article will review the CSCs drug and apoptosis resistance mechanisms and how to target CSCs.
    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
    叉头盒M1(FoxM1)蛋白,转录因子,在调节肿瘤生长和耐药性方面发挥着关键作用,而细胞FLICE抑制蛋白(c-FLIP),抗凋亡调节剂,参与泛素-蛋白酶体途径。在这项研究中,我们研究了c-FLIP对非小细胞肺癌(NSCLC)细胞中FoxM1的表达和泛素化水平以及药物敏感性的影响.我们首先发现FoxM1和c-FLIP在90例NSCLC样本中的表达水平升高并呈正相关(R2=0.1106,P<0.0001)。预后分析的生存数据表明,c-FLIP和/或FoxM1的高表达与NSCLC患者的不良预后有关,并且FoxM1和c-FLIP的组合可能是比单独使用更精确的预后生物标志物。然后,我们探讨了c-FLIP/FoxM1在NSCLC细胞系和异种移植小鼠体内耐药中的功能。我们发现c-FLIP通过抑制FoxM1的泛素化来稳定FoxM1,从而在转录后水平上调FoxM1的表达。此外,由FoxM1、β-catenin和p65组成的正反馈环也参与了c-FLIP-FoxM1轴。我们发现c-FLIP通过上调FoxM1促进NSCLC细胞对硫链菌素和奥希替尼的耐药性。一起来看,这些结果揭示了c-FLIP调控FoxM1的新机制以及这种相互作用在硫链菌素和奥希替尼耐药发展中的作用.这项研究为靶向c-FLIP-FoxM1轴治疗肺癌的潜在治疗益处提供了实验证据。
    The forkhead box M1 (FoxM1) protein, a transcription factor, plays critical roles in regulating tumor growth and drug resistance, while cellular FLICE-inhibitory protein (c-FLIP), an anti-apoptotic regulator, is involved in the ubiquitin-proteasome pathway. In this study, we investigated the effects of c-FLIP on the expression and ubiquitination levels of FoxM1 along with drug susceptibility in non-small-cell lung cancer (NSCLC) cells. We first showed that the expression levels of FoxM1 and c-FLIP were increased and positively correlated (R2 = 0.1106, P < 0.0001) in 90 NSCLC samples. The survival data from prognostic analysis demonstrated that high expression of c-FLIP and/or FoxM1 was related to poor prognosis in NSCLC patients and that the combination of FoxM1 and c-FLIP could be a more precise prognostic biomarker than either alone. Then, we explored the functions of c-FLIP/FoxM1 in drug resistance in NSCLC cell lines and a xenograft mouse model in vivo. We showed that c-FLIP stabilized FoxM1 by inhibiting its ubiquitination, thus upregulated the expression of FoxM1 at post-transcriptional level. In addition, a positive feedback loop composed of FoxM1, β-catenin and p65 also participated in c-FLIP-FoxM1 axis. We revealed that c-FLIP promoted the resistance of NSCLC cells to thiostrepton and osimertinib by upregulating FoxM1. Taken together, these results reveal a new mechanism by which c-FLIP regulates FoxM1 and the function of this interaction in the development of thiostrepton and osimertinib resistance. This study provides experimental evidence for the potential therapeutic benefit of targeting the c-FLIP-FoxM1 axis for lung cancer 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
    人类癌症来自癌症干细胞(CSCs),对癌症化疗药物有抗性,辐射,细胞死亡。此外,自噬提供细胞保护作用,有助于这些细胞的耐药性。此外,许多证据表明,CSC导致肿瘤的发生,programming,转移,和癌症复发。各种信号通路,包括磷脂酰肌醇3-激酶(PI3K)/Akt/哺乳动物雷帕霉素靶蛋白(mTOR),母体胚胎亮氨酸拉链激酶(MELK),NOTCH1和Wnt/β-连环蛋白以及CSC标记保持CSC特性。几种机制,包括ABC多药耐药转运蛋白的过表达,线粒体介导的细胞凋亡缺乏,c-FLIP的上调,抗凋亡Bcl-2家族成员和凋亡蛋白(IAP)抑制剂的过表达,和PI3K/AKT信号传导有助于增强各种癌症中CSC对化疗药物的抗性和细胞死亡诱导。研究这些途径可能有助于详细了解CSC对化学治疗剂的抗性和细胞凋亡的机制,并可能导致开发有效的治疗方法来根除CSC。
    Human cancers emerge from cancer stem cells (CSCs), which are resistant to cancer chemotherapeutic agents, radiation, and cell death. Moreover, autophagy provides the cytoprotective effect which contributes to drug resistance in these cells. Furthermore, much evidence shows that CSCs cause tumor initiation, progression, metastasis, and cancer recurrence. Various signaling pathways including the phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR), maternal embryonic leucine zipper kinase (MELK), NOTCH1, and Wnt/β-catenin as well as the CSC markers maintain CSC properties. Several mechanisms including overexpression of ABC multidrug resistance transporters, a deficiency in mitochondrial-mediated apoptosis, upregulation of c-FLIP, overexpression of anti-apoptotic Bcl-2 family members and inhibitors of apoptosis proteins (IAPs), and PI3K/AKT signaling contribute to enhancing resistance to chemotherapeutic drugs and cell death induction in CSCs in various cancers. Studying such pathways may help provide detailed understanding of CSC mechanisms of resistance to chemotherapeutic agents and apoptosis and may lead to the development of effective therapeutics to eradicate CSCs.
    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
    TNF刺激通过NF-κB的激活产生促存活信号,所述NF-κB限制由TNF触发的内建死亡信号传导。TNF诱导的存活和死亡信号之间的竞争最终决定了细胞的命运。这里,我们报道了Bclaf1作为TNF抗凋亡程序的新成分的鉴定。多个细胞中的Bclaf1耗竭使细胞对TNF诱导的凋亡敏感,但对坏死性凋亡不敏感。Bclaf1通过促进CFLAR的转录发挥其抗凋亡功能,caspase8拮抗剂,NF-κB激活的下游。Bclaf1与NF-κB的p50亚基结合,这是Bclaf1刺激CFLAR转录所必需的。最后,在Bclaf1siRNA给药小鼠中,TNF诱导的小肠损伤比具有加重的凋亡和焦亡迹象的对照小鼠严重得多。这些结果表明Bclaf1是TNF诱导的细胞凋亡的关键调节因子。在体外和体内。
    TNF stimulation generates pro-survival signals through activation of NF-κB that restrict the build-in death signaling triggered by TNF. The competition between TNF-induced survival and death signals ultimately determines the fate of a cell. Here, we report the identification of Bclaf1 as a novel component of the anti-apoptotic program of TNF. Bclaf1 depletion in multiple cells sensitizes cells to TNF-induced apoptosis but not to necroptosis. Bclaf1 exerts its anti-apoptotic function by promoting the transcription of CFLAR, a caspase 8 antagonist, downstream of NF-κB activation. Bclaf1 binds to the p50 subunit of NF-κB, which is required for Bclaf1 to stimulate CFLAR transcription. Finally, in Bclaf1 siRNA administered mice, TNF-induced small intestine injury is much more severe than in control mice with aggravated signs of apoptosis and pyroptosis. These results suggest Bclaf1 is a key regulator in TNF-induced apoptosis, both in vitro and in vivo.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号