c-FLIP

c - FLIP
  • 文章类型: Journal Article
    三种色霉素衍生物,色霉素A3(1,CA3),A5(2,CA5),和单脱乙酰色霉素A3(3,MDA-CA3),从土壤衍生的链霉菌中鉴定出。CGMCC26516。报告了对CA5结构的重新研究,根据核磁共振(NMR)数据分析以及NMR和电子圆二色性计算,首次明确确定其绝对构型与CA3相同。化合物1-3对非小细胞肺癌(NSCLC)细胞(A549,H460,H157-c-FLIP,和H157-LacZ),并下调A549细胞中c-FLIP的蛋白表达。H157-c-FLIP中的色霉素的IC50值高于H157-LacZ。此外,si-c-FLIP对非小细胞肺癌细胞中的色霉素抗增殖作用。在裸鼠异种移植模型中,图1和2对H157-lacZ异种移植物的生长均显示出比H157-c-FLIP异种移植物更有效的抑制。这些结果验证了c-FLIP介导了在NSCLC中的色霉素的抗癌作用。
    Three chromomycin derivatives, chromomycins A3 (1, CA3), A5 (2, CA5), and monodeacetylchromomycin A3 (3, MDA-CA3), were identified from the soil-derived Streptomyces sp. CGMCC 26516. A reinvestigation of the structure of CA5 is reported, of which the absolute configuration was unambiguously determined for the first time to be identical with that of CA3 based on nuclear magnetic resonance (NMR) data analysis as well as NMR and electronic circular dichroism calculations. Compounds 1-3 showed potent cytotoxicity against the non-small-cell lung cancer (NSCLC) cells (A549, H460, H157-c-FLIP, and H157-LacZ) and down-regulated the protein expression of c-FLIP in A549 cells. The IC50 values of chromomycins in H157-c-FLIP were higher than that in H157-LacZ. Furthermore, si-c-FLIP promoted anti-proliferation effect of chromomycins in NSCLC cells. In nude mice xenograft model, 1 and 2 both showed more potent inhibition on the growth of H157-lacZ xenografts than that of H157-c-FLIP xenografts. These results verify that c-FLIP mediates the anticancer effects of chromomycins in NSCLC.
    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
    三阴性乳腺癌(TNBC)是一种侵袭性亚型,预后不良。硫链菌素对包括TNBC在内的几种癌症具有抗肿瘤活性。本文讨论了硫链菌素在TNBC中的新分子机制。硫链菌素抑制MDA-MB-231细胞活力,伴随着c-FLIP和p-SMAD2/3的减少。c-FLIP过表达降低MDA-MB-231细胞对硫链菌素的敏感性,SMAD2/3敲除增加MDA-MB-231细胞对硫链菌素的敏感性。此外,c-FLIP过表达显著增加SMAD2/3蛋白的表达和磷酸化,反之亦然。总之,我们的研究揭示了c-FLIP/SMAD2/3信号通路是硫链菌素抗肿瘤活性的新机制。
    Triple-negative breast cancer (TNBC) is an aggressive subtype with poor prognosis of breast cancer. Thiostrepton exerts anti-tumor activities against several cancers including TNBC. Herein we discussed the new molecular mechanisms of thiostrepton in TNBC. Thiostrepton inhibited MDA-MB-231 cell viability, accompanied by a decrease of c-FLIP and p-SMAD2/3. c-FLIP overexpression reduced the sensitivity of MDA-MB-231 cells to thiostrepton, while SMAD2/3 knockdown increased the sensitivity of MDA-MB-231 cells to thiostrepton. Moreover, c-FLIP overexpression significantly increased the expression and phosphorylation of SMAD2/3 proteins and vice versa. In conclusion, our study reveals c-FLIP/SMAD2/3 signaling pathway as a novel mechanism of antitumor activity of thiostrepton.
    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
    转移性肺腺癌(LuAC)由于潜伏期短和缺乏有效的治疗选择而提出了重大的临床挑战。因此,识别转移性LuAC中的分子脆弱性对于开发针对该疾病的治疗药物非常重要。在这项研究中,我们使用低脑转移和高度脑转移的LuAC细胞系进行了全基因组siRNA筛选.使用这种方法,我们发现,与低转移性LuAC(LuAC-Par)细胞相比,脑转移LuAC(LuAC-BrM)细胞对c-FLIP(caspase-8的抑制剂)耗竭诱导的凋亡表现出更高的脆弱性。此外,体内研究表明,c-FLIP敲除特异性抑制LuAC-BrM的生长,但不是LuAC-Par,肿瘤,表明LuAC-BrM对c-FLIP的存活功能成瘾。我们的体外和体内分析还表明,由于ER应激诱导的c-JUN激活以及随后诱导的包括ATF4和DDIT3的应激基因,LuAC-BrM对c-FLIP消耗更敏感。最后,我们发现c-JUN不仅使LuAC-BrM对c-FLIP耗竭诱导的细胞死亡敏感,而且在体内促进脑转移,为c-JUN在LuAC-BrM中作为一把双刃剑的功能提供了有力的证据。总的来说,我们的发现不仅揭示了C-JUN之间的一种新颖的联系,脑转移瘤,LuAC-BrM中的c-FLIP成瘾,但也为潜在的治疗干预提供了机会。
    Metastatic lung adenocarcinoma (LuAC) presents a significant clinical challenge due to the short latency and the lack of efficient treatment options. Therefore, identification of molecular vulnerabilities in metastatic LuAC holds great importance in the development of therapeutic drugs against this disease. In this study, we performed a genome-wide siRNA screening using poorly and highly brain-metastatic LuAC cell lines. Using this approach, we discovered that compared to poorly metastatic LuAC (LuAC-Par) cells, brain-metastatic LuAC (LuAC-BrM) cells exhibited a significantly higher vulnerability to c-FLIP (an inhibitor of caspase-8)-depletion-induced apoptosis. Furthermore, in vivo studies demonstrated that c-FLIP knockdown specifically inhibited growth of LuAC-BrM, but not the LuAC-Par, tumors, suggesting the addiction of LuAC-BrM to the function of c-FLIP for their survival. Our in vitro and in vivo analyses also demonstrated that LuAC-BrM is more sensitive to c-FLIP-depletion due to ER stress-induced activation of the c-JUN and subsequent induction of stress genes including ATF4 and DDIT3. Finally, we found that c-JUN not only sensitized LuAC-BrM to c-FLIP-depletion-induced cell death but also promoted brain metastasis in vivo, providing strong evidence for c-JUN\'s function as a double-edged sword in LuAC-BrM. Collectively, our findings not only reveal a novel link between c-JUN, brain metastasis, and c-FLIP addiction in LuAC-BrM but also present an opportunity for potential therapeutic intervention.
    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: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
    目的:进一步研究肿瘤坏死因子相关凋亡诱导配体(TRAIL)在非小细胞肺癌中的作用机制,将有助于对TRAIL耐药的认识,并为制定相应的治疗策略提供新的线索。
    背景:细胞FLICE抑制蛋白(c-FLIP)是TRAIL诱导的细胞凋亡的关键抑制剂。我们先前的研究表明,糖原合酶激酶3β(GSK3β)正调节人肺腺癌细胞中c-FLIP的表达。同时,其他研究报道,c-FLIP通过蛋白酶体途径被HECT型E3连接酶ITCH(发痒的E3泛素蛋白连接酶)降解。
    目的:我们将探讨ITCH在TRAIL治疗过程中是否参与GSK3β阳性控制的c-FLIP的表达调控。
    方法:使用人肺腺癌细胞稳定过表达和敲低GSK3β。采用实时定量PCR(qRT-PCR)检测基因mRNA的表达水平。蛋白质印迹分析用于检测蛋白质水平的蛋白质表达。ITCH的siRNA用于敲低其表达。TRAIL处理用于引起细胞凋亡。
    结果:在本研究中,我们已经证实了肺腺癌细胞中c-FLIP的ITCH蛋白降解和GSK3β下调ITCH表达。此外,ITCH沉默逆转了细胞中GSK3β敲低引起的c-FLIP蛋白的下调。因此,TRAIL诱导的GSK3β敲低诱导的细胞凋亡被ITCH的联合干扰阻断。
    结论:这些结果表明,GSK3β/ITCH轴调节c-FLIP的稳定性,并影响TRAIL诱导的细胞凋亡。一起来看,我们的研究揭示了GSK3β/ITCH/c-FLIP轴可以抵消TRAIL诱导的人肺腺癌细胞凋亡。
    OBJECTIVE: Further investigation on the mechanism of action of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in NSCLC would shed light on the understanding of TRAIL resistance and provide new clues for the counter-strategy.
    BACKGROUND: Cellular FLICE-inhibitory protein (c-FLIP) is a critical inhibitor of TRAIL-induced apoptosis. Our previous study suggested that glycogen synthase kinase 3β (GSK3β) positively regulated c-FLIP expression in human lung adenocarcinoma cells. Meanwhile, other studies reported that c-FLIP was degraded by HECT-type E3 ligase ITCH (Itchy E3 Ubiquitin Protein Ligase) via the proteasome pathway.
    OBJECTIVE: We will explore whether ITCH is involved in the expression regulation of c-FLIP positively controlled by GSK3β during the treatment of TRAIL.
    METHODS: Human lung adenocarcinoma cells were used to stably overexpress and knockdown GSK3β. Quantitative real-time PCR (qRT-PCR) assay was used to test the expressional level of mRNA of genes. Western blot analysis was employed to detect the expression of proteins at the protein level. siRNA of ITCH was used to knock down its expression. TRAIL treatment was used to cause apoptosis.
    RESULTS: In the present study, we have confirmed the degradation of c-FLIP by ITCH protein and the downregulation of ITCH expression by GSK3β in lung adenocarcinoma cells. Moreover, ITCH silencing reversed the downregulation of c-FLIP protein caused by GSK3β-knockdown in the cells. Accordingly, TRAIL-induced apoptosis facilitated by GSK3β knockdown was blocked by the combined interference of ITCH.
    CONCLUSIONS: These results suggested that GSK3β/ITCH axis regulated the stability of c-FLIP and influenced TRAIL-induced apoptosis. Taken together, our study revealed a GSK3β/ITCH/c-FLIP axis, which counteracts TRAIL-induced apoptosis in human lung adenocarcinoma cells.
    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