Doublecortin-like kinase 1

  • 文章类型: Preprint
    头颈部鳞状细胞癌(HNSCC)是一个主要的健康问题,因为它的高死亡率来自不良的治疗反应和局部肿瘤侵入头颈部的生命维持结构。对HNSCC侵袭机制的更深入理解具有告知可能增强患者生存的靶向治疗的潜力。我们先前报道了双皮质素样激酶1(DCLK1)调节HNSCC细胞的侵袭。这里,我们检验了DCLK1调节invadopodia内的蛋白质以促进HNSCC入侵的假设。Invadopodia是分泌降解细胞外基质(ECM)的基质金属蛋白酶的特化亚细胞突起。通过比较DCLK1对照和shDCLK1条件的综合蛋白质组分析,我们的研究结果表明,DCLK1在调节细胞骨架和ECM重塑的蛋白质中起着关键作用,有助于细胞入侵。Further,我们在TCGA数据集中证明DCLK1水平与组织学分级和淋巴结转移增加相关.我们发现DCLK1在HNSCC组织的前缘有较高的表达。在HNSCC中敲除DCLK1减少了invadopodia的数量,细胞粘附和集落形成。使用超分辨率显微镜,我们证明了DCLK1在侵袭足中的定位以及与成熟的侵袭足病标记TKS4,TKS5,cortactin和MT1-MMP的共定位。我们进行了磷酸蛋白质组学,并使用免疫荧光和邻近连接试验进行了验证,DCLK1与运动蛋白KIF16B之间的相互作用。药物抑制或敲低DCLK1减少与KIF16B的相互作用,MMPs的分泌,和细胞入侵。这项研究揭示了DCLK1在invadopodia中的新功能,以调节基质降解货物的贩运。这项工作强调了靶向DCLK1抑制局部区域入侵的影响,HNSCC的威胁生命的属性。
    Head and neck squamous cell carcinoma (HNSCC) is a major health concern due to its high mortality from poor treatment responses and locoregional tumor invasion into life sustaining structures in the head and neck. A deeper comprehension of HNSCC invasion mechanisms holds the potential to inform targeted therapies that may enhance patient survival. We previously reported that doublecortin like kinase 1 (DCLK1) regulates invasion of HNSCC cells. Here, we tested the hypothesis that DCLK1 regulates proteins within invadopodia to facilitate HNSCC invasion. Invadopodia are specialized subcellular protrusions secreting matrix metalloproteinases that degrade the extracellular matrix (ECM). Through a comprehensive proteome analysis comparing DCLK1 control and shDCLK1 conditions, our findings reveal that DCLK1 plays a pivotal role in regulating proteins that orchestrate cytoskeletal and ECM remodeling, contributing to cell invasion. Further, we demonstrate in TCGA datasets that DCLK1 levels correlate with increasing histological grade and lymph node metastasis. We identified higher expression of DCLK1 in the leading edge of HNSCC tissue. Knockdown of DCLK1 in HNSCC reduced the number of invadopodia, cell adhesion and colony formation. Using super resolution microscopy, we demonstrate localization of DCLK1 in invadopodia and colocalization with mature invadopodia markers TKS4, TKS5, cortactin and MT1-MMP. We carried out phosphoproteomics and validated using immunofluorescence and proximity ligation assays, the interaction between DCLK1 and motor protein KIF16B. Pharmacological inhibition or knockdown of DCLK1 reduced interaction with KIF16B, secretion of MMPs, and cell invasion. This research unveils a novel function of DCLK1 within invadopodia to regulate the trafficking of matrix degrading cargo. The work highlights the impact of targeting DCLK1 to inhibit locoregional invasion, a life-threatening attribute of HNSCC.
    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
    Doublecortin样激酶1(DCLK1)是一种微管相关蛋白激酶,与神经发生和人类癌症有关。最近的研究揭示了DCLK1在炎症信号中的新功能作用,因此将其定位为呼吸道炎性疾病治疗的新型靶激酶。在这项研究中,我们设计并合成了一系列基于NVP-TAE684的衍生物作为靶向DCLK1的新型抗炎药。NVP-TAE684衍生物的生物层干涉结合筛选和激酶测定导致发现了有效的DCLK1抑制剂(a24),IC50为179.7nM。化合物a24有效抑制巨噬细胞中脂多糖(LPS)诱导的炎症,效力高于先导化合物。机械上,化合物a24通过抑制DCLK1介导的IKKβ磷酸化来抑制LPS诱导的炎症。此外,化合物a24在LPS攻击的急性肺损伤模型中显示出体内抗炎活性。这些发现表明化合物a24可作为开发DCLK1抑制剂的新候选物和用于治疗炎性疾病的潜在治疗剂。
    Doublecortin-like kinase 1 (DCLK1) is a microtubule-associated protein kinase involved in neurogenesis and human cancer. Recent studies have revealed a novel functional role for DCLK1 in inflammatory signaling, thus positioning it as a novel target kinase for respiratory inflammatory disease treatment. In this study, we designed and synthesized a series of NVP-TAE684-based derivatives as novel anti-inflammatory agents targeting DCLK1. Bio-layer interferometry binding screening and kinase assays of the NVP-TAE684 derivatives led to the discovery of an effective DCLK1 inhibitor (a24), with an IC50 of 179.7 nM. Compound a24 effectively inhibited lipopolysaccharide (LPS)-induced inflammation in macrophages with higher potency than the lead compound. Mechanistically, compound a24 inhibited LPS-induced inflammation by inhibiting DCLK1-mediated IKKβ phosphorylation. Furthermore, compound a24 showed in vivo anti-inflammatory activity in an LPS-challenged acute lung injury model. These findings suggest that compound a24 may serve as a novel candidate for the development of DCLK1 inhibitors and a potential therapeutic agent for the treatment of inflammatory diseases.
    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
    少突胶质细胞通过两步机制从多能神经干细胞(NSC)产生:在NSC分化为少突胶质细胞前体/NG2细胞(OPCs)后,它们进一步发展成成熟的少突胶质细胞。这种分化过程的第一步还没有完全理解。在这项研究中,我们利用神经球测定法通过基于质谱(磷酸)的蛋白质组学,以时间过程依赖性方式研究了NSC到OPC的分化.我们确定了双皮质素样激酶1(Dclk1)是两个数据集中最突出的调节蛋白之一。并表明它在NSC到OPC分化期间经历了其短/长同工型之间的逐渐过渡。这是由其富含SP的区域的磷酸化调节,导致抑制蛋白水解Dclk1长切割,因此Dclk1短代。通过邻近生物素化对不同Dclk1同工型的相互作用分析,我们描述了他们各自假定的相互作用伙伴和底物。所有数据均可通过具有标识符PXD040652的ProteomeXchange获得。
    Oligodendrocytes are generated via a two-step mechanism from pluripotent neural stem cells (NSCs): after differentiation of NSCs to oligodendrocyte precursor/NG2 cells (OPCs), they further develop into mature oligodendrocytes. The first step of this differentiation process is only incompletely understood. In this study, we utilized the neurosphere assay to investigate NSC to OPC differentiation in a time course-dependent manner by mass spectrometry-based (phospho-) proteomics. We identify doublecortin-like kinase 1 (Dclk1) as one of the most prominently regulated proteins in both datasets, and show that it undergoes a gradual transition between its short/long isoform during NSC to OPC differentiation. This is regulated by phosphorylation of its SP-rich region, resulting in inhibition of proteolytic Dclk1 long cleavage, and therefore Dclk1 short generation. Through interactome analyses of different Dclk1 isoforms by proximity biotinylation, we characterize their individual putative interaction partners and substrates. All data are available via ProteomeXchange with identifier PXD040652.
    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:表皮生长因子受体-酪氨酸激酶抑制剂(EGFR-TKI)是EGFR敏感突变肺腺癌的一线治疗药物,但对EGFR-TKIs的获得性耐药仍是临床实践中的一个问题.上皮-间质转化(EMT)的发展是诱导对TKI的获得性抗性的关键机制。通过靶向驱动EMT的关键分子来逆转对EGFR-TKIs的获得性抗性为患者提供了一种替代选择。我们,因此,目的探讨双皮质素样激酶1(DCLK1)作为EMT驱动基因在肺腺癌对EGFR-TKIs获得性耐药中的作用。
    方法:使用细胞计数试剂盒-8(CCK8)测定法测量吉非替尼或奥希替尼在PC9/HCC827细胞中的IC50。采用RT-PCR和Westernblot检测PC9和HCC827细胞中EMT相关基因的表达水平。通过transwell测定评估细胞迁移和侵袭能力。对于体内实验,将PC9细胞皮下注射到BALB/c裸鼠中以形成肿瘤。收获时,保留肿瘤组织进行RT-PCR,蛋白质印迹,和多色荧光染色以检测EMT过程中生物标志物的变化。
    结果:吉非替尼耐药的PC9(PC9/GR)和奥希替尼耐药的HCC827(HCC827/OR)细胞与TKI敏感的细胞相比,显示出显着的EMT活化和增强的迁移和侵袭能力。此外,DCLK1在EGFR-TKI耐药的肺腺癌细胞中表达显著升高。DCLK1的靶向敲除可有效逆转TKI耐药细胞的EMT表型,提高EGFR-TKI敏感性,通过体内实验进一步验证。
    结论:DCLK1通过诱导EMT并加速TKI耐药细胞的迁移和侵袭能力,促进了肺腺癌对EGFR-TKI的获得性耐药。
    OBJECTIVE: Epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI) is a first-line treatment for lung adenocarcinoma with EGFR-sensitive mutations, but acquired resistance to EGFR-TKIs remains a problem in clinical practice. The development of epithelial-mesenchymal transition (EMT) is a critical mechanism that induces acquired resistance to TKIs. Reversing acquired resistance to EGFR-TKIs through targeting the key molecules driving EMT provides an alternative choice for patients. We, therefore, aimed to explore the role of doublecortin-like kinase 1 (DCLK1) as an EMT driver gene in the acquired resistance of lung adenocarcinoma to EGFR-TKIs.
    METHODS: The IC50 of Gefitinib or Osimertinib in PC9/HCC827 cells was measured using a cell counting kit-8 (CCK8) assay. The expression levels of EMT-related genes in PC9 and HCC827 cells were detected using RT-PCR and Western blot. Cell migration and invasion abilities were assessed via a transwell assay. For the in vivo experiments, PC9 cells were subcutaneously injected into BALB/c nude mice to form tumors. Upon harvesting, tumor tissues were retained for RT-PCR, Western blot, and polychromatic fluorescence staining to detect biomarker changes in the EMT process.
    RESULTS: Gefitinib-resistant PC9 (PC9/GR) and Osimertinib-resistant HCC827 (HCC827/OR) cells showed remarkable activation of EMT and enhanced migration and invasion abilities compared to TKI-sensitive cells. In addition, DCLK1 expression was markedly increased in EGFR-TKI-resistant lung adenocarcinoma cells. The targeted knockout of DCLK1 effectively reversed the EMT phenotype in TKI-resistant cells and improved EGFR-TKI sensitivity, which was further validated by the in vivo experiments.
    CONCLUSIONS: DCLK1 facilitates acquired resistance to EGFR-TKI in lung adenocarcinoma by inducting EMT and accelerating the migration and invasion abilities of TKI-resistant cells.
    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
    原理:Doublecortin样激酶1(DCLK1)是一种丝氨酸/苏氨酸激酶,可选择性标记癌症干细胞(CSC)并促进结直肠癌(CRC)的恶性进展。然而,DCLK1驱动癌细胞侵袭性表型的确切分子机制尚不完全确定.方法:这里,我们进行了全面的基因组学和蛋白质组学分析,以鉴定DCLK1的结合蛋白,并发现了X线修复交叉互补蛋白5(XRCC5).因此,我们探讨了DCLK1/XRCC5轴在人类CRC细胞和CRC小鼠模型中的生物学作用和下游事件.结果:综合生物信息学分析的结果表明,DCLK1驱动的CRC侵袭性与炎症有关。机械上,DCLK1结合并磷酸化XRCC5,进而转录激活环氧合酶2的表达并增强前列腺素E2的产生;这些事件共同产生了炎性肿瘤微环境并增强了CRC细胞的攻击行为。与发现的机制一致,DCLK1激酶活性的抑制强烈损害了CRC小鼠模型中的肿瘤接种和生长能力。结论:我们的研究阐明了介导CSCs在驱动CRC侵袭性表型中的促炎功能的新机制。扩大DCLK1在CRC中的生物学功能。
    Rationale: Doublecortin-like kinase 1 (DCLK1) is a serine/threonine kinase that selectively marks cancer stem-like cells (CSCs) and promotes malignant progression in colorectal cancer (CRC). However, the exact molecular mechanism by which DCLK1 drives the aggressive phenotype of cancer cells is incompletely determined. Methods: Here, we performed comprehensive genomics and proteomics analyses to identify binding proteins of DCLK1 and discovered X-ray repair cross-complementing 5 (XRCC5). Thus, we explored the biological role and downstream events of the DCLK1/XRCC5 axis in human CRC cells and CRC mouse models. Results: The results of comprehensive bioinformatics analyses suggested that DCLK1-driven CRC aggressiveness is linked to inflammation. Mechanistically, DCLK1 bound and phosphorylated XRCC5, which in turn transcriptionally activated cyclooxygenase-2 expression and enhanced prostaglandin E2 production; these events collectively generated the inflammatory tumor microenvironment and enhanced the aggressive behavior of CRC cells. Consistent with the discovered mechanism, inhibition of DCLK1 kinase activity strongly impaired the tumor seeding and growth capabilities in CRC mouse models. Conclusion: Our study illuminates a novel mechanism that mediates the pro-inflammatory function of CSCs in driving the aggressive phenotype of CRC, broadening the biological function of DCLK1 in CRC.
    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:最近在功能性消化不良(FD)患者中发现了低度十二指肠炎症。据报道,化学感觉簇细胞与胃肠道疾病有关。因此,我们评估了FD患者的十二指肠簇细胞密度和微炎症,以确定这些措施是否可以作为有用的生物标志物。并与FD患者的簇绒细胞密度和微炎症相关。
    方法:十二指肠活检标本取自FD患者和对照组。簇绒细胞,嗜酸性粒细胞,肥大细胞用特异性抗体免疫化学染色。通过免疫染色对双皮质素样激酶1(DCLK1)鉴定簇绒细胞;通过双重染色对胆碱乙酰转移酶(ChAT)和DCLK1评估胆碱能簇绒细胞。使用实时PCR通过IL-25mRNA表达评估免疫型簇细胞。
    结果:FD患者十二指肠粘膜内嗜酸性粒细胞和肥大细胞的密度明显高于对照组。FD患者十二指肠的簇绒细胞密度明显高于对照组,并且与FD患者和对照组的十二指肠嗜酸性粒细胞密度显着相关。此外,一部分ChAT阳性细胞为DCLK1阳性;所有十二指肠DCLK1簇细胞在FD和对照组中均具有ChAT免疫反应性。
    结论:FD患者十二指肠胆碱能簇细胞密度较高,与嗜酸性粒细胞密度显著相关。需要进一步的研究来研究FD中簇绒细胞的病理生理学意义,并可能为FD的病理生理学提供有价值的线索。
    BACKGROUND: Low-grade duodenal inflammation has recently been identified in patients with functional dyspepsia (FD). Chemosensory tuft cells were reported to be associated with gastrointestinal diseases. We therefore assessed duodenal tuft cell density and microinflammation in patients with FD to determine whether these measures could serve as useful biomarkers, and also correlated tuft cell density and microinflammation in FD patients.
    METHODS: Duodenal biopsy specimens were obtained from patients with FD and from controls. Tuft cells, eosinophils, and mast cells were immunochemically stained with specific antibodies. Tuft cells were identified by immunostaining for doublecortin-like kinase 1 (DCLK1); cholinergic tuft cells were assessed by double staining for choline acetyltransferase (ChAT) and DCLK1. Immune-type tuft cells were assessed by IL-25 mRNA expression using real-time PCR.
    RESULTS: The density of intramucosal eosinophils and mast cells was significantly higher in the duodenum of FD patients than in controls. The density of tuft cells was significantly higher in the duodenum of FD patients compared with controls, and significantly correlated with eosinophil density in the duodenum of FD patients and controls. Moreover, a fraction of ChAT-positive cells was DCLK1 positive; all duodenal DCLK1+ tuft cells were ChAT-immunoreactive in FD and in control subjects.
    CONCLUSIONS: Cholinergic tuft cell density was higher in the duodenum of patients with FD and significantly correlated with eosinophil density. Further studies are needed to investigate the pathophysiological significance of tuft cells in FD and may provide valuable clues to the pathophysiology of FD.
    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
    禽白血病病毒J亚组(ALV-J)诱导骨髓细胞瘤,可以转移到病鸡的多个器官。虽然转移是这种情况下死亡的主要原因,其机制尚不清楚。这里,我们发现ALV-J表面蛋白(SU)和双皮质素样激酶1(DCLK1)之间的相互作用促进上皮-间质转化(EMT)和细胞增殖.我们发现ALV-J可以激活感染细胞中的EMT。随后,蛋白质组学分析显示,DCLK1是一种公认的肿瘤干细胞标志物,在ALV-J感染的DF-1细胞和鸡中高表达,可能是介导EMT的潜在因素。此外,使用免疫荧光和免疫沉淀,我们验证了SU与DCLK1相互作用。功能研究表明,DCLK1的过表达通过加速细胞从G0/G1期到S期的细胞周期进程来增加病毒复制并促进细胞增殖。而在ALV-J感染的细胞中,DCLK1的RNA干扰通过延缓细胞周期从G1期晚期进入S期来减少病毒复制并阻止细胞增殖.此外,我们证明DCLK1的积累增加通过增加N-cadherin的表达促进EMT,波形蛋白,MMP2和转录因子Snail1降低上皮标志物E-cadherin的表达。这些结果表明,ALV-JSU与DCLK1相互作用,并加速细胞增殖,导致病毒复制增加并最终激活EMT,这为肿瘤转移铺平了道路。重要性肿瘤转移是癌症研究的主要挑战,因为其系统性和播散性肿瘤细胞对现有治疗剂的抗性。据估计,来自癌症的>90%的死亡率归因于转移。我们发现ALV-J能激活EMT,在癌症转移中起着至关重要的作用。随后,我们确定了肿瘤干细胞标记物,DCLK1,在ALV-J感染的细胞中,与ALV-J的表面蛋白(SU)相互作用以促进病毒复制,激活EMT,并加速细胞增殖,使ALV-J获得转移能力。了解ALV-J参与EMT的过程和转移途径将有助于阐明病毒诱导肿瘤转移的机制,并有助于确定有希望的分子靶标和ALV-J控制和临床技术发展的关键障碍。
    Avian leukosis virus subgroup J (ALV-J) induces myelocytomas, which can metastasize to multiple organs in diseased chickens. Although metastasis is the primary cause of death in such cases, the mechanism for it remains unclear. Here, we found that interaction between ALV-J surface protein (SU) and doublecortin-like kinase 1 (DCLK1) promotes epithelial-mesenchymal transition (EMT) and cell proliferation. We found that ALV-J can activate EMT in infected cells. Subsequently, proteomics analysis revealed that DCLK1, a well-established putative tumor stem cell marker, which is highly expressed in ALV-J-infected DF-1 cells and chickens, might be a potential factor mediating EMT. Furthermore, using immunofluorescence and immunoprecipitation, we verified that SU interacts with DCLK1. Functional studies suggested that overexpression of DCLK1 increased viral replication and promoted cell proliferation by accelerating the progression of cells from the G0/G1 phase to the S phase of cell cycle, whereas RNA interference of DCLK1 reduced viral replication and arrested cell proliferation by retarding cell cycle progression from the late G1 phase into the S phase in ALV-J-infected cells. Moreover, we demonstrate that the increased accumulation of DCLK1 promotes EMT by increasing the expression of N-cadherin, vimentin, MMP2, and transcription factor Snail1 and decreasing the expression of epithelial marker E-cadherin. These results suggest that ALV-J SU interacts with DCLK1, and accelerates cell proliferation, leading to increased viral replication and ultimately activating EMT, which paves the way for tumor metastasis. IMPORTANCE Tumor metastasis is a major challenge in cancer research, because of its systemic nature and the resistance of disseminated tumor cells to existing therapeutic agents. It is estimated that >90% of mortality from cancer is attributable to metastases. We found that ALV-J can activate EMT, which plays a critical role in cancer metastasis. Subsequently, we identified a tumor stem cell marker, DCLK1, in ALV-J infected cells, which interacts with surface protein (SU) of ALV-J to promote virus replication, activate EMT, and accelerate cell proliferation enabling ALV-J to obtain metastatic ability. Understanding the process of participation of ALV-J in EMT and the route of metastasis will help elucidate the mechanism of virus-induced tumor metastasis and help identify promising molecular targets and key obstacles for ALV-J control and clinical technology 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
    Given the functional attributes of Doublecortin-like kinase 1 (DCLK1) in tumor growth, invasion, metastasis, cell motility, and tumor stemness, it is emerging as a therapeutic target in gastrointestinal cancers. Although a series of specific or nonspecific ATP-competitive inhibitors were identified against DCLK1, different types of scaffolds that can be utilized for the development of highly selective inhibitors or structural understanding of binding specificities of the compounds remain limited. Here, we present our work to repurpose a Janus kinase 1 inhibitor, ruxolitinib as a DCLK1 inhibitor, showing micromolar binding affinity and inhibitory activity. Furthermore, to gain an insight into its interaction mode with DCLK1, a crystal structure of the ruxolitinib-complexed DCLK1 has been determined and analyzed. Ruxolitinib as a nonspecific DCLK1 inhibitor characterized in this work is anticipated to provide a starting point for the structure-guided discovery of selective DCLK1 inhibitors.
    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
    微管相关蛋白,双皮质素样激酶1(DCLK1),在一系列癌症中高度表达,并且是激酶抑制剂的突出治疗靶标。DCLK1激酶活性的生理作用以及它是如何被调节的仍然难以捉摸。这里,我们分析了哺乳动物DCLK1激酶活性在调节微管结合中的作用。我们发现DCLK1自磷酸化其C末端尾部内的残基以限制其激酶活性并防止其微管结合域内的异常过度磷酸化。去除C末端尾部或该残基的突变会导致doublecortin结构域内磷酸化的增加,从而消除了微管结合。因此,DCLK1内特定位点的自磷酸化对分子与微管的缔合具有直径效应。我们的结果表明DCLK1调节其激酶活性以调节其微管结合亲和力的机制。这些结果为未来与DCLK1在癌症发展和进展中的作用相关的治疗工作提供了分子见解。
    The microtubule-associated protein, doublecortin-like kinase 1 (DCLK1), is highly expressed in a range of cancers and is a prominent therapeutic target for kinase inhibitors. The physiological roles of DCLK1 kinase activity and how it is regulated remain elusive. Here, we analyze the role of mammalian DCLK1 kinase activity in regulating microtubule binding. We found that DCLK1 autophosphorylates a residue within its C-terminal tail to restrict its kinase activity and prevent aberrant hyperphosphorylation within its microtubule-binding domain. Removal of the C-terminal tail or mutation of this residue causes an increase in phosphorylation within the doublecortin domains, which abolishes microtubule binding. Therefore, autophosphorylation at specific sites within DCLK1 has diametric effects on the molecule\'s association with microtubules. Our results suggest a mechanism by which DCLK1 modulates its kinase activity to tune its microtubule-binding affinity. These results provide molecular insights for future therapeutic efforts related to DCLK1\'s role in cancer development and progression.
    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
    和厚朴酚(HNK)是一种双酚类化合物,已在传统医学中用于治疗各种疾病,包括癌症。在这项研究中,我们在培养物和结肠炎相关癌症模型中测定了HNK对结肠癌细胞的影响.HNK处理抑制增殖和集落形成,同时诱导细胞凋亡。此外,HNK抑制了结肠层的形成。分子对接表明HNK与储备干细胞标记蛋白DCLK1相互作用,结合能为-7.0Kcal/mol。体外激酶测定证明HNK抑制DCLK1激酶活性。HNK还抑制了其他癌症干细胞标记蛋白LGR5和CD44的表达。Hippo信号通路在肠干细胞中具有活性。在典型途径中,YAP1在Ser127处被上游Mst1/2和Lats1/2磷酸化。这导致YAP1在细胞质中的隔离,从而不允许YAP1易位到细胞核并与TEAD1-4转录因子相互作用以诱导基因表达。然而,HNK抑制了YAP1中Ser127的磷酸化,但该蛋白仍被隔离在细胞质中。我们进一步确定,这是通过YAP1与PUMA相互作用而发生的。为了确定这是否也发生在体内,我们在AOM/DSS诱导的结肠炎相关癌症模型中进行了研究.通过以5mg/kgbw的剂量口服管饲法给予HNK24周,证明YAP1和TEAD1的表达以及茎标记蛋白的表达显着降低。一起,这些数据表明,HNK部分通过诱导PUMA-YAP1相互作用和胞质隔离来阻止结肠癌的发生,从而抑制致癌YAP1活性。
    Honokiol (HNK) is a biphenolic compound that has been used in traditional medicine for treating various ailments, including cancers. In this study, we determined the effect of HNK on colon cancer cells in culture and in a colitis-associated cancer model. HNK treatment inhibited proliferation and colony formation while inducing apoptosis. In addition, HNK suppressed colonosphere formation. Molecular docking suggests that HNK interacts with reserve stem cell marker protein DCLK1, with a binding energy of -7.0 Kcal/mol. In vitro kinase assays demonstrated that HNK suppressed the DCLK1 kinase activity. HNK also suppressed the expression of additional cancer stem cell marker proteins LGR5 and CD44. The Hippo signaling pathway is active in intestinal stem cells. In the canonical pathway, YAP1 is phosphorylated at Ser127 by upstream Mst1/2 and Lats1/2. This results in the sequestration of YAP1 in the cytoplasm, thereby not allowing YAP1 to translocate to the nucleus and interact with TEAD1-4 transcription factors to induce gene expression. However, HNK suppressed Ser127 phosphorylation in YAP1, but the protein remains sequestered in the cytoplasm. We further determined that this occurs by YAP1 interacting with PUMA. To determine if this also occurs in vivo, we performed studies in an AOM/DSS induced colitis-associated cancer model. HNK administered by oral gavage at a dose of 5mg/kg bw for 24 weeks demonstrated a significant reduction in the expression of YAP1 and TEAD1 and in the stem marker proteins. Together, these data suggest that HNK prevents colon tumorigenesis in part by inducing PUMA-YAP1 interaction and cytoplasmic sequestration, thereby suppressing the oncogenic YAP1 activity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号