LKB1

LKB1
  • 文章类型: Journal Article
    Exo70,一个关键的外囊复合物成分,在癌症转移中,细胞运动和细胞外基质(ECM)重塑至关重要。尽管它有作为药物靶标的潜力,Exo70的翻译后修饰(PTM)的特征不佳。这里,我们报道,通过转谷氨酰胺酶TGM1和TGM3,Exo70在Gln5上与胱抑素A的Lys56转酰胺化,促进肿瘤转移。这种修饰增强了Exo70与其他外囊亚基的关联,分泌基质金属蛋白酶所必需的,形成invadopodia,并将集成蛋白传递到前沿。抑癌肝激酶B1(LKB1),其失活加速了转移,分别在Thr386和Thr282处磷酸化TGM1和TGM3,抑制它们与Exo70的相互作用以及随后的转酰胺作用。斑潜素,美国食品和药物管理局(FDA)批准的药物,抑制Exo70转酰胺化以抑制肿瘤细胞迁移和侵袭。一起,我们的研究结果强调了Exo70转酰胺作用作为一个关键的分子机制和靶点,并提出了作为一种治疗策略,对转移性癌症具有直接的临床转化价值,尤其是那些LKB1损失。
    Exo70, a key exocyst complex component, is crucial for cell motility and extracellular matrix (ECM) remodeling in cancer metastasis. Despite its potential as a drug target, Exo70\'s post-translational modifications (PTMs) are poorly characterized. Here, we report that Exo70 is transamidated on Gln5 with Lys56 of cystatin A by transglutaminases TGM1 and TGM3, promoting tumor metastasis. This modification enhances Exo70\'s association with other exocyst subunits, essential for secreting matrix metalloproteinases, forming invadopodia, and delivering integrins to the leading edge. Tumor suppressor liver kinase B1 (LKB1), whose inactivation accelerates metastasis, phosphorylates TGM1 and TGM3 at Thr386 and Thr282, respectively, to inhibit their interaction with Exo70 and the following transamidation. Cantharidin, a US Food and Drug Administration (FDA)-approved drug, inhibits Exo70 transamidation to restrain tumor cell migration and invasion. Together, our findings highlight Exo70 transamidation as a key molecular mechanism and target and propose cantharidin as a therapeutic strategy with direct clinical translational value for metastatic cancers, especially those with LKB1 loss.
    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
    与BRAFV600E癌基因一起在黑色素瘤发展中起作用的机制,除了紫外线(UV)辐射(UVR),非常感兴趣。人黑素瘤肿瘤的分析[来自癌症基因组图谱(TCGA)的数据]显示50%或更多的样品不表达或表达少量的丝氨酸/苏氨酸蛋白激酶STK11(也称为LKB1)蛋白。这里,我们报告说,在老鼠模型中,伴随的新生儿BrafV600E激活和黑素细胞中的Lkb1肿瘤抑制物消融导致完全黑色素瘤发展.与BrafV600E照射的小鼠相比,出生后单一剂量的UVB照射对Lkb1耗尽小鼠的黑色素瘤发作没有影响,但增加了肿瘤的多样性。根据这些发现和以前的报告,Lkb1无效的辐照小鼠表现出DNA损伤修复缺陷(DDR)。组织学上,缺乏Lkb1的肿瘤富含神经样肿瘤形态。肿瘤样品突变基因的遗传谱分析和基因集富集分析表明,Lkb1的缺失促进了与神经分化过程相关的改变基因的选择。因此,这些结果表明,Lkb1的丢失与BrafV600E和UVR合作,损害DDR并增加黑色素瘤的多样性和神经样去分化。
    The mechanisms that work alongside BRAFV600E oncogene in melanoma development, in addition to ultraviolet (UV) radiation (UVR), are of great interest. Analysis of human melanoma tumors [data from The Cancer Genome Atlas (TCGA)] revealed that 50% or more of the samples expressed no or low amounts of serine/threonine protein kinase STK11 (also known as LKB1) protein. Here, we report that, in a mouse model, concomitant neonatal BrafV600E activation and Lkb1 tumor suppressor ablation in melanocytes led to full melanoma development. A single postnatal dose of UVB radiation had no effect on melanoma onset in Lkb1-depleted mice compared with BrafV600E-irradiated mice, but increased tumor multiplicity. In concordance with these findings and previous reports, Lkb1-null irradiated mice exhibited deficient DNA damage repair (DDR). Histologically, tumors lacking Lkb1 were enriched in neural-like tumor morphology. Genetic profiling and gene set enrichment analyses of tumor sample mutated genes indicated that loss of Lkb1 promoted the selection of altered genes associated with neural differentiation processes. Thus, these results suggest that the loss of Lkb1 cooperates with BrafV600E and UVR, impairing the DDR and increasing melanoma multiplicity and neural-like dedifferentiation.
    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
    改变的蛋白质泛素化与癌症有关。据报道,E3泛素连接酶的新型三方基序(TRIM)家族在发育中起着至关重要的作用,增长,和各种肿瘤的转移。TRIM家族成员TRIM27在多种癌症中充当肿瘤发展的潜在启动子。然而,关于胶质母细胞瘤(GBM)中TRIM27的生物学特征和临床相关性知之甚少。这里,我们报告了GBM组织和GBM细胞系中TRIM27表达升高的发现。进一步的功能分析显示,TRIM27缺失在体外和体内均抑制GBM细胞生长。此外,我们发现TRIM27通过增强Warburg效应促进GBM细胞的生长。此外,LKB1/AMPK/mTOR通路的失活对于TRIM27在GBM中的致癌作用至关重要.机械上,TRIM27可以直接与LKB1结合,促进LKB1的泛素化和降解,进而增强Warburg效应和GBM进程。总的来说,这些数据提示TRIM27通过抑制LKB1/AMPK/mTOR轴参与GNM发病机制,可能成为GBM患者潜在的诊断和治疗标志物.
    Altered protein ubiquitination is associated with cancer. The novel tripartite motif (TRIM) family of E3 ubiquitin ligases have been reported to play crucial roles in the development, growth, and metastasis of various tumors. The TRIM family member TRIM27 acts as a potential promoter of tumor development in a wide range of cancers. However, little is known regarding the biological features and clinical relevance of TRIM27 in glioblastoma (GBM). Here, we report findings of elevated TRIM27 expression in GBM tissues and GBM cell lines. Further functional analysis showed that TRIM27 deletion inhibited GBM cell growth both in vitro and in vivo. Furthermore, we found that TRIM27 promoted the growth of GBM cells by enhancing the Warburg effect. Additionally, the inactivation of the LKB1/AMPK/mTOR pathway was critical for the oncogenic effects of TRIM27 in GBM. Mechanistically, TRIM27 could directly bind to LKB1 and promote the ubiquitination and degradation of LKB1, which in turn enhanced the Warburg effect and GBM progression. Collectively, these data suggest that TRIM27 contributes to GNM pathogenesis by inhibiting the LKB1/AMPK/mTOR axis and may be a promising candidate as a potential diagnostic and therapeutic marker for patients with GBM.
    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
    Erianin,在石斛提取物中发现的一种二苄基化合物,具有广泛的抗癌活性。然而,其在胃癌(GC)中的作用机制尚不清楚。LKB1是一种抑癌基因,它的突变是各种癌症的重要驱动因素。然而,一些研究报告了相互矛盾的发现。在这项研究中,我们结合生物信息学和体内外实验来研究Erianin治疗GC的作用和潜在机制。结果表明,LKB1在患者肿瘤组织和GC细胞中高表达,与患者预后不良有关。Erianin能促进GC细胞凋亡,抑制划痕修复,迁移,入侵,和上皮-间质转化(EMT)特征。Erianin剂量依赖性地抑制LKB1,SIK2,SIK3和PARD3的表达,但对SIK1没有显着影响。Erianin还抑制CDX小鼠模型中的肿瘤生长。出乎意料的是,5-FU对LKB1也表现出一定的抑制作用。Erianin和5-FU的组合显著提高了5-FU在GC细胞和异种移植小鼠模型的生长中的抗肿瘤功效。总之,Erianin是一种潜在的抗GC化合物,可以通过靶向LKB1-SIK2/3-PARD3信号轴来抑制GC生长和EMT特性。Erianin和5-FU的协同作用提示了用于GC治疗的有希望的治疗策略。
    Erianin, a bibenzyl compound found in dendrobium extract, has demonstrated broad anticancer activity. However, its mechanism of action in gastric cancer (GC) remains poorly understood. LKB1 is a tumor-suppressor gene, and its mutation is an important driver of various cancers. Yet some studies have reported contradictory findings. In this study, we combined bioinformatics and in vitro and in vivo experiments to investigate the effect and potential mechanism of Erianin in the treatment of GC. The results show that LKB1 was highly expressed in patients\' tumor tissues and GC cells, and it was associated with poor patient prognosis. Erianin could promote GC cell apoptosis and inhibit the scratch repair, migration, invasion, and epithelial-mesenchymal transition (EMT) characteristics. Erianin dose-dependently inhibited the expression of LKB1, SIK2, SIK3, and PARD3 but had no significant effect on SIK1. Erianin also inhibited tumor growth in CDX mice model. Unexpectedly, 5-FU also exhibited a certain inhibitory effect on LKB1. The combination of Erianin and 5-FU significantly improved the anti-tumor efficacy of 5-FU in the growth of GC cells and xenograft mouse models. In summary, Erianin is a potential anti-GC compound that can inhibit GC growth and EMT properties by targeting the LKB1-SIK2/3-PARD3-signaling axis. The synergistic effect of Erianin and 5-FU suggests a promising therapeutic strategy for GC 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
    脂质代谢紊乱是几种严重影响公众健康的慢性代谢性疾病的主要原因。Salusin-α,血管活性肽,已被证明可以减轻脂质代谢紊乱,尽管其作用机制尚未报道。为了研究Salusin-α对脂质代谢的影响和潜在机制,使用慢病毒载体过表达或敲低Salusin-α。慢病毒转染HepG2细胞后,游离脂肪酸(FFA)诱导肝细胞脂肪变性。使用油红O染色并通过测量几种生化指标来评估脂质积累的程度。随后,生物信息学用于分析可能参与脂质代谢紊乱的信号通路。最后,半定量PCR和免疫印迹用于验证肝激酶B1(LKB1)/AMPK通路的参与。化合物C,AMPK的抑制剂,被用来进一步证实这种机制的参与。结果表明,Salusin-α显着减弱脂质积累,炎症和氧化应激。此外,Salusin-α增加了LKB1和AMPK的水平,抑制固醇调节元件结合蛋白-1c的表达,脂肪酸合成酶和乙酰辅酶A羧化酶。化合物C的添加消除了Salusin-α介导的AMPK对下游信号分子的调节。总之,Salusin-α的过表达激活了LKB1/AMPK途径,这反过来又抑制了HepG2细胞中的脂质积累。这提供了对Salusin‑α改善脂质代谢紊乱的潜在机制的见解,同时确定了潜在的治疗靶标。
    Lipid metabolism disorders are a major cause of several chronic metabolic diseases which seriously affect public health. Salusin‑α, a vasoactive peptide, has been shown to attenuate lipid metabolism disorders, although its mechanism of action has not been reported. To investigate the effects and potential mechanisms of Salusin‑α on lipid metabolism, Salusin‑α was overexpressed or knocked down using lentiviral vectors. Hepatocyte steatosis was induced by free fatty acid (FFA) after lentiviral transfection into HepG2 cells. The degree of lipid accumulation was assessed using Oil Red O staining and by measuring several biochemical indices. Subsequently, bioinformatics was used to analyze the signaling pathways that may have been involved in lipid metabolism disorders. Finally, semi‑quantitative PCR and western blotting were used to verify the involvement of the liver kinase B1 (LKB1)/AMPK pathway. Compound C, an inhibitor of AMPK, was used to confirm this mechanism\'s involvement further. The results showed that Salusin‑α significantly attenuated lipid accumulation, inflammation and oxidative stress. In addition, Salusin‑α increased the levels of LKB1 and AMPK, which inhibited the expression of sterol regulatory element binding protein‑1c, fatty acid synthase and acetyl‑CoA carboxylase. The addition of Compound C abrogated the Salusin‑α‑mediated regulation of AMPK on downstream signaling molecules. In summary, overexpression of Salusin‑α activated the LKB1/AMPK pathway, which in turn inhibited lipid accumulation in HepG2 cells. This provides insights into the potential mechanism underlying the mechanism by which Salusin‑α ameliorates lipid metabolism disorders while identifying a potential therapeutic target.
    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
    尽管证实了LKB1在抑制肺癌进展中的作用,它对细胞衰老的确切影响是未知的。本研究旨在阐明LKB1在抑制肺腺癌端粒酶活性中的作用及机制。结果表明,LKB1在体外或体内均可诱导细胞衰老和凋亡。LKB1在LKB1缺陷型A549细胞中的过表达通过调节端粒酶逆转录酶(TERT)的表达而导致端粒酶活性的抑制和端粒功能障碍的诱导。作为转录因子,LKB1过表达后Sp1介导TERT抑制。LKB1诱导乳酸产生并抑制组蛋白H4(Lys8)和H4(Lys16)的乳酸化,这进一步改变了Sp1相关的转录活性。端粒酶抑制剂BIBR1532有助于实现传统化疗药物联合糖酵解抑制剂2DG的最佳疗效。这些数据揭示了LKB1通过依赖乳酸化的转录抑制调节端粒酶活性的新机制。因此,为LKB1介导的衰老在肺腺癌中的作用提供了新的见解。我们的研究为创造新的癌症治疗方法开辟了新的可能性。
    Despite the confirmed role of LKB1 in suppressing lung cancer progression, its precise effect on cellular senescence is unknown. The aim of this research was to clarify the role and mechanism of LKB1 in restraining telomerase activity in lung adenocarcinoma. The results showed that LKB1 induced cellular senescence and apoptosis either in vitro or in vivo. Overexpression of LKB1 in LKB1-deficient A549 cells led to the inhibition of telomerase activity and the induction of telomere dysfunction by regulating telomerase reverse transcriptase (TERT) expression in terms of transcription. As a transcription factor, Sp1 mediated TERT inhibition after LKB1 overexpression. LKB1 induced lactate production and inhibited histone H4 (Lys8) and H4 (Lys16) lactylation, which further altered Sp1-related transcriptional activity. The telomerase inhibitor BIBR1532 was beneficial for achieving the optimum curative effect of traditional chemotherapeutic drugs accompanied by the glycolysis inhibitor 2DG. These data reveal a new mechanism by which LKB1 regulates telomerase activity through lactylation-dependent transcriptional inhibition, and therefore, provide new insights into the effects of LKB1-mediated senescence in lung adenocarcinoma. Our research has opened up new possibilities for the creation of new cancer treatments.
    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
    探讨LKB1表达缺失在非小细胞肺癌(NSCLC)中的发生率和预后相关性及协同作用。我们分析了来自188例转移性和60例非转移性可手术I-IIIA期NSCLC患者的手术标本,以评估其与各种过程相关的LKB1和pAMPK蛋白的表达.研究的因素包括抗肿瘤免疫反应调节剂STING和PD-L1;促血管生成,EMT和细胞周期目标,以及与转移相关的(VEGFC,PDGFRα,PDGFRβ,p53,p16,细胞周期蛋白D1,ZEB1,CD24)靶标;和细胞粘附(β-连环蛋白)分子。通过免疫组织化学评估蛋白质表达水平;通过PCR评估LKB1和NEDD9的RNA水平,而KRAS外显子2和BRAFV600E突变通过Sanger测序进行评估。总的来说,在21%(51/248)的患者中观察到LKB1蛋白表达丢失,并且与组织型显着相关(p<0.001),KRAS突变(p<0.001),KC状态(伴随KRAS突变和p16下调)(p<0.001),STING损失(p<0.001),和高CD24表达(p<0.001)。STING丢失也与转移背景下的LKB1表达丢失显著相关(p=0.014)和肺腺癌(LUACs)(p=0.005)。此外,LKB1缺失与仅在LUACs中缺乏或低β-catenin膜表达显著相关,与转移状态(p=0.019)和转移情况(p=0.007)无关。出现LKB1丢失和伴随不存在或低β-catenin膜表达的肿瘤患者的中位总生存期明显低于20.50。52.99个月;p<0.001以及显著更大的死亡风险(HR:3.32,95%c.i.:1.71-6.43;p<0.001)。我们的发现强调了LKB1与STING和β-catenin在NSCLC中的协同作用的影响。在预后方面。
    To investigate the incidence and prognostically significant correlations and cooperations of LKB1 loss of expression in non-small cell lung cancer (NSCLC), surgical specimens from 188 metastatic and 60 non-metastatic operable stage I-IIIA NSCLC patients were analyzed to evaluate their expression of LKB1 and pAMPK proteins in relation to various processes. The investigated factors included antitumor immunity response regulators STING and PD-L1; pro-angiogenic, EMT and cell cycle targets, as well as metastasis-related (VEGFC, PDGFRα, PDGFRβ, p53, p16, Cyclin D1, ZEB1, CD24) targets; and cell adhesion (β-catenin) molecules. The protein expression levels were evaluated via immunohistochemistry; the RNA levels of LKB1 and NEDD9 were evaluated via PCR, while KRAS exon 2 and BRAFV600E mutations were evaluated by Sanger sequencing. Overall, loss of LKB1 protein expression was observed in 21% (51/248) patients and correlated significantly with histotype (p < 0.001), KRAS mutations (p < 0.001), KC status (concomitant KRAS mutation and p16 downregulation) (p < 0.001), STING loss (p < 0.001), and high CD24 expression (p < 0.001). STING loss also correlated significantly with loss of LKB1 expression in the metastatic setting both overall (p = 0.014) and in lung adenocarcinomas (LUACs) (p = 0.005). Additionally, LKB1 loss correlated significantly with a lack of or low β-catenin membranous expression exclusively in LUACs, both independently of the metastatic status (p = 0.019) and in the metastatic setting (p = 0.007). Patients with tumors yielding LKB1 loss and concomitant nonexistent or low β-catenin membrane expression experienced significantly inferior median overall survival of 20.50 vs. 52.99 months; p < 0.001 as well as significantly greater risk of death (HR: 3.32, 95% c.i.: 1.71-6.43; p <0.001). Our findings underscore the impact of the synergy of LKB1 with STING and β-catenin in NSCLC, in prognosis.
    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
    线粒体电子传递链(ETC)在调节铁死亡中的作用尚未完全阐明。这里,我们发现,ETC复合物I的药理学抑制降低泛醇水平,同时降低ATP水平并激活AMP激活的蛋白激酶(AMPK),这两种效应以其在促进和抑制铁性凋亡中的作用而闻名,分别。因此,复合物I抑制剂对谷胱甘肽过氧化物酶4(GPX4)抑制诱导的铁凋亡的影响有限。复合物I在LKB1-AMPK灭活细胞中的药理学抑制作用,或复合物I的遗传消融(不会触发明显的AMPK激活),消除AMPK介导的铁凋亡抑制作用,并使癌细胞对GPX4失活诱导的铁凋亡敏感。此外,复合物I抑制与放射疗法(RT)协同作用,通过在小鼠模型中诱导铁凋亡来选择性抑制LKB1缺陷型肿瘤的生长。我们的数据证明了复合物I在调节铁凋亡中的多方面作用,并提出了针对LKB1缺陷型癌症的铁凋亡诱导治疗策略。
    The role of the mitochondrial electron transport chain (ETC) in regulating ferroptosis is not fully elucidated. Here, we reveal that pharmacological inhibition of the ETC complex I reduces ubiquinol levels while decreasing ATP levels and activating AMP-activated protein kinase (AMPK), the two effects known for their roles in promoting and suppressing ferroptosis, respectively. Consequently, the impact of complex I inhibitors on ferroptosis induced by glutathione peroxidase 4 (GPX4) inhibition is limited. The pharmacological inhibition of complex I in LKB1-AMPK-inactivated cells, or genetic ablation of complex I (which does not trigger apparent AMPK activation), abrogates the AMPK-mediated ferroptosis-suppressive effect and sensitizes cancer cells to GPX4-inactivation-induced ferroptosis. Furthermore, complex I inhibition synergizes with radiotherapy (RT) to selectively suppress the growth of LKB1-deficient tumors by inducing ferroptosis in mouse models. Our data demonstrate a multifaceted role of complex I in regulating ferroptosis and propose a ferroptosis-inducing therapeutic strategy for LKB1-deficient cancers.
    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
    臭氧(O3)是直接威胁呼吸系统的主要空气污染物,肺脂肪酸代谢紊乱是肺部炎症性疾病的重要分子事件。肝激酶B1(LKB1)和核苷酸结合域富含亮氨酸的重复蛋白3(NLRP3)炎症小体不仅调节炎症,还与脂肪酸代谢密切相关。然而,LKB1和NLRP3炎性体在肺脂肪酸代谢中的作用及机制,这可能会导致臭氧引起的肺部炎症,仍不清楚,缺乏预防O3诱导的肺部炎症损伤的有效策略。为了探索这些,小鼠暴露于1.00ppmO3(3小时/天,5days),肺部炎症是由气道高反应性决定的,组织病理学检查,支气管肺泡灌洗液(BALF)中的总细胞和细胞因子。采用靶向脂肪酸代谢组学技术检测肺组织中、长脂肪酸。然后,利用LKB1过表达腺病毒和NLRP3基因敲除(NLRP3-/-)小鼠探讨O3诱导肺脂肪酸代谢紊乱的机制。结果表明,O3暴露引起肺部炎症损伤和肺中长链脂肪酸代谢紊乱,尤其是降低二高-γ-亚麻酸(DGLA)。同时,LKB1表达降低,O3暴露后,小鼠肺部的NLRP3炎性体被激活。此外,LKB1过表达减轻O3诱导的肺部炎症并抑制NLRP3炎性体的活化。我们发现,与O3暴露后的宽型小鼠相比,NLRP3-/-小鼠的肺脂肪酸代谢紊乱得到了改善。此外,在暴露于O3之前气管内施用DGLA可显着减轻O3诱导的肺部炎症损伤。一起来看,这些发现表明脂肪酸代谢紊乱与O3诱导的肺部炎症有关,受LKB1介导的NLRP3通路调节,DGLA补充剂可能是改善臭氧相关肺部炎症损伤的有效预防策略。
    Ozone (O3) is a major air pollutant that directly threatens the respiratory system, lung fatty acid metabolism disorder is an important molecular event in pulmonary inflammatory diseases. Liver kinase B1 (LKB1) and nucleotide-binding domain leucine-rich repeat-containing protein 3 (NLRP3) inflammasome not only regulate inflammation, but also have close relationship with fatty acid metabolism. However, the role and mechanism of LKB1 and NLRP3 inflammasome in lung fatty acid metabolism, which may contribute to ozone-induced lung inflammation, remain unclear, and effective strategy for preventing O3-induced pulmonary inflammatory injury is lacking. To explore these, mice were exposed to 1.00 ppm O3 (3 h/d, 5 days), and pulmonary inflammation was determined by airway hyperresponsiveness, histopathological examination, total cells and cytokines in bronchoalveolar lavage fluid (BALF). Targeted fatty acids metabolomics was used to detect medium and long fatty acid in lung tissue. Then, using LKB1-overexpressing adenovirus and NLRP3 knockout (NLRP3-/-) mice to explore the mechanism of O3-induced lung fatty acid metabolism disorder. Results demonstrated that O3 exposure caused pulmonary inflammatory injury and lung medium and long chain fatty acids metabolism disorder, especially decreased dihomo-γ-linolenic acid (DGLA). Meanwhile, LKB1 expression was decreased, and NLRP3 inflammasome was activated in lung of mice after O3 exposure. Additionally, LKB1 overexpression alleviated O3-induced lung inflammation and inhibited the activation of NLRP3 inflammasome. And we found that pulmonary fatty acid metabolism disorder was ameliorated of NLRP3 -/- mice compared with those in wide type mice after O3 exposure. Furthermore, administrating DGLA intratracheally prior to O3 exposure significantly attenuated O3-induced pulmonary inflammatory injury. Taken together, these findings suggest that fatty acids metabolism disorder is involved in O3-induced pulmonary inflammation, which is regulated by LKB1-mediated NLRP3 pathway, DGLA supplement could be a useful preventive strategy to ameliorate ozone-associated lung inflammatory injury.
    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
    肿瘤抑制因子LKB1是一种丝氨酸/苏氨酸蛋白激酶,在人肺腺癌(LUAD)中经常发生突变。LKB1调节已知控制细胞极性和代谢的复杂信号网络;然而,介导LKB1肿瘤抑制活性的途径尚未完全确定.为了确定LKB1介导的生长抑制的机制,我们开发了一种基于球体的细胞培养试验来研究LKB1依赖性生长。然后,我们在球形培养物中进行了全基因组CRISPR筛选,发现LKB1抑制生长,在某种程度上,通过激活PIKFYVE脂质激酶。最后,我们使用化学抑制剂和pH敏感报告子确定LKB1通过以PIKFYVE依赖性方式促进野生型EGFR的内在化而损害生长.
    The tumor suppressor LKB1 is a serine/threonine protein kinase that is frequently mutated in human lung adenocarcinoma (LUAD). LKB1 regulates a complex signaling network that is known to control cell polarity and metabolism; however, the pathways that mediate the tumor-suppressive activity of LKB1 are incompletely defined. To identify mechanisms of LKB1-mediated growth suppression, we developed a spheroid-based cell culture assay to study LKB1-dependent growth. We then performed genome-wide CRISPR screens in spheroidal culture and found that LKB1 suppresses growth, in part, by activating the PIKFYVE lipid kinase. Finally, we used chemical inhibitors and a pH-sensitive reporter to determine that LKB1 impairs growth by promoting the internalization of wild-type EGFR in a PIKFYVE-dependent manner.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号