ras Proteins

ras 蛋白质
  • 文章类型: Journal Article
    背景:RAS基因的致癌突变与不受控制的细胞生长有关,有助于肿瘤发生的标志特征。虽然不同的治疗策略已经努力应用于治疗RAS突变癌症,成功靶向RAS基因仍然是癌症治疗领域的持续挑战.在我们的研究中,我们发现了一个有希望的途径来应对这一挑战。
    方法:在本研究中,我们测试了几种携带致癌NRAS的细胞系的活力,KRAS,和用IkappaBalpha(IκBα)抑制剂BAY11-7082治疗后的HRAS突变。我们进行了基于细胞培养的活力测定和基于体内皮下异种移植物的测定,以证实BAY11-7082的生长抑制作用。我们还进行了大型RNA测序分析,以确定在致癌NRAS的背景下差异调节的基因和途径。KRAS,和用BAY11-7082治疗后的HRAS突变。
    结果:我们证明了致癌NRAS,KRAS,HRAS激活IκBα激酶的表达。BAY11-7082,IκBα激酶抑制剂,减弱NRAS的生长,KRAS,细胞培养和小鼠模型中的HRAS突变癌细胞。机械上,BAY11-7082抑制剂处理导致所有RAS突变细胞系中PI3K-AKT信号传导途径的抑制和细胞凋亡的激活。此外,我们发现BAY11-7082治疗会导致不同生物学途径的下调,这取决于RAS蛋白的类型,这也可能有助于肿瘤生长抑制。
    结论:我们的研究确定BAY11-7082是治疗RAS癌基因的有效抑制剂(HRAS,KRAS,和NRAS)突变的癌细胞。这一发现为有效治疗RAS突变癌症提供了新的治疗机会。
    BACKGROUND: Oncogenic mutations in the RAS gene are associated with uncontrolled cell growth, a hallmark feature contributing to tumorigenesis. While diverse therapeutic strategies have been diligently applied to treat RAS-mutant cancers, successful targeting of the RAS gene remains a persistent challenge in the field of cancer therapy. In our study, we discover a promising avenue for addressing this challenge.
    METHODS: In this study, we tested the viability of several cell lines carrying oncogenic NRAS, KRAS, and HRAS mutations upon treatment with IkappaBalpha (IκBα) inhibitor BAY 11-7082. We performed both cell culture-based viability assay and in vivo subcutaneous xenograft-based assay to confirm the growth inhibitory effect of BAY 11-7082. We also performed large RNA sequencing analysis to identify differentially regulated genes and pathways in the context of oncogenic NRAS, KRAS, and HRAS mutations upon treatment with BAY 11-7082.
    RESULTS: We demonstrate that oncogenic NRAS, KRAS, and HRAS activate the expression of IκBα kinase. BAY 11-7082, an inhibitor of IκBα kinase, attenuates the growth of NRAS, KRAS, and HRAS mutant cancer cells in cell culture and in mouse model. Mechanistically, BAY 11-7082 inhibitor treatment leads to suppression of the PI3K-AKT signaling pathway and activation of apoptosis in all RAS mutant cell lines. Additionally, we find that BAY 11-7082 treatment results in the downregulation of different biological pathways depending upon the type of RAS protein that may also contribute to tumor growth inhibition.
    CONCLUSIONS: Our study identifies BAY 11-7082 to be an efficacious inhibitor for treating RAS oncogene (HRAS, KRAS, and NRAS) mutant cancer cells. This finding provides new therapeutic opportunity for effective treatment of RAS-mutant 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
    鼻咽癌(NPC),主要发现于中国南部地区,是一种以高度转移特性而闻名的恶性肿瘤。由远处转移和疾病复发引起的高死亡率仍然是临床上尚未解决的问题。在临床上,黄连素(BBR)化合物已广泛用于鼻咽癌治疗,以减少转移和疾病复发,并且BBR被记录为具有多种抗NPC作用的主要成分。然而,BBR抑制鼻咽癌生长和转移的机制尚不清楚。在这里,我们表明,BBR有效地抑制了生长,转移,并通过诱导特异性超级增强子(SE)入侵NPC。从机械的角度来看,RNA测序(RNA-seq)结果表明RAS-RAF1-MEK1/2-ERK1/2信号通路,由表皮生长因子受体(EGFR)激活,在BBR诱导的NPC自噬中起重要作用。自噬的阻断显著减弱了BBR介导的NPC细胞生长和转移抑制的作用。值得注意的是,BBR通过转录增加EGFR的表达,和敲除EGFR显著抑制BBR诱导的微管相关蛋白1轻链3(LC3)-II的增加和p62抑制,提示EGFR在BBR诱导的NPC自噬中起关键作用。染色质免疫沉淀测序(ChIP-seq)结果发现,仅在BBR处理的NPC细胞中存在特异性SE。这种SE敲除明显抑制了EGFR和磷酸化EGFR(EGFR-p)的表达,并逆转了BBR对NPC增殖的抑制作用。转移,和入侵。此外,BBR特异性SE可能通过增强EGFR基因转录触发自噬,从而上调RAS-RAF1-MEK1/2-ERK1/2信号通路。此外,体内BBR有效抑制NPC细胞生长和转移,随着LC3和EGFR的增加和p62的减少。总的来说,这项研究确定了一种新的BBR-特殊SE,并建立了一种新的表观遗传范式,BBR调节自噬,抑制增殖,转移,和入侵。它为BBR作为未来NPC治疗中的治疗方案的应用提供了理论基础。
    Nasopharyngeal carcinoma (NPC), primarily found in the southern region of China, is a malignant tumor known for its highly metastatic characteristics. The high mortality rates caused by the distant metastasis and disease recurrence remain unsolved clinical problems. In clinic, the berberine (BBR) compound has widely been in NPC therapy to decrease metastasis and disease recurrence, and BBR was documented as a main component with multiple anti-NPC effects. However, the mechanism by which BBR inhibits the growth and metastasis of nasopharyngeal carcinoma remains elusive. Herein, we show that BBR effectively inhibits the growth, metastasis, and invasion of NPC via inducing a specific super enhancer (SE). From a mechanistic perspective, the RNA sequencing (RNA-seq) results suggest that the RAS-RAF1-MEK1/2-ERK1/2 signaling pathway, activated by the epidermal growth factor receptor (EGFR), plays a significant role in BBR-induced autophagy in NPC. Blockading of autophagy markedly attenuated the effect of BBR-mediated NPC cell growth and metastasis inhibition. Notably, BBR increased the expression of EGFR by transcription, and knockout of EGFR significantly inhibited BBR-induced microtubule associated protein 1 light chain 3 (LC3)-II increase and p62 inhibition, proposing that EGFR plays a pivotal role in BBR-induced autophagy in NPC. Chromatin immunoprecipitation sequencing (ChIP-seq) results found that a specific SE existed only in NPC cells treated with BBR. This SE knockdown markedly repressed the expression of EGFR and phosphorylated EGFR (EGFR-p) and reversed the inhibition of BBR on NPC proliferation, metastasis, and invasion. Furthermore, BBR-specific SE may trigger autophagy by enhancing EGFR gene transcription, thereby upregulating the RAS-RAF1-MEK1/2-ERK1/2 signaling pathway. In addition, in vivo BBR effectively inhibited NPC cells growth and metastasis, following an increase LC3 and EGFR and a decrease p62. Collectively, this study identifies a novel BBR-special SE and established a new epigenetic paradigm, by which BBR regulates autophagy, inhibits proliferation, metastasis, and invasion. It provides a rationale for BBR application as the treatment regime in NPC therapy in future.
    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
    Ras是一种小的GTP酶,对不同细胞类型的重要功能决定至关重要。Ras信号传导的一个重要方面是其表现出双峰或开关样活性的能力。我们描述了在支持的脂质膜微阵列上由SOS和p120-RasGAP催化的受体介导的Ras激活-失活反应的完全重建。结果显示了双峰Ras激活反应,这不是确定性双稳态的结果,而是由Ras活化剂的独特的可加工性驱动的,求救信号.此外,双峰反应由支架蛋白的缩合状态控制,LAT,SOS被招募的人。即使在强烈的去激活条件下,过程性驱动的双峰性也会导致Ras激活的随机爆发。这种行为与确定性双稳态相反,并且可能对药理学抑制更具抵抗力。
    Ras is a small GTPase that is central to important functional decisions in diverse cell types. An important aspect of Ras signaling is its ability to exhibit bimodal or switch-like activity. We describe the total reconstitution of a receptor-mediated Ras activation-deactivation reaction catalyzed by SOS and p120-RasGAP on supported lipid membrane microarrays. The results reveal a bimodal Ras activation response, which is not a result of deterministic bistability but is rather driven by the distinct processivity of the Ras activator, SOS. Furthermore, the bimodal response is controlled by the condensation state of the scaffold protein, LAT, to which SOS is recruited. Processivity-driven bimodality leads to stochastic bursts of Ras activation even under strongly deactivating conditions. This behavior contrasts deterministic bistability and may be more resistant to pharmacological inhibition.
    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
    巨噬细胞吞噬介导细胞外液的非选择性大量摄取,使细胞能够调查环境并获得营养。一组保守的信号蛋白协调了肌动蛋白动力学,导致各种真核生物中的膜皱褶和大克隆体形成。这个信令网络的中心是RasGTPases,其激活有力地刺激巨细胞增多症。然而,在巨噬细胞增多过程中,Ras信号是如何启动和时空调控的,目前尚不清楚。通过使用模型系统Dictyostelium和基于蛋白质组学的方法来鉴定巨细胞增多的调节因子,我们发现了Leep2,由Leep2A和Leep2B组成,作为RasGAP复合体。Leep2复合物专门定位于新兴的大红细胞杯和新生的大红细胞体,其中它通过调节三个Ras家族小GTP酶的活性来调节大克隆体的形成。复合物的缺失或过表达,以及目标RasGTPases的破坏或持续激活,削弱巨噬细胞活动。我们的数据揭示了微调Ras活性在指导巨染色体形成中的关键作用。
    Macropinocytosis mediates the non-selective bulk uptake of extracellular fluid, enabling cells to survey the environment and obtain nutrients. A conserved set of signaling proteins orchestrates the actin dynamics that lead to membrane ruffling and macropinosome formation across various eukaryotic organisms. At the center of this signaling network are Ras GTPases, whose activation potently stimulates macropinocytosis. However, how Ras signaling is initiated and spatiotemporally regulated during macropinocytosis is not well understood. By using the model system Dictyostelium and a proteomics-based approach to identify regulators of macropinocytosis, we uncovered Leep2, consisting of Leep2A and Leep2B, as a RasGAP complex. The Leep2 complex specifically localizes to emerging macropinocytic cups and nascent macropinosomes, where it modulates macropinosome formation by regulating the activities of three Ras family small GTPases. Deletion or overexpression of the complex, as well as disruption or sustained activation of the target Ras GTPases, impairs macropinocytic activity. Our data reveal the critical role of fine-tuning Ras activity in directing macropinosome formation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: English Abstract
    目的:探讨脆性X智力低下蛋白(FMRP)调控结直肠癌(CRC)细胞铁凋亡逃逸的机制。
    方法:我们使用RT-qPCR和Western印迹检测了CRC细胞系中FMRP的表达水平,并使用TCGA数据库分析了FMRP介导的CRC进展调控中涉及的生物学功能和信号通路。构建慢病毒FMRP过表达载体(Lv-FMRP)和3个敲低载体(siFMRP-1、siFMRP-2和siFMRP-3),并使用CCK8法和平板克隆形成法检测其对HCT116细胞增殖的影响;使用MDA/ROS/GSH/Fe2试剂盒测定细胞铁凋亡水平的变化,使用JC-1荧光染色检测线粒体膜电位变化,免疫印迹法检测铁凋亡相关蛋白和RAS/MAPK信号通路的表达。在裸小鼠中评估转染细胞的皮下致瘤潜力。
    结果:与正常结肠黏膜上皮NCM460细胞相比,CRC细胞系具有显著较高的FMRP表达水平。生物信息学分析提示FMRP参与活性氧的调节,氧化应激诱导的细胞死亡,线粒体呼吸,和谷胱甘肽代谢途径。在细胞实验中,FMRP敲低显著抑制HCT116细胞增殖,细胞GSH含量降低,MDA和ROS水平增加,Fe2+荧光强度,和线粒体膜电位,SLC7A11/GPX4蛋白表达和ERK磷酸化水平降低,MEK,MAPK,和RAS蛋白;FMRP过表达导致细胞发生相反的变化。在荷瘤裸鼠中,具有FMRP敲低的HCT116细胞显示出减弱的致瘤潜能,在异种移植物中降低的异种移植物生长速率和降低的SLC7A11表达。
    结论:FMRP高表达抑制CRC细胞铁凋亡,通过激活RAS/MAPK信号通路促进CRC进展。
    OBJECTIVE: To investigate the mechanism by which fragile X mental retardation protein (FMRP) regulates ferroptosis evasion in colorectal cancer (CRC) cells.
    METHODS: We examined FMRP expression levels in CRC cell lines using RT-qPCR and Western blotting and analyzed the biological functions and signaling pathways involved in FMRP-mediated regulation of CRC progression using the TCGA database. A lentiviral FMRP overexpression vector (Lv-FMRP) and 3 knockdown vectors (siFMRP-1, siFMRP-2, and siFMRP-3) were constructed, and their effects on proliferation of HCT116 cells were examined using CCK8 assay and plate clone formation assay; the changes in cell ferroptosis level was determined using MDA/ROS/GSH/Fe2+ kits, mitochondrial membrane potential changes were detected using JC-1 fluorescence staining, and the expressions of proteins associated with ferroptosis and the RAS/MAPK signaling pathway were detected using Western blotting. The subcutaneous tumorigenic potential of the transfected cells was evaluated in nude mice.
    RESULTS: Compared with normal colonic mucosal epithelial NCM460 cells, the CRC cell lines had significantly higher FMRP expression level. Bioinformatics analysis suggested the involvement of FMRP in regulation of reactive oxygen, oxidative stress-induced cell death, mitochondrial respiration, and glutathione metabolism pathways. In the cell experiments, FMRP knockdown significantly inhibited proliferation of HCT116 cells, lowered cellular GSH content, increased MDA and ROS levels, Fe2+ fluorescence intensity, and mitochondrial membrane potential, and decreased SLC7A11/GPX4 protein expressions and the phosphorylation levels of ERK, MEK, MAPK, and RAS proteins; FMRP overexpression resulted in the opposite changes in the cells. In the tumor-bearing nude mice, HCT116 cells with FMRP knockdown showed attenuated tumorigenic potential with lowered xenograft growth rate and reduced SLC7A11 expression in the xenograft.
    CONCLUSIONS: The high expression of FMRP inhibits ferroptosis in CRC cells and promotes progression of CRC by activating the RAS/MAPK signaling pathway.
    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
    放射病是由编码RAS/丝裂原活化蛋白激酶(MAPK)信号转导途径成分的基因中的种系致病变异引起的罕见发育遗传综合征。尽管每种RASopathy综合征的发生率很少,集体,它们代表了多种先天性异常综合征的最大群体之一,并具有严重的发育后果。这里,我们回顾了我们对RAS/MAPK失调如何影响骨骼肌发育的理解,以及RAS/MAPK通路调节对胚胎肌生成的重要性。我们还讨论了该途径与其他细胞内信号通路在骨骼肌发育和生长调节中的复杂相互作用,以及RASopathy动物模型为探索通路抑制剂的使用提供的机会,通常用于癌症治疗,纠正由该通路失调引起的独特骨骼肌病。
    RASopathies are rare developmental genetic syndromes caused by germline pathogenic variants in genes that encode components of the RAS/mitogen-activated protein kinase (MAPK) signal transduction pathway. Although the incidence of each RASopathy syndrome is rare, collectively, they represent one of the largest groups of multiple congenital anomaly syndromes and have severe developmental consequences. Here, we review our understanding of how RAS/MAPK dysregulation in RASopathies impacts skeletal muscle development and the importance of RAS/MAPK pathway regulation for embryonic myogenesis. We also discuss the complex interactions of this pathway with other intracellular signaling pathways in the regulation of skeletal muscle development and growth, and the opportunities that RASopathy animal models provide for exploring the use of pathway inhibitors, typically used for cancer treatment, to correct the unique skeletal myopathy caused by the dysregulation of this pathway.
    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
    维生素B6是一种水溶性维生素,具有抗氧化特性。其催化活性形式,吡哆醛5'-磷酸(PLP),是DNA和氨基酸代谢的关键辅因子。在几项研究中已经观察到维生素B6与癌症风险之间的负相关,尽管膳食维生素B6的摄入有时未能证实这种关联。然而,维生素B6和癌症之间的分子联系仍然难以捉摸。先前的工作表明,维生素B6缺乏会导致果蝇和人类细胞的染色体畸变(CAB),这表明基因组不稳定可能将这种维生素的缺乏与癌症联系起来。在这里,我们提供了支持这一假设的证据。首先,我们表明PLP缺乏,由PLP拮抗剂4-脱氧吡哆醇(4DP)或银杏毒素(GT)诱导,促进眼部幼虫椎间盘的肿瘤发生,将良性RasV12肿瘤转化为侵袭性形式。相比之下,PLP补充减少了肿瘤的发展。我们还表明,低PLP水平,由4DP诱导或通过沉默参与PLP生物合成的sgllPNPO基因,在伴随激活RasV12和下调Discs-large(Dlg)基因而产生的另一种果蝇癌症模型中,肿瘤表型恶化。此外,我们发现在4DP上饲养的幼虫的RasV12眼盘显示CAB,活性氧(ROS)和丝氨酸羟甲基转移酶(SHMT)的低催化活性,一种参与胸苷酸(dTMP)生物合成的PLP依赖性酶,反过来又需要DNA复制和修复。用PLP或抗坏血酸(AA)加dTMP饲喂RasV124DP的幼虫,拯救了CAB和肿瘤。通过在RasV12DlgRNAi4DP喂养的幼虫中过表达过氧化氢酶产生相同的效果,从而允许建立PLP缺乏之间的关系,CAB,和癌症。总的来说,我们的数据提供了第一个体内证明PLP缺乏可以通过增加基因组不稳定性来影响癌症,其又由ROS和降低的dTMP水平介导。
    Vitamin B6 is a water-soluble vitamin which possesses antioxidant properties. Its catalytically active form, pyridoxal 5\'-phosphate (PLP), is a crucial cofactor for DNA and amino acid metabolism. The inverse correlation between vitamin B6 and cancer risk has been observed in several studies, although dietary vitamin B6 intake sometimes failed to confirm this association. However, the molecular link between vitamin B6 and cancer remains elusive. Previous work has shown that vitamin B6 deficiency causes chromosome aberrations (CABs) in Drosophila and human cells, suggesting that genome instability may correlate the lack of this vitamin to cancer. Here we provide evidence in support of this hypothesis. Firstly, we show that PLP deficiency, induced by the PLP antagonists 4-deoxypyridoxine (4DP) or ginkgotoxin (GT), promoted tumorigenesis in eye larval discs transforming benign RasV12 tumors into aggressive forms. In contrast, PLP supplementation reduced the development of tumors. We also show that low PLP levels, induced by 4DP or by silencing the sgllPNPO gene involved in PLP biosynthesis, worsened the tumor phenotype in another Drosophila cancer model generated by concomitantly activating RasV12 and downregulating Discs-large (Dlg) gene. Moreover, we found that RasV12 eye discs from larvae reared on 4DP displayed CABs, reactive oxygen species (ROS) and low catalytic activity of serine hydroxymethyltransferase (SHMT), a PLP-dependent enzyme involved in thymidylate (dTMP) biosynthesis, in turn required for DNA replication and repair. Feeding RasV12 4DP-fed larvae with PLP or ascorbic acid (AA) plus dTMP, rescued both CABs and tumors. The same effect was produced by overexpressing catalase in RasV12 DlgRNAi 4DP-fed larvae, thus allowing to establish a relationship between PLP deficiency, CABs, and cancer. Overall, our data provide the first in vivo demonstration that PLP deficiency can impact on cancer by increasing genome instability, which is in turn mediated by ROS and reduced dTMP levels.
    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
    转染期间重排(RET)重排癌蛋白介导的Ras/MAPK信号级联在癌症中组成性激活。这里,我们展示了一个独特的信号生态位。生态位是基于嵌合RET液-液相分离的三元复合物。复合物包含重排的激酶(RET融合);衔接子(GRB2),和效应子(SHC1)。一起,他们协调Ras/MAPK信号级联,依赖于酪氨酸激酶。CCDC6-RET融合经历LLPS,需要其激酶结构域和其融合配偶体。CCDC6-RET融合LLPS促进RET融合的自磷酸化,具有增强的激酶活性,这是信号生态位形成所必需的。在信号利基内,组成成分之间的相互作用得到加强,并且信号转导效率被放大。特定的RET融合相关信号生态位阐明了Ras/MAPK信号通路组成型激活的机制。除了关注RET融合本身,三元复合物的探索可能为设计旨在治疗RET融合驱动疾病的治疗策略提供了有希望的途径。
    Rearranged during transfection (RET) rearrangement oncoprotein-mediated Ras/MAPK signaling cascade is constitutively activated in cancers. Here, we demonstrate a unique signal niche. The niche is a ternary complex based on the chimeric RET liquid-liquid phase separation. The complex comprises the rearranged kinase (RET fusion); the adaptor (GRB2), and the effector (SHC1). Together, they orchestrate the Ras/MAPK signal cascade, which is dependent on tyrosine kinase. CCDC6-RET fusion undergoes LLPS requiring its kinase domain and its fusion partner. The CCDC6-RET fusion LLPS promotes the autophosphorylation of RET fusion, with enhanced kinase activity, which is necessary for the formation of the signaling niche. Within the signal niche, the interactions among the constituent components are reinforced, and the signal transduction efficiency is amplified. The specific RET fusion-related signal niche elucidates the mechanism of the constitutive activation of the Ras/MAPK signaling pathway. Beyond just focusing on RET fusion itself, exploration of the ternary complex potentially unveils a promising avenue for devising therapeutic strategies aimed at treating RET fusion-driven diseases.
    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
    大约18%的人类癌症携带KRAS基因突变,使其成为最受欢迎的抗癌靶标之一。然而,突变的KRas蛋白已被证明是非常不可用的。因此,第一代RAS抑制剂的最近批准标志着癌症研究史上的一个开创性里程碑。然而不可避免的是,它还提出了可预测的挑战,即有限的药物疗效和获得性耐药性。因此,在更多的生理环境中提高我们对致癌RAS的致瘤机制的理解的新方法仍然是必不可少的。这里,我们利用近二倍体人hTERTRPE-1细胞产生等基因细胞系,其中一个内源性KRAS等位基因在甘氨酸12处携带致癌KRAS突变.带有KRASG12V/+的细胞,KRASG12C/+,或KRASG12D/+基因型,连同野生型KRASG12G(WT)/+细胞,揭示了致癌KRAS。G12X突变增加KRASG12V/+细胞中的细胞增殖率和细胞运动性并减少粘着斑。EGF诱导的ERK和AKT磷酸化在KRASG12V/+之间相当,KRASG12C/+,KRASG12D/+,和KRASG12G(WT)/+细胞。有趣的是,KRASG12X/+细胞对不同的抑制剂表现出不同的反应,KRASG12V/+和KRASG12D/+细胞对羟基脲和MEK抑制剂更敏感,U0126和曲美替尼,但对PI3K抑制剂更有抗性,PIK-90,比KRASG12G(WT)/+细胞。低剂量的羟基脲和U0126的组合显示出在KRASG12V/+中对生长速率的相加抑制大于野生型细胞。总的来说,这些细胞系将是研究致癌RAS信号传导和开发对野生型细胞具有最小细胞毒性的有效抗KRAS试剂的宝贵资源。
    About 18% of all human cancers carry a mutation in the KRAS gene making it among the most sought-after anticancer targets. However, mutant KRas protein has proved remarkably undruggable. The recent approval of the first generation of RAS inhibitors therefore marks a seminal milestone in the history of cancer research. It also raises the predictable challenges of limited drug efficacies and acquired resistance. Hence, new approaches that improve our understanding of the tumorigenic mechanisms of oncogenic RAS within more physiological settings continue to be essential. Here, we have used the near-diploid hTERT RPE-1 cells to generate isogenic cell lines in which one of the endogenous KRAS alleles carries an oncogenic KRAS mutation at glycine 12. Cells with a KRASG12V/+, KRASG12C/+, or KRASG12D/+ genotype, together with WT KRASG12G(WT)/+ cells, reveal that oncogenic KRAS.G12X mutations increase cell proliferation rate and cell motility and reduced focal adhesions in KRASG12V/+ cells. Epidermal growth factor -induced phosphorylation of ERK and AKT was comparable between KRASG12V/+, KRASG12C/+, KRASG12D/+, and KRASG12G(WT)/+ cells. Interestingly, KRASG12X/+ cells showed varying responses to distinct inhibitors with the KRASG12V/+ and KRASG12D/+ cells more sensitive to hydroxyurea and MEK inhibitors, U0126 and trametinib, but more resistant to PI3K inhibitor, PIK-90, than the KRASG12G(WT)/+ cells. A combination of low doses of hydroxyurea and U0126 showed an additive inhibition on growth rate that was greater in KRASG12V/+ than WT cells. Collectively, these cell lines will be a valuable resource for studying oncogenic RAS signaling and developing effective anti-KRAS reagents with minimum cytotoxicity on WT 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
    前列腺癌谱系可塑性是向神经内分泌前列腺癌(NEPC)过渡的关键驱动因素,和RTK/RAS信号通路是一个成熟的癌症通路。然而,RTK/RAS信号通路与谱系可塑性之间的综合联系受到了有限的研究.特别是,控制RTK/RAS和谱系可塑性之间相互作用的复杂监管网络在很大程度上仍未被探索。采用自变量系数和邻居连接算法对多组数据进行聚类。随后,利用GSEA对聚类结果进行了分析,与干性相关的基因集,多谱系状态数据集,和典型的癌症通路基因集。最后,基于ssGSEA的数据的全面探索,WGCNA,GSEA,VIPER,前列腺癌scRNA-seq数据,并进行了GPSAdb数据库。在聚类结果的六个模块中,有300个重叠的基因,包括3个以前未报告的前列腺癌基因,这些基因通过RT-qPCR验证在前列腺癌中上调。功能模块6显示与前列腺癌细胞干细胞呈正相关,多谱系状态,和RTK/RAS信号通路。此外,RTK/RAS信号通路的19个前沿基因通过转录调控和拷贝数变异的复杂网络促进前列腺癌谱系可塑性.在转录调控网络中,TP63和FOXO1作为前列腺癌谱系可塑性的抑制剂,而RORC发挥促进作用。本研究为RTK/RAS通路在前列腺癌谱系可塑性中的作用提供了全面的视角,并为NEPC的治疗提供了新的线索。
    Prostate cancer lineage plasticity is a key driver in the transition to neuroendocrine prostate cancer (NEPC), and the RTK/RAS signaling pathway is a well-established cancer pathway. Nevertheless, the comprehensive link between the RTK/RAS signaling pathway and lineage plasticity has received limited investigation. In particular, the intricate regulatory network governing the interplay between RTK/RAS and lineage plasticity remains largely unexplored. The multi-omics data were clustered with the coefficient of argument and neighbor joining algorithm. Subsequently, the clustered results were analyzed utilizing the GSEA, gene sets related to stemness, multi-lineage state datasets, and canonical cancer pathway gene sets. Finally, a comprehensive exploration of the data based on the ssGSEA, WGCNA, GSEA, VIPER, prostate cancer scRNA-seq data, and the GPSAdb database was conducted. Among the six modules in the clustering results, there are 300 overlapping genes, including 3 previously unreported prostate cancer genes that were validated to be upregulated in prostate cancer through RT-qPCR. Function Module 6 shows a positive correlation with prostate cancer cell stemness, multi-lineage states, and the RTK/RAS signaling pathway. Additionally, the 19 leading-edge genes of the RTK/RAS signaling pathway promote prostate cancer lineage plasticity through a complex network of transcriptional regulation and copy number variations. In the transcriptional regulation network, TP63 and FOXO1 act as suppressors of prostate cancer lineage plasticity, whereas RORC exerts a promoting effect. This study provides a comprehensive perspective on the role of the RTK/RAS pathway in prostate cancer lineage plasticity and offers new clues for the treatment of NEPC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号