ras Proteins

ras 蛋白质
  • 文章类型: Journal Article
    Ras超家族的Ras和Rap小GTP酶作为分子开关来控制作为不同信号传导途径的一部分的不同细胞过程。Dictyostelium表达几种Ras和Rap蛋白,他们的研究已经并将继续大大有助于我们理解他们在真核生物中的作用。为了研究网菌属Ras和Rap蛋白的活性,已经开发了几种基于它们与已知真核Ras/Rap效应子的Ras结合域相互作用的测定法,并证明对研究它们的调节和细胞作用非常有用。这里,我们描述了使用下拉测定法和使用荧光报道分子通过活细胞成像生化评估Ras/Rap活性的方法。
    Ras and Rap small GTPases of the Ras superfamily act as molecular switches to control diverse cellular processes as part of different signaling pathways. Dictyostelium expresses several Ras and Rap proteins, and their study has and continues to greatly contribute to our understanding of their role in eukaryote biology. To study the activity of Ras and Rap proteins in Dictyostelium, several assays based on their interaction with the Ras binding domain of known eukaryotic Ras/Rap effectors have been developed and proved extremely useful to study their regulation and cellular roles. Here, we describe methods to assess Ras/Rap activity biochemically using a pull-down assay and through live-cell imaging using fluorescent reporters.
    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
    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
    EGFR和HER2/neu作为生长因子受体的改变以及RAS/RAF/MAP激酶的细胞质信号转导蛋白,包括其末端效应分子(ERK)在许多肿瘤的致癌作用中很重要。前列腺癌中这些原癌基因的激活仍在研究中。这项工作的目的是研究EGFR,HER2-neu,与临床和病理参数相关的前列腺腺癌中无活性(非磷酸化)和活性(磷酸化)ERK表达。
    方法:免疫组织化学-使用组织微阵列-用于EGFR,HER2/neu,非磷酸化,和荧光粉ERK,对166例未经治疗的原发性前列腺腺癌患者的组织进行了研究。将不同标志物表达结果与临床病理参数进行相关性分析。
    结果:前列腺组织显示EGFR,HER2neu,磷酸化和非磷酸化ERK表达为8.4%,1.4%,78.2%,无论是低表达(斑片状)还是高表达(弥漫性),分别为83.4%。在患者特征和测试标志物的表达之间没有发现显著的相关性。非磷酸化ERK和EGFR-的负免疫反应性与高肿瘤分期显着相关(p值分别为0.03和0.01)。
    结论:EGFR和HER2/neu可能在前列腺腺癌中起着有限的作用,因为它们在有限数量的检查组织中显示出阳性表达,特别是HER2neu。在大多数情况下,非磷酸化ERK(主要是弱到中等)和磷酸化ERK(主要是中等到强)的表达受到赞赏。因此,我们认为抗EGFR药物在去势抵抗性前列腺癌的治疗中可能具有有限的作用,但抗MEK/ERK药物作为靶向治疗可能具有更有希望的作用.建议进行进一步的分子测试以阐明这些标记的确切机制和意义。
    The alterations of EGFR and HER2/neu as growth factor receptors and the cytoplasmic signal transduction proteins of RAS/RAF/MAP kinases including its end effector molecule (ERK) are important in the carcinogenesis of many tumors. The activation of these protooncogenes in prostate cancer is still under investigation. The aim of this work was to study EGFR, HER2- neu, inactive (non-phosphorylated) and active (phosphorylated) ERK expression in prostatic adenocarcinomas in correlation to the clinical and pathological parameters.
    METHODS: Immunohistochemistry- using tissue microarrays- for EGFR, HER2/neu, non-phosphorylated, and phosphor-ERK, was performed on tissues from 166 patients- with primary prostatic adenocarcinoma with no prior treatment-. The results of different markers expression were correlated with the clinical and pathological parameters and were analyzed statistically.
    RESULTS: The prostatic tissue showed EGFR, HER2 neu, phosphorylated and non-phosphorylated ERK expression in 8.4%, 1.4%, 78.2%, and 83.4% respectively whether low (patchy) or high expression (diffuse).  There were no significant correlations found between patient characteristics and expression of the tested markers. The negative immune reactivity for non-phosphorylated ERK and EGFR- was significantly correlated with high tumor stage (p values 0.03 and 0.01, respectively).
    CONCLUSIONS: EGFR and HER2/neu may play a limited role in prostatic adenocarcinoma as they showed positive expression in a limited number of the examined tissues specifically HER2neu. The expression of non-phosphorylated ERK (mostly weak to moderate) and phosphorylated ERK (mostly moderate to strong)- was appreciated in most cases. Thus, we suggest that anti-EGFR drugs may have a limited role in the treatment of castrate-resistant prostate cancer, but anti-MEK/ERK drugs may have more promising role as a target therapy. It is recommended to perform further molecular testing to elucidate the exact mechanism and significance of these markers.
    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
    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: Journal Article
    Ras小GTP酶在各种细胞信号通路中充当分子开关,包括细胞迁移,扩散,和差异化。三种Rap蛋白存在于网藻中;RapA,拉普,还有RapC.据报道RapA和RapC在控制细胞粘附和迁移方面具有相反的功能。这里,我们调查了RapB的作用,网菌属RasGTPase亚家族的成员,专注于它参与细胞粘附,迁移,和发展过程。这项研究显示RapB,类似于RapA,在调节细胞形态中起着至关重要的作用,附着力,和移民。rapB空细胞,它们是由CRISPR/Cas9基因编辑产生的,显示更改的单元格大小,减少细胞-基质粘附,趋化过程中的迁移速度增加。通过RapB和RapA的表达恢复了这些rapB无效细胞的表型,但不是RapC.与这些结果一致,拉普,类似于RapA,未能挽救rapC无效细胞的表型,扩散形态,细胞粘附增加,趋化过程中迁移速度降低。rapB无效细胞的多细胞发育未受影响。这些结果表明RapB参与控制细胞形态和细胞粘附。重要的是,RapB在趋化过程中似乎在调节迁移速度中起抑制作用,可能通过控制细胞-基质粘附,类似RapA的功能。这些发现有助于理解Ras亚家族蛋白之间的功能关系。
    Ras small GTPases act as molecular switches in various cellular signaling pathways, including cell migration, proliferation, and differentiation. Three Rap proteins are present in Dictyostelium; RapA, RapB, and RapC. RapA and RapC have been reported to have opposing functions in the control of cell adhesion and migration. Here, we investigated the role of RapB, a member of the Ras GTPase subfamily in Dictyostelium, focusing on its involvement in cell adhesion, migration, and developmental processes. This study revealed that RapB, similar to RapA, played a crucial role in regulating cell morphology, adhesion, and migration. rapB null cells, which were generated by CRISPR/Cas9 gene editing, displayed altered cell size, reduced cell-substrate adhesion, and increased migration speed during chemotaxis. These phenotypes of rapB null cells were restored by the expression of RapB and RapA, but not RapC. Consistent with these results, RapB, similar to RapA, failed to rescue the phenotypes of rapC null cells, spread morphology, increased cell adhesion, and decreased migration speed during chemotaxis. Multicellular development of rapB null cells remained unaffected. These results suggest that RapB is involved in controlling cell morphology and cell adhesion. Importantly, RapB appears to play an inhibitory role in regulating the migration speed during chemotaxis, possibly by controlling cell-substrate adhesion, resembling the functions of RapA. These findings contribute to the understanding of the functional relationships among Ras subfamily proteins.
    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
    鸟嘌呤核苷酸交换因子(GEF)调节控制细胞功能的Ras超家族蛋白的小GTP酶(G蛋白)的激活。Ras超家族蛋白质充当“分子开关”,通过鸟嘌呤交换打开。有五组主要的Ras家族GTPases:Ras,Ran,Rho,Rab和Arf,用多种不同的GEF调节它们的GTP负载。GEF与包括癌症在内的各种疾病有关。这使得GEF具有吸引力的靶标来调节由小GTP酶控制的信号网络。
    在这篇评论中,概述了GEF在恶性肿瘤中的作用和机制。说明了GEF对小GTP酶的鸟嘌呤交换活性的机制。然后,介绍了一些在癌症中具有重要意义的GEF的例子,并讨论了使用各种方法进行靶向治疗的最新进展.
    最近,GEF已成为新型癌症药物开发的潜在治疗靶标。瞄准小的GTPases是具有挑战性的;因此,通过GEF靶向它们的激活是一个有前途的策略。大多数GEF靶向药物仍处于临床前开发中。对GEF活动的潜在机制和利用先进技术的更深入的生物学理解对于增强癌症中GEF的药物发现是必要的。
    UNASSIGNED: Guanine nucleotide exchange factors (GEFs) regulate the activation of small GTPases (G proteins) of the Ras superfamily proteins controlling cellular functions. Ras superfamily proteins act as \'molecular switches\' that are turned \'ON\' by guanine exchange. There are five major groups of Ras family GTPases: Ras, Ran, Rho, Rab and Arf, with a variety of different GEFs regulating their GTP loading. GEFs have been implicated in various diseases including cancer. This makes GEFs attractive targets to modulate signaling networks controlled by small GTPases.
    UNASSIGNED: In this review, the roles and mechanisms of GEFs in malignancy are outlined. The mechanism of guanine exchange activity by GEFs on a small GTPase is illustrated. Then, some examples of GEFs that are significant in cancer are presented with a discussion on recent progress in therapeutic targeting efforts using a variety of approaches.
    UNASSIGNED: Recently, GEFs have emerged as potential therapeutic targets for novel cancer drug development. Targeting small GTPases is challenging; thus, targeting their activation by GEFs is a promising strategy. Most GEF-targeted drugs are still in preclinical development. A deeper biological understanding of the underlying mechanisms of GEF activity and utilizing advanced technology are necessary to enhance drug discovery for GEFs in cancer.
    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: 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
    突变选择性KRAS抑制剂的开发代表了重大的治疗进展;然而,患者可以通过RAS途径的反馈机制和遗传改变产生耐药性.Nature和CancerDiscovery中的三份出版物描述了一种有前途的RAS(ON)多选择性抑制剂,该抑制剂同时靶向致癌RAS和多种潜在的抗性机制,同时保留正常组织。
    The development of mutant-selective KRAS inhibitors represents a major therapeutic advance; however, patients can develop resistance through feedback mechanisms and genetic alterations in the RAS pathway. Three publications in Nature and Cancer Discovery describe a promising RAS(ON) multi-selective inhibitor that simultaneously targets oncogenic RAS and multiple potential resistance mechanisms while sparing normal tissue.
    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
    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)

公众号