ERK

ERK
  • 文章类型: Journal Article
    从Eutremajaponicum中分离出的异硫氰酸甲基亚磺基己酯(6-MSITC)是治疗乳腺癌的有希望的候选药物,结直肠癌和胃癌,代谢综合征,心脏病,糖尿病,和肥胖,由于其抗炎和抗氧化特性。此外,它的神经保护特性,改善认知功能和保护多巴胺能神经元,使其成为治疗神经退行性疾病如痴呆症的优秀候选药物,老年痴呆症,和帕金森病。6-MSITC作用于许多信号通路,如PPAR,AMPK,PI3K/AKT/mTOR,Nrf2/Keap1-ARE,ERK1/2-ELK1/CHOP/DR5和MAPK。然而,尽管体外和体内动物研究以及一些人体研究取得了非常有希望的结果,该分子尚未在人群中进行彻底测试。尽管如此,芥末应被归类为人类疾病一级和二级预防的“超级食品”。本文回顾了当前关于6-MSITC的最新研究及其潜在的临床应用,详细讨论了该分子激活的信号通路及其相互作用。
    Methylsulfinyl hexyl isothiocyanate (6-MSITC) isolated from Eutrema japonicum is a promising candidate for the treatment of breast cancer, colorectal and stomach cancer, metabolic syndrome, heart diseases, diabetes, and obesity due to its anti-inflammatory and antioxidant properties. Also, its neuroprotective properties, improving cognitive function and protecting dopaminergic neurons, make it an excellent candidate for treating neurodegenerative diseases like dementia, Alzheimer\'s, and Parkinson\'s disease. 6-MSITC acts on many signaling pathways, such as PPAR, AMPK, PI3K/AKT/mTOR, Nrf2/Keap1-ARE, ERK1/2-ELK1/CHOP/DR5, and MAPK. However, despite the very promising results of in vitro and in vivo animal studies and a few human studies, the molecule has not yet been thoroughly tested in the human population. Nonetheless, wasabi should be classified as a \"superfood\" for the primary and secondary prevention of human diseases. This article reviews the current state-of-the-art research on 6-MSITC and its potential clinical uses, discussing in detail the signaling pathways activated by the molecule and their interactions.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Retraction of Publication
    在本文发表之后,一位关心的读者注意到,图中所示的某些西方印迹数据引起了编辑的注意。第1952页上的图4B和C,以及图4中的Transwell入侵测定数据。2F和4I,已经出现在以前发表的文章中,这些文章是由不同的作者在不同的研究机构撰写的(其中一些已经被撤回)。由于上述文章中的有争议的数据在提交肿瘤学报告之前已经发表,编辑已经决定这篇论文应该从期刊上撤回。作者被要求解释这些担忧,但编辑部没有收到回复。编辑对读者造成的不便表示歉意。[肿瘤学报告42:1946-1956,2019;DOI:10.3892/or.2019.7302]。
    Following the publication of this paper, it was drawn to the Editor\'s attention by a concerned reader that certain of the western blotting data shown in Fig. 4B and C on p. 1952, and the Transwell invasion assay data in Fig. 2F and 4I, had already appeared in previously published articles written by different authors at different research institutes (a number of which have been retracted). Owing to the fact that the contentious data in the above article had already been published prior to its submission to Oncology Reports, the Editor has decided that this paper should be retracted from the Journal. The authors were asked for an explanation to account for these concerns, but the Editorial Office did not receive a reply. The Editor apologizes to the readership for any inconvenience caused. [Oncology Reports 42: 1946‑1956, 2019; DOI: 10.3892/or.2019.7302].
    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
    表皮生长因子受体(EGFR)是一种跨膜酪氨酸激酶,通常通过翻译后糖基化进行修饰。在癌症中,在很大一部分非小细胞肺癌和乳腺腺癌中检测到EGFR扩增和促进增殖的热点突变如L858R。分子动力学模拟表明,天冬酰胺残基361(N361)处的糖基化促进二聚化和配体结合。我们稳定表达糖基化缺陷突变EGFRN361A,有或没有致癌突变L858R。免疫荧光和流式细胞术证明突变体各自在细胞膜上良好表达。相对于野生型EGFR,N361A降低增殖以及对配体的敏感性降低。测量EGFR与其结合配偶体HER2在细胞中的共定位的邻近连接测定揭示N361A突变增加共定位。N361A,位于EGFR抑制剂necitumumab的结合界面附近,表达致癌EGFRL858R的脱敏细胞对基于抗体的抑制。这些发现强调了翻译后修饰对癌基因功能的关键相关性。
    结论:EGFR将生长因子的信号传导到细胞增殖中,并且在肿瘤中经常被过度激活。N361的EGFR糖基化调节EGFR二聚化,增殖信号的生长因子刺激,和对靶向抑制的敏感性。对EGFR糖基化的见解可能会扩大治疗机会,使癌症患者受益。
    Epidermal growth factor receptor (EGFR) is a transmembrane tyrosine kinase that is frequently modified by glycosylation post-translationally. In cancer, EGFR amplifications and hotspot mutations such as L858R that promote proliferation have been detected in a significant fraction of non-small cell lung carcinomas and breast adenocarcinomas. Molecular dynamic simulations suggested that glycosylation at asparagine residue 361 (N361) promotes dimerization and ligand binding. We stably expressed glycosylation-deficient mutant EGFR N361A, with or without the oncogenic mutation L858R. Immunofluorescence and flow cytometry demonstrated that the mutants were each well expressed at the cell membrane. N361A decreased proliferation relative to wild-type EGFR as well as decreased sensitivity to ligands. Proximity ligation assays measuring co-localization of EGFR with its binding partner HER2 in cells revealed that N361A mutations increased co-localization. N361A, located near the binding interface for the EGFR inhibitor necitumumab, desensitized cells expressing the oncogenic EGFR L858R to antibody-based inhibition. These findings underline the critical relevance of post-translational modifications on oncogene function.
    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:本研究旨在研究7-酮甾醇(7-KSS)的作用,对人乳腺癌MCF-7和人肝癌HepG2细胞鞘磷脂/神经酰胺代谢产物和凋亡的影响。方法:评估7-KSS治疗在不同浓度和时期的抗增殖作用。通过MTT分析评估细胞活力,而鞘氨醇-1-磷酸(S1P)的水平,鞘磷脂(SM),使用LC-MS/MS测量神经酰胺(CER)。通过蛋白质印迹分析和免疫荧光染色测量磷酸化44/42ERK1/2和NF-κBp65(Ser536)蛋白水平。通过TUNEL染色和膜联蛋白V和碘化丙啶(PI)标记的流式细胞术评估来评估细胞凋亡。结果:7-KSS处理显著降低细胞存活率和S1P,与对照组相比,癌细胞中p-44/42ERK1/2和p-NF-κBp65蛋白水平。在与7-KSS孵育的癌细胞中,C16-C24CER的细胞内量和凋亡检测到了显着升高。结论:7-KSS通过下调S1P,刺激神经酰胺积累和凋亡,同时降低细胞增殖,p-44/42ERK1/2和p-NF-κBp65蛋白水平。
    Background: This study aimed to examine the effect of 7-Ketositosterol (7-KSS), on sphingomyelin/ceramide metabolites and apoptosis in human breast MCF-7 and human liver HepG2 cancer cells. Methods: Anti-proliferative effects of 7-KSS treatment were assessed at different concentrations and periods. Cell viability was assessed through MTT analysis, whereas the levels of sphingosine-1-phosphate (S1P), sphingomyelins (SMs), and ceramides (CERs) were measured using LC-MS/MS. Phosphorylated 44/42 ERK1/2 and NF-κB p65 (Ser536) protein levels were measured by Western blot analysis and immunofluorescence staining. Apoptosis was evaluated by TUNEL staining and flow cytometric assessment of annexin-V and propidium iodide (PI) labeling. Results: Treatment with 7-KSS significantly decreased cell survival and S1P, p-44/42 ERK1/2, and p-NF-κB p65 protein levels in cancer cells compared to controls. A substantial rise was detected in intracellular amounts of C16-C24 CERs and apoptosis in cancer cells incubated with 7-KSS. Conclusions: 7-KSS stimulated ceramide accumulation and apoptosis while decreasing cell proliferation via downregulating S1P, p-44/42 ERK1/2, and p-NF-κB p65 protein 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
    视网膜母细胞瘤(Rb)蛋白的信号通路,Akt激酶,Erk激酶(细胞外信号调节激酶)在急性髓系白血病的发病机制中具有重要作用。这些蛋白质通过磷酸化的组成型激活通过调节细胞周期来促进细胞存活,增殖和促凋亡信号过程。根据先前的数据,这些蛋白质的磷酸化形式对于癌症患者代表更差的结果。我们研究了磷酸化Rb(P-Rb)的存在,Akt(P-Akt)和Erk(P-Erk)蛋白通过蛋白质印迹技术,使用69例AML患者的骨髓或外周血样本中的磷酸特异性抗体,36例骨髓增生异常综合征(MDS)患者和10例健康志愿者。PTEN(磷酸酶和张力蛋白同源物)和PHLPP(PH结构域和富含亮氨酸的重复蛋白磷酸酶)磷酸酶的表达水平,也检测了Akt激酶通路的负调节因子.我们测试了这些蛋白质对存活的影响以及与AML中已知预后特征的相关性。我们发现46.3%的AML患者有可检测的P-Rb,34.7%具有P-Akt,28.9%具有P-Erk蛋白。66.1%的患者表达PTEN,38.9%PHLPP,PTEN和PHLPP均为37.2%,PTEN和PHLPP磷酸酶均为32.2%。与核磷蛋白突变(NPMc)阴性样品相比,核磷蛋白突变患者的P-Erk明显减少,根据诊断,外周血白细胞计数超过30G/L的患者组的P-Rb明显减少。PHLPP显著存在于FAB型M5中。P-Rb的表达代表显著更好的总生存期(OS),而P-Akt代表不良细胞遗传学患者的无事件生存率(EFS)明显较差。PHLPP和PTEN磷酸酶的存在有助于更好的OS和EFS,尽管差异无统计学意义。我们证实P-Akt与PHLPP之间存在显著正相关。评估Rb的磷酸化,Akt和Erk可以定义AML患者的一个亚组,他们将受益于新的靶向治疗方案补充标准化疗。它可能有助于监测缓解,AML的复发或进展。
    Signaling pathways of Retinoblastoma (Rb) protein, Akt-kinase, and Erk-kinase (extracellular signal-regulated kinase) have an important role in the pathogenesis of acute myeloid leukemia. Constitutive activation of these proteins by phosphorylation contributes to cell survival by regulation of cell cycle, proliferation and proapoptotic signaling processes. According to previous data phosphorylated forms of these proteins represent a worse outcome for cancer patients. We investigated the presence of phosphorylated Rb (P-Rb), Akt (P-Akt) and Erk (P-Erk) proteins by Western blot technique using phospho-specific antibodies in bone marrow or peripheral blood samples of 69 AML patients, 36 patients with myelodysplastic syndrome (MDS) and 10 healthy volunteers. Expression level of PTEN (Phosphatase and tensin homolog) and PHLPP (PH domain and leucine-rich repeat Protein Phosphatase) phosphatases, the negative regulators of Akt kinase pathway were also examined. We tested the effect of these proteins on survival and on the correlation with known prognostic features in AML. We found 46.3% of AML patients had detectable P-Rb, 34.7% had P-Akt and 28.9% had P-Erk protein. 66.1% of patients expressing PTEN, 38.9% PHLPP, 37.2% both PTEN and PHLPP and 32.2% neither PTEN nor PHLPP phosphatases. Compared to nucleophosmin mutation (NPMc) negative samples P-Erk was significantly less in nucleophosmin mutated patients, P-Rb was significantly less in patients\' group with more than 30 G/L peripheral leukocyte count by diagnosis. PHLPP was significantly present in FAB type M5. The expression of P-Rb represented significant better overall survival (OS), while P-Akt represented significantly worse event-free survival (EFS) in unfavorable cytogenetics patients. The presence of both PHLPP and PTEN phosphatases contributes to better OS and EFS, although the differences were not statistically significant. We confirmed significant positive correlation between P-Akt and PHLPP. Assessing the phosphorylation of Rb, Akt and Erk may define a subgroup of AML patients who would benefit especially from new targeted treatment options complemented the standard chemotherapy, and it may contribute to monitoring remission, relapse or progression of AML.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Published Erratum
    上述文章发表后,作者意识到,在图中。在图1D的第7363页上,为“0.5mM琥珀酸盐”组选择的数据面板在图1D中重复。1B(对照)在他们发表在FASEBJ上的另一篇文章中(“α-酮戊二酸通过PHD3/ADRB2途径防止骨骼肌蛋白降解和肌肉萎缩”:doi:10.1096/fj.201700670R),因为他们无意中混淆了两个数字的布局。作者为这个错误道歉。其次,就图中所示的印迹的定量而言。2A,β-肌动蛋白实际上没有用作加载对照;磷蛋白相对于相对总蛋白的水平进行归一化,和无花果的布局。已对2A进行了修订以反映这一点(请注意,图的图例。2也修改了:最后一句不再是,“β-肌动蛋白用作加载对照。\")。无花果的修订版。1和2显示在下一页。请注意,这些错误不会影响研究中报告的结果或主要结论,并且不需要对文本中的描述或这些图中所示的直方图进行更正。所有作者都赞成本更正的出版,作者感谢《分子医学报告》的编辑让他们有机会发表这篇文章。作者感到遗憾的是,他们的疏忽允许这些错误被包括在论文中,并对造成的不便向读者道歉。[分子医学报告16:7361-7366,2017;DOI:10.3892/mmr.2017.7554]。
    Following the publication of the above article, the authors realized that, in Fig. 1D on p. 7363, the data panel selected for the \'0.5 mM Succinate\' group was duplicated in Fig. 1B (Control) in another article of theirs published in FASEB J (\"α‑Ketoglutarate prevents skeletal muscle protein degradation and muscle atrophy through PHD3/ADRB2 pathway\": doi: 10.1096/fj.201700670R) due to the fact that they had inadvertently confused the layout of the two figures. The authors apologize for this error. Secondly, in terms of the quantification of the blots shown in Fig. 2A, β‑actin was not in fact used as a loading control; the phosphoproteins were normalized against the levels of the relative total protein, and the layout of Fig. 2A has been revised to reflect this (note that the the figure legend for Fig. 2 has also been revised: The last sentence no longer reads, \"β‑actin was used as a loading control.\"). The revised versions of Figs. 1 and 2 are shown on the next page. Note that these errors did not affect the results or the main conclusions reported in the study, and no corrections were required either to the descriptions in the text or to the histograms shown in these figures. All the authors approve of the publication of this corrigendum, and the authors are grateful to the Editor of Molecular Medicine Reports for allowing them the opportunity to publish this. The authors regret their oversight in allowing these errors to be included in the paper, and apologize to the readership for any inconvenience caused. [Molecular Medicine Reports 16: 7361‑7366, 2017; DOI: 10.3892/mmr.2017.7554].
    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
    心血管疾病(CVD)是全世界死亡和残疾的主要原因。确定新的靶向治疗方法已成为生物医学研究的优先事项,以改善患者的预后和生活质量。RAS-RAF-MEK(丝裂原活化蛋白激酶激酶)-ERK(细胞外信号调节激酶)途径作为与CVD发病机理有关的潜在信号级联,越来越受到人们的关注。该通路在调节细胞过程如增殖中起着关键作用,增长,迁移,分化,和生存,这对维持心血管稳态至关重要。此外,ERK信号参与控制血管生成,血管张力,心肌收缩力,和氧化应激。这种信号级联的失调与细胞功能障碍和血管和心脏病理重塑有关。这有助于CVD的发生和进展。最近和正在进行的研究提供了针对RAS-RAF-MEK-ERK途径改善心血管病理的潜在治疗干预措施的见解。临床前研究表明,在动物模型中,MEK抑制剂(MEKI)的靶向治疗在减弱ERK激活和缓解CVD进展中的功效。在这篇文章中,我们首先描述ERK信号如何有助于保持心血管健康.然后,我们总结了ERK在心脏和血管疾病的发展和进展中所起的作用的最新知识。包括动脉粥样硬化,心肌梗塞,心脏肥大,心力衰竭,和主动脉瘤。我们最后报告了包括MEKI在内的这些心血管疾病的新治疗策略,并讨论了其优势,挑战,以及MEKI治疗学的未来发展。
    Cardiovascular disease (CVD) represents the leading cause of mortality and disability all over the world. Identifying new targeted therapeutic approaches has become a priority of biomedical research to improve patient outcomes and quality of life. The RAS-RAF-MEK (mitogen-activated protein kinase kinase)-ERK (extracellular signal-regulated kinase) pathway is gaining growing interest as a potential signaling cascade implicated in the pathogenesis of CVD. This pathway is pivotal in regulating cellular processes like proliferation, growth, migration, differentiation, and survival, which are vital in maintaining cardiovascular homeostasis. In addition, ERK signaling is involved in controlling angiogenesis, vascular tone, myocardial contractility, and oxidative stress. Dysregulation of this signaling cascade has been linked to cell dysfunction and vascular and cardiac pathological remodeling, which contribute to the onset and progression of CVD. Recent and ongoing research has provided insights into potential therapeutic interventions targeting the RAS-RAF-MEK-ERK pathway to improve cardiovascular pathologies. Preclinical studies have demonstrated the efficacy of targeted therapy with MEK inhibitors (MEKI) in attenuating ERK activation and mitigating CVD progression in animal models. In this article, we first describe how ERK signaling contributes to preserving cardiovascular health. We then summarize current knowledge of the roles played by ERK in the development and progression of cardiac and vascular disorders, including atherosclerosis, myocardial infarction, cardiac hypertrophy, heart failure, and aortic aneurysm. We finally report novel therapeutic strategies for these CVDs encompassing MEKI and discuss advantages, challenges, and future developments for MEKI therapeutics.
    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
    药物耐受性是癌症治疗后复发的主要原因。尽管付出了巨大的努力,它的分子基础仍然知之甚少,妨碍可操作的干预。我们报道了一种以前未被识别的信号机制,支持用BRAF抑制剂治疗的BRAF突变黑色素瘤的药物耐受性,这可能与其他癌症普遍相关。其关键特征是由P2X7受体(嘌呤能配体门控阳离子通道)启动的细胞内在细胞内Ca2信号传导,以及这些Ca2信号在药物耐受状态下重新激活ERK1/2的能力增强。细胞外ATP,在生命系统中几乎无处不在,是可以通过P2X7通道启动Ca2+尖峰的配体。ATP在肿瘤微环境中丰富,并由垂死的细胞释放,具有讽刺意味的是,治疗引发的癌细胞死亡是导致ERK再激活和药物耐受性的营养刺激来源。这种机制立即解释了BRAF突变黑色素瘤中BRAFi治疗后不可避免的复发,并指出了克服它的可行策略。
    Drug tolerance is a major cause of relapse after cancer treatment. Despite intensive efforts, its molecular basis remains poorly understood, hampering actionable intervention. We report a previously unrecognized signaling mechanism supporting drug tolerance in BRAF-mutant melanoma treated with BRAF inhibitors that could be of general relevance to other cancers. Its key features are cell-intrinsic intracellular Ca2+ signaling initiated by P2X7 receptors (purinergic ligand-gated cation channels) and an enhanced ability for these Ca2+ signals to reactivate ERK1/2 in the drug-tolerant state. Extracellular ATP, virtually ubiquitous in living systems, is the ligand that can initiate Ca2+ spikes via P2X7 channels. ATP is abundant in the tumor microenvironment and is released by dying cells, ironically implicating treatment-initiated cancer cell death as a source of trophic stimuli that leads to ERK reactivation and drug tolerance. Such a mechanism immediately offers an explanation of the inevitable relapse after BRAFi treatment in BRAF-mutant melanoma and points to actionable strategies to overcome it.
    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:磷酸果糖激酶-血小板(PFKP)的异常表达通过修饰多种生物学功能在多种人类癌症的发展中起着至关重要的作用。然而,PFKP在头颈部鳞状细胞癌(HNSCC)中的作用的确切分子机制尚未完全阐明。
    方法:我们评估了120例HNSCC患者的肿瘤和邻近正常组织中PFKP和c-Myc的表达水平。进行了一系列体外和体内实验以探索PFKP和c-Myc之间的反馈回路对HNSCC进展的影响。此外,我们探索了使用患者来源的类器官(PDO)在HNSCC中靶向PFKP和c-Myc的治疗效果,细胞系来源的异种移植物,和患者来源的异种移植物。
    结果:我们的发现表明,PFKP在HNSCC组织和细胞系中经常上调,与预后不良有关。我们的体外和体内实验表明,升高的PFKP促进细胞增殖,血管生成,和HNSCC的转移。机械上,PFKP增加了ERK介导的c-Myc的稳定性,从而推动HNSCC的进展。此外,c-Myc在转录水平刺激PFKP表达,从而在PFKP和c-Myc之间形成正反馈回路。此外,我们的多个模型表明,共同靶向PFKP和c-Myc在HNSCC中触发协同抗肿瘤作用。
    结论:我们的研究证明了PFKP/c-Myc正反馈回路在驱动HNSCC进展中的关键作用,并提示同时靶向PFKP和c-Myc可能是HNSCC的一种新颖有效的治疗策略。
    BACKGROUND: The aberrant expression of phosphofructokinase-platelet (PFKP) plays a crucial role in the development of various human cancers by modifying diverse biological functions. However, the precise molecular mechanisms underlying the role of PFKP in head and neck squamous cell carcinoma (HNSCC) are not fully elucidated.
    METHODS: We assessed the expression levels of PFKP and c-Myc in tumor and adjacent normal tissues from 120 HNSCC patients. A series of in vitro and in vivo experiments were performed to explore the impact of the feedback loop between PFKP and c-Myc on HNSCC progression. Additionally, we explored the therapeutic effects of targeting PFKP and c-Myc in HNSCC using Patient-Derived Organoids (PDO), Cell Line-Derived Xenografts, and Patients-Derived Xenografts.
    RESULTS: Our findings indicated that PFKP is frequently upregulated in HNSCC tissues and cell lines, correlating with poor prognosis. Our in vitro and in vivo experiments demonstrate that elevated PFKP facilitates cell proliferation, angiogenesis, and metastasis in HNSCC. Mechanistically, PFKP increases the ERK-mediated stability of c-Myc, thereby driving progression of HNSCC. Moreover, c-Myc stimulates PFKP expression at the transcriptional level, thus forming a positive feedback loop between PFKP and c-Myc. Additionally, our multiple models demonstrate that co-targeting PFKP and c-Myc triggers synergistic anti-tumor effects in HNSCC.
    CONCLUSIONS: Our study demonstrates the critical role of the PFKP/c-Myc positive feedback loop in driving HNSCC progression and suggests that simultaneously targeting PFKP and c-Myc may be a novel and effective therapeutic strategy for 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
    BRAFV600E突变是结直肠癌锯齿状途径中的驱动突变。BRAFV600E通过组成型下游细胞外信号调节激酶(ERK)激活驱动肿瘤发生,但高强度ERK激活也可引发肿瘤抑制。是否以及如何将致癌ERK信号传导内在地调整到肿瘤发生的最佳水平尚不确定。在这项研究中,我们发现,在小鼠和患者的BRAFV600E突变型腺瘤/息肉中,FAK(粘着斑激酶)表达降低.在Vil1-Cre;BRAFLSL-V600E/+;Ptk2fl/fl小鼠,Fak缺失使BRAFV600E的致癌活性最大化,盲肠肿瘤发病率增加至100%。机械上,我们的结果表明,Fak损失,在不危害BRAFV600E诱导的ERK通路转录输出的情况下,降低EGFR(表皮生长因子受体)依赖性ERK磷酸化。ERK磷酸化的减少增加了Lgr4的水平,促进了肠干性和盲肠肿瘤的形成。我们的发现表明,通过Fak丢失介导的ERK磷酸化下调,可以实现BRAFV600E诱导的盲肠肿瘤形成的最佳ERK信号传导。
    BRAFV600E mutation is a driver mutation in the serrated pathway to colorectal cancers. BRAFV600E drives tumorigenesis through constitutive downstream extracellular signal-regulated kinase (ERK) activation, but high-intensity ERK activation can also trigger tumor suppression. Whether and how oncogenic ERK signaling can be intrinsically adjusted to a \'just-right\' level optimal for tumorigenesis remains undetermined. In this study, we found that FAK (Focal adhesion kinase) expression was reduced in BRAFV600E-mutant adenomas/polyps in mice and patients. In Vil1-Cre;BRAFLSL-V600E/+;Ptk2fl/fl mice, Fak deletion maximized BRAFV600E\'s oncogenic activity and increased cecal tumor incidence to 100%. Mechanistically, our results showed that Fak loss, without jeopardizing BRAFV600E-induced ERK pathway transcriptional output, reduced EGFR (epidermal growth factor receptor)-dependent ERK phosphorylation. Reduction in ERK phosphorylation increased the level of Lgr4, promoting intestinal stemness and cecal tumor formation. Our findings show that a \'just-right\' ERK signaling optimal for BRAFV600E-induced cecal tumor formation can be achieved via Fak loss-mediated downregulation of ERK phosphorylation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号