Akt

Akt
  • 文章类型: Journal Article
    高水平的血管内皮生长因子(VEGF),成纤维细胞生长因子(FGF)-2和血管生成素(ANG)-2存在于口腔鳞状细胞癌(OSCC)和口腔潜在恶性疾病(OPMD)的组织中。正如预期的那样,VEGF,FGF-2和ANG-2过表达与滋养生长的OPMD或OSCC并为后者提供转移途径的新血液和淋巴管的发育平行。值得注意的是,VEGF,FGF-2和ANG-2也与上皮-间质转化(EMT)有关,转分化过程,分别促进或加剧正常和肿瘤性口腔上皮细胞的侵袭性。这里,我们总结了已发表的关于VEGF之间相互作用的影响的工作,FGF-2,ANG-2,血管生成,EMT对口腔癌有影响。审查的研究结果表明,VEGF,FGF-2和ANG-2激发蛋白激酶B(AKT)或丝裂原活化蛋白激酶(MAPK),两种信号通路可以促进OPMDs和OSCC中的EMT和新血管形成。由于EMT和血管生成是OSCC发生和进展的关键,以及它的放射和化学抗性,这些数据鼓励在该恶性肿瘤的治疗中包括AKT或MAPK抑制剂和/或抗血管生成药物.
    High levels of vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF)-2 and angiopoietin (ANG)-2 are found in tissues from oral squamous cell carcinoma (OSCC) and oral potentially malignant disorders (OPMDs). As might be expected, VEGF, FGF-2, and ANG-2 overexpression parallels the development of new blood and lymphatic vessels that nourish the growing OPMDs or OSCCs and provide the latter with metastatic routes. Notably, VEGF, FGF-2, and ANG-2 are also linked to the epithelial-to-mesenchymal transition (EMT), a trans-differentiation process that respectively promotes or exasperates the invasiveness of normal and neoplastic oral epithelial cells. Here, we have summarized published work regarding the impact that the interplay among VEGF, FGF-2, ANG-2, vessel generation, and EMT has on oral carcinogenesis. Results from the reviewed studies indicate that VEGF, FGF-2, and ANG-2 spark either protein kinase B (AKT) or mitogen-activated protein kinases (MAPK), two signaling pathways that can promote both EMT and new vessels\' formation in OPMDs and OSCCs. Since EMT and vessel generation are key to the onset and progression of OSCC, as well as to its radio- and chemo-resistance, these data encourage including AKT or MAPK inhibitors and/or antiangiogenic drugs in the treatment of this malignancy.
    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
    雷沙吉兰(Azilect®)是一种选择性单胺氧化酶B(MAO-B)抑制剂,可在帕金森病(PD)治疗中提供症状益处,并已发现具有临床前神经保护作用。这里,我们研究了急性雷沙吉兰治疗22小时的PC12神经元培养物暴露于氧-葡萄糖剥夺(OGD)4小时的神经保护性信号通路,随后是18小时的复氧(R),导致40%的细胞死亡.在这项研究中,3-10µM雷沙吉兰诱导的20-80%的剂量依赖性神经保护,减少15%的神经毒性活性氧的产生,并将甘油醛-3-磷酸脱氢酶(GAPDH)的核易位减少75-90%。此外,10µM雷沙吉兰使蛋白激酶B(Akt)磷酸化增加50%,并使缺血诱导的α-突触核蛋白的蛋白表达减少50%,与神经保护作用相关。用1-5µM雷沙吉兰处理可诱导转录因子Nrf2的核穿梭达40-90%,并增加了抗氧化酶血红素加氧酶-1(NAD(P)H-醌脱氢酶,过氧化氢酶是OGD/R损伤的1.8-2.0倍。这些结果表明,雷沙吉兰通过抑制α-突触核蛋白和GAPDH介导的无核细胞死亡为缺血性神经元培养物提供神经保护,以及通过线粒体保护,通过涉及Akt/Nrf2氧化还原信号通路的机制增加线粒体特异性抗氧化酶。这些发现可用于PD和中风治疗中的神经保护药物开发。
    Rasagiline (Azilect®) is a selective monoamine oxidase B (MAO-B) inhibitor that provides symptomatic benefits in Parkinson\'s disease (PD) treatment and has been found to exert preclinical neuroprotective effects. Here, we investigated the neuroprotective signaling pathways of acute rasagiline treatment for 22 h in PC12 neuronal cultures exposed to oxygen-glucose deprivation (OGD) for 4 h, followed by 18 h of reoxygenation (R), causing 40% aponecrotic cell death. In this study, 3-10 µM rasagiline induced dose-dependent neuroprotection of 20-80%, reduced the production of the neurotoxic reactive oxygen species by 15%, and reduced the nuclear translocation of glyceraldehyde-3-phosphate dehydrogenase (GAPDH) by 75-90%. In addition, 10 µM rasagiline increased protein kinase B (Akt) phosphorylation by 50% and decreased the protein expression of the ischemia-induced α-synuclein protein by 50% in correlation with the neuroprotective effect. Treatment with 1-5 µM rasagiline induced nuclear shuttling of transcription factor Nrf2 by 40-90% and increased the mRNA levels of the antioxidant enzymes heme oxygenase-1, (NAD (P) H- quinone dehydrogenase, and catalase by 1.8-2.0-fold compared to OGD/R insult. These results indicate that rasagiline provides neuroprotection to the ischemic neuronal cultures through the inhibition of α-synuclein and GAPDH-mediated aponecrotic cell death, as well as via mitochondrial protection, by increasing mitochondria-specific antioxidant enzymes through a mechanism involving the Akt/Nrf2 redox-signaling pathway. These findings may be exploited for neuroprotective drug development in PD and stroke therapy.
    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
    三阴性乳腺癌(TNBC)细胞通常对FAS(CD95)介导的凋亡具有抗性,但潜在的分子机制还没有完全理解。值得注意的是,II型跨膜蛋白的表达,CD74通过未知的机制与化疗抗性和更具侵袭性的癌症形式相关。这里,我们分析了癌症患者和/或患者来源的异种移植(PDX)模型的基因表达模式,发现CD74的mRNA和蛋白水平在TNBC中高表达,并与肿瘤干细胞(CSCs)和上皮-间质转化(EMT)特性相关.机械上,我们发现AKT的激活可能对于维持CD74的表达和蛋白质稳定性以促进其致癌功能至关重要。生理学上,表皮生长因子(EGF)和CD74可以激活AKT信号,可能是通过磷酸化AKT(S473)与CD74结合,而抑制AKT可能会损害CD74的稳定性。我们还揭示了CD74与FAS结合并干扰FAS介导的细胞凋亡的内在信号传导。因此,使用AKT抑制剂和CD74衍生肽对CD74/FAS复合物的选择性靶向可以协同恢复和激活FAS介导的细胞凋亡。因此,我们动员细胞凋亡途径的方法可能为靶向CD74/FAS和CD74-AKT轴治疗TNBC提供了理论基础.
    Triple-negative breast cancer (TNBC) cells are often resistant to FAS (CD95)-mediated apoptosis, but the underlying molecular mechanism(s) is not fully understood yet. Notably, the expression of the type II transmembrane protein, CD74, is correlated with chemotherapy-resistant and more invasive forms of cancers via unknown mechanisms. Here, we analyzed gene expression pattern of cancer patients and/or patient-derived xenograft (PDX) models and found that mRNA and protein levels of CD74 are highly expressed in TNBC and correlated with cancer stem cells (CSCs) and epithelial-mesenchymal transition (EMT) properties. Mechanistically, we found that AKT activation is likely critical for maintaining CD74 expression and protein stability to favor its oncogenic functions. Physiologically, epidermal growth factor (EGF) along with CD74 could activate AKT signaling, likely through binding of phosphorylated AKT (S473) to CD74, whereas inhibition of AKT could impair stability of CD74. We also revealed that CD74 binds to FAS and interferes with the intrinsic signaling of FAS-mediated apoptosis. As such, selective targeting of the CD74/FAS complex using the AKT inhibitor along with the CD74-derived peptide could synergistically restore and activate FAS-mediated apoptosis. Therefore, our approach of mobilizing apoptosis pathways likely provides a rationale for TNBC treatment by targeting the CD74/FAS and CD74-AKT axes.
    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
    食管癌中化疗耐药性的发展代表了癌症治疗中的重大挑战。因此,我们的研究旨在通过检查与这种化学耐药有关的分子来确定有效的治疗策略.我们一致观察到Mcl-1在暴露于短期和长期顺铂治疗的细胞中的表达增加。一种常用于食道癌治疗的药物。功能分析显示Mcl-1调节食管癌细胞对顺铂治疗的反应。值得注意的是,Mcl-1的这种上调不依赖于真核起始因子4E(eIF4E)。相反,它与由于Akt的激活而增加的稳定性相关。Capivasertib,一种有效的泛Akt激酶药物,通过抑制化疗耐药细胞中的Akt信号通路显著降低Mcl-1水平。此外,capivasertib不仅降低了化疗耐药食管癌细胞的活力,而且协同增强了顺铂的作用。在多个鼠标模型中,代表化疗耐药和化疗敏感的食管癌,以无毒剂量给药的capivasertib显示出明显的疗效。它显著延长了小鼠的总生存期。我们的研究强调了Akt相关的Mcl-1上调在食管癌细胞化疗耐药发展中的关键作用。此外,它强调了capivasertib逆转这种抗性机制的潜力。
    The development of resistance to chemotherapy in esophageal cancer represents a significant challenge in cancer treatment. Therefore, our study aimed to identify effective therapeutic strategies by examining the molecules involved in this chemoresistance. We consistently observed an increase in the expression of Mcl-1 in cells exposed to both short and long-term treatment with cisplatin, a drug commonly used in esophageal cancer therapy. Functional analysis showed that Mcl-1 regulates esophageal cancer cell response to cisplatin treatment. Notably, this upregulation of Mcl-1 was not dependent on eukaryotic initiation factor 4E (eIF4E). Instead, it was associated with increased stability due to the activation of Akt. Capivasertib, a potent pan-Akt kinase drug, significantly decreased Mcl-1 level via inhibiting Akt signaling pathway in chemo-resistant cells. In addition, capivasertib not only decreased the viability of chemo-resistant esophageal cancer cells but also synergistically enhanced the effects of cisplatin. In multiple mouse models, representing both chemo-resistant and chemo-sensitive esophageal cancer, capivasertib administered at non-toxic doses demonstrated remarkable efficacy. It significantly extended the overall survival of the mice. Our research underscores the pivotal role of Akt-associated Mcl-1 upregulation in the development of chemo-resistance in esophageal cancer cells. Furthermore, it highlights the potential of capivasertib to reverse this resistance mechanism.
    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
    在这篇文章发表之后,一位感兴趣的读者提请作者注意,图中的西方印迹中有两对蛋白质带。第679页和681页上的图3A和5D分别看起来非常相似。在重新检查了他们的原始数据后,作者意识到,无花果。5D组装不正确。图的修订版。现在包括图5的正确数据。5D,显示在下一页上。请注意,在图中组装数据时出现的错误。5对本文报告的结果或结论没有很大影响,所有作者都同意出版本更正。作者感到遗憾的是,这些错误在他们的文章发表之前没有被注意到,感谢《肿瘤学报告》编辑允许他们有机会发表本更正。对于由此造成的不便,他们也向读者道歉。[肿瘤学报告33:675-684,2015;DOI:10.3892/or.2014.3653]。
    Following the publication of this article, an interested reader drew to the authors\' attention that two pairs of protein bands featured in the western blots in Fig. 3A and 5D on p. 679 and 681 respectively appeared to be strikingly similar. After having re‑examined their original data, the authors realized that Fig. 5D had been assembled incorrectly. The revised version of Fig. 5, now including the correct data for Fig. 5D, is shown on the next page. Note that the errors made in terms of assembling the data in Fig. 5 did not greatly affect either the results or the conclusions reported in this paper, and all the authors agree to the publication of this corrigendum. The authors regret that these errors went unnoticed prior to the publication of their article, are grateful to the Editor of Oncology Reports for allowing them this opportunity to publish this corrigendum. They also apologize to the readership for any inconvenience caused. [Oncology Reports  33: 675‑684, 2015; DOI: 10.3892/or.2014.3653].
    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
    一直在搜索在临床上延长健康寿命的药物。硝唑尼特是FDA批准的临床抗原虫药物。硝唑尼特在体内吸收后迅速代谢为替唑尼特。我们以前的研究发现,硝唑尼特及其代谢产物替唑尼特诱导轻度线粒体解偶联并激活细胞AMPK,口服硝唑尼特可预防实验性高脂血症,肝脂肪变性,和动脉粥样硬化。这里,我们证明,硝唑尼特和替唑尼特均可通过Akt/AMPK/sir2.1/daf16途径延长秀丽隐杆线虫的寿命和健康时间.此外,硝唑尼特和替唑尼特均可改善高糖诱导的C.elegans寿命缩短。硝唑尼特是一种具有良好安全性的临床药物,我们认为它是一种新型的抗衰老药物。
    The drugs extending healthspan in clinic have always been searched. Nitazoxanide is an FDA-approved clinical antiprotozoal drug. Nitazoxanide is rapidly metabolized to tizoxanide after absorption in vivo. Our previous studies find that nitazoxanide and its metabolite tizoxanide induce mild mitochondrial uncoupling and activate cellular AMPK, oral nitazoxanide protects against experimental hyperlipidemia, hepatic steatosis, and atherosclerosis. Here, we demonstrate that both nitazoxanide and tizoxanide extend the lifespan and healthspan of Caenorhabditis elegans through Akt/AMPK/sir 2.1/daf16 pathway. Additionally, both nitazoxanide and tizoxanide improve high glucose-induced shortening of C. elegans lifespan. Nitazoxanide has been a clinical drug with a good safety profile, we suggest that it is a novel anti-aging drug.
    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
    尽管亮氨酸拉链肿瘤抑制因子1(LZTS1)被认为是潜在的肿瘤抑制因子,越来越多的证据表明,LZTS1在许多癌症类型中高表达。为了阐明LZTS1在大肠癌发生中的确切作用,我们进行了LZTS1的生物信息学分析,包括表达差异,表达水平和生存率之间的相关性,基于TCGA数据集的LZTS1启动子和相关细胞通路的甲基化状态,GEO数据库和我们自己的CRC患者队列。此外,我们证实了LZTS1在人类哺乳动物细胞中的致癌功能,采用了一系列的检测方法,包括组织芯片,免疫印迹,细胞增殖和迁移测定。我们发现LZTS1在结直肠癌及其不同临床亚型中的表达高于配对正常组织,也就是说,至少在某种程度上,由于CRC肿瘤样品中LZTS1启动子的低甲基化状态。功能分析确定了LZTS1和PI3K-AKT途径的高表达与上皮-间质转化(EMT)过程之间的密切关系。始终如一,我们发现LZTS1的表达与PIK3CD的表达呈正相关,CRC肿瘤样本中的N-钙粘蛋白,而LZTS1的表达与CRC肿瘤样本中E-cadherin和PTEN的表达呈负相关。实验数据进一步证实LZTS1的过表达上调AKT的活性并促进EMT过程。此外,LZTS1的消耗抑制了CRC细胞的增殖和迁移速率。因此,这项研究表明,LZTS1在大肠癌发生中起致癌作用。
    Although leucine zipper tumour suppressor 1 (LZTS1) has been considered a potential tumour suppressor, accumulating evidence suggests that LZTS1 is highly expressed in many cancer types. To unravel the exact role of LZTS1 in colorectal carcinogenesis, we performed the bioinformatic analysis of LZTS1, including expression differences, correlations between expression levels and survival, methylation status of LZTS1 promoter and related cellular pathways based on TCGA dataset, GEO databases and our own CRC patient cohort. Furthermore, we confirmed the oncogenic function of LZTS1 in human mammalian cells by employing a series of assays including tissue microarray, immunoblotting, cell proliferation and migration assay. We found that the expression of LZTS1 is higher in tumour samples compared to paired normal tissue in CRC cancer and its different clinical subtypes, which is, at least in part, due to the low methylation status of LZTS1 promoter in CRC tumour samples. Functional analysis identified the close relationship between high expression of LZTS1 and PI3K-AKT pathway and the epithelial-mesenchymal transition (EMT) process. Consistently, we found that the expression of LZTS1 positively correlated with the expression PIK3CD, N-cadherin in CRC tumour samples, while the expression of LZTS1 negatively correlated with the expression of E-cadherin and PTEN in CRC tumour samples. Experimental data further confirmed that overexpression of LZTS1 upregulated activity of AKT and promoted EMT process. Furthermore, depletion of LZTS1 repressed the proliferation and migration rate of CRC cells. Thus, this study indicates that LZTS1 plays an oncogenic role in colorectal carcinogenesis.
    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
    再上皮化是皮肤伤口愈合的重要步骤,提到迁移,扩散,和伤口周围角质形成细胞的分化。在这个过程中,伤口的边缘开始形成新的上皮细胞,从伤口的外围向中心迁移,逐渐覆盖整个伤口区域。这些新形成的上皮细胞增殖和分化,最终在暴露的皮肤表面上形成保护层。已知伤口内源性电场(EF)是促进表皮向伤口中心迁移的主要因素。然而,EF促进表皮迁移的确切机制仍然难以捉摸。这里,我们发现在体外培养的角质形成细胞单层模型中,EF的应用逆转了细胞的分化,如早期分化标记K1和K10的减少所示。遗传操作证实,EF通过下调E-cadherin介导的粘附来逆转角质形成细胞的分化。通过RNA测序分析,我们筛选出蜗牛作为E-cadherin的转录抑制因子。蜗牛敲除消除了E-cadherin的下调和EF诱导的分化逆转。KEGG分析鉴定了EF下蜗牛诱导的PI3K/AKT信号传导。LY294002对PI3K的抑制作用减少了EF诱导的AKT激活和蜗牛增强,很大程度上恢复了EF降低的E-cadherin水平。最后,在猪全层皮肤伤口模型中,我们发现,方向逆转的外源性EF削弱了伤口内源性EF,导致E-cadherin上调,并在体内新形成的表皮中早期分化。我们的研究表明,电场(EF)通过抑制PI3K/AKT/Snail途径降低E-cadherin的表达,从而逆转角质形成细胞的分化。这一发现为我们提供了关于电场在伤口愈合中的作用的新见解。EF干预细胞内信号通路,抑制E-cadherin的表达,这导致角质形成细胞的较低分化状态。在这种状态下,角质形成细胞表现出增加的迁移能力,促进表皮细胞的迁移和伤口再上皮化。
    Re-epithelialization is an important step in skin wound healing, referring to the migration, proliferation, and differentiation of keratinocytes around the wound. During this process, the edges of the wound begin to form new epithelial cells, which migrate from the periphery of the wound towards the center, gradually covering the entire wound area. These newly formed epithelial cells proliferate and differentiate, ultimately forming a protective layer over the exposed dermal surface. Wound endogenous electric fields (EFs) are known as the dominant factor to facilitate the epidermal migration to wound center. However, the precise mechanisms by which EFs promote epidermal migration remains elusive. Here, we found that in a model of cultured keratinocyte monolayer in vitro, EFs application reversed the differentiation of cells, as indicated by the reduction of the early differentiation markers K1 and K10. Genetic manipulation confirmed that EFs reversed keratinocyte differentiation through down-regulating the E-cadherin-mediated adhesion. By RNA-sequencing analysis, we screened out Snail as the transcription suppressor of E-cadherin. Snail knockdown abolished the down-regulation of E-cadherin and the reversal of differentiation induced by EFs. KEGG analysis identified PI3K/AKT signaling for Snail induction under EFs. Inhibition of PI3K by LY294002 diminished the EFs-induced AKT activation and Snail augmentation, largely restoring the level of E-cadherin reduced by EFs. Finally, in model of full-thickness skin wounds in pigs, we found that weakening of the wound endogenous EFs by the direction-reversed exogenous EFs resulted in an up-regulation of E-cadherin and earlier differentiation in newly formed epidermis in vivo. Our research suggests that electric fields (EFs) decrease E-cadherin expression by suppressing the PI3K/AKT/Snail pathway, thereby reversing the differentiation of keratinocytes. This discovery provides us with new insights into the role of electric fields in wound healing. EFs intervene in intracellular signaling pathways, inhibiting the expression of E-cadherin, which results in a lower differentiation state of keratinocytes. In this state, keratinocytes exhibit increased migratory capacity, facilitating the migration of epidermal cells and wound reepithelialization.
    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
    谷氨酰-氨甲酰-tRNA合成酶(EPRS1)是一种双功能氨酰基-tRNA合成酶(aaRS),对于解码遗传密码至关重要。EPRS1驻留,与其他七个aaRS和三个非催化蛋白,在细胞质多tRNA合成酶复合物(MSC)中。多个MSC驻留aaRS,包括EPRS1,表现出从MSC的刺激依赖性释放,以执行与其在蛋白质合成中的主要功能不同的非规范活动。这里,我们显示EPRS1存在于组成型低磷酸酶和张力蛋白同源物(PTEN)表达的乳腺癌细胞的细胞质和细胞核中。EPRS1主要是表达PTEN的细胞,但是对PTEN的化学或遗传抑制作用,或其靶标的化学或应激介导的激活,AKT,诱导EPRS1核定位。同样,在浸润性导管癌中观察到EPRS1的优先核定位,也是P-Ser473-AKT。EPRS1核转运需要连接催化谷氨酰-tRNA合成酶和脯氨酸酰-tRNA合成酶结构域的接头区域内的核定位信号(NLS)。核EPRS1与聚(ADP-核糖)聚合酶1(PARP1)相互作用,一种DNA损伤传感器,可指导蛋白质的聚(ADP-核糖基)化(PARylation)。EPRS1是PARP1活性的关键调节因子,如EPRS1敲低细胞中ADP-核糖基化显著降低所示。此外,EPRS1和PARP1敲低可比较地改变多个肿瘤相关基因的表达,抑制DNA损伤修复,降低肿瘤细胞存活率,并减少乳腺癌细胞形成的肿瘤球。EPRS1介导的PARP1活性调节提供了乳腺癌细胞中PTEN缺失之间的机制联系,PARP1激活,细胞存活和肿瘤生长。靶向EPRS1的非规范活性,而不抑制规范的tRNA连接酶活性,提供了一种潜在补充现有PARP1抑制剂的治疗方法。
    Glutamyl-prolyl-tRNA synthetase (EPRS1) is a bifunctional aminoacyl-tRNA-synthetase (aaRS) essential for decoding the genetic code. EPRS1 resides, with seven other aaRSs and three noncatalytic proteins, in the cytoplasmic multi-tRNA synthetase complex (MSC). Multiple MSC-resident aaRSs, including EPRS1, exhibit stimulus-dependent release from the MSC to perform noncanonical activities distinct from their primary function in protein synthesis. Here, we show EPRS1 is present in both cytoplasm and nucleus of breast cancer cells with constitutively low phosphatase and tensin homolog (PTEN) expression. EPRS1 is primarily cytosolic in PTEN-expressing cells, but chemical or genetic inhibition of PTEN, or chemical or stress-mediated activation of its target, AKT, induces EPRS1 nuclear localization. Likewise, preferential nuclear localization of EPRS1 was observed in invasive ductal carcinoma that were also P-Ser473-AKT+. EPRS1 nuclear transport requires a nuclear localization signal (NLS) within the linker region that joins the catalytic glutamyl-tRNA synthetase and prolyl-tRNA synthetase domains. Nuclear EPRS1 interacts with poly(ADP-ribose) polymerase 1 (PARP1), a DNA-damage sensor that directs poly(ADP-ribosyl)ation (PARylation) of proteins. EPRS1 is a critical regulator of PARP1 activity as shown by markedly reduced ADP-ribosylation in EPRS1 knockdown cells. Moreover, EPRS1 and PARP1 knockdown comparably alter the expression of multiple tumor-related genes, inhibit DNA-damage repair, reduce tumor cell survival, and diminish tumor sphere formation by breast cancer cells. EPRS1-mediated regulation of PARP1 activity provides a mechanistic link between PTEN loss in breast cancer cells, PARP1 activation, and cell survival and tumor growth. Targeting the noncanonical activity of EPRS1, without inhibiting canonical tRNA ligase activity, provides a therapeutic approach potentially supplementing existing PARP1 inhibitors.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号