Peptide Elongation Factor 1

肽延长因子 1
  • 文章类型: Journal Article
    A boy, aged 7 months, presented with severe global developmental delay (GDD), refractory epilepsy, hypotonia, nystagmus, ocular hypertelorism, a broad nasal bridge, everted upper lip, a high palatal arch, and cryptorchidism. Genetic testing revealed a de novo heterozygous missense mutation of c.364G>A(p.E122K) in the EEF1A2 gene, and finally the boy was diagnosed with autosomal dominant developmental and epileptic encephalopathy 33 caused by the EEF1A2 gene mutation. This case report suggests that for children with unexplained infancy-onset severe to profound GDD/intellectual disability and refractory epilepsy, genetic testing for EEF1A2 gene mutations should be considered. This is particularly important for those exhibiting hypotonia, nonverbal communication, and craniofacial deformities, to facilitate a confirmed diagnosis.
    患儿,男,7月龄,表现为重度全面发育落后、难治性癫痫、肌张力降低、眼球震颤、眼距宽、鼻梁塌陷、上唇外翻、高腭弓和隐睾,基因检测发现EEF1A2基因存在c.364G>A(p.E122K)新生杂合错义变异,最终该患儿确诊为EEF1A2基因变异致常染色体显性遗传发育性癫痫性脑病33型。该病例报道提示,对不明原因婴儿期起病的重度-极重度全面发育落后/智力障碍、难治性癫痫患儿,尤其是存在肌张力低下、语言缺失、颅面部畸形者,应考虑EEF1A2基因变异可能,应尽早完善遗传学检测协助诊断。.
    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
    四种粘菌(伪藻,欧洲双皮病,Lycogalaregulare,通过光学显微镜和扫描电子显微镜观察到了中国新出现的Trichiaarmillata),并提供了详细的描述和插图,以及与相关物种的比较。其中,A.在类型局部性之外首次发现了伪数据,欧洲是第一次在欧洲以外的地方被发现,以及L.regular和T.armillata在被命名后再次被报告。进行了基于核18SrDNA和延伸因子1α序列或核18SrDNA和细胞色素氧化酶亚基I序列的系统发育分析,为形态学鉴定提供了分子基础。这些标本保存在南京师范大学真菌标本室。
    Four species of myxomycetes (Arcyria pseudodenudata, Diderma europaeum, Lycogala irregulare, and Trichia armillata) new to China were observed via light microscope and scanning electron microscope, and detailed descriptions and illustrations are provided, along with comparisons with related species. Among them, A. pseudodenudata was discovered for the first time outside of the type locality, D. europaeum was discovered for the first time outside of Europe, and L. irregulare and T. armillata were reported again after being named. Phylogenetic analyses based on nuclear 18S rDNA and elongation factor-1 alpha sequences or nuclear 18S rDNA and cytochrome oxidase subunit I sequences was performed to provide a molecular basis for morphological identification. These specimens were deposited in the Herbarium of Fungi of Nanjing Normal University.
    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
    细胞衰老的特征是蛋白质合成减少,尽管基本过程大多不清楚。转移RNA(tRNA)的化学修饰经常影响tRNA活性,这对翻译至关重要。我们描述了tRNAN7-甲基鸟苷(m7G46)甲基化,由METTL1-WDR4催化,调节翻译并影响衰老表型。Mettl1/Wdr4和m7G随衰老而逐渐减弱。METL1的减少导致tRNA的减少,尤其是那些修改了m7G的人,通过快速tRNA降解(RTD)途径。减少导致核糖体在某些密码子处停滞,阻碍在Wnt信号传导和核糖体生物发生等途径中必需的mRNA的翻译。此外,慢性核糖体停滞刺激利波毒性和综合应激反应,诱导衰老相关的分泌表型。此外,恢复eEF1A蛋白通过降低RTD减轻METTL1缺乏引起的衰老表型。我们的发现表明,通过实现有效的mRNA翻译,tRNAm7G修饰对于预防过早衰老和衰老至关重要。
    Cellular senescence is characterized by a decrease in protein synthesis, although the underlying processes are mostly unclear. Chemical modifications to transfer RNAs (tRNAs) frequently influence tRNA activity, which is crucial for translation. We describe how tRNA N7-methylguanosine (m7G46) methylation, catalyzed by METTL1-WDR4, regulates translation and influences senescence phenotypes. Mettl1/Wdr4 and m7G gradually diminish with senescence and aging. A decrease in METTL1 causes a reduction in tRNAs, especially those with the m7G modification, via the rapid tRNA degradation (RTD) pathway. The decreases cause ribosomes to stall at certain codons, impeding the translation of mRNA that is essential in pathways such as Wnt signaling and ribosome biogenesis. Furthermore, chronic ribosome stalling stimulates the ribotoxic and integrative stress responses, which induce senescence-associated secretory phenotype. Moreover, restoring eEF1A protein mitigates senescence phenotypes caused by METTL1 deficiency by reducing RTD. Our findings demonstrate that tRNA m7G modification is essential for preventing premature senescence and aging by enabling efficient mRNA translation.
    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:肝细胞癌(HCC)是全球癌症相关死亡的第二大原因,其特征是预后不良。真核翻译延伸因子1α1(EEF1A1)已被证明在多种人类癌症中发挥重要作用,而对EEF1A1的去泛素化知之甚少。
    方法:使用临床组织样本验证了泛素羧基末端水解酶L3(UCHL3)与EEF1A1之间的结合和调节关系,逆转录定量实时荧光定量PCR(RT-qPCR),西方印迹,免疫共沉淀,和免疫荧光,以及泛素检测和环己酰胺跟踪实验。最后,通过功能实验和裸鼠模型分析了UCHL3/EEF1A1轴对HCC恶性行为的影响。
    结果:发现UCHL3在HCC组织中具有高表达水平。来自60例HCC患者的组织样本用于评估UCHL3和EEF1A1之间的相关性。UCHL3通过赖氨酸位点与EEF1A1结合,这降低了EEF1A1的泛素化水平。功能实验和裸鼠模型已经证明UCHL3/EEF1A1轴促进迁移,stemness,和肝癌细胞的耐药性。降低EEF1A1的表达可以逆转UCHL3对HCC细胞恶性行为的影响。
    结论:我们的发现表明UCHL3通过去泛素化结合并稳定EEF1A1。UCHL3和EEF1A1形成促进HCC恶性进展的功能轴,为肝癌的抗肿瘤靶向治疗提供新的见解。
    BACKGROUND: Hepatocellular carcinoma (HCC) ranks as the second leading cause of global cancer-related deaths and is characterized by a poor prognosis. Eukaryotic translation elongation factor 1 alpha 1 (EEF1A1) have been proved to play important roles in various human cancers, whereas the deubiquitination of EEF1A1 was poorly understood.
    METHODS: The binding and regulatory relationship between Ubiquitin carboxyl-terminal hydrolase L3 (UCHL3) and EEF1A1 was validated using clinical tissue samples, reverse transcription quantitative real-time fluorescence quantitative PCR (RT-qPCR), Western blotting, co-immunoprecipitation, and immunofluorescence, as well as ubiquitin detection and cyclohexamide tracking experiments. Finally, the impact of the UCHL3/EEF1A1 axis on HCC malignant behavior was analyzed through functional experiments and nude mouse models.
    RESULTS: UCHL3 was found to have a high expression level in HCC tissues. Tissue samples from 60 HCC patients were used to evaluate the correlation between UCHL3 and EEF1A1. UCHL3 binds to EEF1A1 through the lysine site, which reduces the ubiquitination level of EEF1A1. Functional experiments and nude mouse models have demonstrated that the UCHL3/EEF1A1 axis promotes the migration, stemness, and drug resistance of HCC cells. Reducing the expression of EEF1A1 can reverse the effect of UCHL3 on the malignant behavior of HCC cells.
    CONCLUSIONS: Our findings revealed that UCHL3 binds and stabilizes EEF1A1 through deubiquitination. UCHL3 and EEF1A1 formed a functional axis in facilitating the malignant progression of HCC, proving new insights for the anti-tumor targeted therapy for HCC.
    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
    骨质疏松症的病理进展是由骨髓间充质干细胞(BMSCs)在成骨和脂肪形成方面的不平衡发育引起的。虽然EEF1B2在智力障碍和肿瘤发生中的作用已经确立,其在BMSCs的骨-脂肪开关中的功能仍未被研究。在成骨分化的过程中,我们观察到EEF1B2的表达增加,而在脂肪形成期间其表达减少。EEF1B2的抑制阻碍了成骨分化和矿化的过程,同时促进成脂分化。相反,EEF1B2的过表达可增强成骨并强烈抑制脂肪生成。此外,EEF1B2在胫骨中的过度表达有可能减轻骨质疏松症小鼠的骨丢失和减少骨髓肥胖。在机制方面,当在成骨过程中EEF1B2功能受到抑制时,β-catenin活性受到抑制。我们的集体发现表明,EEF1B2作为一个调节器,影响BMSCs的分化,维持骨与脂肪的平衡。我们的发现强调了它作为骨代谢相关疾病的治疗靶点的潜力。
    The pathological advancement of osteoporosis is caused by the uneven development of bone marrow-derived mesenchymal stem cells (BMSCs) in terms of osteogenesis and adipogenesis. While the role of EEF1B2 in intellectual disability and tumorigenesis is well established, its function in the bone-fat switch of BMSCs is still largely unexplored. During the process of osteogenic differentiation, we observed an increase in the expression of EEF1B2, while a decrease in its expression was noted during adipogenesis. Suppression of EEF1B2 hindered the process of osteogenic differentiation and mineralization while promoting adipogenic differentiation. On the contrary, overexpression of EEF1B2 enhanced osteogenesis and strongly inhibited adipogenesis. Furthermore, the excessive expression of EEF1B2 in the tibias has the potential to mitigate bone loss and decrease marrow adiposity in mice with osteoporosis. In terms of mechanism, the suppression of β-catenin activity occurred when EEF1B2 function was suppressed during osteogenesis. Our collective findings indicate that EEF1B2 functions as a regulator, influencing the differentiation of BMSCs and maintaining a balance between bone and fat. Our finding highlights its potential as a therapeutic target for diseases related to bone metabolism.
    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:真核延伸因子1A1(eEF1A1)是一种与细胞中PARK2活性相关的RNA结合蛋白,提示在帕金森病(PD)中的可能作用。
    目的:明确eEF1A1是否通过转录或转录后调控在PD中发挥作用。
    方法:从GEO数据库下载GSE68719数据集,所有脑组织尸检的RNA-seq数据来自29例PD患者和44例神经系统正常对照受试者。为了抑制eEF1A1在U251细胞中的表达,siRNA被转染到这些细胞中,和RNA-seq高通量测序用于确定由eEF1A1敲低产生的差异表达基因(DEGs)和差异可变剪接事件(ASEs)。
    结果:eEF1A1在BA9区PD脑组织中显著过表达。GO和KEGG富集分析显示eEF1A1敲低显著上调CXCL10、NGF、PTX3、IL6、ST6GALNAC3、NUPR1、TNFRSF21和CXCL2并上调ACOT7、DDX10、SHMT2、MYEF2和NDUFAF5基因的可变剪接。这些基因富含与PD发病机制相关的通路,如细胞凋亡,炎症反应,和线粒体功能障碍。
    结论:结果表明,eEF1A1通过调节基因的差异表达和可变剪接参与PD的发生,为后续研究提供理论基础。
    BACKGROUND: Eukaryotic elongation factor 1A1 (eEF1A1) is an RNA-binding protein that is associated with PARK2 activity in cells, suggesting a possible role in Parkinson\'s disease (PD).
    OBJECTIVE: To clear whether eEF1A1 plays a role in PD through transcriptional or posttranscriptional regulation.
    METHODS: The GSE68719 dataset was downloaded from the GEO database, and the RNA-seq data of all brain tissue autopsies were obtained from 29 PD patients and 44 neurologically normal control subjects. To inhibit eEF1A1 from being expressed in U251 cells, siRNA was transfected into those cells, and RNA-seq high-throughput sequencing was used to determine the differentially expressed genes (DEGs) and differentially alternative splicing events (ASEs) resulting from eEF1A1 knockdown.
    RESULTS: eEF1A1 was significantly overexpressed in PD brain tissue in the BA9 area. GO and KEGG enrichment analyses revealed that eEF1A1 knockdown significantly upregulated the expression of the genes CXCL10, NGF, PTX3, IL6, ST6GALNAC3, NUPR1, TNFRSF21, and CXCL2 and upregulated the alternative splicing of the genes ACOT7, DDX10, SHMT2, MYEF2, and NDUFAF5. These genes were enriched in pathways related to PD pathogenesis, such as apoptosis, inflammatory response, and mitochondrial dysfunction.
    CONCLUSIONS: The results suggesting that eEF1A1 involved in the development of PD by regulating the differential expression and alternative splicing of genes, providing a theoretical basis for subsequent research.
    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
    胰腺导管腺癌(PDAC)是全球范围内癌症相关死亡的主要原因之一。主要是由于它的快速发展。目前PDAC的治疗方案有限,需要更好地了解PDAC进展的潜在机制,以确定改进的治疗策略.这里,我们确定FBXO32是PDAC的致癌驱动因子。FBXO32在PDAC中异常上调,而FBXO32高表达与PDAC患者的不良预后显著相关。FRG1缺乏促进PDAC中FBXO32的上调。FBXO32在体外促进细胞迁移和侵袭,在体内促进肿瘤的生长和转移。机械上,FBXO32直接与eEF1A1相互作用,并促进其在K273位点的聚泛素化,导致eEF1A1活性增强和PDAC细胞中蛋白质合成增加。此外,FBXO32催化的eEF1A1泛素化促进了ITGB5mRNA的翻译并激活了FAK信号,从而促进焦点粘附组装和驱动PDAC进展。重要的是,干扰FBXO32-eEF1A1轴或defactinib对FAK的药物抑制,FDA批准的FAK抑制剂,显著抑制由异常激活的FBXO32-eEF1A1信号驱动的PDAC生长和转移。总的来说,这项研究揭示了PDAC细胞依赖FBXO32介导的eEF1A1激活来驱动进展和转移的机制.FBXO32可以作为一个有前途的生物标志物,用于选择符合条件的PDAC患者进行defactinib治疗。
    Pancreatic ductal adenocarcinoma (PDAC) is one of the leading causes of cancer-related death worldwide, primarily due to its rapid progression. The current treatment options for PDAC are limited, and a better understanding of the underlying mechanisms responsible for PDAC progression is required to identify improved therapeutic strategies. In this study, we identified FBXO32 as an oncogenic driver in PDAC. FBXO32 was aberrantly upregulated in PDAC, and high FBXO32 expression was significantly associated with an unfavorable prognosis in patients with PDAC. FRG1 deficiency promoted FBXO32 upregulation in PDAC. FBXO32 promoted cell migration and invasion in vitro and tumor growth and metastasis in vivo. Mechanistically, FBXO32 directly interacted with eEF1A1 and promoted its polyubiquitination at the K273 site, leading to enhanced activity of eEF1A1 and increased protein synthesis in PDAC cells. Moreover, FBXO32-catalyzed eEF1A1 ubiquitination boosted the translation of ITGB5 mRNA and activated focal adhesion kinase (FAK) signaling, thereby facilitating focal adhesion assembly and driving PDAC progression. Importantly, interfering with the FBXO32-eEF1A1 axis or pharmaceutical inhibition of FAK by defactinib, an FDA-approved FAK inhibitor, substantially inhibited PDAC growth and metastasis driven by aberrantly activated FBXO32-eEF1A1 signaling. Overall, this study uncovers a mechanism by which PDAC cells rely on FBXO32-mediated eEF1A1 activation to drive progression and metastasis. FBXO32 may serve as a promising biomarker for selecting eligible patients with PDAC for treatment with defactinib. Significance: FBXO32 upregulation in pancreatic cancer induced by FRG1 deficiency increases eEF1A1 activity to promote ITGB5 translation and stimulate FAK signaling, driving cancer progression and sensitizing tumors to the FAK inhibitor defactinib.
    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
    结直肠癌(CRC)是第三大常见癌症,在全球范围内导致高死亡率。虽然CRC已被广泛研究,分子机制尚不完全清楚。真核翻译延伸因子1δ(EEF1D)参与各种肿瘤的进展,然而,EEF1D对CRC的影响尚不清楚.这里,我们旨在确定EEF1D在CRC中的潜在机制。在CRC样品中评估EEF1D的表达水平。在体外和体内检测CRC中EEF1D的功能分析。通过RNA免疫沉淀鉴定了EEF1D的调控机制,RNA下拉法,和蛋白质组学分析。我们的发现证实EEF1D在人类CRC组织中上调。功能上,EEF1D过表达加速细胞增殖和转移,而EEF1D敲除在体外和体内CRC模型中均抑制细胞增殖和转移。此外,我们显示,通过与EEF1DmRNA的3UTR结合,SRSF9上调EEF1D。EEF1D敲低逆转了SRSF9过表达诱导的恶性表型。这些发现表明EEF1D促进CRC进展,EEF1D可能是抗CRC的分子靶标。
    Colorectal cancer (CRC) is the third most common cancer and causes high mortality worldwide. Although CRC has been studied widely, the molecular mechanism is not completely known. Eukaryotic translation elongation factor 1 delta (EEF1D) participates in the progression of various tumors, however, the effect of EEF1D on CRC remains unclear. Here, we aimed to identify the potential mechanism of EEF1D in CRC. The expression levels of EEF1D were assessed in CRC samples. Functional analysis of EEF1D in CRC was detected in vitro and in vivo. The regulatory mechanism of EEF1D was identified with RNA immunoprecipitation, RNA pull-down assay, and proteomics analysis. Our findings confirmed that EEF1D was upregulated in human CRC tissues. Functionally, EEF1D overexpression accelerated cell proliferation and metastasis, whereas EEF1D knockdown inhibited cell proliferation and metastasis both in vitro and in vivo CRC models. Furthermore, we showed that EEF1D was upregulated by SRSF9 via binding to 3\'UTR of EEF1D mRNA. EEF1D knockdown reversed the malignant phenotype induced by SRSF9 overexpression. These findings demonstrated that EEF1D promotes CRC progression, and EEF1D may be a molecular target against CRC.
    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
    骨骼肌肌生成取决于基因调控,由分子机制精心策划。虽然转录因子和非编码RNA在肌肉发生中的作用是众所周知的,RNA结合蛋白(RBP)的作用至今仍不清楚.因此,研究转录后调节因子在肌生成中的功能,并发现调节肌生成的新功能RBPs,我们采用CRISPR高通量RBP-KO(RBP全基因敲除)文库筛选.通过这种方法,我们成功地确定了Eef1a1是一种新的调节因子。使用CRISPR敲除(CRISPRko)和CRISPR干扰(CRISPRi)技术,我们成功建立了CRISPRko和CRISPRi的细胞模型。我们的发现表明,Eef1a1在促进C2C12成肌细胞的增殖中起着至关重要的作用。通过siRNA抑制和过表达方法,我们进一步阐明了Eef1a1在促进增殖和抑制分化过程中的作用。RIP(RNA免疫沉淀),miRNA下拉,和双荧光素酶报告基因测定证实miR-133a-3p靶向Eef1a1。共转染实验表明miR-133a-3p可以挽救Eef1a1对C2C12成肌细胞的作用。总之,我们的研究利用CRISPR文库高通量筛选来揭示一种新型RBP,Eef1a1,参与调节肌生成。Eef1a1在抑制分化过程的同时促进成肌细胞的增殖。此外,它作为miR-133a-3p的拮抗剂,从而调节肌生成的过程。
    Skeletal muscle myogenesis hinges on gene regulation, meticulously orchestrated by molecular mechanisms. While the roles of transcription factors and non-coding RNAs in myogenesis are widely known, the contribution of RNA-binding proteins (RBPs) has remained unclear until now. Therefore, to investigate the functions of post-transcriptional regulators in myogenesis and uncover new functional RBPs regulating myogenesis, we employed CRISPR high-throughput RBP-KO (RBP-wide knockout) library screening. Through this approach, we successfully identified Eef1a1 as a novel regulatory factor in myogenesis. Using CRISPR knockout (CRISPRko) and CRISPR interference (CRISPRi) technologies, we successfully established cellular models for both CRISPRko and CRISPRi. Our findings demonstrated that Eef1a1 plays a crucial role in promoting proliferation in C2C12 myoblasts. Through siRNA inhibition and overexpression methods, we further elucidated the involvement of Eef1a1 in promoting proliferation and suppressing differentiation processes. RIP (RNA immunoprecipitation), miRNA pull-down, and Dual-luciferase reporter assays confirmed that miR-133a-3p targets Eef1a1. Co-transfection experiments indicated that miR-133a-3p can rescue the effect of Eef1a1 on C2C12 myoblasts. In summary, our study utilized CRISPR library high-throughput screening to unveil a novel RBP, Eef1a1, involved in regulating myogenesis. Eef1a1 promotes the proliferation of myoblasts while inhibiting the differentiation process. Additionally, it acts as an antagonist to miR-133a-3p, thus modulating the process of myogenesis.
    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
    氨基酸对于激活雷帕霉素(mTOR)的机制靶标至关重要,但相应的分子机制尚未完全了解。我们先前发现Met刺激牛乳腺上皮细胞(MEC)中的真核延伸因子α(eEF1Bα)核定位。在这里,我们探讨了eEF1Bα在蛋氨酸(Met)和亮氨酸(Leu)刺激的mTOR基因转录和MECs牛奶合成中的作用和分子机制。eEF1Bα敲低降低了牛奶蛋白质和脂肪的合成,细胞增殖,和mTORmRNA表达和磷酸化,而eEF1Bα过表达具有相反的作用。QE-MS分析检测到eEF1Bα在细胞核中的Ser106处被磷酸化,Met和Leu刺激p-eEF1Bα核定位。eEF1Bα敲低消除了mTORmRNA表达和磷酸化对Met和Leu的刺激,这种调节作用依赖于它的磷酸化。Akt敲除阻断了eEF1Bα和p-eEF1Bα表达对Met和Leu的刺激。ChIP-PCR检测到p-eEF1Bα仅与mTOR启动子中的-548至-793nt位点结合,和ChIP-qPCR进一步检测到Met和Leu刺激了这种结合。eEF1Bα通过诱导富含AT的相互作用域1A(ARID1A)泛素化降解介导Met和Leu对mTORmRNA表达和磷酸化的刺激,这个过程依赖于eEF1Bα的磷酸化。p-eEF1Bα与ARID1A和泛素蛋白连接酶E3模块N-识别5(UBR5)相互作用,UBR5敲低通过eEF1Bα过表达挽救了ARID1A蛋白水平的降低。在哺乳期,eEF1Bα和p-eEF1Bα在小鼠乳腺组织中均高表达。总之,我们发现Met和Leu通过eEF1α-UBR5-ARID1A信号传导刺激mTOR转录激活和MECs中乳蛋白和脂肪的合成。
    Amino acids are essential for the activation of the mechanistic target of rapamycin (mTOR), but the corresponding molecular mechanism is not yet fully understood. We previously found that Met stimulated eukaryotic elongation factor α (eEF1Bα) nuclear localization in bovine mammary epithelial cells (MECs). Herein, we explored the role and molecular mechanism of eEF1Bα in methionine (Met)- and leucine (Leu)-stimulated mTOR gene transcription and milk synthesis in MECs. eEF1Bα knockdown decreased milk protein and fat synthesis, cell proliferation, and mTOR mRNA expression and phosphorylation, whereas eEF1Bα overexpression had the opposite effects. QE-MS analysis detected that eEF1Bα was phosphorylated at Ser106 in the nucleus and Met and Leu stimulated p-eEF1Bα nuclear localization. eEF1Bα knockdown abrogated the stimulation of Met and Leu by mTOR mRNA expression and phosphorylation, and this regulatory role was dependent on its phosphorylation. Akt knockdown blocked the stimulation of Met and Leu by eEF1Bα and p-eEF1Bα expression. ChIP-PCR detected that p-eEF1Bα bound only to the -548 to -793 nt site in the mTOR promoter, and ChIP-qPCR further detected that Met and Leu stimulated this binding. eEF1Bα mediated Met and Leu\' stimulation on mTOR mRNA expression and phosphorylation through inducing AT-rich interaction domain 1A (ARID1A) ubiquitination degradation, and this process depended on eEF1Bα phosphorylation. p-eEF1Bα interacted with ARID1A and ubiquitin protein ligase E3 module N-recognition 5 (UBR5), and UBR5 knockdown rescued the decrease of the ARID1A protein level by eEF1Bα overexpression. Both eEF1Bα and p-eEF1Bα were highly expressed in mouse mammary gland tissues during the lactating period. In summary, we reveal that Met and Leu stimulate mTOR transcriptional activation and milk protein and fat synthesis in MECs through eEF1Bα-UBR5-ARID1A signaling.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号