Akt

Akt
  • 文章类型: Journal Article
    非洲猪瘟(ASF)由非洲猪瘟病毒(ASFV)引起,是最重要的传染病之一,由于缺乏有效的疫苗,导致猪的高发病率和死亡率以及受影响国家的猪肉行业的巨大经济损失。需要开发替代的强大的抗病毒对策,尤其是抗ASFV药物,是最紧迫的。这项研究表明,芳奇洛林(FAN),一种双苄基异喹啉生物碱,存在于半子科的Stephaniatetrandra的根中,在微摩尔浓度(IC50=1.66µM)下显著抑制猪肺泡巨噬细胞(PAMs)中的ASFV复制。机械上,ASFV感染可触发AKT/mTOR/NF-κB信号通路。FAN显著抑制ASFV诱导的这些途径的激活,从而抑制病毒复制。使用AKT抑制剂MK2206证实了这种机制,因为它抑制了PAMs中的AKT磷酸化和ASFV复制。本研究结果提示AKT/mTOR通路有可能成为对抗ASFV感染的治疗策略,FAN有可能成为对抗ASFV感染的有效新型抗病毒药物,值得进一步进行体内抗病毒评价.
    African swine fever (ASF), caused by the African swine fever virus (ASFV), is one of the most important infectious diseases that cause high morbidity and mortality in pigs and substantial economic losses to the pork industry of affected countries due to the lack of effective vaccines. The need to develop alternative robust antiviral countermeasures, especially anti-ASFV agents, is of the utmost urgency. This study shows that fangchinoline (FAN), a bisbenzylisoquinoline alkaloid found in the roots of Stephania tetrandra of the family Menispermaceae, significantly inhibits ASFV replication in porcine alveolar macrophages (PAMs) at micromolar concentrations (IC50 = 1.66 µM). Mechanistically, the infection of ASFV triggers the AKT/mTOR/NF-κB signaling pathway. FAN significantly inhibits ASFV-induced activation of such pathways, thereby suppressing viral replication. Such a mechanism was confirmed using an AKT inhibitor MK2206 as it inhibited AKT phosphorylation and ASFV replication in PAMs. Altogether, the results suggest that the AKT/mTOR pathway could potentially serve as a treatment strategy for combating ASFV infection and that FAN could potentially emerge as an effective novel antiviral agent against ASFV infections and deserves further in vivo antiviral evaluations.
    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
    富含肌肉的A型层粘连蛋白相互作用蛋白(MLIP)是一种新兴的蛋白质,参与细胞稳态和压力适应。真核细胞调节各种细胞过程,包括新陈代谢,DNA修复,和细胞周期进程,维持细胞内稳态.这种稳态的破坏会导致癌症等疾病,以不受控制的细胞生长和分裂为特征。本综述旨在首次探讨MLIP在癌症发生发展中的独特作用。鉴于其与PI3K/Akt/mTOR通路的相互作用,p53、MAPK9和FOXO转录因子,细胞稳态和肿瘤抑制的所有关键调节剂。我们讨论了目前对MLIP参与促生存途径的理解及其在癌细胞代谢重塑和体内稳态失调中的潜在意义。此外,我们研究了MLIP作为癌症治疗新治疗靶点的潜力.这篇综述旨在阐明MLIP对癌症生物学的潜在影响,并有助于开发创新的治疗策略。
    Muscle-enriched A-type lamin-interacting protein (MLIP) is an emerging protein involved in cellular homeostasis and stress adaptation. Eukaryotic cells regulate various cellular processes, including metabolism, DNA repair, and cell cycle progression, to maintain cellular homeostasis. Disruptions in this homeostasis can lead to diseases such as cancer, characterized by uncontrolled cell growth and division. This review aims to explore for the first time the unique role MLIP may play in cancer development and progression, given its interactions with the PI3K/Akt/mTOR pathway, p53, MAPK9, and FOXO transcription factors, all critical regulators of cellular homeostasis and tumor suppression. We discuss the current understanding of MLIP\'s involvement in pro-survival pathways and its potential implications in cancer cells\' metabolic remodeling and dysregulated homeostasis. Additionally, we examine the potential of MLIP as a novel therapeutic target for cancer treatment. This review aims to shed light on MLIP\'s potential impact on cancer biology and contribute to developing innovative therapeutic strategies.
    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:N-乙酰转移酶10(NAT10),人类唯一已知的RNA胞嘧啶乙酰转移酶,有助于癌症的发生和发展。本研究旨在探讨NAT10对胃癌恶性生物学特性的影响及其机制。
    方法:使用癌症基因组图谱(TCGA)和中山大学(SYSU)队列分析NAT10在GC中的表达和预后意义。用细胞计数试剂盒(CCK-8)法检测NAT10对GC恶性生物学行为的影响,平板集落形成试验,5-乙炔基-2'-脱氧尿苷(EdU),Transwell迁移和入侵测定,划痕伤口试验,流式细胞仪分析,和动物研究。通过液相色谱-串联质谱(LC-MS/MS)检测GC中N4乙酰胞苷(ac4C)的总体水平。NAT10的下游信号通路通过基因集富集分析(GSEA)进行分析,并通过蛋白质印迹(WB)和免疫荧光(IF)进行验证。
    结果:GC中NAT10表达的显著上调与不良预后相关。NAT10的敲减显著抑制GC细胞增殖,迁移,入侵,和细胞周期进程。下调NAT10可降低ac4C水平并抑制GC中AKT磷酸化和上皮-间质转化(EMT)。
    结论:NAT10作为癌基因发挥作用,可能为GC提供新的治疗靶点。
    BACKGROUND: N-acetyltransferase 10 (NAT10), the only RNA cytosine acetyltransferase known in humans, contributes to cancer tumorigenesis and progression. This study aims to investigate the effect of NAT10 on the malignant biological properties of gastric cancer (GC) and its underlying mechanism.
    METHODS: The expression and prognostic significance of NAT10 in GC were analyzed using The Cancer Genome Atlas (TCGA) and Sun Yat-sen University (SYSU) cohorts. The influence of NAT10 on the malignant biological behaviors of GC was detected by Cell Counting Kit-8 (CCK-8) assay, plate colony formation assay, 5-ethynyl-2\'-deoxyuridine (EdU), Transwell migration and invasion assays, scratch wound assay, flow cytometric analysis, and animal studies. The overall level of N4 acetylcytidine (ac4C) in GC was detected by liquid chromatography with tandem mass spectrometry (LC-MS/MS). The downstream signal pathways of NAT10 were analyzed by Gene Set Enrichment Analysis (GSEA) and verified by Western blot (WB) and immunofluorescence (IF).
    RESULTS: The significant upregulation of NAT10 expression in GC was associated with a poor prognosis. The knockdown of NAT10 markedly suppressed GC cell proliferation, migration, invasion, and cell cycle progression. Downregulating NAT10 reduced ac4C levels and inhibited AKT phosphorylation and epithelial-mesenchymal transition (EMT) in GC.
    CONCLUSIONS: NAT10 functions as an oncogene and may provide a new therapeutic target in GC.
    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
    人参C.A.Mey。(五花香科;人参),一种在东亚常用的传统植物,已证明在治疗神经损害性疾病和糖尿病方面有效。然而,其在缓解2型糖尿病(T2DM)中的确切作用和机制有待进一步研究。本研究的目的是探索人参提取物的药理作用,阐明其保护胰岛和促进β细胞再生的潜在机制。
    通过链脲佐菌素联合高脂饮食诱导T2DM小鼠模型。在施用人参提取物后的第7天和第28天处死两批小鼠。体重,空腹血糖水平,并检测到葡萄糖耐量。通过H&E染色检查胰岛的形态变化。血清胰岛素水平,胰高血糖素,使用ELISA测量GLP-1和炎症因子。通过胰岛素和PCNA双重免疫荧光染色评价人参提取物促进胰岛β细胞再生的能力。此外,通过胰岛素和胰高血糖素双重免疫荧光染色探讨了β细胞再生的机制,伴随着免疫组织化学染色和蛋白质印迹分析。
    本研究表明,人参提取物可缓解小鼠2型糖尿病的症状,包括降低血糖水平和改善葡萄糖耐量。血清胰岛素水平,服用人参提取物后,GLP-1和IL-10增加,而胰高血糖素的水平,TNF-α,IL-1β下降。人参提取物保存正常胰岛形态,新生β细胞群增加,并抑制胰岛内的炎症浸润,此外,α细胞比例降低,β细胞比例增加。机械上,人参提取物可能抑制ARX和MAFB的表达,增加MAFA水平以帮助α细胞向β细胞转化,并激活AKT-FOXM1/细胞周期蛋白D2以增强β细胞增殖。我们的研究表明,人参提取物可能是治疗T2DM的一种有前途的疗法,尤其是那些有胰岛损伤的人。
    UNASSIGNED: Panax ginseng C. A. Mey. (Araliaceae; Ginseng Radix et Rhizoma), a traditional plant commonly utilized in Eastern Asia, has demonstrated efficacy in treating neuro-damaging diseases and diabetes mellitus. However, its precise roles and mechanism in alleviating type 2 diabetes mellitus (T2DM) need further study. The objective of this study is to explore the pharmacological effects of ginseng extract and elucidate its potential mechanisms in protecting islets and promoting β-cell regeneration.
    UNASSIGNED: The T2DM mouse model was induced through streptozotocin combined with a high-fat diet. Two batches of mice were sacrificed on the 7th and 28th days following ginseng extract administration. Body weight, fasting blood glucose levels, and glucose tolerance were detected. Morphological changes in the pancreatic islets were examined via H & E staining. Levels of serum insulin, glucagon, GLP-1, and inflammatory factors were measured using ELISA. The ability of ginseng extract to promote pancreatic islet β-cell regeneration was evaluated through insulin & PCNA double immunofluorescence staining. Furthermore, the mechanism behind β-cells regeneration was explored through insulin & glucagon double immunofluorescence staining, accompanied by immunohistochemical staining and western blot analyses.
    UNASSIGNED: The present research revealed that ginseng extract alleviates symptoms of T2DM in mice, including decreased blood glucose levels and improved glucose tolerance. Serum levels of insulin, GLP-1, and IL-10 increased following the administration of ginseng extract, while levels of glucagon, TNF-α, and IL-1β decreased. Ginseng extract preserved normal islet morphology, increased nascent β-cell population, and inhibited inflammatory infiltration within the islets, moreover, it decreased α-cell proportion while increasing β-cell proportion. Mechanistically, ginseng extract might inhibit ARX and MAFB expressions, increase MAFA level to aid in α-cell to β-cell transformation, and activate AKT-FOXM1/cyclin D2 to enhance β-cell proliferation. Our study suggests that ginseng extract may be a promising therapy in treating T2DM, especially in those with islet injury.
    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
    SIRT6,一种进化保守的组蛋白脱乙酰酶,在我们先前的研究中,已被确定为Akt/FoxO3a的新的直接下游靶标和结肠癌的肿瘤抑制因子。然而,SIRT6阻碍肿瘤发展的确切机制尚不清楚.为了确定SIRT6是否直接影响Survivin转录,使用抗SIRT6抗体进行ChIP测定以分离DNA。合成YM155以探讨Survivin在线粒体凋亡中的作用,自噬与肿瘤进展。我们对Survivin调节的研究涉及活细胞中的实时荧光成像,实时PCR,免疫组织化学,流式细胞术,和异种移植小鼠试验。在目前的研究中,我们深入研究了SIRT6在结肠癌中的作用,并确定激活的SIRT6通过降低Survivin表达触发线粒体凋亡.随后的检查显示SIRT6直接与Survivin启动子结合,阻碍它的转录。值得注意的是,Survivin的直接抑制通过诱导线粒体凋亡和自噬在体外和体内显着阻碍结肠癌的增殖。更有趣的是,Survivin抑制重新激活Akt/FoxO3a通路和升高的SIRT6水平,建立正反馈循环。我们的结果将Survivin确定为SIRT6的新型下游转录靶标,可促进肿瘤生长,并有望成为结肠癌治疗的前瞻性靶标。
    SIRT6, an evolutionarily conserved histone deacetylase, has been identified as a novel direct downstream target of Akt/FoxO3a and a tumor suppressor in colon cancer in our previous research. Nevertheless, the precise mechanisms through which SIRT6 hinders tumor development remain unclear. To ascertain whether SIRT6 directly impacts Survivin transcription, a ChIP assay was conducted using an anti-SIRT6 antibody to isolate DNA. YM155 was synthesized to explore Survivin\'s role in mitochondrial apoptosis, autophagy and tumor progression. Our investigation into the regulation of Survivin involved real-time fluorescence imaging in living cells, real-time PCR, immunohistochemistry, flow cytometry, and xenograft mouse assays. In this current study, we delved into the role of SIRT6 in colon cancer and established that activated SIRT6 triggers mitochondrial apoptosis by reducing Survivin expression. Subsequent examinations revealed that SIRT6 directly binds to the Survivin promoter, impeding its transcription. Notably, direct inhibition of Survivin significantly impeded colon cancer proliferation by inducing mitochondrial apoptosis and autophagy both in vitro and in vivo. More interestingly, Survivin inhibition reactivated the Akt/FoxO3a pathway and elevated SIRT6 levels, establishing a positive feedback loop. Our results identify Survivin as a novel downstream transcriptional target of SIRT6 that fosters tumor growth and holds promise as a prospective target for colon cancer therapy.
    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
    肝细胞核因子-1(HNF1α)是多种肿瘤进展的转录因子。然而,关于HNF1α活性的机制知之甚少。本研究旨在探讨HNF1α在结直肠癌(CRC)进展中的作用及其分子机制。CRC标本中HNF1α表达上调,高表达与CRC患者预后不良相关。HNF1α敲低和过表达抑制和促进增殖,CRC细胞在体外和体内的迁移和侵袭。机械上,HNF1α增加己糖激酶结构域组分1(HKDC1)启动子的转录活性,从而激活AKT/AMPK信号。同时,HKDC1上调对增殖很重要,HKDC1基因敲除后可显著逆转CRC细胞的增殖、迁移和侵袭,HNF1α过表达诱导的迁移和侵袭。一起来看,HNF1α通过与HKDC1结合并激活AKT/AMPK信号参与CRC的进展和转移。靶向HNF1α可能是CRC患者的潜在治疗策略。
    The hepatocyte nuclear factor-1 (HNF1ɑ) is a transcription factor that contributes to several kinds of cancer progression. However, very little is known regarding the mechanisms underlying the activity of HNF1ɑ. We aimed to explore the role of HNF1ɑ in the progress of colorectal cancer (CRC) and elucidate its molecular mechanism. HNF1ɑ expression was upregulated in CRC samples and high expression of HNF1ɑ was associated with poor prognosis of CRC patients. HNF1α knockdown and overexpression inhibited and promoted proliferation, migration and invasion of CRC cells both in vitro and in vivo respectively. Mechanistically, HNF1ɑ increased the transcriptional activity of hexokinase domain component 1(HKDC1)promoter, thus activated AKT/AMPK signaling. Meanwhile, HKDC1 upregulation was important for the proliferation, migration and invasion of CRC cells and knockdown of HKDC1 significantly reversed the proliferation, migration and invasion induced by HNF1α overexpression. Taken together, HNF1ɑ contributes to CRC progression and metastasis through binding to HKDC1 and activating AKT/AMPK signaling. Targeting HNF1ɑ could be a potential therapeutic strategy for CRC patients.
    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
    尽管抗HER2治疗在减少HER2阳性乳腺癌的转移和复发方面取得了重大进展,对曲妥珠单抗等药物的耐药性,帕妥珠单抗,拉帕替尼经常在接受治疗的患者中发展。先前的研究表明,PIK3CA/PTEN基因突变对PI3K-AKT信号通路的过度激活与HER2抗性有关。在这项研究中,我们介绍了一种新型的PI3K-p110αPROteasolsisTAretting嵌合体(PROTAC),该蛋白通过降解PI3K-p110α有效抑制乳腺癌细胞的增殖。在两种拉帕替尼耐药细胞系中进行测试时,JIMT1和MDA-MB-453,两者都有PIK3CA突变,PI3KPROTAC显著降低细胞增殖并诱导G1期细胞周期阻滞。重要的是,即使浓度很低,PI3KPROTAC恢复对拉帕替尼的敏感性。此外,PI3KPROTAC的功效超过了Alpelisib,一种临床上选择性的PI3K-p110α激酶抑制剂。PI3KPROTAC的优异性能也在拉帕替尼耐药乳腺癌异种移植肿瘤和患者来源的乳腺癌类器官(PDO)中得到证实。总之,这项研究表明,我们合成的新型PI3KPROTAC可作为克服拉帕替尼耐药的有效药物.
    Although anti-HER2 therapy has made significant strides in reducing metastasis and relapse in HER2-positive breast cancer, resistance to agents like trastuzumab, pertuzumab, and lapatinib frequently develops in patients undergoing treatment. Previous studies suggest that the hyperactivation of the PI3K-AKT signaling pathway by PIK3CA/PTEN gene mutations is implicated in HER2 resistance. In this study, we introduce a novel PI3K-p110α Proteolysis TAargeting Chimera (PROTAC) that effectively inhibits the proliferation of breast cancer cells by degrading PI3K-p110α. When tested in two lapatinib-resistant cell lines, JIMT1 and MDA-MB-453, both of which harbor PIK3CA mutations, the PI3K PROTAC notably reduced cell proliferation and induced G1 phase cell cycle arrest. Importantly, even at very low concentrations, PI3K PROTAC restored sensitivity to lapatinib. Furthermore, the efficacy of PI3K PROTAC surpassed that of Alpelisib, a selective PI3K-p110α kinase inhibitor in clinic. The superior performance of PI3K PROTAC was also confirmed in lapatinib-resistant breast cancer xenograft tumors and patient-derived breast cancer organoids (PDOs). In conclusion, this study reveals that the novel PI3K PROTAC we synthesized could serve as an effective agent to overcome lapatinib resistance.
    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
    环磷酰胺(CP)是最有效的免疫抑制剂之一。为了了解CP和MSCs在肾脏中使用的机制,我们调查了它们对某些途径的影响。
    使用4组雌性大鼠。GI:是用盐水溶液处理的正常对照组。GII组,GIII,和GIV用CP治疗。GI组和GII组在治疗后第4天处死。,GIII(自动愈合组)在CP注射6天后不进行治疗。在CP注射后的第4天用MSCs处理GIV组。GIII和GIV组在治疗后6天处死,肾脏被切除和处理。
    CP诱导CD14和CD21阳性细胞以及胱天蛋白酶的上调。组III和IV中先前标志物的显著下调。CP对AKT/PI3K有下调作用,在III组和IV组中得到改善。IV组P53、BCL2和VEGF显著升高(P<005)。
    MSCs通过PI3K/AKT通路在CP处理大鼠的免疫抑制中发挥重要作用。
    UNASSIGNED: Cyclophosphamide (CP) is one of the most effective immunosuppressive agents. To understand the mechanisms used by the CP and MSCs in the kidney, we investigated their effects on some pathways.
    UNASSIGNED: 4 groups of female rats were used. GI: was the normal control group treated with saline solution. Groups G II, G III, and G IV were treated with CP. G I and G II groups were sacrificed on the fourth day after treatment., G III (Auto healing group) was left without treatment after the CP injection for six days. The G IV group was treated with MSCs on the fourth day after the CP injection. G III and G IV groups were sacrificed six days after treatment, and the kidney was removed and processed.
    UNASSIGNED: CP induced up-regulation in CD14 and CD21 positive cells and caspase. Significant down-regulation of previous markers in groups III and IV. CP exerted a downregulation effect on AKT/ PI3K, that were ameliorated in groups III and IV. A significant increase in P53, BCL2, as well as VEGF in Group IV (P < 0 05).
    UNASSIGNED: MSCs play a vital function in the immune inhibition in CP-treated rats through PI3K/AKT 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
    背景:骨骼肌缺血再灌注损伤(IRI)是临床实践中遇到的普遍情况,以肌营养不良为特征。由于有限的治疗选择和不良预后,它会导致运动障碍,组织损伤,和残疾。本研究旨在确定和验证瞬时受体电位规范6(TRPC6)对骨骼肌IRI的影响,探讨TRPC6在骨骼肌IRI发生中的作用及TRPC6激活的信号转导通路,为骨骼肌IRI的治疗和干预提供新的见解。
    方法:建立体内缺血/再灌注(I/R)和体外缺氧/复氧(H/R)模型,数据在组织病理学上进行了全面分析,细胞,和分子水平,以及对小鼠运动能力的评价。
    结果:通过比较TRPC6基因敲除小鼠和野生型小鼠,我们发现TRPC6敲除TRPC6可以减轻I/R或H/R后骨骼肌损伤,骨骼肌,从而恢复小鼠的一些运动能力。TRPC6敲低可降低细胞内Ca2+过载,therebyo减少细胞凋亡。此外,我们还发现TRPC6不仅是参与骨骼肌IRII/R损伤的关键离子通道,但也可以影响Ca2水平,然后影响磷脂酰肌醇3-激酶/蛋白激酶B/哺乳动物雷帕霉素靶蛋白(PI3K/Akt/mTOR)信号通路。因此,通过击倒,抑制TRPC6,从而减轻骨骼肌I/R或H/R引起的损伤。
    结论:这些发现数据表明,TRPC6exacerbates的存在可以加重I/Rischemia/再灌注后骨骼肌损伤的损伤,导致不仅导致Ca2+过载和细胞凋亡。,此外,它通过抑制PI3K/Akt/mTOR信号通路的表达而削弱细胞的自我修复能力。探讨TRPC6在骨骼肌中的功能和作用可能为骨骼肌缺血/再灌注损伤的治疗提供了新的方法。
    BACKGROUND: Skeletal muscle ischaemia-reperfusion injury (IRI) is a prevalent condition encountered in clinical practice, characterised by muscular dystrophy. Owing to limited treatment options and poor prognosis, it can lead to movement impairments, tissue damage, and disability. This study aimed to determine and verify the influence of transient receptor potential canonical 6 (TRPC6) on skeletal muscle IRI, and to explore the role of TRPC6 in the occurrence of skeletal muscle IRI and the signal transduction pathways activated by TRPC6 to provide novel insights for the treatment and intervention of skeletal muscle IRI.
    METHODS: In vivo ischaemia/reperfusion (I/R) and in vitro hypoxia/reoxygenation (H/R) models were established, and data were comprehensively analysed at histopathological, cellular, and molecular levels, along with the evaluation of the exercise capacity in mice.
    RESULTS: By comparing TRPC6 knockout mice with wild-type mice, we found that TRPC6 knockout of TRPC6 could reduced skeletal muscle injury after I/R or H/R, of skeletal muscle, so as therebyto restoringe some exercise capacity inof mice. TRPC6 knockdown can reduced Ca2+ overload in cells, therebyo reducinge apoptosis. In additionAdditionally, we also found that TRPC6 functionsis not only a key ion channel involved in skeletal muscle I/R injury, but also can affects Ca2+ levels and then phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/Akt/mTOR) signalling pathway. by knocking downTherefore, knockdown of TRPC6, so as to alleviated the injury inducedcaused by skeletal muscle I/R or and H/R.
    CONCLUSIONS: These findingsdata indicate that the presence of TRPC6 exacerbatescan aggravate the injury of skeletal muscle injury after I/Rischemia/reperfusion, leading towhich not only causes Ca2+ overload and apoptosis., Additionally, it impairsbut also reduces the self- repair ability of cells by inhibiting the expression of the PI3K/Akt/mTOR signalling pathway. ETo exploringe the function and role of TRPC6 in skeletal muscle maycan presentprovide a novelew approachidea for the treatment of skeletal muscle ischemia/reperfusion injury.
    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
    微粒体谷胱甘肽转移酶3(MGST3)调节类花生酸和谷胱甘肽代谢。这些过程与氧化应激和细胞凋亡有关,提示MGST3可能在阿尔茨海默病(AD)的病理生理中起作用。这里,我们报道,细胞系中MGST3的敲除(KD)降低了β-位点淀粉样前体蛋白裂解酶1(BACE1)的蛋白水平,并导致淀粉样蛋白生成.有趣的是,MGST3KD不改变细胞内ROS水平,但选择性地降低了可能与半胱氨酰白三烯(cysLTs)受体相关的凋亡指标的表达,MGST3的下游代谢产物参与花生四烯酸通路。然后,我们表明MGST3对BACE1的作用与cysLTs无关,但涉及翻译机制。进一步的RNA-seq分析鉴定G蛋白信号传导4(RGS4)的调节因子是MGST3的靶基因。RGS4的沉默抑制BACE1翻译并阻止MGST3KD介导的BACE1还原。潜在机制与AKT活性有关,由于MGST3和RGS4的沉默显著降低了磷酸化AKT(p-AKT)的蛋白水平,并且AKT抑制剂消除了MGST3/RGS4对p-AKT和BACE1的作用.一起,MGST3通过控制BACE1蛋白表达调节淀粉样蛋白生成,由RGS4和下游AKT信号通路介导。
    Microsomal glutathione transferase 3 (MGST3) regulates eicosanoid and glutathione metabolism. These processes are associated with oxidative stress and apoptosis, suggesting that MGST3 might play a role in the pathophysiology of Alzheimer\'s disease (AD). Here, we report that knockdown (KD) of MGST3 in cell lines reduced the protein level of beta-site amyloid precursor protein cleaving enzyme 1 (BACE1) and the resulting amyloidogenesis. Interestingly, MGST3 KD did not alter intracellular ROS level but selectively reduced the expression of apoptosis indicators which could be associated with the receptor of cysteinyl leukotrienes (cysLTs), the downstream metabolites of MGST3 in arachidonic acid pathway. We then showed that the effect of MGST3 on BACE1 was independent of cysLTs but involved a translational mechanism. Further RNA-seq analysis identified that regulator of G-protein signaling 4 (RGS4) was a target gene of MGST3. Silencing of RGS4 inhibited BACE1 translation and prevented MGST3 KD-mediated reduction of BACE1. The potential mechanism was related to AKT activity, as the protein level of phosphorylated AKT (p-AKT) was significantly reduced by silencing of MGST3 and RGS4, and the AKT inhibitor abolished the effect of MGST3/RGS4 on p-AKT and BACE1. Together, MGST3 regulated amyloidogenesis by controlling BACE1 protein expression, which was mediated by RGS4 and downstream AKT signaling pathway.
    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