Rictor

Rictor
  • 文章类型: Journal Article
    慢性疼痛的发展和维持涉及脊髓伤害性回路的重组。雷帕霉素复合物2(mTORC2)的机制靶点,调节肌动蛋白依赖性结构变化和mTORC1依赖性mRNA翻译的中心信号中枢,在海马突触可塑性和记忆形成中起关键作用。然而,其在脊柱可塑性和慢性疼痛中的功能知之甚少。在这里,我们显示脊髓mTORC2的药理激活诱导疼痛超敏反应,而它的抑制作用,使用mTORC2定义组件Rictor的下调,缓解炎症和神经性疼痛。Rictor的细胞类型特异性缺失表明,在一部分兴奋性神经元中选择性抑制mTORC2会损害脊髓突触增强作用,并减轻炎症诱导的机械和热超敏反应,和神经损伤引起的热痛觉过敏。抑制性中间神经元中mTORC2的消融强烈减轻了神经损伤引起的机械性超敏反应。我们的发现揭示了mTORC2在慢性疼痛中的作用,并强调了其在介导外周炎症和神经损伤后的疼痛超敏反应中的细胞类型特异性功能。
    The development and maintenance of chronic pain involves the reorganization of spinal nociceptive circuits. The mechanistic target of rapamycin complex 2 (mTORC2), a central signaling hub that modulates both actin-dependent structural changes and mTORC1-dependent mRNA translation, plays key roles in hippocampal synaptic plasticity and memory formation. However, its function in spinal plasticity and chronic pain is poorly understood. Here we show that pharmacological activation of spinal mTORC2 induces pain hypersensitivity, whereas its inhibition, using downregulation of the mTORC2-defining component Rictor, alleviates both inflammatory and neuropathic pain. Cell-type-specific deletion of Rictor showed that the selective inhibition of mTORC2 in a subset of excitatory neurons impairs spinal synaptic potentiation and alleviates inflammation-induced mechanical and thermal hypersensitivity, and nerve injury-induced heat hyperalgesia. The ablation of mTORC2 in inhibitory interneurons strongly alleviated nerve injury-induced mechanical hypersensitivity. Our findings reveal the role of mTORC2 in chronic pain and highlight its cell-type-specific functions in mediating pain hypersensitivity in response to peripheral inflammation and nerve 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
    高脂血症性胰腺炎(HP)是由血清甘油三酯(TG)水平升高引发的胰腺炎症损伤。雷帕霉素(mTOR)信号通路在调节脂质稳态和炎症中起着至关重要的作用。本研究旨在探讨mTOR复合物2(mTORC2)的活性是否影响HP的进展及其潜在机制。在体内,采用高脂饮食和牛磺胆酸钠逆行给药建立大鼠HP模型,与胰腺组织病理评估。检查了Rictor和过氧化物酶体增殖物激活物受体(PPAR)的表达。血清TG、脂肪酸代谢物,确定炎症和脂质代谢相关因子.体外,胰腺腺泡细胞(PACs)暴露于棕榈酸和胆囊收缩素-8。PAC细胞凋亡,焦亡,和铁性凋亡进行了评估。在HP型号中,大鼠和PAC表现出Rictor上调和PPARα下调,Rictor敲除促进PPARα表达。在体内,Rictor敲除降低血清TG水平,α-淀粉酶,总胆固醇,低密度脂蛋白胆固醇,乳酸脱氢酶,和炎症因子,同时增加高密度脂蛋白胆固醇水平。Rictor敲低增加了ACOX1和CPT1α,降低了SREBP-1,CD36,SCD1,ACLY,和ACACA。Rictor敲除减少对胰腺组织结构的损伤。体外,Rictor敲低抑制PAC细胞凋亡,焦亡,和铁中毒。用PPARα拮抗剂GW6471治疗消除了Rictor敲低的有益作用。Rictor/mTORC2缺乏降低血清TG水平,维持脂质稳态,并通过抑制PPARα表达来抑制炎症。减弱mTORC2活性有望成为HP的新型治疗策略。
    Hyperlipidemic pancreatitis (HP) is an inflammatory injury of the pancreas triggered by elevated serum triglyceride (TG) levels. The mechanistic target of rapamycin (mTOR) signaling pathway plays a crucial role in regulating lipid homeostasis and inflammation. This study aimed to investigate whether the activity of mTOR complex 2 (mTORC2) affects the progression of HP and its underlying mechanisms. In vivo, a high-fat diet and retrograde administration of sodium taurocholate were employed to establish the HP models in rats, with pancreatic tissue pathology evaluated. The expression of Rictor and peroxisome proliferator-activator receptor (PPAR) was examined. The serum levels of TG, fatty acid metabolites, inflammatory and lipid metabolism-related factors were determined. In vitro, pancreatic acinar cells (PACs) were exposed to palmitic acid and cholecystokinin-8. PAC apoptosis, pyroptosis, and ferroptosis were assessed. In the HP models, rats and PACs exhibited upregulated Rictor and downregulated PPARα, and Rictor knockdown promoted PPARα expression. In vivo, Rictor knockdown decreased the serum levels of TG, α-amylase, total cholesterol, low-density lipoprotein cholesterol, lactate dehydrogenase, and inflammatory factors, while increasing high-density lipoprotein cholesterol levels. Rictor knockdown increased ACOX1 and CPT1α and decreased SREBP-1, CD36, SCD1, ACLY, and ACACA. Rictor knockdown reduced damage to pancreatic tissue structure. In vitro, Rictor knockdown inhibited PAC apoptosis, pyroptosis, and ferroptosis. Treatment with the PPARα antagonist GW6471 abolished the beneficial effects of Rictor knockdown. Rictor/mTORC2 deficiency reduces serum TG levels, maintains lipid homeostasis, and suppresses inflammation by inhibiting PPARα expression. Weakening mTORC2 activity holds promise as a novel therapeutic strategy for HP.
    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
    肥胖与肥胖相关的胰岛素抵抗一直是研究热点。垂体腺苷酸环化酶激活多肽(PACAP)在能量代谢中起重要作用,具有减轻胰岛素抵抗的潜力。然而,确切的机制还没有完全理解。用棕榈酸和高脂饮食(HFD)建立α小鼠肝12细胞系和C57BL/6小鼠胰岛素抵抗模型,分别。随后,我们评估了PACAP在体内和体外的作用。使用慢病毒载体来探索PACAP可以改善胰岛素抵抗的信号通路。发现PACAP选择性结合PACAPI型受体受体并改善胰岛素抵抗,其特征是在胰岛素抵抗细胞模型和HFD喂养的小鼠中糖原合成增加和糖异生抑制。这些作用与哺乳动物雷帕霉素靶/RAC-α丝氨酸/苏氨酸蛋白激酶(FAIM/Rictor/AKT)轴的Fas凋亡抑制分子/雷帕霉素不敏感伴侣的激活有关。此外,PACAP通过增加溶质载体家族2,促进葡萄糖转运蛋白成员2/4和抑制糖异生相关蛋白葡萄糖6-磷酸酶催化亚基1和磷酸烯醇丙酮酸羧激酶2表达来改善胰岛素抵抗。同时,PACAP在体内和体外均可促进肝AKT/糖原合酶激酶3β的磷酸化。此外,PACAP治疗降低体重,肥胖小鼠的食物摄入量和血糖水平。我们的研究表明,PACAP通过FAIM/Rictor/AKT轴改善胰岛素抵抗,将其作为治疗肥胖相关胰岛素抵抗的有希望的候选药物。
    Obesity and obesity-related insulin resistance have been a research hotspot. Pituitary adenylate cyclase activating polypeptide (PACAP) has emerged as playing a significant role in energy metabolism, holding promising potential for attenuating insulin resistance. However, the precise mechanism is not fully understood. Palmitic acid and a high-fat diet (HFD) were used to establish insulin resistance model in Alpha mouse liver 12 cell line and C57BL/6 mice, respectively. Subsequently, we assessed the effects of PACAP both in vivo and in vitro. Lentivirus vectors were used to explore the signaling pathway through which PACAP may ameliorate insulin resistance. PACAP was found to selectively bind to the PACAP type I receptor receptor and ameliorate insulin resistance, which was characterized by increased glycogen synthesis and the suppression of gluconeogenesis in the insulin-resistant cell model and HFD-fed mice. These effects were linked to the activation of the Fas apoptotic inhibitory molecule/rapamycin-insensitive companion of mammalian target of rapamycin/RAC-alpha serine/threonine-protein kinase (FAIM/Rictor/AKT) axis. Furthermore, PACAP ameliorated insulin resistance by increasing solute carrier family 2, facilitated glucose transporter members 2/4 and inhibiting gluconeogenesis-related proteins glucose 6-phosphatase catalytic subunit 1 and phosphoenolpyruvate carboxykinase 2 expression. Meanwhile, the phosphorylation of hepatic AKT/glycogen synthase kinase 3β was promoted both in vivo and in vitro by PACAP. Additionally, PACAP treatment decreased body weight, food intake and blood glucose levels in obese mice. Our study shows that PACAP ameliorated insulin resistance through the FAIM/Rictor/AKT axis, presenting it as a promising drug candidate for the treatment of obesity-related insulin 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
    为了解决日益增长的能源需求,肿瘤细胞经历代谢重编程,包括氧化磷酸化(OXPHOS)和有氧糖酵解。本研究调查了Kruppel样因子4(KLF4)的作用,转录因子,通过调节ATP合成在肝细胞癌(HCC)中作为肿瘤抑制因子。进行免疫组织化学以评估HCC组织中的KLF4表达。功能测定,如CCK-8,EdU,和殖民地的形成,以及体内测定,包括皮下肿瘤形成和肝脏原位异种移植小鼠模型,进行以确定KLF4对HCC增殖的影响。荧光素酶报告基因测定和染色质免疫沉淀测定用于评估KLF4、miR-206和RICTOR之间的相互作用。研究结果表明,肝癌中KLF4表达较低,这与不良预后有关。体外和体内功能测定均表明KLF4抑制HCC细胞增殖。机械上,研究表明,KLF4通过抑制RICTOR的表达来减少HCC中的ATP合成,mTORC2的核心组件。这种抑制促进谷氨酰胺分解以补充TCA循环并增加ATP水平,促进miR-206转录。总之,这项研究增强了对KLF4在HCCATP合成中作用的理解,并提示靶向KLF4/miR-206/RICTOR轴可能是一种有前景的抗HCC治疗方法.
    To address the increased energy demand, tumor cells undergo metabolic reprogramming, including oxidative phosphorylation (OXPHOS) and aerobic glycolysis. This study investigates the role of Kruppel-like factor 4 (KLF4), a transcription factor, as a tumor suppressor in hepatocellular carcinoma (HCC) by regulating ATP synthesis. Immunohistochemistry was performed to assess KLF4 expression in HCC tissues. Functional assays, such as CCK-8, EdU, and colony formation, as well as in vivo assays, including subcutaneous tumor formation and liver orthotopic xenograft mouse models, were conducted to determine the impact of KLF4 on HCC proliferation. Luciferase reporter assay and chromatin immunoprecipitation assay were utilized to evaluate the interaction between KLF4, miR-206, and RICTOR. The findings reveal low KLF4 expression in HCC, which is associated with poor prognosis. Both in vitro and in vivo functional assays demonstrate that KLF4 inhibits HCC cell proliferation. Mechanistically, it was demonstrated that KLF4 reduces ATP synthesis in HCC by suppressing the expression of RICTOR, a core component of mTORC2. This suppression promotes glutaminolysis to replenish the TCA cycle and increase ATP levels, facilitated by the promotion of miR-206 transcription. In conclusion, this study enhances the understanding of KLF4\'s role in HCC ATP synthesis and suggests that targeting the KLF4/miR-206/RICTOR axis could be a promising therapeutic approach for anti-HCC 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
    背景:肠道炎症和屏障功能受损是胃肠道疾病发病的关键因素。本研究旨在探讨miR-192-5p在肠上皮屏障(IEB)完整性调节中的作用及其与自噬的关系。
    方法:使用DSS诱导的结肠炎模型来评估miR-192-5p对肠道炎症的影响。体外实验涉及细胞培养和瞬时转染技术。各种化验,包括双荧光素酶报告基因测定,实时定量PCR,西方印迹,以及跨上皮电阻的测量,进行评估miR-192-5p表达的变化,Rictor水平,和自噬通量。免疫荧光染色,H&E染色,TEER测量,和FITC-葡聚糖分析也被采用。
    结果:我们的发现揭示了炎症肠组织中miR-192-5p的表达降低,与IEB功能受损相关。miR-192-5p的过表达通过靶向Rictor减轻TNF诱导的IEB功能障碍,导致增强的自噬通量在肠上皮细胞(ECs)。此外,miR-192-5p的治疗潜力在结肠炎小鼠中得到证实,其中增加的miR-192-5p表达通过调节Rictor增强ECs中的自噬通量来改善肠道炎症损伤。
    结论:我们的研究通过证明miR-192-5p在调节自噬和维持IEB功能中的作用,强调了其在肠炎中的治疗潜力。靶向miR-192-5p/Rictor轴是缓解肠炎患者肠道炎症损伤和改善屏障完整性的有希望的方法。
    Intestinal inflammation and compromised barrier function are critical factors in the pathogenesis of gastrointestinal disorders. This study aimed to investigate the role of miR-192-5p in modulating intestinal epithelial barrier (IEB) integrity and its association with autophagy. A DSS-induced colitis model was used to assess the effects of miR-192-5p on intestinal inflammation. In vitro experiments involved cell culture and transient transfection techniques. Various assays, including dual-luciferase reporter gene assays, quantitative real-time PCR, Western blotting, and measurements of transepithelial electrical resistance, were performed to evaluate changes in miR-192-5p expression, Rictor levels, and autophagy flux. Immunofluorescence staining, H&E staining, TEER measurements, and FITC-dextran analysis were also used. Our findings revealed a reduced expression of miR-192-5p in inflamed intestinal tissues, correlating with impaired IEB function. Overexpression of miR-192-5p alleviated TNF-induced IEB dysfunction by targeting Rictor, resulting in enhanced autophagy flux in enterocytes (ECs). Moreover, the therapeutic potential of miR-192-5p was substantiated in colitis mice, wherein increased miR-192-5p expression ameliorated intestinal inflammatory injury by enhancing autophagy flux in ECs through the modulation of Rictor. Our study highlights the therapeutic potential of miR-192-5p in enteritis by demonstrating its role in regulating autophagy and preserving IEB function. Targeting the miR-192-5p/Rictor axis is a promising approach for mitigating gut inflammatory injury and improving barrier integrity in patients with enteritis.NEW & NOTEWORTHY We uncover the pivotal role of miR-192-5p in fortifying intestinal barriers amidst inflammation. Reduced miR-192-5p levels correlated with compromised gut integrity during inflammation. Notably, boosting miR-192-5p reversed gut damage by enhancing autophagy via suppressing Rictor, offering a potential therapeutic strategy for fortifying the intestinal barrier and alleviating inflammation in patients with enteritis.
    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
    背景:基于BRAF/MEK抑制剂(BRAF/MEKi)的靶向治疗在BRAF突变的皮肤转移性黑色素瘤(MM)的治疗中的主要缺点是治疗耐药性的发展。我们旨在评估mTORC2的作用,mTORC2是由必需RICTOR亚基的存在定义的信号复合物,被认为是几种肿瘤类型的致癌驱动因素,包括MM。
    方法:在分析癌症基因组图谱MM患者的数据库后,探索作为肿瘤内RICTOR水平的函数的总生存期和分子特征,我们研究了RICTOR在BRAFV600EMM细胞系中下调对其对BRAF/MEKi反应的影响。我们进行了蛋白质组学筛选,以鉴定由RICTOR表达变化调节的蛋白质,和海马分析评估RICTOR耗竭对线粒体呼吸的影响。BRAFi与蛋白质组学筛选中出现的靶向蛋白质和过程的药物的组合在体外和体内异种移植设置中对RICTOR缺陷细胞进行。
    结果:在BRAF突变的MM中,低RICTOR水平与较差的临床结果相关。低RICTOR肿瘤的基因集富集分析显示提示线粒体电子运输链(ETC)能量产生的激活的基因特征。RICTOR缺陷型BRAFV600E细胞对BRAF/MEKi具有内在的耐受性,并在长期药物暴露后预测对BRAFi的耐药性。此外,在药物幼稚细胞中,我们观察到RICTOR表达在BRAFi暴露后不久下降。在RICTOR耗尽的细胞中,线粒体呼吸和烟酰胺磷酸核糖基转移酶(NAMPT)的表达均增强,和它们的药理学抑制恢复对BRAFi的敏感性。
    结论:我们的工作揭示了mTORC2在BRAFV600E黑色素瘤细胞对靶向治疗的早期适应阶段的不可预见的肿瘤抑制作用,并将NAMPT-ETC轴确定为低RICTOR肿瘤的潜在治疗脆弱性。重要的是,我们的研究结果表明,肿瘤内RICTOR水平的评估对转移性黑色素瘤具有预后价值,可能有助于以个性化方式指导治疗策略.
    BACKGROUND: The main drawback of BRAF/MEK inhibitors (BRAF/MEKi)-based targeted therapy in the management of BRAF-mutated cutaneous metastatic melanoma (MM) is the development of therapeutic resistance. We aimed to assess in this context the role of mTORC2, a signaling complex defined by the presence of the essential RICTOR subunit, regarded as an oncogenic driver in several tumor types, including MM.
    METHODS: After analyzing The Cancer Genome Atlas MM patients\' database to explore both overall survival and molecular signatures as a function of intra-tumor RICTOR levels, we investigated the effects of RICTOR downregulation in BRAFV600E MM cell lines on their response to BRAF/MEKi. We performed proteomic screening to identify proteins modulated by changes in RICTOR expression, and Seahorse analysis to evaluate the effects of RICTOR depletion on mitochondrial respiration. The combination of BRAFi with drugs targeting proteins and processes emerged in the proteomic screening was carried out on RICTOR-deficient cells in vitro and in a xenograft setting in vivo.
    RESULTS: Low RICTOR levels in BRAF-mutated MM correlate with a worse clinical outcome. Gene Set Enrichment Analysis of low-RICTOR tumors display gene signatures suggestive of activation of the mitochondrial Electron Transport Chain (ETC) energy production. RICTOR-deficient BRAFV600E cells are intrinsically tolerant to BRAF/MEKi and anticipate the onset of resistance to BRAFi upon prolonged drug exposure. Moreover, in drug-naïve cells we observed a decline in RICTOR expression shortly after BRAFi exposure. In RICTOR-depleted cells, both mitochondrial respiration and expression of nicotinamide phosphoribosyltransferase (NAMPT) are enhanced, and their pharmacological inhibition restores sensitivity to BRAFi.
    CONCLUSIONS: Our work unveils an unforeseen tumor-suppressing role for mTORC2 in the early adaptation phase of BRAFV600E melanoma cells to targeted therapy and identifies the NAMPT-ETC axis as a potential therapeutic vulnerability of low RICTOR tumors. Importantly, our findings indicate that the evaluation of intra-tumor RICTOR levels has a prognostic value in metastatic melanoma and may help to guide therapeutic strategies in a personalized manner.
    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
    盐皮质激素受体(MR)是介导多种基因的转录因子,细胞特异性功能,包括营养作用以及促进液体/电解质稳态。据报道,在插入的细胞中,ULK1对丝氨酸843(S843)处MR的磷酸化抑制MR激活,而mTOR对ULK1的磷酸化使ULK1失活,从而防止MR失活。我们通过对稳定表达大鼠MR和报告基因的M1小鼠皮质收集管细胞的研究扩展了这些发现。ULK1剂量依赖性增加配体诱导的MR反式激活的药理学抑制,而ULK1激活没有影响。Raptor或Rictor的mTOR和CRISPR/gRNA基因敲低的药理学抑制降低了磷酸化的ULK1和配体诱导的MR报告基因的激活,以及内源性MR靶基因的转录。正如预测的那样,ULK1抑制对具有突变MR-S843A(丙氨酸不能磷酸化)的M1细胞中醛固酮介导的转录没有影响。相比之下,mTOR抑制剂量依赖性地降低MR-S843A细胞中的转录,尽管不如使用wtMR-S843的细胞完全。mTOR,猛禽,Rictor与MR共沉淀,醛固酮的加入增加了它们的磷酸化,活跃,状态。这些结果表明,mTOR至少以两种方式显着调节MR活性:通过抑制ULK1引起的MR失活,以及通过与MR直接相关的尚不明确的机制。他们还为ULK1和mTOR的不同功能提供了新的见解,监测细胞能量状态的两种关键酶。
    The mineralocorticoid receptor (MR) is a transcription factor for genes mediating diverse, cell-specific functions, including trophic effects as well as promoting fluid/electrolyte homeostasis. It was reported that in intercalated cells, phosphorylation of the MR at serine 843 (S843) by Unc-51-like kinase (ULK1) inhibits MR activation and that phosphorylation of ULK1 by mechanistic target of rapamycin (mTOR) inactivates ULK1, and thereby prevents MR inactivation. We extended these findings with studies in M1 mouse cortical collecting duct cells stably expressing the rat MR and a reporter gene. Pharmacological inhibition of ULK1 dose-dependently increased ligand-induced MR transactivation, while ULK1 activation had no effect. Pharmacological inhibition of mTOR and CRISPR/gRNA gene knockdown of rapamycin-sensitive adapter protein of mTOR (Raptor) or rapamycin-insensitive companion of mTOR (Rictor) decreased phosphorylated ULK1 and ligand-induced activation of the MR reporter gene, as well as transcription of endogenous MR-target genes. As predicted, ULK1 inhibition had no effect on aldosterone-mediated transcription in M1 cells with the mutated MR-S843A (alanine cannot be phosphorylated). In contrast, mTOR inhibition dose-dependently decreased transcription in the MR-S843A cells, though not as completely as in cells with the wild-type MR-S843. mTOR, Raptor, and Rictor coprecipitated with the MR and addition of aldosterone increased their phosphorylated, active state. These results suggest that mTOR significantly regulates MR activity in at least 2 ways: by suppressing MR inactivation by ULK1, and by a yet ill-defined mechanism that involves direct association with MR. They also provide new insights into the diverse functions of ULK1 and mTOR, 2 key enzymes that monitor the cell\'s energy status.
    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
    心肌细胞凋亡和心肌纤维化是缺血性心脏病患者死亡的主要原因。因此,这些过程代表了治疗缺血性损伤所致心力衰竭的潜在治疗靶点.我们先前证明了线粒体乙酰转移酶蛋白GCN5L1在体内缺血-再灌注损伤和体外缺氧-复氧损伤中调节心肌细胞细胞保护信号。目前的研究调查了GCN5L1介导的Akt/mTORC2心脏保护性信号通路调节的潜在机制。人类缺血性心脏病患者心脏组织中的Rictor蛋白水平相对于非缺血性对照组显着降低。缺氧应激后,心脏AC16细胞中的Rictor蛋白水平类似地降低,而mRNA水平保持不变。缺氧后Rictor蛋白水平的降低通过GCN5L1的敲低而增强,并被GCN5L1过表达所阻断。这些发现与Rictor赖氨酸乙酰化的变化相关,由GCN5L1乙酰转移酶活性介导。Rictor降解受蛋白酶体活性调节,这是由增加的Rictor乙酰化拮抗。最后,我们发现GCN5L1敲低限制了细胞保护Akt信号,与mTOR丰度和活性降低有关。总之,这些研究表明,GCN5L1通过增强赖氨酸乙酰化维持Rictor蛋白水平,从而促进心脏保护性Akt/mTORC2信号传导.
    Cardiomyocyte apoptosis and cardiac fibrosis are the leading causes of mortality in patients with ischemic heart disease. As such, these processes represent potential therapeutic targets to treat heart failure resulting from ischemic insult. We previously demonstrated that the mitochondrial acetyltransferase protein GCN5L1 regulates cardiomyocyte cytoprotective signaling in ischemia-reperfusion injury in vivo and hypoxia-reoxygenation injury in vitro. The current study investigated the mechanism underlying GCN5L1-mediated regulation of the Akt/mTORC2 cardioprotective signaling pathway. Rictor protein levels in cardiac tissues from human ischemic heart disease patients were significantly decreased relative to non-ischemic controls. Rictor protein levels were similarly decreased in cardiac AC16 cells following hypoxic stress, while mRNA levels remained unchanged. The reduction in Rictor protein levels after hypoxia was enhanced by the knockdown of GCN5L1, and was blocked by GCN5L1 overexpression. These findings correlated with changes in Rictor lysine acetylation, which were mediated by GCN5L1 acetyltransferase activity. Rictor degradation was regulated by proteasomal activity, which was antagonized by increased Rictor acetylation. Finally, we found that GCN5L1 knockdown restricted cytoprotective Akt signaling, in conjunction with decreased mTOR abundance and activity. In summary, these studies suggest that GCN5L1 promotes cardioprotective Akt/mTORC2 signaling by maintaining Rictor protein levels through enhanced lysine acetylation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Review
    雷帕霉素的机制靶点,mTOR,控制细胞代谢以响应生长信号和应激刺激。mTOR的细胞功能由两种不同的蛋白质复合物介导,mTOR复合物1(mTORC1)和mTORC2。雷帕霉素及其类似物目前在临床上用于治疗多种疾病,并有助于描述其直接靶标的功能。mTORC1.尽管缺乏特异性的mTORC2抑制剂,破坏mTORC2表达的遗传研究揭示了这种更难以捉摸的mTOR复合物的功能。就像mTORC1响应生长信号一样,mTORC2也被合成代谢信号激活,但也被压力触发。mTORC2介导来自生长因子受体和G蛋白偶联受体的信号。营养限制等应激条件如何调节mTORC2激活以允许代谢重编程并确保细胞存活仍然知之甚少。已经确定了多种mTORC2的下游效应物,但最充分表征的mTORC2底物包括Akt,PKC,SGK,它们是AGC蛋白激酶家族的成员。这里,我们综述了mTORC2是如何被细胞刺激调节的,包括复杂成分的分隔和调节如何影响mTORC2信号传导.我们详细阐述了它的底物是如何磷酸化的,特别是AGC激酶,调解其在增长中的多种功能,扩散,生存,和差异化。我们讨论了与mTORC2串扰的其他信号和代谢成分以及这些信号的细胞输出。最后,我们考虑如何更有效地靶向mTORC2通路来治疗mTOR信号下调的疾病.
    The mechanistic target of rapamycin, mTOR, controls cell metabolism in response to growth signals and stress stimuli. The cellular functions of mTOR are mediated by two distinct protein complexes, mTOR complex 1 (mTORC1) and mTORC2. Rapamycin and its analogs are currently used in the clinic to treat a variety of diseases and have been instrumental in delineating the functions of its direct target, mTORC1. Despite the lack of a specific mTORC2 inhibitor, genetic studies that disrupt mTORC2 expression unravel the functions of this more elusive mTOR complex. Like mTORC1 which responds to growth signals, mTORC2 is also activated by anabolic signals but is additionally triggered by stress. mTORC2 mediates signals from growth factor receptors and G-protein coupled receptors. How stress conditions such as nutrient limitation modulate mTORC2 activation to allow metabolic reprogramming and ensure cell survival remains poorly understood. A variety of downstream effectors of mTORC2 have been identified but the most well-characterized mTORC2 substrates include Akt, PKC, and SGK, which are members of the AGC protein kinase family. Here, we review how mTORC2 is regulated by cellular stimuli including how compartmentalization and modulation of complex components affect mTORC2 signaling. We elaborate on how phosphorylation of its substrates, particularly the AGC kinases, mediates its diverse functions in growth, proliferation, survival, and differentiation. We discuss other signaling and metabolic components that cross-talk with mTORC2 and the cellular output of these signals. Lastly, we consider how to more effectively target the mTORC2 pathway to treat diseases that have deregulated mTOR signaling.
    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
    胃肠道肿瘤占所有癌症死亡的前10个原因中的5个(结直肠癌,肝脏,胃,食管癌和胰腺癌)。哺乳动物雷帕霉素靶标(mTOR)信号传导在各种人类癌症中通常失调。作为mTOR复合物2(mTORC2)的核心成分,Rictor是PI3K/Akt途径的关键效应分子。在胃肠道肿瘤中观察到Rictor的高改变率,此类Rictor改变通常与化疗耐药和相关不良临床结局相关.然而,Rictor在胃肠道肿瘤中的确切作用仍然难以捉摸。本研究的目的是批判性地讨论以下内容:i)肿瘤中Rictor的突变和生物学特征,并详细概述了Rictor在细胞增殖中的作用,血管生成,凋亡,自噬和耐药性;ii)Rictor在消化系统肿瘤中的作用,尤其是结直肠,肝胆,胃,食管癌和胰腺癌和胆管癌;以及iii)通过抑制Akt激活靶向治疗Rictor的现状和前景。尽管人们越来越意识到Rictor/mTORC2在癌症中的重要性,潜在的机制细节仍然知之甚少;这需要改变,以便开发有效的靶向疗法和治疗抗性癌症的再敏化成为可能.
    Gastrointestinal tumors account for five of the top 10 causes of mortality from all cancers (colorectal, liver, stomach, esophageal and pancreatic cancer). Mammalian target of rapamycin (mTOR) signaling is commonly dysregulated in various human cancers. As a core component of the mTOR complex 2 (mTORC2), Rictor is a key effector molecule of the PI3K/Akt pathway. A high alteration rate of Rictor has been observed in gastrointestinal tumors, and such Rictor alterations are often associated with resistance to chemotherapy and related adverse clinical outcomes. However, the exact roles of Rictor in gastrointestinal tumors remain elusive. The aim of the present study was to critically discuss the following: i) Mutation and biological characteristics of Rictor in tumors with a detailed overview of Rictor in cell proliferation, angiogenesis, apoptosis, autophagy and drug resistance; ii) the role of Rictor in tumors of the digestive system, particularly colorectal, hepatobiliary, gastric, esophageal and pancreatic cancer and cholangiocarcinoma; and iii) the current status and prospects of targeted therapy for Rictor by inhibiting Akt activation. Despite the growing realization of the importance of Rictor/mTORC2 in cancer, the underlying mechanistic details remain poorly understood; this needs to change in order for the development of efficient targeted therapies and re‑sensitization of therapy‑resistant cancers to be made possible.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号