p66Shc

p66Shc
  • 文章类型: Journal Article
    尽管表达富含脯氨酸的p66shc,但仍需要适当研究癌细胞对ROS介导的凋亡的冗余。P66shc,一种衔接蛋白,对于启动ROS介导的凋亡和随后通过Rac-1激活的ROS产生都是必不可少的。P66shc在Ser-36处被磷酸化,从而触发其易位到线粒体,并随后响应于氧化应激而释放细胞色素c。它还有助于Rac-1依赖性NADPH氧化酶激活,导致胞质ROS的产生,可以根据其浓度执行不同的功能。这项研究已经确定了多方面的抗凋亡蛋白BAG3作为p66shc的相互作用伴侣。BAG3利用其WW结构域结合p66shc的富含脯氨酸的基序。BAG3通过其WW域,拮抗p66shc介导的细胞凋亡,通过在正常和氧化应激条件下抑制p66shc的表达和磷酸化。这导致针对ROS介导的细胞凋亡的显著保护。BAG3介导的p66shc表达减少增加了细胞增殖和转移。细胞增殖的增加归因于在正常条件下BAG3对Rac-1活化和ROS产生的影响。这项研究揭示了p66shc的相互作用剂,该相互作用剂增强了促生存作用,同时抑制了其凋亡作用。
    Redundancy of cancer cells towards ROS-mediated apoptosis despite expressing proline-rich p66shc abundantly needs to be investigated properly. P66shc, an adapter protein, is indispensable both for initiating ROS-mediated apoptosis and subsequent ROS generation through Rac-1 activation. P66shc gets phosphorylated at Ser-36 that triggers its translocation to the mitochondria and subsequent release of Cytochrome c in response to oxidative stress. It also aids in Rac-1 dependent NADPH oxidase activation, leading to the generation of cytosolic ROS that can perform diverse functions depending on its concentration. This study has identified the multi-faceted anti-apoptotic protein BAG3 as an interacting partner of p66shc. BAG3 utilizes its WW domain to bind to the proline-rich motifs of p66shc. BAG3, through its WW domain, antagonizes p66shc mediated apoptosis, by inhibiting both the expression and phosphorylation of p66shc under normal and oxidative stress conditions. This results in significant protection against ROS-mediated apoptosis. BAG3-mediated reduction in p66shc expression increases cell proliferation and metastasis. The increase in cell proliferation is attributed to the impact of BAG3 on Rac-1 activation and ROS production under normal conditions. This study has unraveled an interactor of p66shc that enhances pro-survival role while simultaneously suppressing its apoptotic role.
    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
    许多研究强调了线粒体相关基因(MRGs)在胶质母细胞瘤(GBM)的发生和发展中的关键作用。然而,MRGs编码蛋白对GBM病理的具体贡献仍未完全阐明.GBM中预后性MRGs的鉴定为开发个性化靶向治疗和增强患者预后提供了希望。我们将差异表达与单变量Cox回归分析相结合,以筛选GBM中与预后相关的MRGs。根据九个MRG,风险比模型采用多变量Cox回归算法.SHC相关生存,通路,GBM队列中的免疫分析来自实体瘤数据库的生物标志物探索。通过CCK-8和transwell测定法测量U87细胞的增殖和迁移。使用流式细胞术评估U87细胞中的凋亡。使用共聚焦显微镜来测量线粒体活性氧(ROS)水平和形态。通过蛋白质印迹检查SHC1和其他相关蛋白的表达水平。我们筛选了15个与预后相关的MRGs,并构建了一个基于9个MRGs的模型。模型风险评分的验证证实了其在预测GBM患者预后方面的有效性。此外,分析表明,SHC1是预后模型的一个组成MRG,被上调并参与了进展,迁移,和GBM的免疫浸润。体外实验阐明了p66Shc,SHC1的最长同工型,调节线粒体ROS的产生和形态,从而促进U87细胞的增殖和迁移。基于9个MRGs的预后模型可以预测GBM的预后。通过参与免疫浸润,SHC1上调并与患者预后相关。此外,体外实验表明p66Shc通过介导线粒体ROS的产生促进U87细胞增殖和迁移。因此,p66Shc可以作为GBM的有希望的生物标志物和治疗靶标。
    Numerous studies have highlighted the pivotal role of mitochondria-related genes (MRGs) in the initiation and progression of glioblastoma (GBM). However, the specific contributions of MRGs coding proteins to GBM pathology remain incompletely elucidated. The identification of prognostic MRGs in GBM holds promise for the development of personalized targeted therapies and the enhancement of patient prognosis. We combined differential expression with univariate Cox regression analysis to screen prognosis-associated MRGs in GBM. Based on the nine MRGs, the hazard ratio model was conducted using a multivariate Cox regression algorithm. SHC-related survival, pathway, and immune analyses in GBM cohorts were obtained from the Biomarker Exploration of the Solid Tumor database. The proliferation and migration of U87 cells were measured by CCK-8 and transwell assay. Apoptosis in U87 cells was evaluated using flow cytometry. Confocal microscopy was employed to measure mitochondrial reactive oxygen species (ROS) levels and morphology. The expression levels of SHC1 and other relevant proteins were examined via western blotting. We screened 15 prognosis-associated MRGs and constructed a 9 MRGs-based model. Validation of the model\'s risk score confirmed its efficacy in predicting the prognosis of patients with GBM. Furthermore, analysis revealed that SHC1, a constituent MRG of the prognostic model, was upregulated and implicated in the progression, migration, and immune infiltration of GBM. In vitro experiments elucidated that p66Shc, the longest isoform of SHC1, modulates mitochondrial ROS production and morphology, consequently promoting the proliferation and migration of U87 cells. The 9 MRGs-based prognostic model could predict the prognosis of GBM. SHC1 was upregulated and correlated with the prognosis of patients by involvement in immune infiltration. Furthermore, in vitro experiments demonstrated that p66Shc promotes U87 cell proliferation and migration by mediating mitochondrial ROS production. Thus, p66Shc may serve as a promising biomarker and therapeutic target for GBM.
    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:p66Shc,作为氧化还原酶,调节线粒体中活性氧(ROS)的产生和自噬。然而,p66Shc影响自噬体形成的机制尚不完全清楚。
    方法:在体内和体外检测p66Shc在滋养细胞中的表达及其定位。小发夹RNA或CRISPR/Cas9,RNA测序,使用共聚焦激光扫描显微镜来阐明p66Shc在调节自噬通量和STING激活中的作用。此外,透射电镜(TEM)观察p66Shc对线粒体相关内质网膜(MAMs)形成的影响。通过检测细胞可塑性线粒体DNA(mtDNA)和线粒体膜电位(MMP)评估线粒体功能。
    结果:高糖诱导p66Shc的表达和线粒体易位,促进MAMs形成并刺激PINK1-PRKN介导的线粒体自噬。此外,线粒体局部p66Shc减少MMP并触发胞浆mtDNA释放,从而激活cGAS/STING信号并最终导致自噬增强和细胞衰老。特别是,我们发现STING和LC3II之间的相互作用需要p66Shc,以及STING和ATG5之间,从而调节cGAS/STING介导的自噬。我们还鉴定了数百种与包括衰老在内的多种生物过程相关的基因被p66Shc和ATG5共同调节,其中任何一种缺失都会导致细胞衰老减少。
    结论:p66Shc不仅通过促进MAMs形成参与自噬的启动,而且还通过激活滋养细胞中的cGAS/STING途径来帮助稳定活性自噬通量。
    BACKGROUND: p66Shc, as a redox enzyme, regulates reactive oxygen species (ROS) production in mitochondria and autophagy. However, the mechanisms by which p66Shc affects autophagosome formation are not fully understood.
    METHODS: p66Shc expression and its location in the trophoblast cells were detected in vivo and in vitro. Small hairpin RNAs or CRISPR/Cas9, RNA sequencing, and confocal laser scanning microscope were used to clarify p66Shc\'s role in regulating autophagic flux and STING activation. In addition, p66Shc affects mitochondrial-associated endoplasmic reticulum membranes (MAMs) formation were observed by transmission electron microscopy (TEM). Mitochondrial function was evaluated by detected cytoplastic mitochondrial DNA (mtDNA) and mitochondrial membrane potential (MMP).
    RESULTS: High glucose induces the expression and mitochondrial translocation of p66Shc, which promotes MAMs formation and stimulates PINK1-PRKN-mediated mitophagy. Moreover, mitochondrial localized p66Shc reduces MMP and triggers cytosolic mtDNA release, thus activates cGAS/STING signaling and ultimately leads to enhanced autophagy and cellular senescence. Specially, we found p66Shc is required for the interaction between STING and LC3II, as well as between STING and ATG5, thereby regulates cGAS/STING-mediated autophagy. We also identified hundreds of genes associated several biological processes including aging are co-regulated by p66Shc and ATG5, deletion either of which results in diminished cellular senescence.
    CONCLUSIONS: p66Shc is not only implicated in the initiation of autophagy by promoting MAMs formation, but also helps stabilizing active autophagic flux by activating cGAS/STING pathway in trophoblast.
    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
    这篇综述研究了p66Shc衔接蛋白和肠道菌群在调节线粒体功能和氧化应激中的多方面作用。以及它们对慢性病发病机制的集体影响。该研究深入研究了p66Shc通过Rac1激活影响细胞应激反应的分子机制,叉头型转录因子失活,线粒体介导的细胞凋亡,肠道菌群衍生的代谢产物和内毒素的调节作用。采用综合方法,这篇综述综合了一系列广泛研究的结果,包括分子生物学技术和微生物代谢物对宿主细胞途径影响的分析。结果强调了微生物代谢物之间复杂的相互作用,p66Shc激活,和线粒体功能障碍,强调肠道微生物组在通过氧化应激途径影响疾病结局方面的重要性。最后,该综述认为,靶向肠道微生物群-p66Shc-线粒体轴可以为减缓代谢性疾病的发展和进展提供新的治疗策略.这强调了饮食干预和微生物群调节在管理氧化应激和炎症方面的潜力,慢性病病因的关键因素。
    This review investigates the multifaceted role of the p66Shc adaptor protein and the gut microbiota in regulating mitochondrial function and oxidative stress, and their collective impact on the pathogenesis of chronic diseases. The study delves into the molecular mechanisms by which p66Shc influences cellular stress responses through Rac1 activation, Forkhead-type transcription factors inactivation, and mitochondria-mediated apoptosis, alongside modulatory effects of gut microbiota-derived metabolites and endotoxins. Employing an integrative approach, the review synthesizes findings from a broad array of studies, including molecular biology techniques and analyses of microbial metabolites\' impacts on host cellular pathways. The results underscore a complex interplay between microbial metabolites, p66Shc activation, and mitochondrial dysfunction, highlighting the significance of the gut microbiome in influencing disease outcomes through oxidative stress pathways. Conclusively, the review posits that targeting the gut microbiota-p66Shc-mitochondrial axis could offer novel therapeutic strategies for mitigating the development and progression of metabolic diseases. This underscores the potential of dietary interventions and microbiota modulation in managing oxidative stress and inflammation, pivotal factors in chronic disease etiology.
    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
    慢性淋巴细胞白血病(CLL)患者的促凋亡蛋白p66Shc及其转录因子STAT4表达缺陷,引起分子异常,细胞凋亡受损,疾病预后和严重程度恶化。p66Shc表达受STAT4的表观遗传控制和转录调节;表观遗传修饰剂在CLL细胞中失调,并且特异性组蛋白脱乙酰酶(HDACs)如HDAC1被过表达。STAT4/p66Shc表达的再激活可能代表了逆转CLL凋亡缺陷的有吸引力和挑战性的策略。新的选择性I类HDAC抑制剂(HDACis,6a-g)比现有药物具有更高的效力,并优先干扰CLL相关的同工型HDAC1,以揭示I类HDACs在STAT4表达上调中的作用,上调p66Shc表达,从而使CLL细胞凋亡正常化。6c(氯吡诺坦)被鉴定为有效的HDAC1i,其特征优于恩替诺坦。与SAHA相比,6c诱导CLL细胞显著凋亡,这与STAT4/p66Shc蛋白表达上调有关。HDAC1而不是HDAC3在STAT4/p66Shc的表观遗传上调中的作用首次在CLL细胞中得到证实,并在siRNA诱导的HDAC1/HDAC3敲低EBV-B细胞中得到验证。总而言之,HDAC1抑制是CLL患者重新激活STAT4/p66Shc表达所必需的。6c是迄今为止已知的最有效的HDAC1之一,代表了一种新的逆转STAT4/p66Shc凋亡机制损伤的药理学工具。
    Chronic Lymphocytic Leukemia (CLL) patients have a defective expression of the proapoptotic protein p66Shc and of its transcriptional factor STAT4, which evoke molecular abnormalities, impairing apoptosis and worsening disease prognosis and severity. p66Shc expression is epigenetically controlled and transcriptionally modulated by STAT4; epigenetic modifiers are deregulated in CLL cells and specific histone deacetylases (HDACs) like HDAC1, are overexpressed. Reactivation of STAT4/p66Shc expression may represent an attractive and challenging strategy to reverse CLL apoptosis defects. New selective class I HDAC inhibitors (HDACis, 6a-g) were developed with increased potency over existing agents and preferentially interfering with the CLL-relevant isoform HDAC1, to unveil the role of class I HDACs in the upregulation of STAT4 expression, which upregulates p66Shc expression and hence normalizes CLL cell apoptosis. 6c (chlopynostat) was identified as a potent HDAC1i with a superior profile over entinostat. 6c induces marked apoptosis of CLL cells compared with SAHA, which was associated with an upregulation of STAT4/p66Shc protein expression. The role of HDAC1, but not HDAC3, in the epigenetic upregulation of STAT4/p66Shc was demonstrated for the first time in CLL cells and was validated in siRNA-induced HDAC1/HDAC3 knock-down EBV-B cells. To sum up, HDAC1 inhibition is necessary to reactivate STAT4/p66Shc expression in patients with CLL. 6c is one of the most potent HDAC1is known to date and represents a novel pharmacological tool for reversing the impairment of the STAT4/p66Shc apoptotic machinery.
    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
    简介:逃避免疫监视是慢性淋巴细胞白血病(CLL)细胞的标志。在淋巴器官的保护性小生境中,白血病细胞抑制T淋巴细胞形成免疫突触(IS)的能力,从而阻碍T细胞介导的抗肿瘤活性。通过在T淋巴细胞表面结合其同源受体PD-1,抑制性配体PD-L1在CLL细胞中过度表达,介导CLL细胞的T细胞抑制活性。然而,CLL细胞中PD-L1过表达的分子机制尚不清楚.我们先前报道了CLL细胞中促凋亡和促氧化接头p66Shc的缺陷表达,这与细胞内活性氧(ROS)产生的损害和对ROS敏感的转录因子NF-κB的激活有因果关系。PD-L1表达受NF-κB调节的事实表明CLL细胞中p66Shc缺乏与PD-L1过表达之间的机制关系。方法:本研究包括62例未经治疗的CLL患者和43例健康供体。通过流式细胞术和qRT-PCR定量B细胞中的PD-L1和p66Shc表达。通过流式细胞术和共聚焦显微镜评估IS结构和局部信号传导。通过流式细胞术评估CD8+细胞杀伤活性。结果:在这里,我们显示从CLL患者和CLL小鼠模型Eμ-TCL1分离的白血病细胞中残留的p66Shc表达与PD-L1表达呈负相关。我们还表明,PD-L1的增加阻止了白血病细胞与T淋巴细胞形成IS。p66Shc的重组,但不是ROS缺陷突变体,在CLL细胞和CLL衍生的细胞系MEC-1中,细胞内ROS增强,PD-L1表达降低。用H2O2作为ROS的外源来源处理CLL细胞后获得了类似的结果,使PD-L1表达正常化并恢复IS形成。讨论:我们的数据提供了直接证据,表明CLL细胞中p66Shc缺乏相关的ROS耗竭可以增强PD-L1的表达,并为免疫抑制性淋巴环境中T细胞介导的抗肿瘤功能的抑制提供了机制基础。
    Introduction: Escape from immunosurveillance is a hallmark of chronic lymphocytic leukemia (CLL) cells. In the protective niche of lymphoid organs, leukemic cells suppress the ability of T lymphocytes to form the immune synapse (IS), thereby hampering T-cell mediated anti-tumoral activities. By binding its cognate receptor PD-1 at the surface of T lymphocytes, the inhibitory ligand PD-L1, which is overexpressed in CLL cells, mediates the T-cell suppressive activities of CLL cells. However, the molecular mechanism underlying PD-L1 overexpression in CLL cells remains unknown. We have previously reported a defective expression of the pro-apoptotic and pro-oxidant adaptor p66Shc in CLL cells, which is causally related to an impairment in intracellular reactive oxygen species (ROS) production and to the activation of the ROS-sensitive transcription factor NF-κB. The fact that PD-L1 expression is regulated by NF-κB suggests a mechanistic relationship between p66Shc deficiency and PD-L1 overexpression in CLL cells. Methods: 62 treatment-naive CLL patients and 43 healthy donors were included in this study. PD-L1 and p66Shc expression was quantified in B cells by flow cytometry and qRT-PCR. IS architecture and local signaling was assessed by flow cytometry and confocal microscopy. CD8+ cell killing activity was assessed by flow cytometry. Results: Here we show that residual p66Shc expression in leukemic cells isolated both from CLL patients and from the CLL mouse model Eμ-TCL1 inversely correlated with PD-L1 expression. We also show that the PD-L1 increase prevented leukemic cells from forming ISs with T lymphocytes. Reconstitution of p66Shc, but not of a ROS-defective mutant, in both CLL cells and the CLL-derived cell line MEC-1, enhanced intracellular ROS and decreased PD-L1 expression. Similar results were obtained following treatment of CLL cells with H2O2 as exogenous source of ROS, that normalized PD-L1 expression and recovered IS formation. Discussion: Our data provide direct evidence that the p66Shc-deficiency-related ROS depletion in CLL cells concurs to enhance PD-L1 expression and provides a mechanistic basis for the suppression of T cell-mediated anti-tumoral functions in the immunosuppressive lymphoid niche.
    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
    P66Shc和Rac1蛋白负责肿瘤相关的炎症,特别是在以氧化应激升高和活性氧(ROS)产生增加为特征的脑肿瘤中。槲皮素,一种天然的多酚类黄酮,是众所周知的具有抗癌性质的氧化还原调节剂。它有能力穿过血脑屏障,因此,可能是一种对抗脑瘤的药物.在这项研究中,我们探索槲皮素对Rac1/p66Shc介导的肿瘤细胞炎症的影响,这是在脑细胞中产生ROS的主要途径。用Rac1,p66Shc转染的胶质瘤细胞,或两者都用不同浓度的槲皮素处理不同的时间点。槲皮素以ROS依赖性方式显着降低细胞的活力和迁移,同时抑制了初始和Rac1/p66Shc转染的细胞系中的Rac1/p66Shc表达和ROS产生,提示阻止Rac1激活。通过分子对接模拟,我们观察到,槲皮素与其他已知的Rac1抑制剂相比显示出最佳的结合,并特异性阻断Rac1的A环中的GTP结合位点,以防止GTP结合,因此,Rac1激活。我们得出结论,槲皮素通过特异性抑制Rac1激活,通过调节Rac1-p66Shc信号发挥其抗癌作用,从而克制ROS的产生和肿瘤的成长。
    P66Shc and Rac1 proteins are responsible for tumor-associated inflammation, particularly in brain tumors characterized by elevated oxidative stress and increased reactive oxygen species (ROS) production. Quercetin, a natural polyphenolic flavonoid, is a well-known redox modulator with anticancer properties. It has the capacity to cross the blood-brain barrier and, thus, could be a possible drug against brain tumors. In this study, we explored the effect of quercetin on Rac1/p66Shc-mediated tumor cell inflammation, which is the principal pathway for the generation of ROS in brain cells. Glioma cells transfected with Rac1, p66Shc, or both were treated with varying concentrations of quercetin for different time points. Quercetin significantly reduced the viability and migration of cells in an ROS-dependent manner with the concomitant inhibition of Rac1/p66Shc expression and ROS production in naïve and Rac1/p66Shc-transfected cell lines, suggestive of preventing Rac1 activation. Through molecular docking simulations, we observed that quercetin showed the best binding compared to other known Rac1 inhibitors and specifically blocked the GTP-binding site in the A-loop of Rac1 to prevent GTP binding and, thus, Rac1 activation. We conclude that quercetin exerts its anticancer effects via the modulation of Rac1-p66Shc signaling by specifically inhibiting Rac1 activation, thus restraining the production of ROS and tumor growth.
    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
    糖尿病是一种慢性代谢性疾病,其患病率在全球范围内不断增加。它通常由致残合并症造成负担,这些合并症降低了受影响个体的生活质量和预期。糖尿病的传统并发症通常被描述为大血管并发症(例如,冠心病,外周动脉疾病,和中风),和微血管并发症(例如,糖尿病肾病,视网膜病变,和神经病)。最近,由于糖尿病管理的进步和糖尿病患者预期寿命的增加,糖尿病和其他病理状况(如肝病,癌症,神经退行性疾病,认知障碍,和睡眠障碍)已经出现。因此,这些合并症被认为是糖尿病的新兴并发症.P66Shc是一种氧化还原蛋白,在氧化应激中起作用,凋亡,葡萄糖代谢,和细胞老化。它可以受到糖尿病环境中典型的各种应激刺激的调节,并参与糖尿病条件下各种类型的器官和组织损伤。尽管其在糖尿病发病机制中的作用仍存在争议,有强有力的证据表明p66Shc参与了糖尿病的传统并发症.在这次审查中,我们将总结支持p66Shc在糖尿病及其并发症发病机制中的作用的证据,首次关注新出现的糖尿病并发症。
    Diabetes mellitus is a chronic metabolic disease, the prevalence of which is constantly increasing worldwide. It is often burdened by disabling comorbidities that reduce the quality and expectancy of life of the affected individuals. The traditional complications of diabetes are generally described as macrovascular complications (e.g., coronary heart disease, peripheral arterial disease, and stroke), and microvascular complications (e.g., diabetic kidney disease, retinopathy, and neuropathy). Recently, due to advances in diabetes management and the increased life expectancy of diabetic patients, a strong correlation between diabetes and other pathological conditions (such as liver diseases, cancer, neurodegenerative diseases, cognitive impairments, and sleep disorders) has emerged. Therefore, these comorbidities have been proposed as emerging complications of diabetes. P66Shc is a redox protein that plays a role in oxidative stress, apoptosis, glucose metabolism, and cellular aging. It can be regulated by various stressful stimuli typical of the diabetic milieu and is involved in various types of organ and tissue damage under diabetic conditions. Although its role in the pathogenesis of diabetes remains controversial, there is strong evidence regarding the involvement of p66Shc in the traditional complications of diabetes. In this review, we will summarize the evidence supporting the role of p66Shc in the pathogenesis of diabetes and its complications, focusing for the first time on the emerging complications of diabetes.
    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
    大量证据表明卵巢氧化应激可导致严重的卵巢功能障碍。
    本文的目的是研究SIRT6通过抑制氧化应激缓解卵巢早衰(POF)的潜力。
    要模仿POF,小鼠每天皮下注射d-半乳糖。E2,FSH,LH,AMH,测定血清中的孕酮,随着卵泡和SIRT6水平的变化。小鼠用SIRT6激动剂MDL-800,SIRT6水平,卵泡,和上述激素被重新评估。随后鉴定了MDL-800对氧化应激和细胞凋亡的影响。原代颗粒细胞从小鼠中分离,以及H2O2和MDL-800对细胞活力的影响,氧化应激,SIRT6级,和细胞凋亡进行评估。此外,SIRT6对H3K9AC/p66SHC的调节通过检测蛋白质水平的变化来验证,启动子活性,和p66SHC过表达的逆转效应。
    MDL-800缓解了激素波动,减少卵巢组织中的卵泡消耗,并减轻小鼠的氧化应激和凋亡。体外实验表明,MDL-800增强了原代颗粒细胞对H2O2的抵抗力,这可以通过增加细胞活力,减少氧化应激和凋亡来证明。此外,SIRT6被发现降低H3K9AC和p66SHC水平,以及减弱p66SHC启动子的活性。p66SHC过表达后,MDL-800对细胞的保护作用被逆转。
    总之,这项研究强调了SIRT6的激活可以通过降解H3K9AC和抑制颗粒细胞中的p66Shc水平来减轻POF和减少氧化应激。
    UNASSIGNED: Substantial evidence suggests that ovarian oxidative stress can result in severe ovarian dysfunction.
    UNASSIGNED: The purpose of this article is to investigate the potential of SIRT6 in alleviating premature ovarian failure (POF) by inhibiting oxidative stress.
    UNASSIGNED: To mimic POF, mice were administered daily subcutaneous injections of d-galactose. The levels of E2, FSH, LH, AMH, and progesterone in serum were measured, along with changes in follicles and SIRT6 levels. Mice were treated with the SIRT6 agonist MDL-800, SIRT6 levels, follicles, and aforementioned hormones were reassessed. The effects of MDL-800 on oxidative stress and apoptosis were subsequently identified. Primary granulosa cells were isolated from mice, and the effects of H2O2 and MDL-800 on cell viability, oxidative stress, SIRT6 level, and apoptosis were evaluated. In addition, the regulation of SIRT6 on H3K9AC/p66SHC was verified by examining changes in protein levels, promoter activity, and the reversal effects of p66SHC overexpression.
    UNASSIGNED: MDL-800 mitigated hormone fluctuations, reduced follicle depletion in ovarian tissue, and attenuated oxidative stress and apoptosis in mice. In vitro experiments demonstrated that MDL-800 enhanced the resilience of primary granulosa cells against H2O2, as evidenced by increased cell viability and reduced oxidative stress and apoptosis. Furthermore, SIRT6 was found to decrease H3K9AC and p66SHC levels, as well as attenuate p66SHC promoter activity. The protective effects of MDL-800 on cells were reversed upon p66SHC overexpression.
    UNASSIGNED: In summary, this study highlights that activation of SIRT6 can alleviate POF and reduce oxidative stress by degrading H3K9AC and suppressing p66Shc levels in granulosa cells.
    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
    目的:衰老诱导血管重塑的潜在机制尚不清楚。这项研究调查了细胞质去乙酰化酶sirtuin2(SIRT2)在衰老诱导的血管重塑中的作用和潜在机制。
    结果:转录组和定量实时PCR数据用于分析沉默酶表达。年轻和年老的野生型和Sirt2基因敲除小鼠用于探索血管功能和病理重塑。RNA-seq,组织化学染色,和生化试验用于评估Sirt2敲除对血管转录组和病理重塑的影响,并探索潜在的生化机制。在sirtuins中,SIRT2在人和小鼠主动脉中具有最高水平。Sirtuin2活性在老年主动脉中降低,SIRT2的丢失加速了血管老化。在老老鼠身上,SIRT2缺乏加重老化引起的动脉僵硬度和收缩-舒张功能障碍,伴有主动脉重塑(增厚的血管中层,弹性蛋白纤维的断裂,胶原蛋白沉积,和炎症)。转录组和生化分析表明,控制衰老的蛋白p66Shc和线粒体活性氧(mROS)的代谢有助于SIRT2在血管衰老中的功能。Sirtuin2通过在赖氨酸81处将p66Shc脱乙酰来抑制p66Shc的活化和mROS的产生。MnTBAP消除活性氧抑制了SIRT2缺乏介导的血管紧张素II攻击和老年小鼠血管重塑和功能障碍的加重。主动脉中的SIRT2共表达模块随着物种的老化而减少,并且是人类年龄相关主动脉疾病的重要预测指标。
    结论:脱乙酰酶SIRT2是一种延缓血管衰老的衰老反应,细胞质-线粒体轴(SIRT2-p66Shc-mROS)对于血管老化很重要。因此,SIRT2可作为血管再生的潜在治疗靶点。
    The mechanisms underlying ageing-induced vascular remodelling remain unclear. This study investigates the role and underlying mechanisms of the cytoplasmic deacetylase sirtuin 2 (SIRT2) in ageing-induced vascular remodelling.
    Transcriptome and quantitative real-time PCR data were used to analyse sirtuin expression. Young and old wild-type and Sirt2 knockout mice were used to explore vascular function and pathological remodelling. RNA-seq, histochemical staining, and biochemical assays were used to evaluate the effects of Sirt2 knockout on the vascular transcriptome and pathological remodelling and explore the underlying biochemical mechanisms. Among the sirtuins, SIRT2 had the highest levels in human and mouse aortas. Sirtuin 2 activity was reduced in aged aortas, and loss of SIRT2 accelerated vascular ageing. In old mice, SIRT2 deficiency aggravated ageing-induced arterial stiffness and constriction-relaxation dysfunction, accompanied by aortic remodelling (thickened vascular medial layers, breakage of elastin fibres, collagen deposition, and inflammation). Transcriptome and biochemical analyses revealed that the ageing-controlling protein p66Shc and metabolism of mitochondrial reactive oxygen species (mROS) contributed to SIRT2 function in vascular ageing. Sirtuin 2 repressed p66Shc activation and mROS production by deacetylating p66Shc at lysine 81. Elimination of reactive oxygen species by MnTBAP repressed the SIRT2 deficiency-mediated aggravation of vascular remodelling and dysfunction in angiotensin II-challenged and aged mice. The SIRT2 coexpression module in aortas was reduced with ageing across species and was a significant predictor of age-related aortic diseases in humans.
    The deacetylase SIRT2 is a response to ageing that delays vascular ageing, and the cytoplasm-mitochondria axis (SIRT2-p66Shc-mROS) is important for vascular ageing. Therefore, SIRT2 may serve as a potential therapeutic target for vascular rejuvenation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号