Sirtuin 1 (SIRT1)

  • 文章类型: Journal Article
    背景:全球过度食用单糖对骨骼健康的影响,在青春期/成年早期达到高峰,并与骨质疏松症(OP)和骨折风险相关数十年,不清楚。间充质基质/干细胞(MSCs)是成骨细胞/骨形成细胞的祖细胞,并且已知随着年龄的增长它们的成骨分化能力降低。令人震惊的是,虽然有相关证据表明,消耗最多单糖的青少年骨量最低,对这种相关性的因果关系没有机械理解。
    方法:使用人细胞信息对MSC分化过程中涉及的能量通路进行生物信息学分析。使用多源非衰老的人和鼠MSCs评估了正常与高葡萄糖(HG)条件对骨/脂肪谱系定型和线粒体功能的体外解剖;用于体内验证,用正常或添加HG的水喂养幼鼠,随后分析骨髓CD45-MSC。
    结果:生物信息学分析显示,线粒体和葡萄糖相关的代谢途径是MSC骨/脂肪谱系确定的组成部分。功能上,由于烟酰胺腺嘌呤二核苷酸(NAD+)的消耗,单独的体外HG降低了MSC线粒体活性和骨生成,同时增强了8小时的脂肪形成,Sirtuin(SIRT)1的重要线粒体辅酶和辅因子,Sirtuin是一种也参与成骨的长寿基因。在体内,年轻小鼠中HG的摄入量耗尽MSCNAD+,口服NAD前体补充剂在1〜5天内以SIRT1依赖性方式迅速逆转线粒体下降和骨/脂肪承诺。
    结论:我们发现了过量葡萄糖的惊人快速影响,单一的饮食因素,在年轻环境中MSCSIRT1功能和成骨,以及NAD+-单分子对MSC线粒体功能和谱系承诺的关键作用。鉴于当前全球单糖的过度消费,这些发现对未来的全球OP和残疾风险具有重要意义。
    BACKGROUND: The impact of global overconsumption of simple sugars on bone health, which peaks in adolescence/early adulthood and correlates with osteoporosis (OP) and fracture risk decades, is unclear. Mesenchymal stromal/stem cells (MSCs) are the progenitors of osteoblasts/bone-forming cells, and known to decrease their osteogenic differentiation capacity with age. Alarmingly, while there is correlative evidence that adolescents consuming greatest amounts of simple sugars have the lowest bone mass, there is no mechanistic understanding on the causality of this correlation.
    METHODS: Bioinformatics analyses for energetics pathways involved during MSC differentiation using human cell information was performed. In vitro dissection of normal versus high glucose (HG) conditions on osteo-/adipo-lineage commitment and mitochondrial function was assessed using multi-sources of non-senescent human and murine MSCs; for in vivo validation, young mice was fed normal or HG-added water with subsequent analyses of bone marrow CD45- MSCs.
    RESULTS: Bioinformatics analyses revealed mitochondrial and glucose-related metabolic pathways as integral to MSC osteo-/adipo-lineage commitment. Functionally, in vitro HG alone without differentiation induction decreased both MSC mitochondrial activity and osteogenesis while enhancing adipogenesis by 8 h\' time due to depletion of nicotinamide adenine dinucleotide (NAD+), a vital mitochondrial co-enzyme and co-factor to Sirtuin (SIRT) 1, a longevity gene also involved in osteogenesis. In vivo, HG intake in young mice depleted MSC NAD+, with oral NAD+ precursor supplementation rapidly reversing both mitochondrial decline and osteo-/adipo-commitment in a SIRT1-dependent fashion within 1 ~ 5 days.
    CONCLUSIONS: We found a surprisingly rapid impact of excessive glucose, a single dietary factor, on MSC SIRT1 function and osteogenesis in youthful settings, and the crucial role of NAD+-a single molecule-on both MSC mitochondrial function and lineage commitment. These findings have strong implications on future global OP and disability risks in light of current worldwide overconsumption of simple sugars.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Case Reports
    背景:Helsmoortel-VanderAa综合征是一种神经发育障碍,智力残疾,和常见的神经外特征,如喂养和胃肠道问题,视觉障碍,和心脏异常。所有患者在活动依赖性神经保护蛋白(ADNP)基因中表现出杂合的从头无义或移码停止突变,占全球所有自闭症病例的0.2%。ADNP在大脑发育过程中具有重要的染色质重塑功能。在这项研究中,我们调查了一名死亡的6岁男性患者的小脑c.1676dupA/p。His559Glnfs*3ADNP突变。
    结果:患者的临床表现为具有代表性的Helsmoortel-VanderAa综合征。在他的一生中,他接受了两次肝移植手术,之后孩子因多器官衰竭而死亡。进行了尸检,和各种组织样本进行进一步分析。我们对小脑进行了分子表征,参与运动协调的大脑区域,以其最高的ADNP表达而闻名,并将其与年龄匹配的对照受试者进行了比较。重要的是,对ADNP小脑的全基因组分析确定了CpG甲基化差异和导致神经发育延迟的多种途径的表达。有趣的是,差异甲基化基因的转录因子基序富集分析表明,ADNP结合基序富集最显著。尸检大脑的RNA测序进一步确定了WNT信号通路的下调和自噬缺陷可能是神经发育迟缓的原因。最终,无标记定量质谱鉴定了参与线粒体应激和沉默调节蛋白信号通路等的差异表达蛋白。蛋白质-蛋白质相互作用分析进一步揭示了一个包括染色质重塑剂(ADNP,SMARCC2、HDAC2和YY1),自噬相关蛋白(LAMP1,BECN1和LC3)以及参与线粒体能量代谢的关键组蛋白去乙酰化酶SIRT1。通过小鼠小脑中的直接共免疫沉淀,通过微管末端结合蛋白EB1/EB3进一步生化验证了ADNP与SIRT1的蛋白质相互作用,表明染色质重塑和线粒体能量代谢之间重要的线粒体表观遗传串扰与自噬应激反应有关。线粒体活性测定和患者来源的成纤维细胞的染色进一步支持了这一点,这表明ADNP缺陷人脑中的线粒体功能障碍。
    结论:这项研究形成了ADNP尸检小脑的基线临床和分子特征,为Helsmoortel-VanderAa综合征的疾病机制提供新的见解。通过结合多维和生化方法,我们发现了一种新的SIRT1-EB1/EB3-ADNP蛋白复合物,该复合物可能导致Helsmoortel-VanderAa综合征的自噬通量改变和线粒体代谢受损,有望成为新的治疗靶点.
    BACKGROUND: Helsmoortel-Van der Aa syndrome is a neurodevelopmental disorder in which patients present with autism, intellectual disability, and frequent extra-neurological features such as feeding and gastrointestinal problems, visual impairments, and cardiac abnormalities. All patients exhibit heterozygous de novo nonsense or frameshift stop mutations in the Activity-Dependent Neuroprotective Protein (ADNP) gene, accounting for a prevalence of 0.2% of all autism cases worldwide. ADNP fulfills an essential chromatin remodeling function during brain development. In this study, we investigated the cerebellum of a died 6-year-old male patient with the c.1676dupA/p.His559Glnfs*3 ADNP mutation.
    RESULTS: The clinical presentation of the patient was representative of the Helsmoortel-Van der Aa syndrome. During his lifespan, he underwent two liver transplantations after which the child died because of multiple organ failure. An autopsy was performed, and various tissue samples were taken for further analysis. We performed a molecular characterization of the cerebellum, a brain region involved in motor coordination, known for its highest ADNP expression and compared it to an age-matched control subject. Importantly, epigenome-wide analysis of the ADNP cerebellum identified CpG methylation differences and expression of multiple pathways causing neurodevelopmental delay. Interestingly, transcription factor motif enrichment analysis of differentially methylated genes showed that the ADNP binding motif was the most significantly enriched. RNA sequencing of the autopsy brain further identified downregulation of the WNT signaling pathway and autophagy defects as possible causes of neurodevelopmental delay. Ultimately, label-free quantification mass spectrometry identified differentially expressed proteins involved in mitochondrial stress and sirtuin signaling pathways amongst others. Protein-protein interaction analysis further revealed a network including chromatin remodelers (ADNP, SMARCC2, HDAC2 and YY1), autophagy-related proteins (LAMP1, BECN1 and LC3) as well as a key histone deacetylating enzyme SIRT1, involved in mitochondrial energy metabolism. The protein interaction of ADNP with SIRT1 was further biochemically validated through the microtubule-end binding proteins EB1/EB3 by direct co-immunoprecipitation in mouse cerebellum, suggesting important mito-epigenetic crosstalk between chromatin remodeling and mitochondrial energy metabolism linked to autophagy stress responses. This is further supported by mitochondrial activity assays and stainings in patient-derived fibroblasts which suggest mitochondrial dysfunctions in the ADNP deficient human brain.
    CONCLUSIONS: This study forms the baseline clinical and molecular characterization of an ADNP autopsy cerebellum, providing novel insights in the disease mechanisms of the Helsmoortel-Van der Aa syndrome. By combining multi-omic and biochemical approaches, we identified a novel SIRT1-EB1/EB3-ADNP protein complex which may contribute to autophagic flux alterations and impaired mitochondrial metabolism in the Helsmoortel-Van der Aa syndrome and holds promise as a new therapeutic target.
    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
    ·诱导的调节性T细胞(iTregs)是具有治疗潜力的免疫抑制性T细胞的异质群体。Treg细胞表现出一定的可塑性,可以获得T效应子样能力,类似于T助手1(Th1)的iTregs。因此,区分功能可塑性和谱系不稳定性是很重要的。再生障碍性贫血(AA)是一种自身免疫性疾病,其特征是骨髓(BM)中造血干细胞和祖细胞的免疫介导破坏。Th1样1iTreg可以是异常Th1介导的免疫应答(例如驱动AA疾病进展的那些)的有效抑制剂。在这里,我们研究了表观遗传酶的功能,蛋白质精氨酸甲基转移酶5(PRMT5),它对iTreg去稳定脱乙酰酶的调节,抑制Th1样iTregs中的沉默蛋白1(Sirt1),以及将Th1样iTregs用作AA的细胞疗法的潜力。
    我们通过用IL-12培养iTregs来产生Th1样iTregs,然后评估它们的抑制能力,iTreg抑制标记的表达,和PRMT5的酶活性,使用组蛋白对称精氨酸二甲基化(H3R2me2s)作为读数。我们在Th1细胞上使用了ChIP测序,iTregs,和Th1样iTregs来鉴定Th1样iTregs特有的H3R2me2s结合基因,然后使用CHiP-qPCR验证靶标。我们取消了PRMT5,以验证其对Th1样iTreg谱系承诺的贡献。最后,我们使用Th1介导的AA小鼠模型测试了Th1样iTregs的治疗潜力。
    将iTregs暴露于Th1细胞因子,白细胞介素-12(IL-12),在分化的早期事件中,表达了抑制功能的增加。我们观察到PRMT5酶活性增加,用H3R2me2s测量,在类似Th1的iTregs中,在iTregs中被下调。使用ChIP测序,我们发现H3R2me2s与Th1样iTreg中的Sirt1启动子区域大量结合,从而负向调节其表达。此外,向AA小鼠施用Th1样iTregs提供了生存益处。
    在Th1样iTregs中击倒PRMT5的同时降低了它们的抑制能力,支持PRMT5对于Th1样iTregs的优异抑制能力和稳定性很重要的观点。最后,Th1样iTregs在AA小鼠模型中的治疗性给药可显着延长其生存期,并且它们可能具有治疗潜力。
    Induced regulatory T cells (iTregs) are a heterogeneous population of immunosuppressive T cells with therapeutic potential. Treg cells show a range of plasticity and can acquire T effector-like capacities, as is the case for T helper 1 (Th1)-like iTregs. Thus, it is important to distinguish between functional plasticity and lineage instability. Aplastic anemia (AA) is an autoimmune disorder characterized by immune-mediated destruction of hematopoietic stem and progenitor cells in the bone marrow (BM). Th1-like 1 iTregs can be potent suppressors of aberrant Th1-mediated immune responses such as those that drive AA disease progression. Here we investigated the function of the epigenetic enzyme, protein arginine methyltransferase 5 (PRMT5), its regulation of the iTreg-destabilizing deacetylase, sirtuin 1 (Sirt1) in suppressive Th1-like iTregs, and the potential for administering Th1-like iTregs as a cell-based therapy for AA.
    We generated Th1-like iTregs by culturing iTregs with IL-12, then assessed their suppressive capacity, expression of iTreg suppression markers, and enzymatic activity of PRMT5 using histone symmetric arginine di-methylation (H3R2me2s) as a read out. We used ChIP sequencing on Th1 cells, iTregs, and Th1-like iTregs to identify H3R2me2s-bound genes unique to Th1-like iTregs, then validated targets using CHiP-qPCR. We knocked down PRMT5 to validate its contribution to Th1-like iTreg lineage commitment. Finally we tested the therapeutic potential of Th1-like iTregs using a Th1-mediated mouse model of AA.
    Exposing iTregs to the Th1 cytokine, interleukin-12 (IL-12), during early events of differentiation conveyed increased suppressive function. We observed increased PRMT5 enzymatic activity, as measured by H3R2me2s, in Th1-like iTregs, which was downregulated in iTregs. Using ChIP-sequencing we discovered that H3R2me2s is abundantly bound to the Sirt1 promoter region in Th1-like iTregs to negatively regulate its expression. Furthermore, administering Th1-like iTregs to AA mice provided a survival benefit.
    Knocking down PRMT5 in Th1-like iTregs concomitantly reduced their suppressive capacity, supporting the notion that PRMT5 is important for the superior suppressive capacity and stability of Th1-like iTregs. Conclusively, therapeutic administration of Th1-like iTregs in a mouse model of AA significantly extended their survival and they may have therapeutic potential.
    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
    迟发性阿尔茨海默病(AD)是一种复杂的多因素疾病。已知晚发性AD的最大危险因素是载脂蛋白E(APOE)的E4等位基因,而年龄增长是已知的最大的非遗传危险因素。神经干细胞(NSC)的细胞类型特异性功能,特别是它们的干细胞可塑性,在AD病理学的背景下仍然缺乏探索。这里,我们描述了一种新的模型,该模型采用源自晚发性AD患者的诱导多能干细胞(iPSCs)来产生神经干细胞,并与携带APOE3的健康受试者相比,研究了APOE4在衰老标志物如sirtuin1(SIRT1)表达中的作用.通过使用来自老年受试者的iPSC衍生的NSC作为健康匹配的对照来研究衰老的影响。转录和蛋白质分析显示,基因在迟发性AD患者的神经干细胞中表达不同,例如,表现出减少的自噬相关蛋白7(ATG7),磷酸酶和张力蛋白同源物(PTEN),和成纤维细胞生长因子2(FGF2)。由于SIRT1表达在APOE3和APOE4NSC之间不同,NSC中APOE功能的抑制也抑制SIRT1的表达。然而,质粒强制表达APOE3并没有恢复不同表达的基因。改变的老化标志物表明NSC的可塑性降低。我们的研究提供了一个合适的体外模型来研究与衰老相关的人类神经干细胞的变化。APOE4和晚发性AD。
    Late-onset Alzheimer\'s disease (AD) is a complex multifactorial disease. The greatest known risk factor for late-onset AD is the E4 allele of the apolipoprotein E (APOE), while increasing age is the greatest known non-genetic risk factor. The cell type-specific functions of neural stem cells (NSCs), in particular their stem cell plasticity, remain poorly explored in the context of AD pathology. Here, we describe a new model that employs late-onset AD patient-derived induced pluripotent stem cells (iPSCs) to generate NSCs and to examine the role played by APOE4 in the expression of aging markers such as sirtuin 1 (SIRT1) in comparison to healthy subjects carrying APOE3. The effect of aging was investigated by using iPSC-derived NSCs from old age subjects as healthy matched controls. Transcript and protein analysis revealed that genes were expressed differently in NSCs from late-onset AD patients, e.g., exhibiting reduced autophagy-related protein 7 (ATG7), phosphatase and tensin homolog (PTEN), and fibroblast growth factor 2 (FGF2). Since SIRT1 expression differed between APOE3 and APOE4 NSCs, the suppression of APOE function in NSCs also repressed the expression of SIRT1. However, the forced expression of APOE3 by plasmids did not recover differently expressed genes. The altered aging markers indicate decreased plasticity of NSCs. Our study provides a suitable in vitro model to investigate changes in human NSCs associated with aging, APOE4, and late-onset AD.
    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
    已显示激活的G蛋白偶联受体39(GPR39)通过与沉默调节蛋白1(SIRT1)和过氧化物酶体增殖物激活受体-γ共激活因子1α(PGC-1α)相互作用来减轻炎症。然而,GPR39是否减轻神经性疼痛尚不清楚.在这项研究中,我们建立了Sprague-Dawley大鼠幸免神经损伤诱导的神经病理性疼痛模型,发现与假手术大鼠相比,脊髓背角神经元和小胶质细胞中GPR39的表达显著降低.鞘内注射GPR39的特异性激动剂TC-G1008可明显减轻保留神经损伤大鼠的机械性异常疼痛。改善脊髓线粒体生物发生,减轻神经炎症.这些变化被GPR39小干扰RNA(siRNA)消除,Ex-527(SIRT1抑制剂),和PGC-1αsiRNA。一起来看,这些发现表明,GPR39激活通过激活SIRT1/PGC-1α通路改善了保留神经损伤大鼠的机械性异常疼痛。
    Activated G-protein-coupled receptor 39 (GPR39) has been shown to attenuate inflammation by interacting with sirtuin 1 (SIRT1) and peroxisome proliferator-activated receptor-γ coactivator 1α (PGC-1α). However, whether GPR39 attenuates neuropathic pain remains unclear. In this study, we established a Sprague-Dawley rat model of spared nerve injury-induced neuropathic pain and found that GPR39 expression was significantly decreased in neurons and microglia in the spinal dorsal horn compared with sham-operated rats. Intrathecal injection of TC-G 1008, a specific agonist of GPR39, significantly alleviated mechanical allodynia in the rats with spared nerve injury, improved spinal cord mitochondrial biogenesis, and alleviated neuroinflammation. These changes were abolished by GPR39 small interfering RNA (siRNA), Ex-527 (SIRT1 inhibitor), and PGC-1α siRNA. Taken together, these findings show that GPR39 activation ameliorates mechanical allodynia by activating the SIRT1/PGC-1α pathway in rats with spared nerve 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
    简介:慢性不愈合伤口是一个相当大的临床挑战和研究发现新的促愈合剂正在进行中,因为现有的治疗方法不能充分满足当前的需求。方法:我们通过Western印迹研究了Corylin在细胞系成纤维细胞和巨噬细胞中的作用,PCR,流式细胞术测定,免疫荧光。结果:我们发现corylin,从补骨脂中提取的主要类黄酮,减少炎症反应,促进胶原蛋白沉积,加速小鼠全层皮肤伤口的愈合。对潜在机制的探索表明,corylin激活了PI3K/AKT信号,导致成纤维细胞迁移,扩散,和划痕愈合。Corylin还激活沉默酶1(SIRT1)信号,增强NF-κBp65的脱乙酰和细胞质易位,因此减少了脂多糖(LPS)诱导的巨噬细胞炎症反应。此外,用LY294002和EX527抑制PI3K/AKT和sirtuin1途径可防止Corylin对慢性伤口的治疗效力。结论:总之,我们的结果表明,corylin可能是开发新的促愈合剂的候选药物。
    Introduction: Chronic non-healing wound is a considerable clinical challenge and research into the discovery of novel pro-healing agents is underway as existing therapeutic approaches cannot sufficiently meet current needs. Method: We studied the effects of corylin in cell line fibroblasts and macrophages by Western blots, PCR, Flow cytometry assay, Immunofluorescence. Results: We showed that corylin, a main flavonoid extracted from Psoralea corylifolia L, reduced inflammatory responses, promoted collagen deposition, and accelerated the healing of full-thickness skin wounds in mice. Exploration of the underlying mechanisms showed that corylin activated the PI3K/AKT signaling, leading to fibroblasts\' migration, proliferation, and scratch healing. Corylin also activated sirtuin 1 (SIRT1) signaling, enhanced the deacetylation and cytoplasmic translocation of NF-κB p65, and therefore reduced lipopolysaccharide (LPS)-induced inflammatory responses in macrophages. Furthermore, inhibition of PI3K/AKT and sirtuin 1 pathway with LY294002 and EX527 prevent the therapeutic potency of corylin against chronic wounds. Conclusion: In summary, our results suggested that corylin may be a candidate for the development of novel pro-healing agents.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Retraction of Publication
    [这撤回了文章DOI:10.3389/fonc.2021.684477。].
    [This retracts the article DOI: 10.3389/fonc.2021.684477.].
    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
    牛皮癣是一种无法治愈的自身免疫性疾病,影响全球2-3%的人口。对其发病机理的有限理解阻碍了该疾病疗法的发展。在这里,我们报道了N-酰基乙醇胺酸酰胺酶(NAAA),一种催化脂肪酸乙醇酰胺(FAEs)水解的半胱氨酸酶,在银屑病患者和咪喹莫特(IMQ)诱导的银屑病小鼠模型中上调。上调的NAAA通过增强树突状细胞(DC)成熟促进银屑病的进展。NAAA在小鼠体内的转基因表达加速了银屑病的发展,而NAAA的遗传消融或NAAA抑制剂F96的局部给药可改善银屑病。NAAA在树突状细胞(DC)中表达,但不是在巨噬细胞中,T细胞,或角质形成细胞在牛皮癣的发展中起关键作用。此外,结果表明,NAAA降解棕榈酰乙醇胺(PEA)并减少PEA-PPARα介导的NF-κBp65从Sirtuin1(SIRT1)的解离,随后,抑制p65的乙酰化并下调IL10的产生。减少的IL10然后导致DCs的成熟,从而促进银屑病的发展。这些结果为银屑病的病理生理机制提供了新的见解,并将NAAA确定为治疗银屑病的新靶点。
    Psoriasis is an incurable autoimmune disease that affects 2-3% of the world\'s population. Limited understanding of its pathogenesis hinders the development of therapies for the disease. Herein, we reported that N-acylethanolamine acid amidase (NAAA), a cysteine enzyme that catalyzes the hydrolysis of fatty acid ethanolamides (FAEs), was upregulated in psoriasis patients and imiquimod (IMQ)-induced mouse model of psoriasis. The upregulated NAAA contributes to the progression of psoriasis via enhancing dendritic cell (DCs) maturation. Transgenic expression of NAAA in mice accelerated the development of psoriasis, whereas genetic ablation of NAAA or local administration of NAAA inhibitor F96 ameliorated psoriasis. NAAA expressed in dendritic cells (DCs), but not in macrophages, T cells, or keratinocytes plays a critical role in psoriasis development. In addition, the results showed that NAAA degrades palmitoylethanolamide (PEA) and reduces PEA-PPARα-mediated dissociation of NF-κB p65 from Sirtuin 1 (SIRT1), subsequently, repressing the acetylation of p65 and down-regulating IL10 production. The decreased IL10 then leads to the maturation of DCs, thus promoting the development of psoriasis. These results provide new insights into the pathophysiological mechanism of psoriasis and identify NAAA as a novel target for the treatment of psoriasis.
    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
    含有非POU结构域的八聚体结合蛋白(NONO,也称为p54nrb)是一种参与转录调控的多功能核蛋白,mRNA剪接,有缺陷的RNA的核保留,DNA修复新的证据表明,p54nrb受到各种翻译后修饰,包括磷酸化和甲基化,这可能是其多功能的重要调节器。然而,在这些修改中,p54nrb乙酰化的直接证据及其潜在机制尚不清楚。在这项研究中,我们报道了p54nrb的赖氨酸371被乙酰转移酶一般对照不可抑制的5(GCN5)和去乙酰化酶沉默酶1(SIRT1)可逆地乙酰化,这对于p54nrb抑制白细胞介素-8(IL-8)表达的活性至关重要。机械上,GCN5介导的乙酰化减弱了IL-8基因启动子内p54nrb在其核心结合基序上的募集,优先增加IL-8基因的表达。相比之下,通过SIRT1的脱乙酰化逆转了该过程。总之,我们的数据表明,可逆乙酰化是p54nrb/NONO多核功能的重要转换.
    The non-POU domain-containing octamer-binding protein (NONO, also referred to as p54nrb) is a multifunctional nuclear protein engaging in transcriptional regulation, mRNA splicing, nuclear retention of defective RNA, and DNA repair. Emerging evidence has demonstrated that p54nrb is subjected to various posttranslational modifications, including phosphorylation and methylation, which may be important regulators of its multifunction. However, among these modifications, direct evidence of p54nrb acetylation and its underlying mechanism remains unclear. In this study, we reported that lysine 371 of p54nrb was reversibly acetylated by the acetyltransferase general control non-depressible 5 (GCN5) and deacetylase sirtuin 1 (SIRT1), which was crucial for activity of p54nrb to inhibit interleukin-8 (IL-8) expression. Mechanistically, GCN5-mediated acetylation attenuates the recruitment of p54nrb on its core binding motif within the IL-8 gene promoter, preferentially increasing the expression of the IL-8 gene. In contrast, deacetylation by SIRT1 reverses this process. Altogether, our data suggest that reversible acetylation is an important switch for the multiple nuclear functions of p54nrb/NONO.
    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RNA(miRNA)作为肿瘤抑制因子或癌基因,被称为oncomiRs,是癌症靶向治疗的一个有希望的新焦点。大约16%的乳腺癌患者患有糖尿病。乳腺癌伴2型糖尿病(BDM)具有其独特的生物学特性和临床特点。这项研究主要调查了miR-29a在BDM患者中的治疗潜力和调节机制。
    通过乳腺组织中的实时逆转录酶聚合酶链反应(qRT-PCR)分析了BDM中miR-29a的重要性。通过使用在高水平的葡萄糖和胰岛素培养的3T3-L1脂肪细胞中培养的MDA-MB-231细胞来建立BDM的细胞模型。通过高脂饮食加低剂量链脲佐菌素(STZ)在雌性BALB/c小鼠中诱导2型糖尿病(T2DM)小鼠模型。通过使用MDA-MB-231细胞在这些T2DM小鼠上建立用于BDM的异种移植小鼠模型。然后研究慢病毒-shRNAs介导的miR-29a敲低对细胞增殖的生物学效应,凋亡,细胞周期,和迁移进行了调查。
    我们的结果表明miR-29a在BDM患者中上调,这与预后较差有关。在人类乳腺癌细胞中,miR-29a敲低降低了BDM中的细胞增殖以及细胞迁移和侵袭。在T2DM异种移植物中,miR-29a敲低抑制MDA-MB-231细胞肿瘤发生和转移。我们还证明miR-29a通过靶向Sirtuin1(SIRT1)促进BDM细胞生长和转移。
    我们的发现表明,抗miR-29a通过靶向SIRT1抑制BDM中的细胞增殖和侵袭。我们相信抗miR-29a可能代表了BDM患者的一种新的治疗方法。
    UNASSIGNED: MicroRNAs (miRNAs) acting as tumour suppressors or oncogenes, known as oncomiRs, are a promising new focus in targeted therapies for cancer. Approximately 16% of breast cancer patients have pre-existing diabetes. Breast cancer with type 2 diabetes mellitus (BDM) is provided with its unique biological characteristics and clinical characteristics. This study primarily investigated the therapeutic potential and regulatory mechanism of miR-29a in patients with BDM.
    UNASSIGNED: The significance of miR-29a in BDM was analyzed by real-time reverse transcriptase polymerase chain reaction (qRT-PCR) in breast tissues. A cell model for BDM was established by using MDA-MB-231 cells cultured in 3T3-L1 adipocytes cultured with high levels of glucose and insulin. A type 2 diabetes mellitus (T2DM) mouse model was induced in female BALB/c mice through a high-fat diet plus low doses of streptozotocin (STZ). The xenograft mouse-model for BDM was established on these T2DM mouse by using MDA-MB-231 cells. Then the biological effects of miR-29a knockdown mediated by lentivirus-shRNAs on cell proliferation, apoptosis, cell cycle, and migration were investigated.
    UNASSIGNED: Our results indicated that miR-29a was upregulated in patients with BDM, which correlated with a worse prognosis. In human breast cancer cells, miR-29a knockdown reduced cell proliferation and cell migration and invasion in BDM. In the T2DM xenograft, miR-29a knockdown suppressed MDA-MB-231 cells tumorigenesis and metastasis. We also demonstrated that miR-29a promoted BDM cell growth and metastasis by targeting Sirtuin 1 (SIRT1).
    UNASSIGNED: Our findings indicated that anti-miR-29a inhibited cell proliferation and invasion in BDM by targeting SIRT1. We believe anti-miR-29a may represent a novel therapeutic approach for the management of patients with BDM.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号