sirtuin 1

Sirtuin 1
  • 文章类型: Journal Article
    尽管肝细胞癌(HCC)的全身治疗有最新进展,乙型肝炎病毒(HBV)诱导的HCC患者的预后仍然很差。通过筛选针对人去泛素酶的sgRNA文库,我们发现泛素特异性肽酶26(USP26)缺乏会损害HBV阳性HCC细胞的增殖。具有Usp26敲除的基因工程小鼠模型证实Usp26驱动HCC肿瘤发生。机械上,我们发现HBV编码蛋白HBx与启动子结合并诱导USP26的产生,USP26是一个X连锁基因,在睾丸中完全表达.因此,HBx促进USP26与SIRT1的联合,通过去泛素化协同稳定SIRT1,促进细胞增殖,阻碍细胞凋亡,加速肝癌的发生。在HBV阳性HCC患者中,USP26是强烈诱导的,其水平与SIRT1水平和不良预后相关。总的来说,我们的研究强调了HBV感染之间的因果关系,去泛素酶诱导和肝癌的发展,识别一个可下药的目标,USP26.
    Despite recent advances in systemic therapy for hepatocellular carcinoma (HCC), the prognosis of hepatitis B virus (HBV)-induced HCC patients remains poor. By screening a sgRNA library targeting human deubiquitinases, we find that ubiquitin-specific peptidase 26 (USP26) deficiency impairs HBV-positive HCC cell proliferation. Genetically engineered murine models with Usp26 knockout confirm that Usp26 drives HCC tumorigenesis. Mechanistically, we find that the HBV-encoded protein HBx binds to the promoter and induces the production of USP26, which is an X-linked gene exclusively expressed in the testis. HBx consequently promotes the association of USP26 with SIRT1 to synergistically stabilize SIRT1 by deubiquitination, which promotes cell proliferation and impedes cell apoptosis to accelerate HCC tumorigenesis. In patients with HBV-positive HCC, USP26 is robustly induced, and its levels correlate with SIRT1 levels and poor prognosis. Collectively, our study highlights a causative link between HBV infection, deubiquitinase induction and development of HCC, identifying a druggable target, USP26.
    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
    心肌纤维化可引发糖尿病性心肌病(DCM)的心力衰竭,还有irisin,一种运动诱导的肌动蛋白,可能对心脏功能有有益的影响。然而,运动与irisin在糖尿病性心脏病中的具体分子机制仍未得到充分探索。本研究旨在探讨miR-34a介导运动性irisin改善心肌纤维化的机制。在成年雄性大鼠中,高脂饮食和链脲佐菌素注射诱导了DCM引起的2型糖尿病(T2DM)。DCM大鼠进行游泳(60分钟/天)和重组irisin(r-irisin,500μg/kg/d)干预8周,分别。心功能,心肌细胞结构,分析心肌纤维化及其相关基因和蛋白表达。游泳干预减轻胰岛素抵抗,心肌纤维化,心肌肥厚,促进T2DM模型大鼠血糖稳态。这种改善与心肌中的irisin上调和miR-34a下调有关,从而增强心脏功能。通过腹膜内注射外源性重组irisin观察到类似的功效。体内抑制miR-34a通过激活沉默调节蛋白1(SIRT1)/过氧化物酶体增殖物激活受体-γ共激活因子-1α(PGC-1α)/纤连蛋白III型结构域-含蛋白5(FNDC5)信号通路和下调心肌纤维化标志物(胶原蛋白I,胶原蛋白III,和转化生长因子-β1)。因此,游泳诱导的irisin通过激活miR-34a介导的SIRT1/PGC-1α/FNDC5信号通路对糖尿病心肌纤维化具有潜在的治疗作用。
    Myocardial fibrosis can trigger heart failure in diabetic cardiomyopathy (DCM), and irisin, an exercise-induced myokine, may have a beneficial effect on cardiac function. However, the specific molecular mechanism between exercise and irisin in the diabetic heart remains not fully explored. This study aimed to investigate how miR-34a mediates exercise-induced irisin to ameliorate myocardial fibrosis and its underlying mechanisms. Type 2 diabetes mellitus (T2DM) with DCM was induced in adult male rats with high-fat diet and streptozotocin injection. The DCM rats were subjected to swimming (60 min/d) and recombinant irisin (r-irisin, 500 μg/kg/d) interventions for 8 weeks, respectively. Cardiac function, cardiomyocyte structure, myocardial fibrosis and its correlated gene and protein expression were analyzed. Swimming intervention alleviated insulin resistance, myocardial fibrosis, and myocardial hypertrophy, and promoted blood glucose homeostasis in T2DM model rats. This improvement was associated with irisin upregulation and miR-34a downregulation in the myocardium, thus enhancing cardiac function. Similar efficacy was observed via intraperitoneal injection of exogenous recombinant irisin. Inhibition of miR-34a in vivo exhibited an anti-myocardial fibrotic effect by promoting irisin secretion through activating sirtuin 1 (SIRT1)/peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α)/fibronectin type III domain-containing protein 5 (FNDC5) signal pathway and downregulating myocardial fibrosis markers (collagen I, collagen III, and transforming growth factor-β1). Therefore, swimming-induced irisin has the potential therapeutic effect on diabetic myocardial fibrosis through activating the miR-34a-mediated SIRT1/PGC-1α/FNDC5 signal pathway.
    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
    GLP-1受体激动剂(GLP-1RA)已被证明可治疗多种代谢疾病;然而,GLP-1RA对多囊卵巢综合征(PCOS)的影响尚不清楚.这里,我们的目的是调查司马鲁肽,一种新型的GLP-1RA,可以减轻PCOS小鼠的卵巢炎症。
    雌性C57BL/6J小鼠皮下注射脱氢表雄酮21天建立PCOS模型。然后将小鼠随机分为三组:PCOS组(n=6),S-0.42组(司马鲁肽0.42mg/kg/w,n=6),和S-0.84组(司马鲁肽0.84mg/kg/w,n=6)。其余6只小鼠用作对照(NC)。经过28天的干预,测定血清性激素和炎性细胞因子水平。采用苏木精、伊红染色观察卵巢形态。免疫组化染色检测CYP19A1、TNF-α、IL-6,IL-1β,和卵巢中的NF-κB。免疫荧光染色检测CYP17A1和StAR。最后,AMPK的相对表达式,pAMPK,SIRT1,NF-κB,IκBα,pIκBα,TNF-α,使用蛋白质印迹法测量IL-6和IL-1β。
    首先,在接受司马鲁肽干预后,老鼠的体重下降了,胰岛素抵抗改善,发情周期恢复正常。血清睾酮和IL-1β水平显著降低,而雌二醇和孕激素水平显著升高。卵泡囊性扩张明显改善。TNF-α的表达,IL-6,IL-1β,NF-κB,CYP17A1和StAR在卵巢中显著下调,而CYP19A1表达在干预后上调。最后,我们证实司马鲁肽通过AMPK/SIRT1/NF-κB信号通路减轻卵巢组织炎症并改善PCOS.
    塞马鲁肽通过AMPK/SIRT1/NF‑κB信号通路减轻PCOS小鼠卵巢炎症。
    UNASSIGNED: GLP-1 receptor agonists (GLP-1 RA) have been proven to treat several metabolic diseases; however, the effects of GLP-1 RA on polycystic ovary syndrome (PCOS) remain unclear. Here, we aimed to investigate whether semaglutide, a novel GLP-1 RA, could alleviate ovarian inflammation in PCOS mice.
    UNASSIGNED: Female C57BL/6J mice were subcutaneously injected with dehydroepiandrosterone for 21 days to establish the PCOS model. Then the mice were randomly divided into three groups: PCOS group (n = 6), S-0.42 group (semaglutide 0.42 mg/kg/w, n = 6), and S-0.84 group (semaglutide 0.84 mg/kg/w, n = 6). The remaining six mice were used as controls (NC). After 28 days of intervention, serum sex hormones and inflammatory cytokine levels were measured. Hematoxylin and eosin staining was used to observe the ovarian morphology. Immunohistochemical staining was used to detect the relative expression of CYP19A1, TNF-α, IL-6, IL-1β, and NF-κB in ovaries. CYP17A1 and StAR were detected using immunofluorescence staining. Finally, the relative expressions of AMPK, pAMPK, SIRT1, NF-κB, IκBα, pIκBα, TNF-α, IL-6, and IL-1β were measured using Western blotting.
    UNASSIGNED: First, after intervention with semaglutide, the weight of the mice decreased, insulin resistance improved, and the estrous cycle returned to normal. Serum testosterone and IL-1β levels decreased significantly, whereas estradiol and progestin levels increased significantly. Follicular cystic dilation significantly improved. The expression of TNF-α, IL-6, IL-1β, NF-κB, CYP17A1, and StAR in the ovary was significantly downregulated, whereas CYP19A1 expression was upregulated after the intervention. Finally, we confirmed that semaglutide alleviates ovarian tissue inflammation and improves PCOS through the AMPK/SIRT1/NF-κB signaling pathway.
    UNASSIGNED: Semaglutide alleviates ovarian inflammation via the AMPK/SIRT1/NF‑κB signaling pathway in PCOS mice.
    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
    由于目前的递送系统,在大脑中使用基于蛋白质的CRISPR系统的基因治疗具有实际局限性。尤其是在动脉闭塞的情况下。克服这些障碍,提高稳定性,我们设计了一种用于鼻内基因治疗的系统,用于治疗缺血性卒中。方法:在缺血性中风小鼠模型中,将含有靶向Sirt1的基于蛋白质的CRISPR/dCas9系统的纳米颗粒鼻内递送至大脑。CRISPR/dCas9系统用磷酸钙(CaP)纳米颗粒封装以防止它们降解。然后将它们与β-羟基丁酸酯(bHb)缀合,以靶向鼻上皮细胞中的单羧酸转运蛋白1(MCT1),以促进其转移到大脑中。结果:体外培养的人鼻上皮细胞能高效地吸收和转移纳米颗粒至人脑内皮细胞。dCas9/CaP/PEI-PEG-bHb纳米颗粒在小鼠中的鼻内给药有效上调靶基因,Sirt1,在大脑中,永久性大脑中动脉闭塞后,脑水肿减少,生存率增加。此外,我们没有观察到显著的体内毒性与纳米颗粒的鼻内给药,强调这种方法的安全性。结论:这项研究表明,提出的基于蛋白质的CRISPR-dCas9系统通常靶向神经保护基因,特别是SIRT1,可能是急性缺血性卒中的潜在新疗法。
    Gene therapy using a protein-based CRISPR system in the brain has practical limitations due to current delivery systems, especially in the presence of arterial occlusion. To overcome these obstacles and improve stability, we designed a system for intranasal administration of gene therapy for the treatment of ischemic stroke. Methods: Nanoparticles containing the protein-based CRISPR/dCas9 system targeting Sirt1 were delivered intranasally to the brain in a mouse model of ischemic stroke. The CRISPR/dCas9 system was encapsulated with calcium phosphate (CaP) nanoparticles to prevent them from being degraded. They were then conjugated with β-hydroxybutyrates (bHb) to target monocarboxylic acid transporter 1 (MCT1) in nasal epithelial cells to facilitate their transfer into the brain. Results: Human nasal epithelial cells were shown to uptake and transfer nanoparticles to human brain endothelial cells with high efficiency in vitro. The intranasal administration of the dCas9/CaP/PEI-PEG-bHb nanoparticles in mice effectively upregulated the target gene, Sirt1, in the brain, decreased cerebral edema and increased survival after permanent middle cerebral artery occlusion. Additionally, we observed no significant in vivo toxicity associated with intranasal administration of the nanoparticles, highlighting the safety of this approach. Conclusion: This study demonstrates that the proposed protein-based CRISPR-dCas9 system targeting neuroprotective genes in general, and SIRT1 in particular, can be a potential novel therapy for acute ischemic stroke.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • DOI:
    文章类型: Journal Article
    血管紧张素II(AngII),肾素-血管紧张素-醛固酮系统(RAAS)的组成部分,与脂肪组织功能失调有关。已显示抑制AngII可改善患有代谢综合征的小鼠的脂肪组织功能。众所周知,血红素氧合酶-1(HO-1),抗氧化剂可改善脂肪细胞的氧化应激和表型变化。高氧化应激的分子效应包括抑制Sirtuin-1(SIRT1),适合氧化还原操作。然而,肾素-血管紧张素-醛固酮系统(RAAS)发挥其代谢作用的潜在机制尚未完全了解。在这项研究中,我们认为AngII诱导的氧化应激可能通过下调HO-1抑制脂肪细胞SIRT1。因此,SIRT1的这种抑制可能导致盐皮质激素受体(MR)的上调。我们进一步假设HO-1的诱导将拯救SIRT1,从而改善氧化应激和脂肪细胞表型。为了建立这个假设,我们用AngII处理的小鼠前脂肪细胞进行了实验,在存在或不存在钴原卟啉(CoPP)的情况下,HO-1的诱导剂和锡的中卟啉(SnMP),HO-1的抑制剂。我们的数据表明,用AngII处理小鼠前脂肪细胞导致脂质积累增加,超氧化物和炎性细胞因子(白细胞介素-6和肿瘤坏死因子α)水平升高,脂联素水平降低。然而,这些效应被HO-1的诱导减弱,这种减弱被SnMP逆转,表明对脂肪细胞表型的有益作用受HO-1调节。此外,我们的研究结果表明,AngII处理的前脂肪细胞表现出上调的MR水平和抑制SIRT1表达,通过HO-1诱导拯救。在小鼠前脂肪细胞中使用CoPP和SIRT1siRNA处理后,脂质积累增加和脂肪酸合酶水平升高,表明HO-1的有益效果是通过SIRT1调制的。我们的研究提供了HO-1恢复细胞氧化还原平衡的证据,挽救SIRT1,并减弱AngII对脂肪细胞和全身代谢谱的有害影响。
    Angiotensin II (AngII), a component of the Renin-Angiotensin-Aldosterone System (RAAS), has been implicated in the dysregulation of adipose tissue function. Inhibition of AngII has been shown to improve adipose tissue function in mice with metabolic syndrome. It is well established that the Heme Oxygenase-1 (HO-1), an antioxidant improves oxidative stress and phenotypic change in adipocytes. Molecular effects of high oxidative stress include suppression of Sirtuin-1 (SIRT1), which is amenable to redox manipulations. However, the underlying mechanisms by which the Renin-Angiotensin-Aldosterone System (RAAS) exerts its metabolic effects are not fully understood. In this study, we propose that AngII-induced oxidative stress may suppress adipocyte SIRT1 through down-regulation of HO-1. Consequently, this suppression of SIRT1 may result in the up-regulation of the Mineralocorticoid Receptor (MR). We further hypothesize that the induction of HO-1 would rescue SIRT1, thereby improving oxidative stress and adipocyte phenotype. To establish this hypothesis, we conducted experiments using mouse preadipocytes treated with AngII, in the presence or absence of Cobalt Protoporphyrin (CoPP), an inducer of HO-1, and Tin Mesoporphyrin (SnMP), an inhibitor of HO-1. Our data demonstrate that treatment of mouse preadipocytes with AngII leads to increased lipid accumulation, elevated levels of superoxide and inflammatory cytokines (Interleukin-6 and Tumor necrosis factor alpha), and reduced levels of adiponectin. However, these effects were attenuated by the induction of HO-1, and this attenuation was reversed by SnMP, indicating that the beneficial effects on adipocyte phenotype are modulated by HO-1. Furthermore, our findings reveal that AngII-treated preadipocytes exhibit upregulated MR levels and suppressed SIRT1 expression, which are rescued by HO-1 induction. Following treatment with CoPP and SIRT1 siRNA in mouse preadipocytes resulted in increased lipid accumulation and elevated levels of fatty acid synthase, indicating that the beneficial effects of HO-1 are modulated through SIRT1. Our study provides evidence that HO-1 restores cellular redox balance, rescues SIRT1, and attenuates the detrimental effects of AngII on adipocytes and systemic metabolic profile.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: English Abstract
    目的:探讨靶向抑制3型脱碘酶(Dio3)对脓毒症骨骼肌线粒体的保护作用及其机制。
    方法:(1)体内实验:采用腺相关病毒(AAV)特异性靶向Dio3在大鼠胫骨前肌的表达,并使用盲肠结扎和穿孔(CLP)制作脓毒症大鼠模型。雄性SD大鼠分为shNC+Sham组,shD3+Sham组,shNC+CLP组,shD3+CLP组采用随机数字表法,每组8只大鼠。CLP建模后,收集胫骨样本并进行蛋白质印迹分析以评估Dio3,过氧化物酶体增殖物激活受体-γ共激活因子-1α(PGC1α)的蛋白质水平,和沉默调节蛋白1(SIRT1)。实时荧光定量聚合酶链反应(RT-qPCR)用于检测包括甲状腺激素受体(THRα,THRβ),单羧酸转运蛋白10(MCT10),线粒体DNA(mtDNA),和PGC1α。采用透射电子显微镜研究线粒体形态。(2)体外实验:涉及培养C2C12成肌细胞,使用慢病毒干扰Dio3表达,用脂多糖(LPS)处理细胞,构建内毒素细胞模型。C2C12细胞分为shNC组,shD3组,shNC+LPS组,和shD3+LPS组。进行免疫荧光共定位分析以确定PGC1α的细胞内分布。进行联合免疫共沉淀测定和Western印迹以评估PGC1α的乙酰化水平。
    结果:(1)体内实验:与shNC+Sham组相比,shNC+CLP组骨骼肌中Dio3蛋白的表达明显增加(Dio3/β-微管蛋白:3.32±0.70vs.1.00±0.49,P<0.05),然而,shD3+Sham组差别无统计学意义。相对于shNC+CLP组,shD3+CLP组中的Dio3表达显着降低(Dio3/β-微管蛋白:1.42±0.54vs.3.32±0.70,P<0.05)。与shNC+CLP组相比,shD3+CLP组T3调节基因表达恢复[THRαmRNA(2-ΔΔCt):0.67±0.05vs.0.33±0.01,THRβmRNA(2-ΔΔCt):0.94±0.05vs.0.67±0.02,MCT10mRNA(2-ΔΔCt):0.65±0.03vs.0.57±0.02,均P<0.05]。电镜形态学分析提示shNC+CLP组骨骼肌线粒体损伤明显,而shD3+CLP组表现出明显的改善。与shNC+Sham组相比,shNC+CLP组显着减少线粒体数量(细胞/HP:10.375±1.375vs.13.750±2.063,P<0.05),而shD3+CLP组与shNC+CLP组相比,线粒体数量显著增加(细胞/HP:11.250±2.063vs.10.375±1.375,P<0.05)。与shNC+Sham组相比,shNC+CLP组mtDNA的表达明显减少(拷贝:0.842±0.035vs.1.002±0.064,P<0.05)。尽管shD3+CLP组和shNC+CLP组之间的mtDNA表达没有检测到差异,但与shD3+Sham组相比,发现显着增加(拷贝:0.758±0.035vs.0.474±0.050,P<0.05)。在shD3+CLP组中,相对于shNCCLP组,PGC1α的表达在转录和蛋白质水平上均显着提高[PGC1αmRNA(2-ΔΔCt):1.49±0.13vs.0.68±0.06,PGC1α/β-微管蛋白:0.76±0.02vs.0.62±0.04,均P<0.05]。(2)体外实验:LPS处理C2C12细胞24小时后,PGC1α的细胞定位变得弥散;干扰Dio3表达促进了PGC1α向核周区域和细胞核的易位。此外,shD3+LPS组的乙酰化PGC1α水平明显低于shNC+LPS组(乙酰化PGC1α/β-微管蛋白:0.59±0.01vs.1.24±0.01,P<0.05),而去乙酰化剂SIRT1的表达在Dio3抑制后显著升高(SIRT1/β-微管蛋白:1.04±0.04vs.0.58±0.03,P<0.05)。当使用EX527抑制SIRT1活性时,与shD3LPS组相比,PGC1α蛋白表达显着降低(PGC1α/β-微管蛋白:0.92±0.03vs.1.58±0.03,P<0.05)。
    结论:抑制骨骼肌中的Dio3通过激活SIRT1减少PGC1α的乙酰化,促进PGC1α的核转位,从而提供针对脓毒症诱导的骨骼肌线粒体损伤的保护。
    OBJECTIVE: To investigate the protective effects and mechanisms of targeted inhibition of type 3 deiodinase (Dio3) on skeletal muscle mitochondria in sepsis.
    METHODS: (1) In vivo experiments: adeno-associated virus (AAV) was employed to specifically target Dio3 expression in the anterior tibial muscle of rats, and a septic rat model was generated using cecal ligation and puncture (CLP). The male Sprague-Dawley (SD) rats were divided into shNC+Sham group, shD3+Sham group, shNC+CLP group, and shD3+CLP group by random number table method, with 8 rats in each group. After CLP modeling, tibial samples were collected and Western blotting analysis was conducted to assess the protein levels of Dio3, peroxisome proliferator-activated receptor-γ coactivator-1α (PGC1α), and silence-regulatory protein 1 (SIRT1). Real-time fluorescence quantitative polymerase chain reaction (RT-qPCR) was utilized to examine mRNA expression of genes including thyroid hormone receptors (THRα, THRβ), monocarboxylate transporter 10 (MCT10), mitochondrial DNA (mtDNA), and PGC1α. Transmission electron microscopy was employed to investigate mitochondrial morphology. (2) In vitro experiments: involved culturing C2C12 myoblasts, interfering with Dio3 expression using lentivirus, and constructing an endotoxin cell model by treating cells with lipopolysaccharide (LPS). C2C12 cells were divided into shNC group, shD3 group, shNC+LPS group, and shD3+LPS group. Immunofluorescence colocalization analysis was performed to determine the intracellular distribution of PGC1α. Co-immunoprecipitation assay coupled with Western blotting was carried out to evaluate the acetylation level of PGC1α.
    RESULTS: (1) In vivo experiments: compared with the shNC+Sham group, the expression of Dio3 protein in skeletal muscle of the shNC+CLP group was significantly increased (Dio3/β-Tubulin: 3.32±0.70 vs. 1.00±0.49, P < 0.05), however, there was no significant difference in the shD3+Sham group. Dio3 expression in the shD3+CLP group was markedly reduced relative to the shNC+CLP group (Dio3/β-Tubulin: 1.42±0.54 vs. 3.32±0.70, P < 0.05). Compared with the shNC+CLP group, the expression of T3-regulated genes in the shD3+CLP group were restored [THRα mRNA (2-ΔΔCt): 0.67±0.05 vs. 0.33±0.01, THRβ mRNA (2-ΔΔCt): 0.94±0.05 vs. 0.67±0.02, MCT10 mRNA (2-ΔΔCt): 0.65±0.03 vs. 0.57±0.02, all P < 0.05]. Morphology analysis by electron microscopy suggested prominent mitochondrial damage in the skeletal muscle of the shNC+CLP group, while the shD3+CLP group exhibited a marked improvement. Compared with the shNC+Sham group, the shNC+CLP group significantly reduced the number of mitochondria (cells/HP: 10.375±1.375 vs. 13.750±2.063, P < 0.05), while the shD3+CLP group significantly increased the number of mitochondria compared to the shNC+CLP group (cells/HP: 11.250±2.063 vs. 10.375±1.375, P < 0.05). The expression of mtDNA in shNC+CLP group was markedly reduced compared with shNC+Sham group (copies: 0.842±0.035 vs. 1.002±0.064, P < 0.05). Although no difference was detected in the mtDNA expression between shD3+CLP group and shNC+CLP group, but significant increase was found when compared with the shD3+Sham group (copies: 0.758±0.035 vs. 0.474±0.050, P < 0.05). In the shD3+CLP group, PGC1α expression was significantly improved at both transcriptional and protein levels relative to the shNC+CLP group [PGC1α mRNA (2-ΔΔCt): 1.49±0.13 vs. 0.68±0.06, PGC1α/β-Tubulin: 0.76±0.02 vs. 0.62±0.04, both P < 0.05]. (2) In vitro experiments: post-24-hour LPS treatment of C2C12 cells, the cellular localization of PGC1α became diffuse; interference with Dio3 expression promoted PGC1α translocation to the perinuclear region and nucleus. Moreover, the acetylated PGC1α level in the shD3+LPS group was significantly lower than that in the shNC+LPS group (acetylated PGC1α/β-Tubulin: 0.59±0.01 vs. 1.24±0.01, P < 0.05), while the expression of the deacetylating agent SIRT1 was substantially elevated following Dio3 inhibition (SIRT1/β-Tubulin: 1.04±0.04 vs. 0.58±0.03, P < 0.05). When SIRT1 activity was inhibited by using EX527, PGC1α protein expression was notably decreased compared to the shD3+LPS group (PGC1α/β-Tubulin: 0.92±0.03 vs. 1.58±0.03, P < 0.05).
    CONCLUSIONS: Inhibition of Dio3 in skeletal muscle reduced the acetylation of PGC1α through activating SIRT1, facilitating nuclear translocation of PGC1α, thereby offering protection against sepsis-induced skeletal muscle mitochondrial damage.
    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
    在小鼠模型中检查了肺缺血再灌注损伤(LIRI)的复杂发病机理,重点研究焦亡的作用及其对肺损伤的加重。我们特别检查了肺内焦亡的水平和细胞定位,显示肺泡巨噬细胞为主要部位。VX-765对焦亡的抑制作用降低了肺损伤的严重程度,强调其在LIRI中的重要作用。此外,研究了β-羟基丁酸酯(β-OHB)在改善LIRI方面的治疗潜力。通过补充酮酯和3-羟基丁酸脱氢酶1(BDH-1)基因敲除评估β-OHB水平的调节,以及使用EX-527和pCMV-SIRT1质粒转染操作SIRT1-FOXO3信号通路。这表明β-OHB具有肺保护和抗发热作用,通过SIRT1的上调和FOXO3脱乙酰的增强介导,导致焦凋亡标志物降低和肺损伤。此外,β-OHB在体外处理MH-S细胞显示浓度依赖性的改善,将其治疗益处与特定的细胞机制联系起来。总的来说,这项研究强调了肺泡巨噬细胞焦亡在LIRI恶化中的意义,并表明β-OHB通过调节SIRT1-FOXO3信号通路减轻损伤的潜力.
    The complex pathogenesis of lung ischemia-reperfusion injury (LIRI) was examined in a murine model, focusing on the role of pyroptosis and its exacerbation of lung injury. We specifically examined the levels and cellular localization of pyroptosis within the lung, which revealed alveolar macrophages as the primary site. The inhibition of pyroptosis by VX-765 reduced the severity of lung injury, underscoring its significant role in LIRI. Furthermore, the therapeutic potential of β-hydroxybutyrate (β-OHB) in ameliorating LIRI was examined. Modulation of β-OHB levels was evaluated by ketone ester supplementation and 3-hydroxybutyrate dehydrogenase 1 (BDH-1) gene knockout, along with the manipulation of the SIRT1-FOXO3 signaling pathway using EX-527 and pCMV-SIRT1 plasmid transfection. This revealed that β-OHB exerts lung-protective and anti-pyroptotic effects, which were mediated through the upregulation of SIRT1 and the enhancement of FOXO3 deacetylation, leading to decreased pyroptosis markers and lung injury. In addition, β-OHB treatment of MH-S cells in vitro showed a concentration-dependent improvement in pyroptosis, linking its therapeutic benefits to specific cell mechanisms. Overall, this study highlights the significance of alveolar macrophage pyroptosis in the exacerbation of LIRI and indicates the potential of β-OHB in mitigating injury by modulating the SIRT1-FOXO3 signaling pathway.
    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NAD+在静止条件下连续降解和合成。在哺乳动物中,NAD+的合成主要是由烟酰胺(Nam)启动的Nam磷酸核糖基转移酶,而聚(ADP-核糖)聚合酶1(PARP1)和2(PARP2),sirtuin1(SIRT1),CD38和含有1(SARM1)的无菌α和TIR基序参与NAD+分解。使用2H标记的Nam的通量分析,我们发现,当哺乳动物细胞在没有Nam的情况下培养时,细胞NAD+水平得以维持,NAD+分解被完全抑制。在Nam面前,NAD+分解率(RB)在PARP1、PARP2、SIRT1或SARM1缺失时没有显著变化,而CD38的稳定表达并没有增加RB。然而,与野生型细胞相比,PARP1缺失细胞中的RB高得多,其中PARP1活性被选择性抑制剂阻断。相比之下,与对照细胞相比,在特异性CD38抑制剂存在下,CD38过表达细胞中的RB要低得多。结果表明,PARP1缺失上调了其他NAD酶的活性,而CD38表达下调内源性NAD酶的活性,包括PARP1和PARP2。细胞NAD+分解的速率和产生的NAD+浓度可以保持在恒定水平。尽管NAD+降解酶表达发生变化,通过对NAD酶活性的补偿调节。
    Cellular NAD+ is continuously degraded and synthesized under resting conditions. In mammals, NAD+ synthesis is primarily initiated from nicotinamide (Nam) by Nam phosphoribosyltransferase, whereas poly(ADP-ribose) polymerase 1 (PARP1) and 2 (PARP2), sirtuin1 (SIRT1), CD38, and sterile alpha and TIR motif containing 1 (SARM1) are involved in NAD+ breakdown. Using flux analysis with 2H-labeled Nam, we found that when mammalian cells were cultured in the absence of Nam, cellular NAD+ levels were maintained and NAD+ breakdown was completely suppressed. In the presence of Nam, the rate of NAD+ breakdown (RB) did not significantly change upon PARP1, PARP2, SIRT1, or SARM1 deletion, whereas stable expression of CD38 did not increase RB. However, RB in PARP1-deleted cells was much higher compared with that in wild-type cells, in which PARP1 activity was blocked with a selective inhibitor. In contrast, RB in CD38-overexpressing cells in the presence of a specific CD38 inhibitor was much lower compared with that in control cells. The results indicate that PARP1 deletion upregulates the activity of other NADases, whereas CD38 expression downregulates the activity of endogenous NADases, including PARP1 and PARP2. The rate of cellular NAD+ breakdown and the resulting NAD+ concentration may be maintained at a constant level, despite changes in the NAD+-degrading enzyme expression, through the compensatory regulation of NADase activity.
    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
    非典型畸胎样/横纹肌样瘤(ATRT)是中枢神经系统的高度恶性胚胎性肿瘤,预后不良。使用新开发和验证的患者来源的ATRT培养和异种移植模型,除了一组主要的ATRT模型,我们发现ATRT对核苷类似物吉西他滨具有选择性敏感性.基因表达和蛋白质分析表明,吉西他滨治疗导致沉默调节蛋白1(SIRT1)降解,通过激活核因子κB(NF-κB)和p53导致细胞死亡。此外,我们发现,吉西他滨诱导的SIRT1缺失导致Sonichedgehog(SHH)信号传导激活因子GLI2的核转至细胞质,这解释了在SHH亚型ATRT中观察到的吉西他滨额外敏感性.在两个独立的患者来源的异种移植物模型中,用吉西他滨处理带有ATRT异种移植物的小鼠导致中位存活增加>30%并产生长期存活者。这些发现表明ATRTs对吉西他滨治疗高度敏感,可能成为ATRTs未来多模式治疗策略的一部分。
    Atypical teratoid/rhabdoid tumors (ATRTs) are highly malignant embryonal tumors of the central nervous system with a dismal prognosis. Using a newly developed and validated patient-derived ATRT culture and xenograft model, alongside a panel of primary ATRT models, we found that ATRTs are selectively sensitive to the nucleoside analog gemcitabine. Gene expression and protein analyses indicate that gemcitabine treatment causes the degradation of sirtuin 1 (SIRT1), resulting in cell death through activation of nuclear factor κB (NF-κB) and p53. Furthermore, we discovered that gemcitabine-induced loss of SIRT1 results in a nucleus-to-cytoplasm translocation of the sonic hedgehog (SHH) signaling activator GLI2, explaining the observed additional gemcitabine sensitivity in SHH-subtype ATRT. Treatment of ATRT xenograft-bearing mice with gemcitabine resulted in a >30% increase in median survival and yielded long-term survivors in two independent patient-derived xenograft models. These findings demonstrate that ATRTs are highly sensitive to gemcitabine treatment and may form part of a future multimodal treatment strategy for ATRTs.
    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
    肌肉减少症,由肌肉生长和损失之间的不平衡引起的疾病,严重影响老年代谢患者的生活质量,炎症,和癌症疾病。Vigeo,三种植物的努鲁克发酵提取物(马克西姆刺五加(ESM),牛膝(Miq。)Nakai(AJN),据报道,白术(AJK)具有抗骨质疏松作用。然而,没有证据证明Vigeo对肌肉萎缩的影响。这里,在地塞米松(Dex)诱导的肌肉萎缩的体内模型中,Vigeo治疗显着逆转Dex诱导的小腿肌肉体积减少,腓肠肌(GA)肌肉重量,和组织学横截面积。此外,在从GA肌肉分离的mRNA和蛋白质分析中,我们观察到Vigeo通过抑制由atrogin和MuRF-1调节的蛋白质降解,显著保护Dex诱导的小鼠肌肉萎缩.此外,我们证明Vigeo显著促进C2C12细胞系分化,肌管的宽度和长度增加证明了这一点,以及具有三个或更多细胞核的融合肌管的数量增加。与对照组相比,Vigeo减轻了肌管的形成。Vigeo还显着增加了肌球蛋白重链(MyHC)的mRNA和蛋白质表达,MyoD,和肌细胞生成素与对照组相比。Vigeo处理在体外显著降低了C2C12细胞系中肌肉降解标志物atrogin-1和肌肉RING指1(MuRF-1)的mRNA和蛋白表达。Vigeo还激活了AMP激活的蛋白激酶(AMPK)/沉默信息调节因子1(Sirt-1)/过氧化物酶体增殖物激活的受体-γ共激活因子-1α(PGC1α)线粒体生物发生途径和Akt/mTOR蛋白合成信号通路Dex诱导的肌管萎缩。这些发现表明Vigeo可能对Dex诱导的肌肉萎缩具有保护作用。因此,我们建议Vigeo作为一种补充剂或潜在的治疗药物来预防或治疗伴有肌肉萎缩和变性的肌肉减少症。
    Sarcopenia, a condition caused by an imbalance between muscle growth and loss, can severely affect the quality of life of elderly patients with metabolic, inflammatory, and cancer diseases. Vigeo, a nuruk-fermented extract of three plants (Eleutherococcus senticosus Maxim (ESM), Achyranthes japonica (Miq.) Nakai (AJN), and Atractylodes japonica Koidzumi (AJK)) has been reported to have anti-osteoporotic effects. However, evidence of the effects of Vigeo on muscle atrophy is not available. Here, in the in vivo model of dexamethasone (Dex)-induced muscle atrophy, Vigeo treatment significantly reversed Dex-induced decreases in calf muscle volume, gastrocnemius (GA) muscle weight, and histological cross-section area. In addition, in mRNA and protein analyses isolated from GA muscle, we observed that Vigeo significantly protected against Dex-induced mouse muscle atrophy by inhibiting protein degradation regulated by atrogin and MuRF-1. Moreover, we demonstrated that Vigeo significantly promoted C2C12 cell line differentiation, as evidenced by the increased width and length of myotubes, and the increased number of fused myotubes with three or more nuclei. Vigeo alleviated the formation of myotubes compared to the control group. Vigeo also significantly increased the mRNA and protein expression of myosin heavy chain (MyHC), MyoD, and myogenin compared to that in the control. Vigeo treatment significantly reduced the mRNA and protein expression of muscle degradation markers atrogin-1 and muscle RING Finger 1 (MuRF-1) in the C2C12 cell line in vitro. Vigeo also activated the AMP-activated protein kinase (AMPK)/silent information regulator 1 (Sirt-1)/peroxisome proliferator-activated receptor-γ co-activator-1α (PGC1α) mitochondrial biogenesis pathway and the Akt/mTOR protein synthesis signaling pathway in Dex-induced myotube atrophy. These findings suggest that Vigeo may have protective effects against Dex-induced muscle atrophy. Therefore, we propose Vigeo as a supplement or potential therapeutic agent to prevent or treat sarcopenia accompanied by muscle atrophy and degeneration.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号