Peroxiredoxin III

过氧化物酶 III
  • 文章类型: Journal Article
    DNA复制过程中的核小体组装依赖于组蛋白伴侣。最近的研究表明,失调的组蛋白伴侣有助于癌症进展,包括胃癌(GC)。需要进一步的研究来探索组蛋白伴侣的预后和治疗意义及其在GC进展中的作用机制。在这里,我们确定组蛋白伴侣ASF1B作为GC增殖和预后的潜在生物标志物。ASF1B在GC中显著上调,这与不良预后有关。体外和体内实验表明,抑制ASF1B抑制GC的恶性特征,而过表达ASF1B则有相反的作用。机械上,转录因子FOXM1直接与ASF1B启动子区结合,从而调节其转录。用硫链菌素治疗,FOXM1抑制剂,不仅抑制了ASF1B的表达,但也抑制GC进展。此外,ASF1B以FOXM1依赖性方式调节线粒体蛋白过氧化物氧还蛋白3(PRDX3)转录。还阐明了ASF1B调节的PRDX3在GC细胞增殖和氧化应激平衡中的关键作用。总之,我们的研究提示FOXM1-ASF1B-PRDX3轴是治疗GC的潜在治疗靶点.
    Nucleosome assembly during DNA replication is dependent on histone chaperones. Recent studies suggest that dysregulated histone chaperones contribute to cancer progression, including gastric cancer (GC). Further studies are required to explore the prognostic and therapeutic implications of histone chaperones and their mechanisms of action in GC progression. Here we identified histone chaperone ASF1B as a potential biomarker for GC proliferation and prognosis. ASF1B was significantly upregulated in GC, which was associated with poor prognosis. In vitro and in vivo experiments demonstrated that the inhibition of ASF1B suppressed the malignant characteristics of GC, while overexpression of ASF1B had the opposite effect. Mechanistically, transcription factor FOXM1 directly bound to the ASF1B-promoter region, thereby regulating its transcription. Treatment with thiostrepton, a FOXM1 inhibitor, not only suppressed ASF1B expression, but also inhibited GC progression. Furthermore, ASF1B regulated the mitochondrial protein peroxiredoxin 3 (PRDX3) transcription in a FOXM1-dependent manner. The crucial role of ASF1B-regulated PRDX3 in GC cell proliferation and oxidative stress balance was also elucidated. In summary, our study suggests that the FOXM1-ASF1B-PRDX3 axis is a potential therapeutic target for treating GC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    硫氧还蛋白还原酶2(Txnrd2)属于硒蛋白的硫氧还蛋白还原酶家族,是哺乳动物细胞中调节氧化还原稳态的关键抗氧化酶。这里,我们报道,Txnrd2通过抑制内质网(ER)应激氧化应激并通过Trx2/Prx3途径对脑出血(ICH)引起的脑损伤产生重要影响.此外,我们证明,药理硒(Se)通过增强Txnrd2表达来挽救ICH后的脑损伤。首先,在胶原酶IV诱导的ICH模型中确定Txnrd2,Trx2和Prx3的表达和定位。然后在发现发展更严重的脑水肿和神经缺陷的大鼠中使用siRNA干扰击倒Txnrd2。机械上,我们观察到Txnrd2的缺失导致神经元和星形胶质细胞的脂质过氧化水平和ER应激蛋白表达增加。此外,结果表明,硒在侧脑室给药亚硒酸钠时,通过促进Trx2/Prx3kilter,有效恢复了脑中Txnrd2的表达,并抑制了ER应激蛋白活性和活性氧(ROS)的产生。这项研究揭示了Txnrd2通过Trx2/Prx3途径调节ICH氧化应激和ER应激的作用及其作为ICH治疗靶标的潜力。
    Thioredoxin-reductase 2 (Txnrd2) belongs to the thioredoxin-reductase family of selenoproteins and is a key antioxidant enzyme in mammalian cells to regulate redox homeostasis. Here, we reported that Txnrd2 exerted a major influence in brain damage caused by Intracerebral hemorrhage (ICH) by suppressing endoplasmic reticulum (ER) stress oxidative stress and via Trx2/Prx3 pathway. Furthermore, we demonstrated that pharmacological selenium (Se) rescued the brain damage after ICH by enhancing Txnrd2 expression. Primarily, expression and localization of Txnrd2, Trx2 and Prx3 were determined in collagenase IV-induced ICH model. Txnrd2 was then knocked down using siRNA interference in rats which were found to develop more severe encephaledema and neurological deficits. Mechanistically, we observed that loss of Txnrd2 leads to increased lipid peroxidation levels and ER stress protein expression in neurons and astrocytes. Additionally, it was revealed that Se effectively restored the expression of Txnrd2 in brain and inhibited both the activity of ER stress protein activity and the generation of reactive oxygen species (ROS) by promoting Trx2/Prx3 kilter when administrating sodium selenite in lateral ventricle. This study shed light on the effect of Txnrd2 in regulating oxidative stress and ER stress via Trx2/Prx3 pathway upon ICH and its promising potential as an ICH therapeutic target.
    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
    急性肺损伤(ALI)是脓毒症危及生命的并发症之一。巨噬细胞极化在脓毒症相关ALI中起着至关重要的作用。然而,ALI和炎症发展中巨噬细胞极化的调节机制尚不清楚.在这项研究中,我们证明,巨噬细胞极化发生在脓毒症相关的ALI中,并伴有线粒体功能障碍和炎症,PRDX3的减少促进巨噬细胞极化和线粒体功能障碍的启动。机械上,PRDX3过表达促进M1巨噬细胞向M2巨噬细胞分化,并通过降低糖酵解水平和增加TCA循环活性来增强损伤后线粒体功能恢复。总之,我们确定PRDX3是整合氧化应激的关键枢纽,炎症,和巨噬细胞极化中的代谢重编程。这些发现说明了巨噬细胞极化和脓毒症相关ALI之间联系的适应性机制。
    Acute lung injury (ALI) is one of the life-threatening complications of sepsis, and macrophage polarization plays a crucial role in the sepsis-associated ALI. However, the regulatory mechanisms of macrophage polarization in ALI and in the development of inflammation are largely unknown. In this study, we demonstrated that macrophage polarization occurs in sepsis-associated ALI and is accompanied by mitochondrial dysfunction and inflammation, and a decrease of PRDX3 promotes the initiation of macrophage polarization and mitochondrial dysfunction. Mechanistically, PRDX3 overexpression promotes M1 macrophages to differentiate into M2 macrophages, and enhances mitochondrial functional recovery after injury by reducing the level of glycolysis and increasing TCA cycle activity. In conclusion, we identified PRDX3 as a critical hub integrating oxidative stress, inflammation, and metabolic reprogramming in macrophage polarization. The findings illustrate an adaptive mechanism underlying the link between macrophage polarization and sepsis-associated ALI.
    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:心肌缺血/再灌注损伤(MIRI)严重威胁着人们的健康。心肌细胞线粒体功能障碍可促进MIRI的进展。右美托咪定(Dex)可减轻心肌损伤,已知可以逆转肺损伤中的线粒体功能障碍。然而,Dex在MIRI期间线粒体功能障碍中的功能尚不清楚.
    目的:评估Dex在MIRI期间线粒体功能障碍中的功能。
    方法:为了研究Dex在MIRI中的功能,将H9C2细胞置于缺氧/复氧(H/R)条件下。进行CCK8测定以测试细胞活力,并通过JC-1染色评估线粒体膜电位。此外,通过Co-IP分析探索Sirt3和Prdx3之间的结合关系。此外,蛋白质表达采用蛋白质印迹法检测。
    结果:Dex可以消除H/R诱导的H9C2细胞线粒体功能障碍。此外,H/R处理显著抑制了Sirt3的表达,而Dex部分恢复了这一现象。敲除Sirt3或Prdx3可明显降低Dex对H/R诱导的线粒体损伤的保护作用。同时,Sirt3可通过Prdx3的脱乙酰作用增强Prdx3的功能。
    结论:发现Dex通过激活Sirt3/Prdx3通路减轻H/R诱导的心肌细胞线粒体功能障碍。因此,这项研究可能为探索MIRI治疗的新策略提供新的思路.
    BACKGROUND: Myocardial ischemia/reperfusion injury (MIRI) seriously threatens the health of people. The mitochondrial dysfunction in cardiomyocytes can promote the progression of MIRI. Dexmedetomidine (Dex) could alleviate the myocardial injury, which was known to reverse mitochondrial dysfunction in lung injury. However, the function of Dex in mitochondrial dysfunction during MIRI remains unclear.
    OBJECTIVE: To assess the function of Dex in mitochondrial dysfunction during MIRI.
    METHODS: To investigate the function of Dex in MIRI, H9C2 cells were placed in condition of hypoxia/reoxygenation (H/R). CCK8 assay was performed to test the cell viability, and the mitochondrial membrane potential was evaluated by JC-1 staining. In addition, the binding relationship between Sirt3 and Prdx3 was explored by Co-IP assay. Furthermore, the protein expressions were examined using western blot.
    RESULTS: Dex could abolish H/R-induced mitochondrial dysfunction in H9C2 cells. In addition, H/R treatment significantly inhibited the expression of Sirt3, while Dex partially restored this phenomenon. Knockdown of Sirt3 or Prdx3 obviously reduced the protective effect of Dex on H/R-induced mitochondrial injury. Meanwhile, Sirt3 could enhance the function of Prdx3 via deacetylation of Prdx3.
    CONCLUSIONS: Dex was found to attenuate H/R-induced mitochondrial dysfunction in cardiomyocytes via activation of Sirt3/Prdx3 pathway. Thus, this study might shed new lights on exploring new strategies for the treatment of MIRI.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Letter
    暂无摘要。
    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
    奥希替尼耐药被认为是限制接受表皮生长因子受体(EGFR)突变的非小细胞肺癌(NSCLC)治疗的患者生存益处的主要障碍。然而,获得性耐药的潜在机制尚不清楚.在这项研究中,我们报道,雌激素受体β(ERβ)在奥希替尼耐药的NSCLC中高表达,并在促进奥希替尼耐药中发挥关键作用.我们进一步确定泛素特异性蛋白酶7(USP7)是去泛素化和上调NSCLC中ERβ的关键结合伴侣。ERβ通过减轻活性氧(ROS)积累促进奥希替尼耐药。我们发现ERβ在机制上抑制了过氧化物酶3(PRDX3)去氧化,从而赋予NSCLC奥希替尼耐药性。此外,我们提供的证据表明,ERβ的耗竭在体外和体内均能诱导NSCLC中的ROS积累并逆转奥希替尼耐药.因此,我们的结果表明,USP7介导的ERβ稳定抑制PRDX3SUMO化,以减轻ROS积累并促进奥希替尼耐药,提示靶向ERβ可能是克服NSCLC奥希替尼耐药的有效治疗策略.
    Osimertinib resistance is regarded as a major obstacle limiting survival benefits for patients undergoing treatment of epidermal growth factor receptor (EGFR)-mutant non-small cell lung cancer (NSCLC). However, the underlying mechanisms of acquired resistance remain unclear. In this study, we report that estrogen receptor β (ERβ) is highly expressed in osimertinib-resistant NSCLC and plays a pivotal role in promoting osimertinib resistance. We further identified ubiquitin-specific protease 7 (USP7) as a critical binding partner that deubiquitinates and upregulates ERβ in NSCLC. ERβ promotes osimertinib resistance by mitigating reactive oxygen species (ROS) accumulation. We found that ERβ mechanistically suppresses peroxiredoxin 3 (PRDX3) SUMOylation and thus confers osimertinib resistance onto NSCLC. Furthermore, we provide evidence showing that depletion of ERβ induces ROS accumulation and reverses osimertinib resistance in NSCLC both in vitro and in vivo. Thus, our results demonstrate that USP7-mediated ERβ stabilization suppresses PRDX3 SUMOylation to mitigate ROS accumulation and promote osimertinib resistance, suggesting that targeting ERβ may be an effective therapeutic strategy to overcome osimertinib resistance in NSCLC.
    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:甲醛(FA)与白血病的发生有关,氧化应激被认为是一个主要原因。作为氧化应激的内源性生物标志物,很少有研究关注过氧化物氧化还原蛋白III(PrxIII)与FA毒性之间的关系。我们前期的研讨不雅察到PrxIII的高表达在FA引诱的骨髓细胞(BMCs)凋亡进程中,然而,确切的机制尚不清楚。因此,本文旨在探讨FA毒性与PrxIII基因的可能关联。
    方法:我们首先,使用细胞计数试剂盒-8(CCK-8)检测BMC暴露于不同剂量的FA(50、100、200μmol/L)不同暴露时间(12、24、48h)后的活力,然后选择24小时作为暴露时间,通过定量逆转录PCR(qRT-PCR)和Westernblot分析检测暴露不同剂量FA的PrxIII的表达。根据我们的初步实验结果,我们选择100μmol/LFA作为暴露剂量,暴露24小时,并使用小干扰RNA(siRNA)沉默PrxIII,通过CCK-8检查细胞活力,通过DCFH-DA检查活性氧(ROS)水平,通过膜联蛋白V/PI双重染色和流式细胞术(FCM)检测细胞周期,以探讨PrxIII沉默对FA诱导的骨髓毒性的可能调节作用。
    结果:PrxIII的高表达发生在FA诱导的氧化应激过程中。沉默PrxIII可防止FA诱导氧化应激,从而增加细胞活力,降低ROS水平,拯救G0-G1和G2-M逮捕,减少细胞凋亡。
    结论:PrxIII沉默可能是减轻FA诱导的氧化损伤的潜在目标。
    BACKGROUND: Formaldehyde (FA) is associated with the occurrence of leukemia, and oxidative stress is considered to be a major reason. As an endogenous biomarker of oxidative stress, few studies focus on the relationship between peroxiredoxin III (PrxIII) and FA toxicity. Our previous research observed high expression of PrxIII occurred in the process of apoptosis of bone marrow cells (BMCs) induced by FA, however the exact mechanism is unclear. Therefore, this paper aimed to explore the possible association between FA toxicity and PrxIII gene.
    METHODS: We first, used a Cell Counting Kit-8 (CCK-8) to detect the viability of BMCs after they were exposed to different doses of FA (50, 100, 200 μmol/L) for different exposure time (12, 24, 48 h), then chose 24 h as an exposure time to detect the expression of PrxIII for exposing different doses of FA by Quantitative reverse transcription-PCR (qRT-PCR) and Western blot analysis. Based on our preliminary experimental results, we chose 100 μmol/L FA as an exposure dose to expose for 24 h, and used a small interfering RNA (siRNA) to silenced PrxIII to examine the cell viability by CCK-8, reactive oxygen species (ROS) level by DCFH-DA, apoptosis by Annexin V/PI double staining and cell cycle by flow cytometry (FCM) so as to explore the possible regulatory effect of PrxIII silencing on FA-induced bone marrow toxicity.
    RESULTS: High expression of PrxIII occurred in the process of FA-induced oxidative stress. Silencing of PrxIII prevented FA from inducing oxidative stress, thus increasing cell viability, decreasing ROS level, rescuing G0 -G1 and G2 -M arrest, and reducing cell apoptosis.
    CONCLUSIONS: PrxIII silencing might be a potential target for alleviating FA-induced oxidative 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: Letter
    暂无摘要。
    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:尽管已经报道了环状RNA(circularRNAs,circRNAs)与多种病理状况进展的关系,骨关节炎(OA)中的circRNA参与者几乎没有研究。
    方法:在本研究中,我们招募了25例接受关节成形术的OA患者进行软骨组织收集.检索来自基因表达Omnibus的公共circRNA微阵列数据用于circRNA鉴定。通过用IL-1β处理人软骨细胞(CHON-001细胞系),构建了OA相关损伤的体外细胞模型,用circSOD2siRNA沉默circSOD2表达,研究其在细胞凋亡中的作用,炎症反应,和细胞外基质(ECM)降解。此外,我们调查了circSOD2,miR-224-5p,和过氧化物酶3(PRDX3)通过荧光素酶报告基因测定,RNA免疫沉淀测定,和定量逆转录聚合酶链反应。
    结果:我们的发现揭示了circSOD2在OA软骨和细胞样品中的过度表达,和circSOD2敲除减轻ECM降解,炎症,CHON-001细胞模型中的凋亡。此外,我们的研究结果表明circSOD2敲低对miR-224-5p表达的调节功能,而miR-224-5p能够下调PRDX3表达。miR-224-5p抑制剂或pcDNA-PRDX3的共转染可以防止circSOD2敲低的作用。
    结论:因此,我们的结果表明,circSOD2敲低可作为通过调节miR-224-5p/PRDX3信号轴缓解OA进展的干预策略.
    BACKGROUND: Although the implications of circular RNAs (circRNAs) with the progression of diverse pathological conditions have been reported, the circRNA players in osteoarthritis (OA) are barely studied.
    METHODS: In this study, twenty-five OA patients who received arthroplasty were recruited for cartilage tissue collection. Public circRNA microarray data from Gene Expression Omnibus was retrieved for circRNA identification. An in vitro cell model of OA-related damages was constructed by treating human chondrocytes (CHON-001 cell line) with IL-1β, and circSOD2 siRNA was used to silence circSOD2 expression to study its functional role in apoptosis, inflammatory responses, and extracellular matrix (ECM) degradation. Besides, we investigated the functional interactions among circSOD2, miR-224-5p, and peroxiredoxin 3 (PRDX3) by luciferase reporter assay, RNA-immunoprecipitation assay, and quantitative reverse transcription polymerase chain reaction.
    RESULTS: Our findings revealed the overexpression of circSOD2 in the OA cartilage and cell samples, and circSOD2 knockdown alleviated ECM degradation, inflammation, and apoptosis in CHON-001 cell model. In addition, our findings suggested the regulatory function of circSOD2 knockdown on miR-224-5p expression, while miR-224-5p was capable of downregulating PRDX3 expression. The co-transfection of miR-224-5p inhibitor or pcDNA-PRDX3 could prevent the effect of circSOD2 knockdown.
    CONCLUSIONS: Hence, our results demonstrated that knockdown of circSOD2 may serve as an intervention strategy to alleviate OA progression through modulating miR-224-5p/PRDX3 signaling axis.
    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:突触后密度(PSD)是一种电子致密结构,包含各种支架和信号蛋白。Shank1是位于谷氨酸能突触的突触支架的主要调节因子,并被认为与多种神经系统疾病有关。
    方法:在本研究中,我们研究了shank1在神经元SN4741细胞中6-OHDA治疗模拟的体外帕金森病(PD)模型中的作用。用westernblot和免疫染色检测相关分子的表达。
    结果:我们发现6-OHDA在SN4741细胞中显著增加shank1的mRNA和蛋白水平,但是亚细胞分布没有改变。通过小干扰RNA(siRNA)敲除shank1,防止6-OHDA处理,乳酸脱氢酶(LDH)释放减少和细胞凋亡减少。RT-PCR和Westernblot结果表明,敲低shank1可明显抑制6-OHDA暴露后内质网(ER)应激相关因子的激活。此外,shank1的下调使PRDX3的表达明显增加,并伴随着线粒体功能的保留。机械上,通过siRNA下调PRDX3部分阻止了shank1敲低诱导的SN4741细胞对6-OHDA的保护作用。
    结论:总之,本研究提供了第一个证据,证明敲低shank1通过激活PRDX3通路保护6-OHDA诱导的内质网应激和线粒体功能障碍.
    Postsynaptic density (PSD) is an electron-dense structure that contains various scaffolding and signaling proteins. Shank1 is a master regulator of the synaptic scaffold located at glutamatergic synapses, and has been proposed to be involved in multiple neurological disorders.
    In this study, we investigated the role of shank1 in an in vitro Parkinson\'s disease (PD) model mimicked by 6-OHDA treatment in neuronal SN4741 cells. The expression of related molecules was detected by western blot and immunostaining.
    We found that 6-OHDA significantly increased the mRNA and protein levels of shank1 in SN4741 cells, but the subcellular distribution was not altered. Knockdown of shank1 via small interfering RNA (siRNA) protected against 6-OHDA treatment, as evidenced by reduced lactate dehydrogenase (LDH) release and decreased apoptosis. The results of RT-PCR and western blot showed that knockdown of shank1 markedly inhibited the activation of endoplasmic reticulum (ER) stress associated factors after 6-OHDA exposure. In addition, the downregulation of shank1 obviously increased the expression of PRDX3, which was accompanied by the preservation of mitochondrial function. Mechanically, downregulation of PRDX3 via siRNA partially prevented the shank1 knockdowninduced protection against 6-OHDA in SN4741 cells.
    In summary, the present study has provided the first evidence that the knockdown of shank1 protects against 6-OHDA-induced ER stress and mitochondrial dysfunction through activating the PRDX3 pathway.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号