Prdx3

PRDX3
  • 文章类型: Journal Article
    目的:过氧化物酶3(Prdx3)是一种细胞内抗氧化酶,特异性定位于线粒体中,通过去除线粒体活性氧(ROS)来抵抗氧化应激。肠上皮提供物理和生化屏障,其将宿主组织与共生细菌隔离以维持肠稳态。细胞抗氧化防御系统和氧化应激之间的不平衡与炎症性肠病(IBD)的发病机理有关。然而,Prdx3在肠道炎症下肠上皮中的作用尚未阐明.探讨Prdx3在肠道炎症中的潜在作用。我们使用肠上皮细胞(IEC)特异性Prdx3基因敲除小鼠。
    结果:IEC特异性Prdx3缺陷小鼠表现出更严重的结肠炎表型,体重减轻程度更高,结肠缩短,屏障破坏,线粒体损伤,和IECs中的ROS生成。此外,在Prdx3敲低的结肠上皮细胞中,外泌体miR-1260b显着增加。机械上,Prdx3缺乏通过P38-MAPK/NFκB信号传导促进肠屏障破坏和炎症。
    方法:这是首次报道使用IEC特异性条件敲除小鼠的急性结肠炎中Prdx3的保护作用的研究。
    结论:我们的研究揭示了外泌体负载miRNA的作用,特别是miR-1260b,在IBD。靶向miR-1260b或调节外泌体介导的细胞间通讯可能有望成为治疗IBD和恢复肠屏障完整性的潜在治疗策略.
    Aims: Peroxiredoxin3 (Prdx3) is an intracellular antioxidant enzyme that is specifically localized in mitochondria and protects against oxidative stress by removing mitochondrial reactive oxygen species (ROS). The intestinal epithelium provides a physical and biochemical barrier that segregates host tissues from commensal bacteria to maintain intestinal homeostasis. An imbalance between the cellular antioxidant defense system and oxidative stress has been implicated in the pathogenesis of inflammatory bowel disease (IBD). However, the role of Prdx3 in the intestinal epithelium under intestinal inflammation has not been elucidated. To investigate the potential role of Prdx3 in intestinal inflammation, we used intestinal epithelial cell (IEC)-specific Prdx3-knockout mice. Results: IEC-specific Prdx3-deficient mice showed more severe colitis phenotypes with greater degrees of body weight loss, colon shortening, barrier disruption, mitochondrial damage, and ROS generation in IECs. Furthermore, exosomal miR-1260b was dramatically increased in Prdx3-knockdown colonic epithelial cells. Mechanistically, Prdx3 deficiency promoted intestinal barrier disruption and inflammation via P38-mitogen-activated protein kinase/NFκB signaling. Innovation: This is the first study to report the protective role of Prdx3 in acute colitis using IEC-specific conditional knockout mice. Conclusion: Our study sheds light on the role of exosome-loaded miRNAs, particularly miR-1260b, in IBD. Targeting miR-1260b or modulating exosome-mediated intercellular communication may hold promise as potential therapeutic strategies for managing IBD and restoring intestinal barrier integrity.
    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
    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
    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
    铁死亡的可靠和特异性标记的缺乏阻碍了与该细胞死亡机制相关的治疗向临床应用的进展。最近发表在《分子细胞》杂志上的一项研究发现,过氧化过氧化物氧还蛋白3(PRDX3)是铁凋亡的一个有希望的标记,为疾病治疗中监测和靶向铁性凋亡开辟了新的途径。
    The lack of a reliable and specific marker for ferroptosis has hindered the advancement of treatments related to this cell death mechanism toward clinical application. A recent study published in Molecular Cell has identified hyperoxidized peroxiredoxin 3 (PRDX3) as a promising marker for ferroptosis, opening up new avenues for monitoring and targeting ferroptosis in disease treatment.
    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
    Ferroptosis,由磷脂过氧化物积累驱动的调节细胞死亡途径,由于缺乏特定的标记,在生理条件下进行鉴定一直具有挑战性。这里,我们在体外和体内都确定了高氧化过氧化物氧还蛋白3(PRDX3)作为铁凋亡的标志物。在铁死亡期间,线粒体脂质过氧化物引发PRDX3过度氧化,将Cys硫醇转化为亚磺酸或磺酸的翻译后修饰。一旦过度氧化,PRDX3从线粒体转移到质膜,抑制胱氨酸的摄取,从而导致铁中毒。应用高氧化PRDX3作为标记,我们确定,在酒精性和非酒精性脂肪性肝病小鼠模型中,铁死亡是肝细胞死亡的原因,最常见的慢性肝脏疾病。我们的研究强调了铁性凋亡在病理生理条件下的重要性,并开辟了用抑制铁性凋亡的药物治疗这些肝脏疾病的可能性。
    Ferroptosis, a regulated cell death pathway driven by accumulation of phospholipid peroxides, has been challenging to identify in physiological conditions owing to the lack of a specific marker. Here, we identify hyperoxidized peroxiredoxin 3 (PRDX3) as a marker for ferroptosis both in vitro and in vivo. During ferroptosis, mitochondrial lipid peroxides trigger PRDX3 hyperoxidation, a posttranslational modification that converts a Cys thiol to sulfinic or sulfonic acid. Once hyperoxidized, PRDX3 translocates from mitochondria to plasma membranes, where it inhibits cystine uptake, thereby causing ferroptosis. Applying hyperoxidized PRDX3 as a marker, we determined that ferroptosis is responsible for death of hepatocytes in mouse models of both alcoholic and nonalcoholic fatty liver diseases, the most prevalent chronic liver disorders. Our study highlights the importance of ferroptosis in pathophysiological conditions and opens the possibility to treat these liver diseases with drugs that inhibit ferroptosis.
    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
    目的:线粒体功能障碍是肝脏缺血/再灌注(I/R)损伤的主要机制。赖氨酸去琥珀酰酶5(SIRT5)是线粒体琥珀酰基因组的全局调节因子,在线粒体代谢和功能中起着关键作用;然而,其在肝脏I/R中的保肝能力尚不清楚。在这项研究中,我们在SIRT5沉默和SIRT5过表达小鼠中建立了肝脏I/R模型,以研究SIRT5在肝脏I/R损伤中的作用和确切机制。结果:I/R期间肝线粒体中琥珀酰化强烈富集,并抑制线粒体琥珀酰化可显着减轻肝脏I/R损伤。重要的是,肝脏移植患者的脱琥珀酶SIRT5水平明显下降,以及接受I/R的小鼠和暴露于缺氧/复氧的AML12细胞。此外,SIRT5显著改善肝脏I/R诱导的氧化损伤,凋亡,和炎症通过调节线粒体氧化应激和功能。有趣的是,SIRT5的保肝作用由PRDX3介导。机械上,SIRT5在K84位点特异性地脱琥珀酰化PRDX3,使PRDX3能够减轻肝脏I/R过程中的线粒体氧化应激。创新点:本研究指出了SIRT5通过赖氨酸脱琥珀酰化调节线粒体氧化应激的新作用和机制。从而防止肝脏I/R损伤。结论:我们的发现首次证明SIRT5是肝脏I/R的关键介质,通过PRDX3的去琥珀酰化调节线粒体氧化应激,这提供了一种预防肝脏I/R损伤的新策略。
    Aims: Mitochondrial dysfunction is the primary mechanism of liver ischemia/reperfusion (I/R) injury. The lysine desuccinylase sirtuin 5 (SIRT5) is a global regulator of the mitochondrial succinylome and has pivotal roles in mitochondrial metabolism and function; however, its hepatoprotective capacity in liver I/R remains unclear. In this study, we established liver I/R model in SIRT5-silenced and SIRT5-overexpressed mice to examine the role and precise mechanisms of SIRT5 in liver I/R injury. Results: Succinylation was strongly enriched in liver mitochondria during I/R, and inhibiting mitochondrial succinylation significantly attenuated liver I/R injury. Importantly, the levels of the desuccinylase SIRT5 were notably decreased in liver transplant patients, as well as in mice subjected to I/R and in AML12 cells exposed to hypoxia/reoxygenation. Furthermore, SIRT5 significantly ameliorated liver I/R-induced oxidative injury, apoptosis, and inflammation by regulating mitochondrial oxidative stress and function. Intriguingly, the hepatoprotective effect of SIRT5 was mediated by PRDX3. Mechanistically, SIRT5 specifically desuccinylated PRDX3 at the K84 site, which enabled PRDX3 to alleviate mitochondrial oxidative stress during liver I/R. Innovation: This study denoted the new effect and mechanism of SIRT5 in regulating mitochondrial oxidative stress through lysine desuccinylation, thus preventing liver I/R injury. Conclusions: Our findings demonstrate for the first time that SIRT5 is a key mediator of liver I/R that regulates mitochondrial oxidative stress through the desuccinylation of PRDX3, which provides a novel strategy to prevent liver I/R injury. Antioxid. Redox Signal. 40, 616-631.
    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