首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 140 毫秒
1.
硫氧还蛋白(thioredoxin,Trx)是广泛存在于原核与真核生物体内的氧化还原调节蛋白。Trx通过对目标蛋白质进行还原,从而调节机体的氧化还原平衡。Trx与硫氧还蛋白还原酶(thioredoxin reductase,TrxR)及NADPH共同组成硫氧还蛋白系统参与众多生理过程。细胞中的活性氧是导致生物氧化胁迫的一个主要方面。Trx可以通过对细胞内被氧化的二硫键的还原来修复机体的氧化损伤,并通过这种方式防止机体衰老。同时,Trx系统可以与其它氧化还原系统如谷胱甘肽(GSH)系统协调配合,并消除体内过多的活性氧。  相似文献   

2.
硫氧还蛋白(Trx)是体内广泛存在的氧化还原蛋白,其家族中两种重要的硫氧还蛋白:硫氧还蛋白1(thioredoxin1,Trx1)和硫氧还蛋白2(thioredoxin2,Trx2)都含有保守的-Cys-Gly-Pro-Cys-还原序列。由于Trx具有调节细胞生长增殖和抗凋亡的作用,因此Trx在凋亡途径中的作用机制就成为了对抗肿瘤的研究热点。  相似文献   

3.
硫醇相关的氧化还原系统(thiol-dependent redox system, TDRS)由硫氧还蛋白系统(thioredoxin system, Trx system)和谷胱甘肽系统(glutaredoxin system, GSH system)组成,存在于多种生物体内,共同维持细胞内氧化还原平衡。原核生物细胞Trx系统中的硫氧还蛋白还原酶(thioredoxin reductase, TrxR)在结构和功能上与哺乳动物TrxR存在着天然差异。这些差异导致了细胞在相同药物作用下发生截然相反的氧化还原反应,使其可以成为潜在的抗菌作用靶点。此外,TrxR作为细菌看家基因trxB编码的蛋白,难以发生突变,故不易产生耐药性,是理想的新型抗菌靶标。本文就TDRS特别是Trx系统靶向性的新型抗菌策略作一综述。  相似文献   

4.
本文旨在使用病毒转染技术,观察正常培养条件下的心肌细胞单纯过表达硫氧还蛋白相互作用蛋白(thioredoxin inter-acting protein,TXNIP)是否可以引起细胞损伤和凋亡的发生,并分析其作用途径。对数生长期的H9C2心肌细胞分为三组:正常培养组、空病毒(Ad-eGFP)组、TXNIP过表达(Ad-TXNIP-eGFP)组,使用正常糖脂浓度(5mmo1/L葡萄糖)的DMEM培养基,均于转染72h后收集细胞和培养基进行指标测定。结果显示,细胞转染成功,72h转染效率达到高峰。与Ad-eGFP组相比,Ad-TXNIP-eGFP转染组TXNIPmRNA(P<0.05)和蛋白表达(P<0.01)均明显升高;心肌caspase-3(P<0.05)和LDH活性明显升高(P<0.01);使用流式细胞仪测得细胞凋亡明显增加(P<0.01)。Trx活性、与Trx相关的自由基损伤以及介导氧化应激损伤凋亡途径的p38激酶的活性检测显示,与Ad-eGFP组相比,TXNIP过表达组的Trx活性明显降低(P<0.01),反映膜脂质过氧化损伤的指标丙二醛(malondial dehyde,MDA)、3-硝基酪氨酸明显升高(P<0.01),p38激酶活性明显升高(P<0.01)。这些结果提示,通过腺病毒转染单纯过表达TXNIP可以引起正常糖脂浓度培养条件下的心肌细胞发生损伤和凋亡,其具体机制与抑制Trx活性、增加自由基损伤和p38激酶介导的凋亡有关。  相似文献   

5.
硫氧还蛋白结合蛋白(thioredoxin interacting protein, TXNIP)在细胞增殖、凋亡、分化的过程以及肿瘤、应激性疾病的发生中具有重要功能. 作为一个氧还反应的调节子,TXNIP能与硫氧还蛋白(thioredoxin, Trx)相结合,下调Trx的表达,而Trx则在DNA的损伤及细胞凋亡机制中有着关键的作用. 本文阐述了TXNIP基因的特征及其蛋白的生物学功能, 并简要总结TXNIP在人类肿瘤中低表达的研究成果. TXNIP基因是一个新的抑癌基因,它在人类乳腺癌、肝癌、肺癌等癌组织细胞中均表达下降, 并且与肿瘤的转移相关. TXNIP的缺失可以促使肿瘤细胞的增殖和抑制细胞凋亡的进程. 而在抗肿瘤机制中, TXNIP可通过参与细胞周期阻滞、低氧调节、影响NK细胞(natural killer cell, NK cell)发育等过程介导肿瘤的发生发展.  相似文献   

6.
硫氧还蛋白系统是由硫氧还蛋白(thioredoxin,Trx)、硫氧还蛋白还原酶(thioredoxin reductase,TrxR)和还原型辅酶Ⅱ(NADPH)组成的多功能小分子蛋白系统,广泛表达的硫氧还蛋白作为蛋白质二硫键的还原酶,它参与很多生理过程,并发挥重要生物学功能,包括调节机体的氧化还原反应、抑制细胞凋亡、调节转录因子DNA结合活性以及免疫应答等,其中一重要作用是参与调节细胞氧化还原状态以对抗氧化应激。因此在一些炎症性疾病如慢性阻塞性肺疾病、急性呼吸窘迫综合征、肺间质疾病、哮喘、肺结节病等的发生发展中扮演重要角色,本文对硫氧还蛋白系统在慢性阻塞性肺疾病中的抗氧化作用作一综述。  相似文献   

7.
目的:探索硫氧还蛋白(Trx)抗体柱对Trx融合蛋白纯化的可行性。方法与结果:对含有Trx基因的质粒表达载体pTrxFus进行改造,在Trx读框之后加入6×His序列,并在大肠杆菌中表达C端带有6×His标签的Trx,经Ni2+柱亲和纯化后制备多克隆抗体;把经蛋白A纯化后的抗体偶联在溴化氰活化的琼脂糖凝胶上,制成Trx抗体柱;用此抗体柱纯化与Trx融合表达的豇豆胰蛋白酶抑制剂(CpTI),SDS-PAGE结果显示获得了纯度较高的Trx-CpTI。结论:用Trx抗体制成的免疫亲和层析柱可以有效纯化Trx融合蛋白。  相似文献   

8.
线粒体缺陷和氧化应激参与了神经退行性疾病的发病机制.叠氮钠(NaN3)是线粒体细胞色素C氧化酶(COX)的特异性抑制剂,能诱导线粒体缺陷.本实验通过细胞活性检测(MTT法),形态学观察,分析H2O2对原代培养的正常神经元及NaN3诱导的线粒体缺陷神经元的损伤作用的差异.并通过RT-PCR半定量法检测H2O2损伤后两类神经元内硫氧还蛋白(Thioredoxin,Trx)mRNA水平的变化,以阐明细胞内这一重要氧化还原调节蛋白在神经元损伤时的作用机制.实验表明,在正常神经元内,H2O2的损伤对Trx表达量的改变似乎不明显;而线粒体缺陷神经元内Trx的表达量下降,且对于H2O2的损伤具有浓度、时间依赖性.提示在线粒体功能缺陷神经元中,Trx似乎发挥更重要的作用.  相似文献   

9.
目的: 探讨胃癌组织硫氧还蛋白还原酶1(TrxR1)表达与生存时间的关系及其对胃癌细胞生长的影响。方法: 用Real-time PCR法检测76例胃癌组织及癌旁TrxR1 mRNA表达,并分析其与胃癌患者临床病理特征及预后的关系;随机选取3例胃癌组织及癌旁组织,采用免疫组化法、Western blot法检测TrxR1蛋白表达。采用Western blot法和Real-time PCR法检测胃癌细胞系及人胃粘膜上皮细胞中TrxR1的表达。采用小RNA干扰序列(siRNA)处理AGS细胞,根据处理方法不同将AGS细胞分为3组:阴性对照组:转染NC-siRNA、TRXR1 siRNA干扰1组:转染TRXR1-siRNA1、TRXR1 siRNA干扰2组:转染TRXR1-siRNA2。使用Real-time PCR法检测各组AGS细胞中TrxR1 mRNA的表达,克隆形成试验和MTT法检测AGS细胞生长情况。结果: 胃癌组织中TrxR1 mRNA和蛋白表达量均显著性上调,TrxR1主要定位于细胞质中。TrxR1高表达与患者TNM分期及淋巴结转移有关,且TrxR1高表达组患者的中位生存时间短于低表达组(P<0.05)。胃癌细胞中TrxR1表达量高于人胃粘膜上皮细胞系中的表达。TRXR1-siRNA1组AGS细胞和TRXR1-siRNA2组AGS细胞中TrxR1 mRNA和蛋白与NC-siRNA组相比均显著性降低(P<0.05),且AGS细胞克隆形成与增殖能力均降低(P<0.05)。结论: 胃癌组织中TrxR1高表达提示患者预后不良,沉默TrxR1能抑制胃癌细胞的增殖。  相似文献   

10.
硫氧还蛋白1(thioredoxin1,Trx1)是细胞内一种重要的巯基 二硫键氧化还原酶,在细胞内氧化还原状态的调控及抵抗氧化应激损伤过程中发挥重要的作用.为了探讨高糖环境下Trx1过表达对 肾小球系膜细胞(glomerular mesangial cells)HBZY-1中基质金属蛋白酶9(matrix metalloproteinase 9,MMP9)表达水平的影响,本实验采用脂质体介导的瞬时转染实现Trx1蛋白过表达;采用RT-PCR和明胶酶谱法检测HBZY-1中MMP9 mRNA及酶活性的变化;通过流式细胞仪检测细胞内活性氧的含量.实验结果显示,高糖状态下,细胞中MMP9的mRNA和酶活性分别在12 h、24 h、48 h时表达增加(P<0.05);HBZY-1细胞中转染正义Trx1组,MMP9 mRNA水平及MMP9酶活性,高糖组与正常糖组无明显差异(P>0.05),转染反义Trx1组和未转染组中,高糖组均比正常糖组表达增加,差异有统计学意义(P<0.01);细胞中活性氧含量,高糖作用12 h、24 h、48 h均较正常糖组明显增多(P<0.01),高糖环境下转染正义Trx1质粒较转染反义Trx1质粒,细胞中活性氧含量明显减少,差异有统计学意义(P<0.05).实验提示,高糖环境下,Trx1过表达对MMP9的抑制作用是通过减少细胞内活性氧含量来实现的.本实验为Trx1的抗氧化作用提供新的证据,也为继续探讨 Trx1在糖尿病肾病的预防和治疗提供新的思路.  相似文献   

11.
Thioredoxin (Trx) inhibited tissue inhibitor of metalloproteinase (TIMP)-1 and TIMP-2 activity with an approximate IC50 of 0.3 microM, matrix metalloproteinase (MMP)-2 activity with an approximate IC50 of 2 microM but did not inhibit MMP-9 activity. This differential capacity of Trx to inhibit TIMP and MMP activity resulted in the promotion of MMP-2 and MMP-9 activity in the presence of molar TIMP excess. Inhibition of TIMP and MMP-2 activity by Trx was dependent upon thioredoxin reductase (TrxR), was abolished by Trx catalytic site mutation and did not result from TIMP or MMP-2 degradation. HepG2 hepatocellular carcinoma cells induced to secrete Trx inhibited TIMP activity in the presence of TrxR. SK-N-SH neuroblastoma cells secreted TrxR, which inhibited TIMP and MMP-2 activity in the presence of Trx. Trx stimulated SK-N-SH invasive capacity in vitro in the absence of exogenous TrxR. This study therefore identifies a novel extracellular role for the thioredoxin/thioredoxin reductase redox system in the differential inhibition of TIMP and MMP activity and provides a novel mechanism for altering the TIMP/MMP balance that is of potential relevance to tumor invasion.  相似文献   

12.
The mammalian cytosolic/nuclear thioredoxin system, comprising thioredoxin (Trx), selenoenzyme thioredoxin reductase (TrxR), and NADPH, is the major protein-disulfide reductase of the cell and has numerous functions. The active site of reduced Trx comprises Cys(32)-Gly-Pro-Cys(35) thiols that catalyze target disulfide reduction, generating a disulfide. Human Trx1 has also three structural Cys residues in positions 62, 69, and 73 that upon diamide oxidation induce a second Cys(62)-Cys(69) disulfide as well as dimers and multimers. We have discovered that after incubation with H(2)O(2) only monomeric two-disulfide molecules are generated, and they are inactive but able to regain full activity in an autocatalytic process in the presence of NADPH and TrxR. There are conflicting results regarding the effects of S-nitrosylation on Trx antioxidant functions and which residues are involved. We found that S-nitrosoglutathione-mediated S-nitrosylation at physiological pH is critically dependent on the redox state of Trx. Starting from fully reduced human Trx, both Cys(69) and Cys(73) were nitrosylated, and the active site formed a disulfide; the nitrosylated Trx was not a substrate for TrxR but regained activity after a lag phase consistent with autoactivation. Treatment of a two-disulfide form of Trx1 with S-nitrosoglutathione resulted in nitrosylation of Cys(73), which can act as a trans-nitrosylating agent as observed by others to control caspase 3 activity (Mitchell, D. A., and Marletta, M. A. (2005) Nat. Chem. Biol. 1, 154-158). The reversible inhibition of human Trx1 activity by H(2)O(2) and NO donors is suggested to act in cell signaling via temporal control of reduction for the transmission of oxidative and/or nitrosative signals in thiol redox control.  相似文献   

13.
Cellular redox balance is maintained by various antioxidative systems. Among those is the thioredoxin system, consisting of thioredoxin, thioredoxin reductase, and NADPH. In the present study, we examined the effects of caloric restriction (2 mo) on the expression of the cytosolic and mitochondrial thioredoxin system in skeletal muscle and heart of senescent and young rats. Mitochondrial thioredoxin reductase (TrxR2) is significantly reduced in aging skeletal and cardiac muscle and renormalized after caloric restriction, while the cytosolic isoform remains unchanged. Thioredoxins (mitochondrial Trx2, cytosolic Trx1) are not influenced by caloric restriction. In skeletal and cardiac muscle of young rats, caloric restriction has no effect on the expression of thioredoxins or thioredoxin reductases. Enforced reduction of TrxR2 (small interfering RNA) in myoblasts under exposure to ceramide or TNF-alpha causes a dramatic enhancement of nucleosomal DNA cleavage, caspase 9 activation, and mitochondrial reactive oxygen species release, together with reduced cell viability, while this TrxR2 reduction is without effect in unstimulated myoblasts under basal conditions. Oxidative stress in vitro (H2O2 in C2C12 myoblasts and myotubes) results in different changes: TrxR2, Trx2, and Trx1 are induced without alterations in the cytosolic thioredoxin reductase isoforms. Thus aging is associated with a TrxR2 reduction in skeletal muscle and heart, which enhances susceptibility to apoptotic stimuli but is renormalized after short-term caloric restriction. Exogenous oxidative stress does not result in these age-related changes of TrxR2.  相似文献   

14.
The antioxidant mechanism of ebselen involves recently discovered reductions by mammalian thioredoxin reductase (TrxR) and thioredoxin (Trx) forming ebselen selenol. Here we describe a previously unknown reaction; ebselen reacts with its selenol forming an ebselen diselenide with a rate constant of 372 m(-1)s(-1). The diselenide also was a substrate of TrxR forming the selenol with K(m) of 40 microm and k(cat) of 79 min(-1) (k(cat)/K(m) of 3.3 x 10(4) m(-1)s(-1)). Trx increased the reduction because of its fast reaction with diselenide (rate constant 1.7 x 10(3) m(-1)s(-1)). Diselenide stimulated the H2O2 reductase activity of TrxR, even more efficiently with Trx present. Because the mechanism of ebselen as an antioxidant has been assumed to involve glutathione peroxidase-like activity, we compared the H2O2 reductase activity of ebselen with the GSH and Trx systems. TrxR at 50 nm, far below the estimated physiological level, gave 8-fold higher activity compared with 1 mm GSH; addition of 5 microm Trx increased this difference to 13-fold. The rate constant of ebselen selenol reacting with H2O2 was estimated to be faster than 350 m(-1)s(-1). We propose novel mechanisms for ebselen antioxidant action involving ebselen selenol and diselenide formation, with the thioredoxin system rather than glutathione as the predominant effector and target.  相似文献   

15.
Thioredoxin systems, composed of thioredoxin reductase (TrxR), thioredoxin (Trx) and NADPH, play important roles in maintaining cellular redox homeostasis and redox signaling. Recently the cytosolic Trx1 system has been shown to be a cellular target of arsenic containing compounds. To elucidate the relationship of the structure of arsenic compounds with their ability of inhibiting TrxR1 and Trx1, and cytotoxicity, we have investigated the reaction of Trx1 system with seven arsenic trithiolates: As(Cys)3, As(GS)3, As(Penicillamine)3, As(Mercaptoethanesulfonate)3, As(Mercaptopurine)3, As(2-mercaptopyridine)3 and As(2-mercaptopyridine N-oxide)3. The cytotoxicity of these arsenicals was consistent with their ability to inhibit TrxR1 in vitro and in cells. Unlike other arsenicals, As(Mercaptopurine)3 which did not show inhibitory effects on TrxR1 had very weak cytotoxicity, indicating that TrxR1 is a reliable drug target for arsenicals. Moreover, the two aromatic compounds As(2-mercaptopyridine)3 and As(2-mercaptopyridine N-oxide)3 showed stronger cytotoxicity than the others. As(2-mercaptopyridine)3 which selectively oxidized two structural cysteines (Cys62 and Cys69) in Trx1 showed mild improvement in cytotoxicity. As(2-mercaptopyridine N-oxide)3 oxidized all the Cys residues in Trx1, exhibiting the strongest cytotoxicity. Oxidation of Trx1 by As(2-mercaptopyridine)3 and As(2-mercaptopyridine N-oxide)3 affected electron transfer from NADPH and TrxR1 to peroxiredoxin 1 (Prx1), which could result in the reactive oxygen species elevation and trigger cell death process. These results suggest that oxidation of structural cysteine residues in Trx1 by aromatic group in TrxR1-targeting drugs may sensitize tumor cells to cell death, providing a novel approach to regulate cellular redox signaling and also a basis for rational design of new anticancer agents.  相似文献   

16.
Thioredoxin reductase 1 (TrxR1) in cytosol is the only known reductant of oxidized thioredoxin 1 (Trx1) in vivo so far. We and others found that aurothioglucose (ATG), a well known active-site inhibitor of TrxR1, inhibited TrxR1 activity in HeLa cell cytosol but had no effect on the viability of the cells. Using a redox Western blot analysis, no change was observed in redox state of Trx1, which was mainly fully reduced with five sulfhydryl groups. In contrast, auranofin killed cells and oxidized Trx1, also targeting mitochondrial TrxR2 and Trx2. Combining ATG with ebselen gave a strong synergistic effect, leading to Trx1 oxidation, reactive oxygen species accumulation, and cell death. We hypothesized that there should exist a backup system to reduce Trx1 when only TrxR1 activity was lost. Our results showed that physiological concentrations of glutathione, NADPH, and glutathione reductase reduced Trx1 in vitro and that the reaction was strongly stimulated by glutaredoxin1. Simultaneous depletion of TrxR activity by ATG and glutathione by buthionine sulfoximine led to overoxidation of Trx1 and loss of HeLa cell viability. In conclusion, the glutaredoxin system and glutathione have a backup role to keep Trx1 reduced in cells with loss of TrxR1 activity. Monitoring the redox state of Trx1 shows that cell death occurs when Trx1 is oxidized, followed by general protein oxidation catalyzed by the disulfide form of thioredoxin.  相似文献   

17.
18.
19.
Molecular oxygen is key to aerobic life but is also converted into cytotoxic byproducts referred to as reactive oxygen species (ROS). Intracellular defense systems that protect cells from ROS-induced damage include glutathione reductase (GR), thioredoxin reductase (TrxR), superoxide dismutase (Sod), and catalase (Cat). Sod and Cat constitute an evolutionary conserved ROS defense system against superoxide; Sod converts superoxide anions to H(2)O(2), and Cat prevents free hydroxyl radical formation by breaking down H(2)O(2) into oxygen and water. As a consequence, they are important effectors in the life span determination of the fly Drosophila. ROS defense by TrxR and GR is more indirect. They transfer reducing equivalents from NADPH to thioredoxin (Trx) and glutathione disulfide (GSSG), respectively, resulting in Trx(SH)(2) and glutathione (GSH), which act as effective intracellular antioxidants. TrxR and GR were found to be molecularly conserved. However, the single GR homolog of Drosophila specifies TrxR activity, which compensates for the absence of a true GR system for recycling GSH. We show that TrxR null mutations reduce the capacity to adequately protect cells from cytotoxic damage, resulting in larval death, whereas mutations causing reduced TrxR activity affect pupal eclosion and cause a severe reduction of the adult life span. We also provide genetic evidence for a functional interaction between TrxR, Sod1, and Cat, indicating that the burden of ROS metabolism in Drosophila is shared by the two defense systems.  相似文献   

20.
Thioredoxin-2 (Trx2) is a mitochondrial protein-disulfide oxidoreductase essential for control of cell survival during mammalian embryonic development. This suggests that mitochondrial thioredoxin reductase-2 (TrxR2), responsible for reducing oxidized Trx2, may also be a key player in the regulation of mitochondria-dependent apoptosis. With this in mind, we investigated the effects of overexpression of TrxR2, Trx2, or both on mammalian cell responses to various apoptotic inducers. Stable transfectants of mouse Neuro2A cells were generated that overexpressed TrxR2 or an EGFP-TrxR2 fusion protein. EGFP-TrxR2 was enzymatically active and was localized in mitochondria. TrxR2 protein level and TrxR activity could be increased up to 6-fold in mitochondria. TrxR2 and EGFP-TrxR2 transfectants showed reduced growth rates as compared with control cells. This growth alteration was not due to cytotoxic effects nor related to changes in basal mitochondrial transmembrane potential (DeltaPsi(m)), reactive oxygen species production, or to other mitochondrial antioxidant components such as Trx2, peroxyredoxin-3, MnSOD, GPx1, and glutathione whose levels were not affected by increased TrxR2 activity. In response to various apoptotic inducers, the extent of DeltaPsi(m) dissipation, reactive oxygen species induction, caspase activation, and loss of viability were remarkably similar in TrxR2 and control transfectants. Excess TrxR2 did not prevent trichostatin A-mediated neuronal differentiation of Neuro2A cells nor did it protect them against beta-amyloid neurotoxicity. Neither massive glutathione depletion nor co-transfection of Trx2 and TrxR2 in Neuro2A (mouse), COS-7 (monkey), or HeLa (human) cells revealed any differential cellular resistance to prooxidant or non-oxidant apoptotic stimuli. Our results suggest that neither Trx2 nor TrxR2 gain of function modified the redox regulation of mitochondria-dependent apoptosis in these mammalian cells.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号