GSNOR

GSNOR
  • 文章类型: Journal Article
    吗啡,典型的阿片类药物,广泛用于控制疼痛,但长期使用会导致各种副作用,包括上瘾,镇痛耐受性,和痛觉过敏。目前,然而,吗啡镇痛耐受发展的潜在机制尚不完全清楚.这种耐受性受各种阿片受体和激酶蛋白修饰的影响,如磷酸化和泛素化。这里,我们建立了小鼠吗啡耐受模型,以研究S-亚硝基谷胱甘肽还原酶(GSNOR)是否以及如何参与吗啡耐受.反复服用吗啡导致GSNOR下调,这增加了前额叶皮质中过多的总蛋白S-亚硝化。GSNOR的敲除或化学抑制促进了吗啡镇痛耐受的发展,GSNOR的神经元特异性过表达减轻了吗啡镇痛耐受。机械上,GSNOR缺陷增强了细胞蛋白激酶α(PKCα)在Cys78和Cys132位点的S-亚硝化,导致PKCα激酶活性的抑制,最终促进了吗啡镇痛耐受性的发展。我们的研究强调了GSNOR作为PKCαS-亚硝化的关键调节因子的重要作用及其在吗啡镇痛耐受中的参与,从而为吗啡耐受提供了一个潜在的治疗靶点.
    Morphine, a typical opiate, is widely used for controlling pain but can lead to various side effects with long-term use, including addiction, analgesic tolerance, and hyperalgesia. At present, however, the mechanisms underlying the development of morphine analgesic tolerance are not fully understood. This tolerance is influenced by various opioid receptor and kinase protein modifications, such as phosphorylation and ubiquitination. Here, we established a murine morphine tolerance model to investigate whether and how S-nitrosoglutathione reductase (GSNOR) is involved in morphine tolerance. Repeated administration of morphine resulted in the down-regulation of GSNOR, which increased excessive total protein S-nitrosation in the prefrontal cortex. Knockout or chemical inhibition of GSNOR promoted the development of morphine analgesic tolerance and neuron-specific overexpression of GSNOR alleviated morphine analgesic tolerance. Mechanistically, GSNOR deficiency enhanced S-nitrosation of cellular protein kinase alpha (PKCα) at the Cys78 and Cys132 sites, leading to inhibition of PKCα kinase activity, which ultimately promoted the development of morphine analgesic tolerance. Our study highlighted the significant role of GSNOR as a key regulator of PKCα S-nitrosation and its involvement in morphine analgesic tolerance, thus providing a potential therapeutic target for morphine tolerance.
    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
    作为信号分子,一氧化氮(NO)调节不同生物体的发育和应激反应。NO的主要生物活性是蛋白质S-亚硝基化,其在真菌中的功能仍不清楚。这里,它在稻瘟病真菌稻瘟病菌中发现,脱亚硝基化过程对于感染过程中的功能性附着层形成至关重要。NO过度积累引起的硝化应激对真菌感染有害。而S-亚硝基谷胱甘肽还原酶GSNOR介导的脱亚硝基化可在附睾形成过程中消除过量的NO毒性以促进感染。通过indoTMT开关标记蛋白质组学技术,鉴定了483种蛋白质中的741S-亚硝基化位点。关键吸食蛋白,如MgB1、MagB、Sps1、Cdc42和隔膜,被GSNOR通过脱亚硝基活化。去除上述蛋白质的S-亚硝基化位点对于适当的蛋白质结构和表观功能至关重要。因此,GSNOR介导的脱亚硝基化是附着层形成的重要调节因子。还表明,NO供体打破NO稳态,没有清除剂,以及GSNOR的化学抑制剂,是控制真菌病的有效方法。
    As a signaling molecule, nitric oxide (NO) regulates the development and stress response in different organisms. The major biological activity of NO is protein S-nitrosylation, whose function in fungi remains largely unclear. Here, it is found in the rice blast fungus Magnaporthe oryzae, de-nitrosylation process is essential for functional appressorium formation during infection. Nitrosative stress caused by excessive accumulation of NO is harmful for fungal infection. While the S-nitrosoglutathione reductase GSNOR-mediated de-nitrosylation removes excess NO toxicity during appressorium formation to promote infection. Through an indoTMT switch labeling proteomics technique, 741 S-nitrosylation sites in 483 proteins are identified. Key appressorial proteins, such as Mgb1, MagB, Sps1, Cdc42, and septins, are activated by GSNOR through de-nitrosylation. Removing S-nitrosylation sites of above proteins is essential for proper protein structure and appressorial function. Therefore, GSNOR-mediated de-nitrosylation is an essential regulator for appressorium formation. It is also shown that breaking NO homeostasis by NO donors, NO scavengers, as well as chemical inhibitor of GSNOR, shall be effective methods for fungal disease control.
    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
    NLRP3炎性体的过度激活与许多疾病的发病机理有关。然而,调控NLRP3转录调控的确切分子机制仍不清楚.在这项研究中,我们证明巨噬细胞中的S-亚硝基谷胱甘肽还原酶(GSNOR)缺乏导致NLRP3炎症小体刺激引起的Nlrp3和IL-1β表达水平和白细胞介素-1β(IL-1β)分泌显著增加。此外,使用Gsnor-/-小鼠的体内实验显示,在脂多糖(LPS)诱导的脓毒性休克和葡聚糖硫酸钠(DSS)诱导的结肠炎模型中,疾病严重程度均增加。此外,我们发现在Gsnor-/-Nlrp3-/-双基因敲除(DKO)小鼠中,LPS诱导的感染性休克和DSS诱导的结肠炎均得到改善。机械上,GSNOR缺乏增加了丝裂原活化蛋白激酶14(MAPK14)在Cys211残基的S-亚硝化,并增加了MAPK14激酶的活性,从而促进Nlrp3和Il-1β转录并刺激NLRP3炎性体活性。我们的研究结果表明,GSNOR是NLRP3炎症小体的调节因子,降低S-亚硝基化的MAPK14水平可能是缓解与NLRP3介导的炎症相关疾病的有效策略。
    Hyperactivation of the NLRP3 inflammasome has been implicated in the pathogenesis of numerous diseases. However, the precise molecular mechanisms that modulate the transcriptional regulation of NLRP3 remain largely unknown. In this study, we demonstrated that S-nitrosoglutathione reductase (GSNOR) deficiency in macrophages leads to significant increases in the Nlrp3 and Il-1β expression levels and interleukin-1β (IL-1β) secretion in response to NLRP3 inflammasome stimulation. Furthermore, in vivo experiments utilizing Gsnor-/- mice revealed increased disease severity in both lipopolysaccharide (LPS)-induced septic shock and dextran sodium sulfate (DSS)-induced colitis models. Additionally, we showed that both LPS-induced septic shock and DSS-induced colitis were ameliorated in Gsnor-/- Nlrp3-/- double-knockout (DKO) mice. Mechanistically, GSNOR deficiency increases the S-nitrosation of mitogen-activated protein kinase 14 (MAPK14) at the Cys211 residue and augments MAPK14 kinase activity, thereby promoting Nlrp3 and Il-1β transcription and stimulating NLRP3 inflammasome activity. Our findings suggested that GSNOR is a regulator of the NLRP3 inflammasome and that reducing the level of S-nitrosylated MAPK14 may constitute an effective strategy for alleviating diseases associated with NLRP3-mediated inflammation.
    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
    嵌合抗原受体T细胞(CAR-T)已被开发为难治性或复发性淋巴瘤和白血病患者的有希望的药物,但并非所有接受者都能获得持久的缓解。激活后体内扩增和记忆分化的能力有限是CAR-T治疗效率欠佳的主要原因之一。一氧化氮(NO)在线粒体动力学和T细胞活化中起着多方面的作用,但其对CAR-T细胞持久性和抗肿瘤疗效的作用仍不清楚。在这里,我们发现来自CAR的连续信号不仅促进过量的NO产生,但也抑制了T细胞中S-亚硝基谷胱甘肽还原酶(GSNOR)的表达,共同导致蛋白质S-亚硝基化增加,导致线粒体适应性受损和T细胞干细胞缺乏。有趣的是,GSNOR的强制表达促进了免疫激活后CAR-T细胞的记忆分化,使CAR-T更好地抵抗线粒体功能障碍,在体外和小鼠肿瘤模型中进一步增强CAR-T细胞扩增和抗肿瘤能力。因此,我们揭示了NO在限制CAR-T细胞持久性和功能性方面的关键作用,并定义GSNOR过表达可以提供对抗NO应激的解决方案,并使患者对CAR-T治疗具有更持久的保护作用。
    Chimeric antigen receptor-T (CAR-T) cell has been developed as a promising agent for patients with refractory or relapsed lymphoma and leukemia, but not all the recipients could achieve a long-lasting remission. The limited capacity of in vivo expansion and memory differentiation post activation is one of the major reasons for suboptimal CAR-T therapeutic efficiency. Nitric oxide (NO) plays multifaceted roles in mitochondrial dynamics and T cell activation, but its function on CAR-T cell persistence and anti-tumor efficacy remains unknown. Herein, we found the continuous signaling from CAR not only promotes excessive NO production, but also suppressed S-nitrosoglutathione reductase (GSNOR) expression in T cells, which collectively led to increased protein S-nitrosylation, resulting in impaired mitochondrial fitness and deficiency of T cell stemness. Intriguingly, enforced expression of GSNOR promoted memory differentiation of CAR-T cell after immune activation, rendered CAR-T better resistance to mitochondrial dysfunction, further enhanced CAR-T cell expansion and anti-tumor capacity in vitro and in a mouse tumor model. Thus, we revealed a critical role of NO in restricting CAR-T cell persistence and functionality, and defined that GSNOR overexpression may provide a solution to combat NO stress and render patients with more durable protection from CAR-T therapy.
    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
    破骨细胞,专有的骨吸收细胞,是骨骼重塑不可或缺的。因此,了解调节破骨细胞生成的新型信号调节剂在临床上很重要。活化T细胞核因子,细胞质1(NFATc1)是破骨细胞形成中的主要转录因子,NF-κBp65亚基与NFATc1启动子的结合是其表达所必需的。众所周知,p65的DNA结合活性可以通过各种翻译后修饰来调节。包括S-亚硝化。最近的研究表明,S-亚硝基谷胱甘肽还原酶(GSNOR)介导的蛋白质脱氮通过调节基因转录参与细胞命运的决定。然而,GSNOR在破骨细胞生成中的作用仍未被探索和神秘。这里,我们研究了GSNOR介导的p65脱氮对破骨细胞生成的影响。我们的结果表明,GSNOR在体外破骨细胞形成过程中上调。此外,用化学抑制剂抑制GSNOR损害破骨细胞分化,足体带形成,和骨吸收活性。此外,GSNOR抑制增强了p65的S-亚硝化水平,排除了p65与NFATc1启动子的结合,并抑制NFATc1表达。此外,采用脂多糖(LPS)诱导的颅骨骨溶解的小鼠模型来评估GSNOR抑制剂的体内治疗效果。我们的结果表明,GSNOR抑制剂治疗通过损害小鼠破骨细胞的形成来减轻炎性骨丢失。一起来看,这些数据表明,GSNOR活性是破骨细胞形成所必需的,通过促进p65脱氮促进p65与NFATc1启动子的结合,提示GSNOR可能是治疗溶骨性疾病的潜在治疗靶点。
    Osteoclasts, the exclusive bone resorptive cells, are indispensable for bone remodeling. Hence, understanding novel signaling modulators regulating osteoclastogenesis is clinically important. Nuclear factor of activated T-cells, cytoplasmic 1 (NFATc1) is a master transcription factor in osteoclastogenesis, and binding of NF-κB p65 subunit to NFATc1 promoter is required for its expression. It is well-established that DNA binding activity of p65 can be regulated by various post-translational modifications, including S-nitrosation. Recent studies have demonstrated that S-nitrosoglutathione reductase (GSNOR)-mediated protein denitrosation participated in cell fate commitment by regulating gene transcription. However, the role of GSNOR in osteoclastogenesis remains unexplored and enigmatic. Here, we investigated the effect of GSNOR-mediated denitrosation of p65 on osteoclastogenesis. Our results revealed that GSNOR was up-regulated during osteoclastogenesis in vitro. Moreover, GSNOR inhibition with a chemical inhibitor impaired osteoclast differentiation, podosome belt formation, and bone resorption activity. Furthermore, GSNOR inhibition enhanced the S-nitrosation level of p65, precluded the binding of p65 to NFATc1 promoter, and suppressed NFATc1 expression. In addition, mouse model of lipopolysaccharides (LPS)-induced calvarial osteolysis was employed to evaluate the therapeutic effect of GSNOR inhibitor in vivo. Our results indicated that GSNOR inhibitor treatment alleviated the inflammatory bone loss by impairing osteoclast formation in mice. Taken together, these data have shown that GSNOR activity is required for osteoclastogenesis by facilitating binding of p65 to NFATc1 promoter via promoting p65 denitrosation, suggesting that GSNOR may be a potential therapeutic target in the treatment of osteolytic diseases.
    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:血管内皮生长因子D(VEGFD),VEGF家族的一员,与血管生成和淋巴管生成有关,并且被认为在癌症中以低水平表达。S-亚硝基化,NO(一氧化氮)介导的翻译后修饰在血管生成中具有关键作用。这里,我们试图剖析S-亚硝基化介导的VEGFD抑制在肺腺癌(LUAD)中的作用和潜在机制.
    方法:通过TCGA和CPTAC数据库分析LUAD中VEGFD的信使RNA和蛋白质表达,分别,和临床生物信息学助理进行复杂分析。建立了尿烷(Ure)诱导的LUAD或LUAD异种移植的小鼠模型,以研究S-亚硝基化在VEGFD表达中的作用以及VEGFD突变体在LUAD肿瘤发生中的作用。分子,细胞,和生化方法被用来探讨S-亚硝基化介导的VEGFD抑制的潜在机制。使用管形成和伤口愈合试验来检查VEGFD对LUAD细胞的血管生成和迁移的作用。并应用分子模型预测VEGFD突变体的蛋白稳定性。
    结果:VEGFDmRNA和蛋白水平在多原发恶性肿瘤中有不同程度的降低,尤其是在LUAD。VEGFD蛋白的低表达与LUAD的发生密切相关,并且是由NO在Cys277诱导的VEGFDS亚硝基化引起的。此外,S-亚硝基谷胱甘肽还原酶的抑制作用持续降低了Cys277处的VEGFD反硝基化,因此促进了LUAD的血管生成。最后,VEGFDC277S突变体通过减弱PC7依赖性蛋白水解而减少成熟VEGFD的分泌,因此VEGFDC277S突变体逆转了VEGFD对LUAD血管生成的影响。
    结论:VEGFD的低表达与LUAD的发生呈正相关。VEGFD的异常S-亚硝基化自身否定以诱导LUAD的肿瘤发生,而VEGFD的正常S-亚硝基化对于其分泌和抑制LUAD的血管生成是必不可少的。
    BACKGROUND: Vascular endothelial growth factor D (VEGFD), a member of the VEGF family, is implicated in angiogenesis and lymphangiogenesis, and is deemed to be expressed at a low level in cancers. S-nitrosylation, a NO (nitric oxide)-mediated post-translational modification has a critical role in angiogenesis. Here, we attempt to dissect the role and underlying mechanism of S-nitrosylation-mediated VEGFD suppression in lung adenocarcinoma (LUAD).
    METHODS: Messenger RNA and protein expression of VEGFD in LUAD were analyzed by TCGA and CPTAC database, respectively, and Assistant for Clinical Bioinformatics was performed for complex analysis. Mouse models with urethane (Ure)-induced LUAD or LUAD xenograft were established to investigate the role of S-nitrosylation in VEGFD expression and of VEGFD mutants in the oncogenesis of LUAD. Molecular, cellular, and biochemical approaches were applied to explore the underlying mechanism of S-nitrosylation-mediated VEGFD suppression. Tube formation and wound healing assays were used to examine the role of VEGFD on the angiogenesis and migration of LUAD cells, and the molecular modeling was applied to predict the protein stability of VEGFD mutant.
    RESULTS: VEGFD mRNA and protein levels were decreased to a different extent in multiple primary malignancies, especially in LUAD. Low VEGFD protein expression was closely related to the oncogenesis of LUAD and resultant from excessive NO-induced VEGFD S-nitrosylation at Cys277. Moreover, inhibition of S-nitrosoglutathione reductase consistently decreased the VEGFD denitrosylation at Cys277 and consequently promoted angiogenesis of LUAD. Finally, the VEGFDC277S mutant decreased the secretion of mature VEGFD by attenuating the PC7-dependent proteolysis and VEGFDC277S mutant thus reversed the effect of VEGFD on angiogenesis of LUAD.
    CONCLUSIONS: Low-expression of VEGFD positively correlates with LUAD development. Aberrant S-nitrosylation of VEGFD negates itself to induce the tumorigenesis of LUAD, whereas normal S-nitrosylation of VEGFD is indispensable for its secretion and repression of angiogenesis of LUAD.
    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
    S-亚硝基谷胱甘肽还原酶(GSNOR)是调节蛋白质S-亚硝化的关键脱氮酶,已发现与帕金森病(PD)的发病机制有关的过程。然而,GSNOR在PD中的生理功能尚不清楚。在1-甲基-4-苯基-1,2,3,6-四氢吡啶(MPTP)诱导的PD小鼠模型中,我们发现GSNOR表达显著增加,并伴有MPTP诱导的细胞周期蛋白依赖性激酶5(CDK5)介导的自噬,行为运动障碍和多巴胺能神经元丢失。然而,敲除GSNOR,或使用GSNOR抑制剂N6022治疗,可减轻MPTP诱导的PD样病理和神经毒性。机械上,GSNOR的缺乏通过在Cys83处增加CDK5的S-亚硝化来抑制MPTP诱导的CDK5激酶活性和CDK5介导的自噬。我们的研究表明,GSNOR是CDK5S-亚硝化的关键调节因子,并积极参与MPTP诱导的CDK5介导的自噬。
    The S-nitrosoglutathione reductase (GSNOR) is a key denitrosating enzyme that regulates protein S-nitrosation, a process which has been found to be involved in the pathogenesis of Parkinson\'s disease (PD). However, the physiological function of GSNOR in PD remains unknown. In a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mouse model, we found that GSNOR expression was significantly increased and accompanied by autophagy mediated by MPTP-induced cyclin dependent kinase 5 (CDK5), behavioral dyskinesias and dopaminergic neuron loss. Whereas, knockout of GSNOR, or treatment with the GSNOR inhibitor N6022, alleviated MPTP-induced PD-like pathology and neurotoxicity. Mechanistically, deficiency of GSNOR inhibited MPTP-induced CDK5 kinase activity and CDK5-mediated autophagy by increasing S-nitrosation of CDK5 at Cys83. Our study indicated that GSNOR is a key regulator of CDK5 S-nitrosation and is actively involved in CDK5-mediated autophagy induced by MPTP.
    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
    S-亚硝基谷胱甘肽还原酶(GSNOR)被认为是植物胁迫耐受性的关键调节剂,因为它通过调节S-亚硝基硫醇(SNO)水平对蛋白质S-亚硝基化产生影响。然而,GSNOR介导的应激耐受机制尚不清楚。这里,我们发现番茄(Solanumlycopersicum)植物的高温诱导GSNOR活性,而SlGSNOR1的mRNA水平几乎没有反应。通过病毒诱导的基因沉默(VIGS)抑制SlGSNOR1表达会增加高温下SNO和亚硝酸盐的积累,并降低耐热性。耐热性受损与脱落酸(ABA)和水杨酸(SA)的积累减少有关,丝裂原活化蛋白激酶(MAPK)的激活减弱,热休克蛋白的表达降低。有趣的是,SlGSNOR1沉默损害了响应于高温的呼吸破裂氧化酶HOMOLOG1(SlRBOH1)和质外生H2O2积累的上调,而SlRBOH1沉默消除了GSNOR的激活,并导致与SlGSNOR1沉默的植物相似的耐热性下降。重要的是,H2O2处理恢复了SlGSNOR1沉默植物的耐热性并提高了抗氧化能力。我们的结果表明,GSNOR在响应高温时调节SlRBOH1依赖的质外体H2O2的产生中起作用,而SNO和H2O2之间的平衡相互作用对于维持细胞氧化还原稳态和耐热性至关重要。
    S-nitrosoglutathione reductase (GSNOR) is considered as a critical regulator of plant stress tolerance for its impacts on protein S-nitrosylation through regulation of the S-nitrosothiol (SNO) level. However, the mechanism of GSNOR-mediated stress tolerance is still obscure. Here, we found that GSNOR activity was induced by high temperature in tomato (Solanum lycopersicum) plants, whereas mRNA level of SlGSNOR1 exhibited little response. Suppressing SlGSNOR1 expression by virus-induced gene silencing (VIGS) increased accumulation of SNO and nitrites under high temperature and reduced thermotolerance. The compromised thermotolerance was associated with less accumulation of abscisic acid (ABA) and salicylic acid (SA), attenuated activation of mitogen-activated protein kinase (MAPK) and reduced expression of heat shock protein. Intriguingly, SlGSNOR1 silencing impaired upregulation of RESPIRATORY BURST OXIDASE HOMOLOG1 (SlRBOH1) and apoplastic H2O2 accumulation in response to high temperature, whereas SlRBOH1 silencing abolished activation of GSNOR and led to a similar decline in thermotolerance as in SlGSNOR1-silenced plants. Importantly, H2O2 treatment recovered the thermotolerance and improved antioxidant capacity in SlGSNOR1-silenced plants. Our results suggest that GSNOR plays a role in regulating the SlRBOH1-dependent apoplastic H2O2 production in response to high temperature, while a balanced interaction between SNO and H2O2 is critical for maintaining the cellular redox homeostasis and thermotolerance.
    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
    蛛网膜下腔出血(SAH)是出血性中风,具有很高的死亡率和致残率。一氧化氮(NO)可以通过血管舒张促进血液供应,导致蛋白质S-亚硝基化。然而,SAH后神经元中S-亚硝基化的功能尚不清楚.病理状态下过量的NO转化为S-亚硝基谷胱甘肽(GSNO)并储存在细胞中,这导致细胞内蛋白质的高S-亚硝基化并引起亚硝基应激。S-亚硝基谷胱甘肽还原酶(GSNOR)促进GSNO降解并保护细胞免受过度的S-亚硝基化。我们进行了体内大鼠颈动脉穿刺模型和体外神经元血红蛋白干预。结果表明,SAH诱导增加NO,GSNO,神经元蛋白S-亚硝基化,和神经元凋亡,同时降低GSNOR的水平和活性。慢病毒过表达GSNOR可降低GSNO,但对NO影响不大。GSNOR过表达还改善了大鼠的短期和长期神经行为结果,并减轻了亚硝基应激。此外,GSNOR通过减轻Drp1S-亚硝基化减少神经元凋亡并发挥神经保护作用,减少线粒体分裂.因此,GSNOR在早期脑损伤中的调控和神经元脱氮糖基化可能在神经保护中起重要作用。
    Subarachnoid hemorrhage (SAH) is a hemorrhagic stroke with a high mortality and disability rate. Nitric oxide (NO) can promote blood supply through vasodilation, leading to protein S-nitrosylation. However, the function of S-nitrosylation in neurons after SAH remains unclear. Excessive NO in the pathological state is converted into S-nitrosoglutathione (GSNO) and stored in cells, which leads to high S-nitrosylation of intracellular proteins and causes nitrosative stress. S-nitrosoglutathione reductase (GSNOR) promotes GSNO degradation and protects cells from excessive S-nitrosylation. We conducted an in vivo rat carotid puncture model and an in vitro neuron hemoglobin intervention. The results showed that SAH induction increased NO, GSNO, neuron protein S-nitrosylation, and neuronal apoptosis, while decreasing the level and activity of GSNOR. GSNOR overexpression by lentivirus decreased GSNO but had little effect on NO. GSNOR overexpression also improved short- and long-term neurobehavioral outcomes in rats and alleviated nitrosative stress. Furthermore, GSNOR reduced neuronal apoptosis and played a neuroprotective role by alleviating Drp1 S-nitrosylation, reducing mitochondrial division. Thus, the regulation of GSNOR in early brain injury and neuronal denitrosylation may play an important role in neuroprotection.
    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
    Maintenance of genome stability is an essential requirement for all living organisms. Formaldehyde and UV-B irradiation cause DNA damage and affect genome stability, growth and development, but the interplay between these two genotoxic factors is poorly understood in plants. We show that Arabidopsis adh2/gsnor1 mutant, which lacks alcohol dehydrogenase 2/S-nitrosoglutathione reductase 1 (ADH2/GSNOR1), are hypersensitive to low fluence UV-B irradiation or UV-B irradiation-mimetic chemicals. Although the ADH2/GSNOR1 enzyme can act on different substrates, notably on S-hydroxymethylglutathione (HMG) and S-nitrosoglutathione (GSNO), our study provides several lines of evidence that the sensitivity of gsnor1 to UV-B is caused mainly by UV-B-induced formaldehyde accumulation rather than other factors such as alteration of the GSNO concentration. Our results demonstrate an interplay between formaldehyde and UV-B that exacerbates genome instability, leading to severe DNA damage and impaired growth and development in Arabidopsis, and show that ADH2/GSNOR1 is a key player in combating these effects.
    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