FBXO3

FBXO3
  • 文章类型: Journal Article
    MALT1已被认为是免疫细胞和肿瘤中NF-κB信号传导的上游调节剂。本研究确定了MALT1在非小细胞肺癌(NSCLC)中的调控机制和生物学功能。在细胞培养和原位异种移植模型中,通过基因表达干扰或蛋白活性抑制的MALT1抑制显著损害NSCLC细胞的恶性表型和增强的放射敏感性。CSN5,COP9信号体的核心亚基,首先验证了通过干扰与E3连接酶FBXO3的相互作用来稳定MALT1。FBXO3在NSCLC细胞中的丢失减少了MALT1的泛素化并促进其积累,被CSN5干扰逆转。CSN5/FBXO3/MALT1调节轴与NSCLC患者的不良预后之间的关联被确定。我们的发现揭示了NF-κB信号传导中持续MALT1激活的详细机制,强调其作为NSCLC预测因子和潜在治疗靶点的意义。
    MALT1 has been implicated as an upstream regulator of NF-κB signaling in immune cells and tumors. This study determined the regulatory mechanisms and biological functions of MALT1 in non-small cell lung cancer (NSCLC). In cell culture and orthotopic xenograft models, MALT1 suppression via gene expression interference or protein activity inhibition significantly impaired malignant phenotypes and enhanced radiation sensitivity of NSCLC cells. CSN5, the core subunit of COP9 signalosome, was firstly verified to stabilize MALT1 via disturbing the interaction with E3 ligase FBXO3. Loss of FBXO3 in NSCLC cells reduced MALT1 ubiquitination and promoted its accumulation, which was reversed by CSN5 interference. An association between CSN5/FBXO3/MALT1 regulatory axis and poor prognosis in NSCLC patients was identified. Our findings revealed the detail mechanism of continuous MALT1 activation in NF-κB signaling, highlighting its significance as predictor and potential therapeutic target in NSCLC.
    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:脓毒症诱导的急性肺损伤(ALI)是严重危及生命的脓毒症并发症之一,在病理上与线粒体功能障碍有关。人参皂苷Rg1对ALI有较好的治疗作用。在这里,研究了Rg1在脓毒症诱导的ALI中的药理作用。
    方法:采用CLP手术和LPS处理建立脓毒症诱导的ALI模型。HE染色分析肺部病理改变。通过RT-qPCR和ELISA检测细胞因子的表达和分泌。通过MTT法评估细胞活力和凋亡,流式细胞术和TUNEL染色。使用DHE探针和JC-1染色分析ROS水平和线粒体膜电位(MMP),分别。使用MeRIP测定法评估FBXO3m6A水平。通过Co-IP或RIP分析了FBXO3,YTHDF1和PGC-1α之间的相互作用。
    结果:Rg1给药可改善LPS诱导的上皮细胞炎症,凋亡,和线粒体功能障碍呈剂量依赖性。机械上,Rg1通过依赖性抑制FBXO3表达m6A-YTHDF1降低PGC-1α泛素化修饰水平。不出所料,Rg1对LPS诱导的炎症的缓解作用,FBXO3过表达消除了肺上皮细胞的凋亡和线粒体功能障碍。此外,FBXO3上调消除了Rg1对CLP诱导的大鼠肺损伤的恢复作用。
    结论:Rg1以m6A-YTHDF1依赖性方式通过降低FBXO3稳定性激活PGC-1α/Nrf2信号通路,从而改善脓毒症诱导的ALI进展过程中肺上皮细胞的线粒体功能。
    Sepsis-induced acute lung injury (ALI) is one of the serious life-threatening complications of sepsis and is pathologically associated with mitochondrial dysfunction. Ginsenoside Rg1 has good therapeutic effects on ALI. Herein, the pharmacological effects of Rg1 in sepsis-induced ALI were investigated.
    Sepsis-induced ALI models were established by CLP operation and LPS treatment. HE staining was adopted to analyze lung pathological changes. The expression and secretion of cytokines were measured by RT-qPCR and ELISA. Cell viability and apoptosis were assessed by MTT assay, flow cytometry and TUNEL staining. ROS level and mitochondrial membrane potential (MMP) were analyzed using DHE probe and JC-1 staining, respectively. FBXO3 m6A level was assessed using MeRIP assay. The interactions between FBXO3, YTHDF1, and PGC-1α were analyzed by Co-IP or RIP.
    Rg1 administration ameliorated LPS-induced epithelial cell inflammation, apoptosis, and mitochondrial dysfunction in a dose-dependent manner. Mechanically, Rg1 reduced PGC-1α ubiquitination modification level by inhibiting FBXO3 expression m6A-YTHDF1 dependently. As expected, Rg1\'s mitigative effect on LPS-induced inflammation, apoptosis and mitochondrial dysfunction in lung epithelial cells was abolished by FBXO3 overexpression. Moreover, FBXO3 upregulation eliminated the restoring effect of Rg1 on CLP-induced lung injury in rats.
    Rg1 activated PGC-1α/Nrf2 signaling pathway by reducing FBXO3 stability in an m6A-YTHDF1-dependent manner to improve mitochondrial function in lung epithelial cells during sepsis-induced ALI progression.
    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
    BC-1215,双吡啶基苄基乙二胺,是仅F-box蛋白3(FBXO3)的抑制剂,并发挥抗炎作用。BC-1215抑制FBXO3-F-box与富含亮氨酸的重复蛋白2(FBXL2)之间的相互作用,导致FBXL2表达上调,FBXL2介导的泛素化和肿瘤坏死因子受体(TNFR)相关因子6(TRAF6)或NOD-降解,LRR-和含pyrin结构域的蛋白3(NLRP3),这随后导致炎症细胞因子产生的下调。在目前的研究中,我们研究了BC-1215是否或如何抑制LPS引发的人巨噬细胞样细胞中ATP诱导的IL-1β分泌的问题,THP-1细胞。我们的结果表明,用BC-1215预处理会减弱ATP诱导的LPS引发的THP-1细胞中IL-1β的分泌。用BC-1215处理LPS引发的THP-1细胞导致蛋白质水平的NLRP3和pro-IL-1β水平降低,但不是在mRNA水平。此外,用MG-132治疗,而不是leupeptin,抑制BC-1215诱导的NLRP3和pro-IL-1β蛋白降解,并恢复了他们的水平,表明BC-1215通过蛋白酶体依赖性降解在蛋白质水平降低NLRP3和pro-IL-1β的稳定性。我们的结果还表明,被BC-1215增加的FBXL2与NLRP3和pro-IL-1β结合并被泛素化,但不是半胱天冬酶-1。这些共同的结果表明,在LPS引发的THP-1细胞中,用FBXO3抑制剂BC-1215通过FBXL2介导的泛素化和pro-IL-1β的降解以及NLRP3抑制ATP诱导的IL-1β分泌。这表明FBXO3是开发抗炎症性疾病药物的潜在治疗靶点。
    BC-1215, bis-pyridinyl benzyl ethanediamine, is an inhibitor of F-box only protein 3 (FBXO3) and exerts anti-inflammatory effects. BC-1215 inhibits interactions between FBXO3-F-box and the leucine rich repeat protein 2 (FBXL2), leading to the upregulation of FBXL2 expression, FBXL2-mediated ubiquitination and the degradation of tumor necrosis factor receptor (TNFR)-associated factor 6 (TRAF6) or NOD-, LRR- and the pyrin domain-containing protein 3 (NLRP3), which subsequently results in the down-regulation of inflammatory cytokine production. In the current study, we investigated the issue of whether or how BC-1215 suppresses the ATP-induced secretion of IL-1β in LPS-primed human macrophage-like cells, THP-1 cells. Our result show that pre-treatment with BC-1215 attenuated the ATP-induced secretion of IL-1β in LPS-primed THP-1 cells. Treatment of the LPS-primed THP-1 cells with BC-1215 resulted in a decrease in the level of NLRP3 and pro-IL-1β at the protein level, but not at the mRNA level. In addition, treatment with MG-132, but not leupeptin, inhibited the BC-1215-induced degradation of NLRP3 and pro-IL-1β proteins, and restored their levels, suggesting that BC-1215 decreases the stability of NLRP3 and pro-IL-1β at the protein level via proteasome-dependent degradation. Our results also show that FBXL2, which is increased by BC-1215, bound to and ubiquitinated NLRP3 and pro-IL-1β, but not pro-caspase-1. These collective results indicate that treatment with BC-1215, an inhibitor of FBXO3, inhibits ATP-induced IL-1β secretion via the FBXL2-mediated ubiquitination and degradation of pro-IL-1β as well as NLRP3 in LPS-primed THP-1 cells, suggesting that FBXO3 is a potential therapeutic target for developing agents against inflammatory 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
    缺血性中风,人类死亡和发病的最普遍原因之一,病理特征是炎症级联反应,特别是在缺血/再灌注(I/R)损伤的进展过程中。F-盒蛋白3(FBXO3),SKP1-cullin1-F-box蛋白(SCF)E3连接酶复合物的底物识别亚基,最近已被证明在多种疾病的病理过程中作为潜在的促炎因子被切断。鉴于这些考虑,本研究旨在调查FBXO3是否对脑I/R损伤中的炎症产生影响。在这项研究中,首先,已证实,在Sprague-Dawley(SD)大鼠中大脑中动脉阻塞/再灌注(MCAO/R)模型后,FBXO3蛋白表达增加,并且在小胶质细胞或星形胶质细胞以外的神经元中特异性表达。同时,在小鼠海马神经元细胞系HT22细胞中,氧和葡萄糖剥夺/复氧(OGD/R)治疗后观察到FBXO3蛋白的升高。还发现用siRNA干扰FBXO3通过抑制体内和体外I/R损伤中的炎症反应而显著减轻神经元损伤。FBXO3抑制剂BC-1215也用于证实FBXO3在OGD/R模型中的促炎作用。此外,通过给予FBXO3siRNA和BC-1215,FBXO3被证实可以降低同源结构域相互作用蛋白激酶2(HIPK2)的蛋白质水平,可能通过泛素-蛋白酶体系统(UPS),加重脑I/R损伤。总的来说,我们的结果强调了FBXO3通过加速炎症反应对脑I/R损伤的不利影响,可能通过泛素化和降解HIPK2。尽管FBXO3和HIPK2之间的特定相互作用需要进一步研究,我们认为,我们的数据表明FBXO3与缺血性卒中的治疗相关性,并为I/R损伤的机制提供了新的视角.
    Ischemic stroke, one of the most universal causes of human mortality and morbidity, is pathologically characterized by inflammatory cascade, especially during the progression of ischemia/reperfusion (I/R) injury. F-Box Protein 3 (FBXO3), a substrate-recognition subunit of SKP1-cullin 1-F-box protein (SCF) E3 ligase complexes, has recently been proven to be severed as an underlying pro-inflammatory factor in pathological processes of diverse diseases. Given these considerations, the current study aims at investigating whether FBXO3 exerts impacts on inflammation in cerebral I/R injury. In this study, first, it was verified that FBXO3 protein expression increased after a middle cerebral artery occlusion/reperfusion (MCAO/R) model in Sprague-Dawley (SD) rats and was specifically expressed in neurons other than microglia or astrocytes. Meanwhile, in mouse hippocampal neuronal cell line HT22 cells, the elevation of FBXO3 protein was observed after oxygen and glucose deprivation/reoxygenation (OGD/R) treatment. It was also found that interference of FBXO3 with siRNA significantly alleviated neuronal damage via inhibiting the inflammatory response in I/R injury both in vivo and in vitro. The FBXO3 inhibitor BC-1215 was used to confirm the pro-inflammatory effect of FBXO3 in the OGD/R model as well. Furthermore, by administration of FBXO3 siRNA and BC-1215, FBXO3 was verified to reduce the protein level of Homeodomain-Interacting Protein Kinase 2 (HIPK2), likely through the ubiquitin-proteasome system (UPS), to aggravate cerebral I/R injury. Collectively, our results underline the detrimental effect FBXO3 has on cerebral I/R injury by accelerating inflammatory response, possibly through ubiquitylating and degrading HIPK2. Despite the specific interaction between FBXO3 and HIPK2 requiring further study, we believe that our data suggest the therapeutic relevance of FBXO3 to ischemic stroke and provide a new perspective on the mechanism of I/R injury.
    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
    目的:这项工作旨在确定miR-219a-5p在调节膝骨关节炎(KOA)中的功能和机制。
    方法:采用脂多糖和三磷酸腺苷处理分离大鼠成纤维样滑膜细胞(FLSs),构建KOA细胞模型。通过转染调节FLS中的miR-219a-5p和FBXO3表达。流式细胞术用于研究FLS细胞凋亡。免疫荧光法研究了Caspase-1和IL-1β在FLSs中的表达。通过双荧光素酶报告基因测定鉴定miR-219a-5p与FBXO3之间的结合。构建KOA年夜鼠模子和miR-219a-5p上调KOA年夜鼠模子。分析大鼠的步长。大鼠膝关节经SafraninO-fast绿色染色评价膝关节损伤情况。FBXO3,焦亡相关蛋白,Westernblot检测大鼠FLS和关节软骨组织中IL-1β和IL-18的表达,qRT-PCR和酶联免疫吸附测定。
    结果:KOA细胞模型有较高的凋亡率,焦亡相关蛋白的表达,IL-1β和IL-18水平。miR-219a-5p上调降低了上述指标,而miR-219a-5p下调增加了上述指标。FBXO3表达被miR-219a-5p直接抑制。FBXO3的损失抑制了上述指标。FBXO3抵消了miR-219a-5p对上述指标的抑制。miR-219a-5pagomir减轻膝关节损伤,KOA大鼠的步长增加,并降低了KOA大鼠关节软骨组织中FBXO3,焦亡相关蛋白和IL-1β和IL-18的水平。
    结论:miR-219a-5p通过靶向FBXO3使NLRP3信号失活而抑制KOA的焦亡,这可能是临床上改善KOA的一个有希望的靶标。
    OBJECTIVE: This work was to identify the function and mechanism of miR-219a-5p in regulating knee osteoarthritis (KOA).
    METHODS: Rat fibroblast-like synoviocytes (FLSs) were isolated to construct KOA cell model by lipopolysaccharide and adenosine triphosphate treatment. miR-219a-5p and FBXO3 expression in FLSs was modulated by transfection. Flow cytometry was executed to research FLSs apoptosis. Caspase-1 and IL-1β expression in FLSs was researched by immunofluorescence. The binding between miR-219a-5p and FBXO3 was identified by dual luciferase reporter gene assay. KOA rat model and miR-219a-5p up-modulation KOA rat model were constructed. Step size of rats was analyzed. Knee joints of rats were experienced Safranin O-fast green staining to evaluate the knee joint injury. FBXO3, pyroptosis-associated proteins, and IL-1β and IL-18 expression in FLSs and articular cartilage tissues of rats were assessed by Western blot, qRT-PCR and Enzyme-linked immunosorbent assay.
    RESULTS: KOA cell model had higher apoptosis percentage, expression of pyroptosis-associated proteins, and IL-1β and IL-18 level. miR-219a-5p up-modulation decreased the above indicators, whereas miR-219a-5p down-modulation increased the above indicators. FBXO3 expression was directly repressed by miR-219a-5p. Loss of FBXO3 suppressed the above indicators. FBXO3 counteracted the suppression of miR-219a-5p on the above indicators. miR-219a-5p agomir attenuated knee joint injury, increased step size of KOA rats, and reduced FBXO3, pyroptosis-associated proteins and level of IL-1β and IL-18 in the articular cartilage tissues of KOA rats.
    CONCLUSIONS: miR-219a-5p suppressed the pyroptosis in KOA by inactivating the NLRP3 signaling via targeting FBXO3, which might be a promising target for ameliorating KOA in the clinic.
    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
    大量证据表明,F-box蛋白3(FBXO3)具有多种生物学功能,包括免疫病理的调节,神经病变和抗病毒反应。在这篇评论文章中,我们关注FBXO3在炎症性疾病和人类恶性肿瘤中的作用。我们还描述了FBXO3的底物,其有助于炎性疾病和癌症。我们强调,在类风湿性关节炎中经常观察到FBXO3的高表达,白血病,垂体腺瘤,和口腔鳞状细胞癌。此外,我们讨论了致癌物和癌症预防剂对FBXO3的调节。我们的综述全面了解了FBXO3在各种生物系统中的作用,并阐明了FBXO3如何在各种生理和病理过程中调节底物泛素化和降解。因此,FBXO3可以是治疗人类疾病包括癌症的新靶标。
    Accumulated evidence shows that the F-box protein 3 (FBXO3) has multiple biological functions, including regulation of immune pathologies, neuropathic diseases and antiviral response. In this review article, we focus on the role of FBXO3 in inflammatory disorders and human malignancies. We also describe the substrates of FBXO3, which contribute to inflammatory disorders and cancers. We highlight that the high expression of FBXO3 is frequently observed in rheumatoid arthritis, leukemia, pituitary adenoma, and oral squamous cell carcinoma. Moreover, we discuss the regulation of FBXO3 by both carcinogens and cancer preventive agents. Our review provides a comprehensive understanding of the role of FBXO3 in various biological systems and elucidates how FBXO3 regulates substrate ubiquitination and degradation during various physiological and pathological processes. Therefore, FBXO3 can be a novel target in the treatment of human diseases including carcinomas.
    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
    许多表观遗传调节因子与疼痛相关的脊柱可塑性有关。共激活剂相关精氨酸甲基转移酶1(CARM1),组蛋白精氨酸甲基化的表观遗传调节剂,是神经可塑性的一个非常有趣的目标。然而,其对脊髓可塑性相关神经性疼痛发展的潜在贡献仍未得到充分研究.这里,我们报道了神经损伤降低脊髓CARM1的表达并引起异常性疼痛。此外,通过Fbxo3介导的CARM1泛素化减少脊髓CARM1表达促进了K通道启动子处的H3R17me2减少,从而引起K+通道表观遗传沉默和神经性疼痛的发展。值得注意的是,在幼稚的老鼠中,使用CARM1siRNA或CARM1抑制剂降低脊髓CARM1导致相似的表观遗传信号和异常性疼痛.此外,鞘内施用BC-1215(一种新型Fbxo3抑制剂)可防止CARM1泛素化阻断K+通道基因沉默并改善神经损伤后的异常性疼痛。总的来说,结果表明,这种新发现的脊髓Fbxo3-CARM1-K通道基因功能轴促进神经性疼痛。这些发现提供了重要的见解,将有助于开发针对神经性疼痛的更有效和特定的疗法。
    Many epigenetic regulators are involved in pain-associated spinal plasticity. Coactivator-associated arginine methyltransferase 1 (CARM1), an epigenetic regulator of histone arginine methylation, is a highly interesting target in neuroplasticity. However, its potential contribution to spinal plasticity-associated neuropathic pain development remains poorly explored. Here, we report that nerve injury decreased the expression of spinal CARM1 and induced allodynia. Moreover, decreasing spinal CARM1 expression by Fbxo3-mediated CARM1 ubiquitination promoted H3R17me2 decrement at the K+ channel promoter, thereby causing K+ channel epigenetic silencing and the development of neuropathic pain. Remarkably, in naïve rats, decreasing spinal CARM1 using CARM1 siRNA or a CARM1 inhibitor resulted in similar epigenetic signaling and allodynia. Furthermore, intrathecal administration of BC-1215 (a novel Fbxo3 inhibitor) prevented CARM1 ubiquitination to block K+ channel gene silencing and ameliorate allodynia after nerve injury. Collectively, the results reveal that this newly identified spinal Fbxo3-CARM1-K+ channel gene functional axis promotes neuropathic pain. These findings provide essential insights that will aid in the development of more efficient and specific therapies against neuropathic pain.
    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
    BACKGROUND: miR-142-3p has been reported to be involved in several diseases, including cardiac hypertrophy and several tumors, but whether it exerts neuroprotective effects against cerebral ischemia/reperfusion (I/R) injury remains unclear.
    METHODS: In this study, the neuronal cell line SH-SY5Y was exposed to 6 hours of oxygen and glucose deprivation followed by 24 hours of reoxygenation (OGD/R) to mimic I/R injury in vitro. MiR-142-3p mimics were used to up-regulate the expression of miR-142-3p in SH-SY5Y cells before OGD/R induction. The changes in cell viability, apoptosis, and inflammation were assessed by CCK-8, lactate dehydrogenase (LDH), flow cytometry, and enzyme-linked immunosorbent assays.
    RESULTS: We found that miR-142-3p expression was decreased after OGD/R induction. miR-142-3p overexpression significantly protected SH-SY5Y cells against OGD/R-induced cell injury, as reflected by improved cell viability and reduced LDH leakage, proinflammatory cytokines, and apoptosis. Mechanistically, bioinformatics analysis and a dual luciferase reporter assay confirmed F-box protein 3 (FBXO3) as the target gene of miR-142-3p. Direct siRNA-mediated silencing of FBXO3 exerted a protective role against OGD/R-induced injury. Moreover, FBXO3 overexpression significantly reversed the protective effects of miR-142-3p against OGD/R-induced cell injury.
    CONCLUSIONS: miR-142-3p-mediated down-regulation of FBXO3 may be a potential agent for protection against cerebral I/R injury.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    Background: Increasing evidence suggests that Fbxo3 signaling has an important impact on the pathophysiology of the inflammatory process. Fbxo3 protein inhibition has reduced cytokine-driven inflammation and improved disease severity in animal model of Pseudomonas-induced lung injury. However, it remains unclear whether inhibition of Fbxo3 protein provides protection in acute lung injury induced by ischemia-reperfusion (I/R). In this study, we investigated the protective effects of BC-1215 administration, a Fbxo3 inhibitor, on acute lung injury induced by I/R in rats. Methods: Lung I/R injury was induced by ischemia (40 min) followed by reperfusion (60 min). The rats were randomly assigned into one of six experimental groups (n = 6 rats/group): the control group, control + BC-1215 (Fbxo3 inhibitor, 0.5 mg/kg) group, I/R group, or I/R + BC-1215 (0.1, 0.25, 0.5 mg/kg) groups. The effects of BC-1215 on human alveolar epithelial cells subjected to hypoxia-reoxygenation (H/R) were also examined. Results: BC-1215 significantly attenuated I/R-induced lung edema, indicated by a reduced vascular filtration coefficient, wet/dry weight ratio, lung injury scores, and protein levels in bronchoalveolar lavage fluid (BALF). Oxidative stress and the level of inflammatory cytokines in BALF were also significantly reduced following administration of BC-1215. Additionally, BC-1215 mitigated I/R-stimulated apoptosis, NF-κB, and mitogen-activated protein kinase activation in the injured lung tissue. BC-1215 increased Fbxl2 protein expression and suppressed Fbxo3 and TNFR associated factor (TRAF)1-6 protein expression. BC-1215 also inhibited IL-8 production and NF-κB activation in vitro in experiments with alveolar epithelial cells exposed to H/R. Conclusions: Our findings demonstrated that Fbxo3 inhibition may represent a novel therapeutic approach for I/R-induced lung injury, with beneficial effects due to destabilizing TRAF proteins.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Spinal plasticity, a key process mediating neuropathic pain development, requires ubiquitination-dependent protein turnover. Presynaptic active zone proteins have a crucial role in regulating vesicle exocytosis, which is essential for synaptic plasticity. Nevertheless, the mechanism for ubiquitination-regulated turnover of presynaptic active zone proteins in the progression of spinal plasticity-associated neuropathic pain remains unclear. Here, after research involving Sprague Dawley rats, we reported that spinal nerve ligation (SNL), in addition to causing allodynia, enhances the Rab3-interactive molecule-1α (RIM1α), a major active zone protein presumed to regulate neural plasticity, specifically in the synaptic plasma membranes (SPMs) of the ipsilateral dorsal horn. Spinal RIM1α-associated allodynia was mediated by Fbxo3, which abates Fbxl2-dependent RIM1α ubiquitination. Subsequently, following deubiquitination, enhanced RIM1α directly binds to CaV2.2, resulting in increased CaV2.2 expression in the SPMs of the dorsal horn. While exhibiting no effect on Fbxo3/Fbxl2 signaling, the focal knockdown of spinal RIM1α expression reversed the SNL-induced allodynia and increased spontaneous EPSC (sEPSC) frequency by suppressing RIM1α-facilitated CaV2.2 expression in the dorsal horn. Intrathecal applications of BC-1215 (a Fbxo3 activity inhibitor), Fbxl2 mRNA-targeting small-interfering RNA, and ω-conotoxin GVIA (a CaV2.2 blocker) attenuated RIM1α upregulation, enhanced RIM1α expression, and exhibited no effect on RIM1α expression, respectively. These results confirm the prediction that spinal presynaptic Fbxo3-dependent Fbxl2 ubiquitination promotes the subsequent RIM1α/CaV2.2 cascade in SNL-induced neuropathic pain. Our findings identify a role of the presynaptic active zone protein in pain-associated plasticity. That is, RIM1α-facilitated CaV2.2 expression plays a role in the downstream signaling of Fbxo3-dependent Fbxl2 ubiquitination/degradation to promote spinal plasticity underlying the progression of nociceptive hypersensitivity following neuropathic injury.
    Ubiquitination is a well known process required for protein degradation. Studies investigating pain pathology have demonstrated that ubiquitination contributes to chronic pain by regulating the turnover of synaptic proteins. Here, we found that the spinal presynaptic active zone protein Rab3-interactive molecule-1α (RIM1α) participates in neuropathic pain development by binding to and upregulating the expression of CaV2.2. In addition, Fbxo3 modifies this pathway by inhibiting Fbxl2-mediated RIM1α ubiquitination, suggesting that presynaptic protein ubiquitination makes a crucial contribution to the development of neuropathic pain. Research in this area, now in its infancy, could potentially provide a novel therapeutic strategy for pain relief.
    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