syntaxin 17

语法蛋白 17
  • 文章类型: Journal Article
    Syntaxin17(STX17)已被确定为介导自噬体和溶酶体融合的关键因素。然而,其在动脉粥样硬化(AS)背景下的具体参与尚不清楚.本研究旨在阐明STX17在AS的开始和进展中的作用和机制贡献。利用体内和体外AS模型系统,我们采用高脂饮食的ApoE基因敲除(KO)小鼠和用氧化低密度脂蛋白(ox-LDL)处理的人脐静脉内皮细胞(HUVECs)评估STX17表达.为了调查潜在的机制,我们使用shRNA-STX17慢病毒敲低STX17表达,然后评估HUVECs中的自噬和炎症。在体内和体外AS模型中,STX17表达显著上调。STX17的击倒加剧了HUVEC的损伤,无论是否使用ox-LDL治疗。此外,我们观察到STX17敲低损害了自噬体降解,阻碍自噬通量,并导致HUVEC中功能失调的溶酶体积累。此外,STX17敲低增强了ox-LDL治疗后HUVECs的炎症反应。进一步的机制探索揭示了STX17和STING之间的关联;减少STX17表达增加STING水平。进一步敲除STING增强自噬通量。总之,我们的研究结果表明,STX17敲低通过阻碍自噬通量和放大炎症反应而使AS恶化.此外,STX17与STING之间的相互作用可能在STX17介导的自噬中起关键作用。
    Syntaxin 17 (STX17) has been identified as a crucial factor in mediating the fusion of autophagosomes and lysosomes. However, its specific involvement in the context of atherosclerosis (AS) remains unclear. This study sought to elucidate the role and mechanistic contributions of STX17 in the initiation and progression of AS. Utilizing both in vivo and in vitro AS model systems, we employed ApoE knockout (KO) mice subjected to a high-fat diet and human umbilical vein endothelial cells (HUVECs) treated with oxidized low-density lipoprotein (ox-LDL) to assess STX17 expression. To investigate underlying mechanisms, we employed shRNA-STX17 lentivirus to knock down STX17 expression, followed by evaluating autophagy and inflammation in HUVECs. In both in vivo and in vitro AS models, STX17 expression was significantly upregulated. Knockdown of STX17 exacerbated HUVEC damage, both with and without ox-LDL treatment. Additionally, we observed that STX17 knockdown impaired autophagosome degradation, impeded autophagy flux and also resulted in the accumulation of dysfunctional lysosomes in HUVECs. Moreover, STX17 knockdown intensified the inflammatory response following ox-LDL treatment in HUVECs. Further mechanistic exploration revealed an association between STX17 and STING; reducing STX17 expression increased STING levels. Further knockdown of STING enhanced autophagy flux. In summary, our findings suggest that STX17 knockdown worsens AS by impeding autophagy flux and amplifying the inflammatory response. Additionally, the interaction between STX17 and STING may play a crucial role in STX17-mediated autophagy.
    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:自噬缺陷与甲基苯丙胺(Meth)诱导的神经毒性有关。语法17(Stx17),SNARE蛋白家族的一员,参与自噬的几个阶段,包括自噬-晚期内体/溶酶体融合。然而,Stx17在Meth诱导的自噬缺陷中的作用和潜在机制仍然知之甚少。
    方法:为了解决Meth引起的认知障碍的机制,将腺病毒(AV)和腺相关病毒(AAV)注射到海马体以进行立体定位,从而在体内过表达Stx17,通过Morris水迷宫和新物体识别来检查认知能力.在分子水平上,评估了突触损伤和自噬缺陷。为了解决Meth引起的神经元损伤,进行表皮生长因子受体(EGFR)降解试验以评估由Meth介导的“货物”的降解性,在机械上,囊泡的成熟,包括自噬体和内体,通过Co-IP和GTP-琼脂糖亲和分离试验验证。
    结果:在海马中Stx17的过表达明显地挽救了Meth诱导的认知障碍和突触丢失。对于内体,甲基暴露上调Rab5表达及其鸟嘌呤-核苷酸交换因子(GEF)(未成熟核内体),与相应的Rab7活性形式(Rab7-GTP)减少,并阻碍Rab7与CCZ1(成熟内体)的结合;对于自噬体,Meth治疗导致Stx17和自噬体之间的重叠显着减少,但增加了ATG5和自噬体(未成熟的自噬体)的共定位。Stx17过表达后,纯化的晚期内体中的Rab7-GTP水平与成熟自噬体升高平行大幅增加,便利货物(Aβ42,p-tau,和EGFR)在囊泡中降解,最终改善了Meth诱导的小鼠突触丧失和记忆障碍。
    结论:Meth介导的Stx17降低导致囊泡融合缺陷,这可能归因于未成熟的自噬体和内体,导致自噬功能障碍,并最终导致神经元损伤和认知障碍。因此,靶向Stx17可能是Meth诱导的神经元损伤的新治疗策略。
    BACKGROUND: Autophagic defects are involved in Methamphetamine (Meth)-induced neurotoxicity. Syntaxin 17 (Stx17), a member of the SNARE protein family, participating in several stages of autophagy, including autophagosome-late endosome/lysosome fusion. However, the role of Stx17 and potential mechanisms in autophagic defects induced by Meth remain poorly understood.
    METHODS: To address the mechanism of Meth-induced cognitive impairment, the adenovirus (AV) and adeno-associated virus (AAV) were injected into the hippocampus for stereotaxis to overexpress Stx17 in vivo to examine the cognitive ability via morris water maze and novel object recognition. In molecular level, the synaptic injury and autophagic defects were evaluated. To address the Meth induced neuronal damage, the epidermal growth factor receptor (EGFR) degradation assay was performed to evaluate the degradability of the \"cargos\" mediated by Meth, and mechanistically, the maturation of the vesicles, including autophagosomes and endosomes, were validated by the Co-IP and the GTP-agarose affinity isolation assays.
    RESULTS: Overexpression of Stx17 in the hippocampus markedly rescued the Meth-induced cognitive impairment and synaptic loss. For endosomes, Meth exposure upregulated Rab5 expression and its guanine-nucleotide exchange factor (GEF) (immature endosome), with a commensurate decreased active form of Rab7 (Rab7-GTP) and impeded the binding of Rab7 to CCZ1 (mature endosome); for autophagosomes, Meth treatment elicited a dramatic reduction in the overlap between Stx17 and autophagosomes but increased the colocalization of ATG5 and autophagosomes (immature autophagosomes). After Stx17 overexpression, the Rab7-GTP levels in purified late endosomes were substantially increased in parallel with the elevated mature autophagosomes, facilitating cargo (Aβ42, p-tau, and EGFR) degradation in the vesicles, which finally ameliorated Meth-induced synaptic loss and memory deficits in mice.
    CONCLUSIONS: Stx17 decrease mediated by Meth contributes to vesicle fusion defects which may ascribe to the immature autophagosomes and endosomes, leading to autophagic dysfunction and finalizes neuronal damage and cognitive impairments. Therefore, targeting Stx17 may be a novel therapeutic strategy for Meth-induced neuronal injury.
    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:未矫正的肥胖伴随着心脏不利的结构和功能变化,被称为肥胖心肌病。最近的证据表明,线粒体相关的内质网膜(MAMs)在肥胖引起的心脏并发症中起着至关重要的作用。Syntaxin17(STX17)充当位于MAM上的支架分子,尽管其在肥胖心脏并发症中的作用仍然难以捉摸。
    方法:本研究使用他莫昔芬诱导的心脏特异性STX17基因敲除(STX17cko)和使用静脉注射重组腺相关病毒血清型9(AAV9-cTNT-STX17)的STX17过表达小鼠,研究了STX17在HFD诱导的肥胖心肌病中的MAMs和线粒体Ca2+稳态中的作用。
    结果:STX17水平在肥胖患者的血浆和HFD喂养小鼠的心脏组织中显著升高。我们的数据显示,心脏STX17基因敲除减轻了肥胖心脏的心脏重塑和功能障碍,本身没有引起任何显著的影响。而STX17过表达加重了肥胖小鼠的心功能障碍。STX17缺失和STX17过表达消失和加剧,分别,HFD诱导的心脏氧化应激(O2-产生)和线粒体损伤。此外,STX17转染促进肥胖诱导的心肌细胞MAMs的形成,并引起过量的线粒体Ca2+流入,依赖于通过Habc结构域与线粒体Ca2+单转体显性阴性β(MCUb)的相互作用。我们的数据还表明,面对棕榈酸酯的挑战,STX17通过E3连接酶parkin促进了MCUb的泛素化和降解。
    结论:综合来看,我们的结果确定了STX17在促进肥胖诱导的MAMs形成中的新作用,随后线粒体Ca2+过载,线粒体O2积累,脂质过氧化,导致心脏损伤。我们的发现表明了在肥胖中靶向STX17的治疗前景。
    Uncorrected obesity is accompanied by unfavorable structural and functional changes in the heart, known as obesity cardiomyopathy. Recent evidence has revealed a crucial role for mitochondria-associated endoplasmic reticulum membranes (MAMs) in obesity-induced cardiac complication. Syntaxin 17 (STX17) serves as a scaffolding molecule localized on MAMs although its role in obesity heart complication remains elusive.
    This study examined the role of STX17 in MAMs and mitochondrial Ca2+ homeostasis in HFD-induced obesity cardiomyopathy using tamoxifen-induced cardiac-specific STX17 knockout (STX17cko) and STX17 overexpression mice using intravenously delivered recombinant adeno-associated virus serotype-9 (AAV9-cTNT-STX17).
    STX17 levels were significantly elevated in plasma from obese patients and heart tissues of HFD-fed mice. Our data revealed that cardiac STX17 knockout alleviated cardiac remodeling and dysfunction in obese hearts without eliciting any notable effect itself, while STX17 overexpression aggravated cardiac dysfunction in obese mice. STX17 deletion and STX17 overexpression annihilated and aggravated, respectively, HFD-induced oxidative stress (O2- production) and mitochondrial injury in the heart. Furthermore, STX17 transfection facilitated obesity-induced MAMs formation in cardiomyocytes and evoked excess mitochondrial Ca2+ influx, dependent upon interaction with mitochondrial Ca2+ uniporter dominant negative β (MCUb) through Habc domain. Our data also suggested that STX17 promoted ubiquitination and degradation of MCUb through the E3 ligase parkin in the face of palmitate challenging.
    Taken together, our results identified a novel role for STX17 in facilitating obesity-induced MAMs formation, and subsequently mitochondrial Ca2+ overload, mitochondrial O2- accumulation, lipid peroxidation, resulting in cardiac impairment. Our findings denoted therapeutic promises of targeting STX17 in obesity.
    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
    猪繁殖与呼吸综合征病毒(PRRSV)是一种经济上重要的病原体,30多年来一直困扰着全球养猪业。自噬是一种进化保守的细胞内溶酶体降解途径,和以前的研究已经证明PRRSV感染促进自噬体积累。然而,PRRSV诱导完全自噬还是不完全自噬仍存在争议.这里,我们证明了PRRSV非结构蛋白5(nsp5)的过表达诱导自噬体的积累,在PRRSV感染的细胞中观察到类似的情况。此外,PRRSV感染和nsp5过表达都激活了不完全自噬,自噬体-溶酶体融合的阻断证明了这一点。机械上,nsp5过表达,以及PRRSV感染,抑制突触体蛋白17(STX17)与突触体相关蛋白29(SNAP29)的相互作用,两种介导自噬体与溶酶体融合的SNARE蛋白,损害自体溶酶体的形成。我们进一步证实nsp5与STX17而不是SANP29相互作用,并且STX17的相互作用结构域是N末端基序和SNARE基序。一起来看,我们的研究结果提示了PRRSV通过阻断自噬体降解诱导不完全自噬的机制,并为开发对抗PRRSV感染的新疗法提供了见解.重要性已经证明大量病毒利用或劫持自噬来促进其复制。在猪繁殖与呼吸综合征病毒(PRRSV)的情况下,先前的研究已经证明了自噬对PRRSV增殖的前病毒作用。因此,对PRRSV调节自噬过程的机制的研究可以为病毒发病机制提供新的见解.自噬通量是由自噬体形成和随后的溶酶体降解组成的动态过程。然而,PRRSV感染对自噬通量的确切影响仍存在争议。在这项研究中,我们证明PRRSV感染,以及PRRSVnsp5过表达,抑制STX17与SNAP29的相互作用,损害自噬体与溶酶体的融合,从而阻断自噬通量。这些信息将帮助我们了解PRRSV与宿主的相互作用,并揭示PRRS预防和控制的新目标。
    Porcine reproductive and respiratory syndrome virus (PRRSV) is an economically important pathogen that has devastated the worldwide swine industry for over 30 years. Autophagy is an evolutionarily conserved intracellular lysosomal degradation pathway, and previous studies have documented that PRRSV infection prompts autophagosome accumulation. However, whether PRRSV induces complete or incomplete autophagy remains controversial. Here, we demonstrated that overexpression of PRRSV nonstructural protein 5 (nsp5) induced the accumulation of autophagosomes, and a similar scenario was observed in PRRSV-infected cells. Moreover, both PRRSV infection and nsp5 overexpression activated incomplete autophagy, as evidenced by the blockage of autophagosome-lysosome fusion. Mechanistically, nsp5 overexpression, as well as PRRSV infection, inhibited the interaction of syntaxin 17 (STX17) with synaptosomal-associated protein 29 (SNAP29), two SNARE proteins that mediate autophagosome fusion with lysosomes, to impair the formation of autolysosomes. We further confirmed that nsp5 interacted with STX17, rather than SANP29, and the interacting domains of STX17 were the N-terminal motif and SNARE motif. Taken together, the findings of our study suggest a mechanism by which PRRSV induces incomplete autophagy by blocking autophagosome degradation and provide insights into the development of new therapeutics to combat PRRSV infection. IMPORTANCE A substantial number of viruses have been demonstrated to utilize or hijack autophagy to benefit their replication. In the case of porcine reproductive and respiratory syndrome virus (PRRSV), previous studies have demonstrated the proviral effects of autophagy on PRRSV proliferation. Thus, an investigation of the mechanism by which PRRSV regulates the autophagy processes can provide new insight into viral pathogenesis. Autophagic flux is a dynamic process that consists of autophagosome formation and subsequent lysosomal degradation. However, the exact effect of PRRSV infection on the autophagic flux remains disputed. In this study, we demonstrated that PRRSV infection, as well as PRRSV nsp5 overexpression, inhibited the interaction of STX17 with SNAP29 to impair the fusion of autophagosomes with lysosomes, thereby blocking autophagic flux. This information will help us to understand PRRSV-host interactions and unravel new targets for PRRS prevention and 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
    Methamphetamine (METH), a psychoactive-stimulant facilitates massive accumulation of autophagosomes and causes autophagy-associated neuronal death. However, the underlying mechanisms involving METH-induced auto-phagosome accumulation remain poorly understood. In the current study, autophagic flux was tracked by mRFP-GFP-LC3 adenovirus, 900 μM METH treatment was found to significantly disrupt autophagic flux, which was further validated by remarkable increase of co-localized of LC3 and SQSTM1/p62, enhancement of LC3-II and SQSTM1/p62 protein levels, and massive autophagosome puncta aggregation. With the cycloheximide (CHX) treatment, METH treatment was displayed a significant inhibition of SQSTM1/p62 degradation. Therefore, the mRNAs associated with vesicle degradation were screened, and syntaxin 17 (Stx17) and dynein-dynactin mRNA levels significantly decreased, an effect was proved in protein level as well. Intriguingly, METH induced autophagosome accumulation and autophagic flux disturbance was incredibly retarded by overexpression of Stx17, which was validated by the restoration of the fusion autophagosome-late endosome/lysosome fusion. Moreover, Stx17 overexpression obviously impeded the METH-induced decrease of co-localization of the retrograded motor protein dynein/dynactin and autophagosome-late endosome, though the dynein/dynactin proteins were not involved in autophagosome-late endosome/lysosome fusion. Collectively, our findings unravel the mechanism of METH-induced autophagosome accumulation involving autophagosome-late endosome/lysosome fusion deficiency and that autophagy-enhancing mechanisms such as the overexpression of Stx17 may be therapeutic strategies for the treatment of METH-induced neuronal 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: Journal Article
    Previous studies have shown that syntaxin 17 (STX17) is involved in mediating the fusion of autophagosomes and lysosomes. This study aimed to investigate the role and mechanism of STX17 in neuronal injury following cerebral ischemia/reperfusion. The ischemia/reperfusion (I/R) models were established by transient middle cerebral artery occlusion (tMCAO) in mice and oxygen glucose deprivation/reperfusion (O/R) in primary cultured cortical neurons and HT22 cells. Cerebral ischemia/reperfusion significantly up-regulated the expression of STX17 in neurons. Lentivirus mediated knockdown of STX17 in neurons reduced neuronal viability and increased LDH leakage. Injection of AAV9-shSTX17 into the brain of mice then subjected to tMCAO also significantly augmented the infarct area and exacerbated neurobehavioral deficits and mortality. Depletion of STX17 caused accumulation of autophagic marker/substrate LC3 II and p62, blockade of the autophagic flux, and the accumulation of dysfunctional lysosomes. Knockdown of STX17 also aggravated endoplasmic reticulum (ER) stress-dependent neuronal apoptosis induced by ischemia/reperfusion. Importantly, induction of autophagy-lysosomal pathway and alleviation of ER stress partially rescued STX17 knockdown-induced neuronal damage. These results suggest that STX17 may ameliorate ischemia/reperfusion-induced neuronal damage by enhancing autophagy flux and reducing ER stress-dependent neuronal apoptosis.
    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
    Picornaviruses have evolved to hijack host cellular machinery, including the autophagic pathway. However, the mechanisms remain largely unclear. We use coxsackievirus B3 (CVB3) as a model organism to explore the possible role of picornavirus subversion of the autophagic pathway in viral infection. Our in vivo and in vitro experiments demonstrate that CVB3 infection causes a significant, albeit incomplete, inhibition of autophagic flux by limiting the fusion of autophagosomes with lysosomes and/or late endosomes. Furthermore, we show that CVB3 specifically targets SNARE protein SNAP29 and adaptor protein PLEKHM1, two critical proteins known to regulate autophagosome fusion, for cleavage through the catalytic activity of viral proteinase 3C, ultimately impairing the formation of SNARE complexes. Finally, we demonstrate that loss of SNAP29/PLEKHM1 inhibits autophagic flux, resulting in increased viral replication. Collectively, our study reveals a mechanism that supports an emerging model whereby CVB3 hijacks the autophagic machinery to facilitate its own propagation.
    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