PIAS1

PIAS1
  • 文章类型: Journal Article
    缺血性中风是一种严重的疾病,发病率很高,致残率和死亡率。缺血性卒中涉及多种发病机制,如炎症和神经元细胞凋亡。激活的信号转导和转录激活因子1的蛋白质抑制剂(PIAS1)在各种生物过程中起着至关重要的作用,包括炎症。PIAS1在缺血再灌注损伤中也下调并参与疾病过程。然而,PIAS1在脑缺血中的作用尚不清楚.
    Sprague-Dawley(SD)大鼠大脑中动脉阻塞(MCAO)。通过Longa试验探讨PIAS1在缺血性脑梗死中的作用及机制,氯化2,3,5-三苯基四唑(TTC)染色,Morris水迷宫(MWM)试验,苏木精-伊红(HE)染色,脑含水量的量化,逆转录-定量聚合酶链反应(RT-qPCR),酶联免疫吸附测定(ELISA),末端脱氧核苷酸转移酶脱氧尿苷三磷酸(dUTP)缺口末端标记(TUNEL),蛋白质印迹和免疫荧光测定。
    与假手术大鼠相比,PIAS1在MCAO诱导大鼠中的表达下降。PIAS1的过表达降低了Longa神经评分,梗死面积的百分比,病理异常,游泳的逃避潜伏期和大脑含水量的百分比,并增加了MCAO诱导的大鼠的平台穿越次数和目标象限的时间。此外,PIAS1的过表达降低了MCAO诱导的IL-1β的含量,IL-6和TNF-α,但进一步升高血清和脑组织中IL-10的浓度。此外,PIAS1的过表达逆转了MCAO诱导的细胞凋亡率和Bax的相对蛋白水平,裂开的caspase3和Bcl-2。PIAS1的过表达也逆转了参与NF-κB途径的蛋白质水平。
    PIAS1减少炎症和细胞凋亡,从而通过调节NF-κB通路减轻MCAO诱导的大鼠缺血性脑梗死。
    UNASSIGNED: Ischemic stroke is a severe disorder with high incidence, disability rate and mortality. Multiple pathogenesis mechanisms are involved in ischemic stroke, such as inflammation and neuronal cell apoptosis. Protein inhibitor of activated signal transducer and activators of transcription 1 (PIAS1) plays a crucial role in various biological processes, including inflammation. PIAS1 is also downregulated in ischemia-reperfusion injury and involved in the disease processes. However, the role of PIAS1 in cerebral ischemia is unclear.
    UNASSIGNED: Sprague-Dawley (SD) rats were induced with middle cerebral artery occlusion (MCAO). The role and mechanisms of PIAS1 in ischemic cerebral infarction were explored by Longa test, 2,3,5-triphenyltetrazolium chloride (TTC) staining, Morris water maze (MWM) test, hematoxylin-eosin (HE) staining, quantification of brain water content, reverse transcription-quantitative polymerase chain reaction (RT-qPCR), enzyme-linked immunosorbent assay (ELISA), terminal deoxynucleotidyl transferase deoxyuridine triphosphate (dUTP) nick end labeling (TUNEL), Western blot and immunofluorescence assays.
    UNASSIGNED: The expression of PIAS1 in MCAO-induced rat was declined compared to sham rats. Overexpression of PIAS1 reduced the Longa neurological scores, the percent of infarction area, the pathological abnormality, the escape latency of swimming and the percent of brain water content, and increased the number of platform crossings and time in the target quadrant in the MCAO-induced rats. Besides, overexpression of PIAS1 decreased the MCAO-induced the contents of IL-1β, IL-6 and TNF-α, but further elevated the concentrations of IL-10 in both sera and brain tissues. Moreover, overexpression of PIAS1 reversed the MCAO-induced apoptosis rate and the relative protein level of Bax, cleaved caspase3 and Bcl-2. Overexpression of PIAS1 also reversed the level of proteins involved in NF-κB pathway.
    UNASSIGNED: PIAS1 reduced inflammation and apoptosis, thereby alleviating ischemic cerebral infarction in MCAO-induced rats through regulation NF-κB pathway.
    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:m6ARNA修饰通路在多种细胞过程和病毒生命周期中发挥重要作用。这里,我们研究了PIAS1与m6A阅读蛋白YTHDF2之间的关系,该蛋白通过与m6A修饰的RNA结合而参与调节RNA稳定性。我们发现YTHDF2的N端和C端区域都与PIAS1相互作用。我们显示PIAS1促进YTHDF2在三个特定赖氨酸残基处的SUMOylation。我们还证明PIAS1增强YTHDF2的抗EBV活性。我们进一步揭示了PIAS1介导其他YTHDF家族成员的SUMOylation,即,YTHDF1和YTHDF3,以限制EBV的复制。这些发现共同阐明了YTHDF蛋白在通过PIAS1介导的SUMO化控制病毒RNA衰变和EBV复制中的重要调节机制。
    YTH N6-methyladenosine RNA-binding protein F2 (YTHDF2) is a member of the YTH protein family that binds to N6-methyladenosine (m6A)-modified RNA, regulating RNA stability and restricting viral replication, including Epstein-Barr virus (EBV). PIAS1 is an E3 small ubiquitin-like modifier (SUMO) ligase known as an EBV restriction factor, but its role in YTHDF2 SUMOylation remains unclear. In this study, we investigated the functional regulation of YTHDF2 by PIAS1. We found that PIAS1 promotes the SUMOylation of YTHDF2 at three specific lysine residues (K281, K571, and K572). Importantly, PIAS1 synergizes with wild-type YTHDF2, but not a SUMOylation-deficient mutant, to limit EBV lytic replication. Mechanistically, YTHDF2 lacking SUMOylation exhibits reduced binding to EBV transcripts, leading to increased viral mRNA stability. Furthermore, PIAS1 mediates SUMOylation of YTHDF2\'s paralogs, YTHDF1 and YTHDF3, to restrict EBV replication. These results collectively uncover a unique mechanism whereby YTHDF family proteins control EBV replication through PIAS1-mediated SUMOylation, highlighting the significance of SUMOylation in regulating viral mRNA stability and EBV replication.IMPORTANCEm6A RNA modification pathway plays important roles in diverse cellular processes and viral life cycle. Here, we investigated the relationship between PIAS1 and the m6A reader protein YTHDF2, which is involved in regulating RNA stability by binding to m6A-modified RNA. We found that both the N-terminal and C-terminal regions of YTHDF2 interact with PIAS1. We showed that PIAS1 promotes the SUMOylation of YTHDF2 at three specific lysine residues. We also demonstrated that PIAS1 enhances the anti-EBV activity of YTHDF2. We further revealed that PIAS1 mediates the SUMOylation of other YTHDF family members, namely, YTHDF1 and YTHDF3, to limit EBV replication. These findings together illuminate an important regulatory mechanism of YTHDF proteins in controlling viral RNA decay and EBV replication through PIAS1-mediated SUMOylation.
    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
    PBRM1在上皮起源的癌症中经常突变。PBRM1如何调节正常上皮稳态,在癌症开始之前,尚不清楚。这里,我们显示PBRM1的基因调控作用在细胞状态之间有很大差异,利用人类皮肤上皮(表皮)作为研究平台。在祖细胞中,PBRM1的主要功能是抑制终末分化,以维持祖细胞的再生潜能;在分化状态下,然而,PBRM1向激活器切换。在这两种细胞状态之间,PBRM1保留其基因组结合,但与差异相互作用蛋白相关。我们的目标屏幕将E3SUMO连接酶PIAS1确定为关键相互作用者。PIAS1与PBRM1共同定位在染色质上,直接抑制祖细胞中的分化基因,和PIAS1的染色质结合在分化中急剧减少。此外,SUMO化有助于PBRM1在祖细胞维持中的抑制功能。因此,我们的研究结果强调了PBRM1的细胞状态特异性调控作用受其蛋白相互作用组的影响,尽管其稳定的染色质结合.
    PBRM1 is frequently mutated in cancers of epithelial origin. How PBRM1 regulates normal epithelial homeostasis, prior to cancer initiation, remains unclear. Here, we show that PBRM1\'s gene regulatory roles differ drastically between cell states, leveraging human skin epithelium (epidermis) as a research platform. In progenitors, PBRM1 predominantly functions to repress terminal differentiation to sustain progenitors\' regenerative potential; in the differentiation state, however, PBRM1 switches toward an activator. Between these two cell states, PBRM1 retains its genomic binding but associates with differential interacting proteins. Our targeted screen identified the E3 SUMO ligase PIAS1 as a key interactor. PIAS1 co-localizes with PBRM1 on chromatin to directly repress differentiation genes in progenitors, and PIAS1\'s chromatin binding drastically diminishes in differentiation. Furthermore, SUMOylation contributes to PBRM1\'s repressive function in progenitor maintenance. Thus, our findings highlight PBRM1\'s cell-state-specific regulatory roles influenced by its protein interactome despite its stable chromatin binding.
    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
    内皮功能障碍(ED)通常被认为是许多心血管疾病发病机理中的关键起始步骤。内皮型一氧化氮合酶(eNOS)的偶联对维持正常的内皮功能很重要。然而,eNOSSUMO化是否以及如何影响eNOS偶联仍然难以捉摸。在研究中,我们研究了激活的STAT1蛋白抑制剂(PIAS1)在ED中的作用和可能的作用机制。建立体外用棕榈酸(PA)处理的人脐静脉内皮细胞(HUVECs)和体内高脂饮食(HFD)喂养的ApoE-/-小鼠的ED模型。我们的体内数据表明,PIAS1通过增加一氧化氮(NO)水平减轻血管内皮功能障碍,降低丙二醛(MDA)水平,并激活ApoE-/-小鼠的磷脂酰肌醇3-激酶-蛋白激酶B-内皮型一氧化氮合酶(PI3K-AKT-eNOS)信号。我们的体外数据还表明,PIAS1可以在内源性条件下超糖基化eNOS;此外,它拮抗PA诱导的eNOS解偶联。结果表明,PIAS1通过促进SUMO化和抑制eNOS的解偶联减轻血管内皮功能障碍,提示PIAS1可能成为动脉粥样硬化的早期预测因子和高脂血症相关心血管疾病新的潜在靶点。
    Endothelial dysfunction (ED) is commonly considered a crucial initiating step in the pathogenesis of numerous cardiovascular diseases. The coupling of endothelial nitric oxide synthase (eNOS) is important in maintaining normal endothelial functions. However, it still remains elusive whether and how eNOS SUMOylation affects the eNOS coupling. In the study, we investigate the roles and possible action mechanisms of protein inhibitor of activated STAT 1 (PIAS1) in ED. Human umbilical vein endothelial cells (HUVECs) treated with palmitate acid (PA) in vitro and ApoE-/- mice fed with high-fat diet (HFD) in vivo were constructed as the ED models. Our in vivo data show that PIAS1 alleviates the dysfunction of vascular endothelium by increasing nitric oxide (NO) level, reducing malondialdehyde (MDA) level, and activating the phosphatidylinositol 3-kinase-protein kinase B-endothelial nitric oxide synthase (PI3K-AKT-eNOS) signaling in ApoE-/- mice. Our in vitro data also show that PIAS1 can SUMOylate eNOS under endogenous conditions; moreover, it antagonizes the eNOS uncoupling induced by PA. The findings demonstrate that PIAS1 alleviates the dysfunction of vascular endothelium by promoting the SUMOylation and inhibiting the uncoupling of eNOS, suggesting that PIAS1 would become an early predictor of atherosclerosis and a new potential target of the hyperlipidemia-related cardiovascular 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:已经推测波形蛋白作为上皮-间质转化(EMT)标志物的表达与非小细胞肺癌(NSCLC)的组织异质性和转移有关。
    方法:这项研究利用了在转化生长因子-β(TGF-β)信号通路中与针对STAT系统1型(PIAS1)或SMAD4蛋白抑制剂的小干扰RNA(siRNA)的体外共免疫沉淀结合SUMO化测定。
    结果:我们成功地证明了PIAS1通过形成复合物PIAS1-SUMO1-SMAD4蛋白复合物来增强SMAD4的SUMO化。这个,根据随后增加的波形蛋白微丝的生产,导致非小细胞肺癌(NSCLC)A549细胞系的迁移能力增强,从伤口愈合试验中观察到。
    结论:我们的结果进一步支持PIAS1介导的SUMO化SMAD4与波形蛋白下游过表达呈正相关。此外,观察到波形蛋白在这种细胞系中的过表达不一定与加速的相对伤口闭合有关,这引起了人们的担忧,即需要进一步探索以确认波形蛋白表达与NSCLC转移之间是否存在因果关系,如果是这样,波形蛋白在多大程度上有助于它。
    The expression of vimentin as a marker of epithelial-to-mesenchymal transition (EMT) has been speculated to be associated with tissue heterogeneity and metastases of non-small cell lung cancer (NSCLC).
    This study utilized in vitro co-immunoprecipitation with small interfering RNAs (siRNAs) against protein inhibitors of STAT system type 1 (PIAS1) or SMAD4 in transforming growth factor-beta (TGF-β) signaling pathway in combination with SUMOylation assay.
    We successfully demonstrated that PIAS1 enhanced SUMOylation of SMAD4 by forming a complex PIAS1-SUMO1-SMAD4 protein complex. This, in accordance with subsequently increased production of vimentin microfilaments, led to enhanced migration ability of non-small cell lung cancer (NSCLC) A549 line, observed from wound healing assay.
    Our results further supported the positive correlation of SUMOylated SMAD4 mediated by PIAS1 and downstream overexpression of vimentin. In addition, the observation that overexpression of vimentin in this certain cell line was not necessarily linked with accelerated relative wound closure raised concerns that further exploration will be needed to confirm if the causal relationship exists between vimentin expression and the metastases of NSCLC, and if so, to what extent vimentin contributes to it.
    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: Preprint
    YTHDF2是YTH蛋白家族的成员,与N6-甲基腺苷(m6A)修饰的RNA结合,调节RNA稳定性和限制病毒复制,包括EB病毒(EBV)。PIAS1是一种E3SUMO连接酶,被称为EBV限制因子,但其在YTHDF2磺酰化中的作用尚不清楚。在这项研究中,我们研究了PIAS1对YTHDF2的功能调节。我们发现PIAS1促进YTHDF2在三个特定赖氨酸残基(K281、K571和K572)处的SUMO化。重要的是,PIAS1增强YTHDF2的抗病毒活性,并且SUMO化缺陷型YTHDF2显示降低的抗EBV活性。机械上,缺乏SUMOylation的YTHDF2表现出与EBV转录本的结合减少,导致病毒mRNA稳定性增加。此外,PIAS1介导YTHDF2旁系同源物的亚甲基化,YTHDF1和YTHDF3。这些结果共同揭示了YTHDF2通过PIAS1介导的SUMOylation控制EBV复制的独特机制,强调SUMO化在调节病毒mRNA稳定性和EBV复制中的意义。
    N6-甲基腺苷(m6A)RNA修饰途径在多种细胞过程和病毒生命周期中起着重要作用。这里,我们研究了PIAS1与m6A阅读蛋白YTHDF2之间的关系,该蛋白通过与m6A修饰的RNA结合而参与调节RNA稳定性。我们发现YTHDF2的N端和C端区域都与PIAS1相互作用。我们显示PIAS1促进YTHDF2在三个特定赖氨酸残基处的SUMOylation。我们还证明PIAS1增强YTHDF2的抗爱泼斯坦-巴尔病毒(EBV)活性。我们进一步揭示了PIAS1介导其他YTHDF家族成员的SUMOylation,即YTHDF1和YTHDF3,因为它们与YTHDF2相似。这些发现共同阐明了YTHDF2通过PIAS1介导的SUMO化控制病毒RNA衰变和EBV复制的重要调节机制。
    YTHDF2 is a member of the YTH protein family that binds to N6-methyladenosine (m6A)-modified RNA, regulating RNA stability and restricting viral replication, including Epstein-Barr virus (EBV). PIAS1 is an E3 SUMO ligase known as an EBV restriction factor, but its role in YTHDF2 SUMOylation remains unclear. In this study, we investigated the functional regulation of YTHDF2 by PIAS1. We found that PIAS1 promotes the SUMOylation of YTHDF2 at three specific lysine residues (K281, K571, and K572). Importantly, PIAS1 enhances the antiviral activity of YTHDF2, and SUMOylation-deficient YTHDF2 shows reduced anti-EBV activity. Mechanistically, YTHDF2 lacking SUMOylation exhibits reduced binding to EBV transcripts, leading to increased viral mRNA stability. Furthermore, PIAS1 mediates SUMOylation of YTHDF2\'s paralogs, YTHDF1 and YTHDF3. These results collectively uncover a unique mechanism whereby YTHDF2 controls EBV replication through PIAS1-mediated SUMOylation, highlighting the significance of SUMOylation in regulating viral mRNA stability and EBV replication.
    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:急性胰腺炎(AP)是一种高死亡率的炎症性疾病。先前的研究表明环状RNA失调并参与AP中炎症反应的调节。本研究旨在探讨mmu_circ_0000037在caerulein诱导的AP细胞模型中的功能和调控机制。
    方法:Caerulein处理的MPC-83细胞用作AP的体外细胞模型。mmu_circ_0000037,microRNA(miR)-92a-3p,通过定量实时聚合酶链反应检测活化的STAT1蛋白抑制剂(Pias1)。细胞活力,淀粉酶活性,凋亡,3-(4,5-二甲基噻唑-2-基)-2,5-二苯基四唑溴化物检测到炎症反应,淀粉酶检测试剂盒,流式细胞术,和酶联免疫吸附测定。通过蛋白质印迹分析定量蛋白质水平。通过StarbaseV3.0预测miR-92a-3p与mmu_circ_0000037或Pias1之间的靶相互作用,并通过双荧光素酶报告基因测定和RNA免疫沉淀测定进行验证。
    结果:Mmu_circ_0000037和Pias1水平下降,而miR-92a-3p在caerulein诱导的MPC-83细胞中表达升高。mmu_circ_0000037的过表达保护了MPC-83细胞免受caerulein诱导的细胞活力降低,以及促进淀粉酶活性,细胞凋亡和炎症。mmu_circ_0000037靶向MiR-92a-3p,miR-92a-3p过表达挽救了mmu_circ_0000037对caerulein诱导的MPC-83细胞损伤的影响。Pias1被证实为miR-92a-3p的靶标,mmu_circ_0000037通过海绵作用miR-92a-3p调节Pias1的表达。
    结论:Mmu_circ_0000037通过靶向miR-92a-3p/Pias1轴来缓解菜青素诱导的MPC-83细胞炎症损伤,为AP的治疗提供理论依据。
    Acute pancreatitis (AP) is an inflammatory disease with high mortality. Previous study has suggested that circular RNAs are dysregulated and involved in the regulation of inflammatory responses in AP. This study aimed to investigate the function and regulatory mechanism underlying mmu_circ_0000037 in caerulein-induced AP cellular model.
    Caerulein-treated MPC-83 cells were used as an in vitro cellular model for AP. The expression levels of mmu_circ_0000037, microRNA (miR)-92a-3p, and protein inhibitor of activated STAT1 (Pias1) were detected by quantitative real-time polymerase chain reaction. Cell viability, amylase activity, apoptosis, and inflammatory response were detected by 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, Amylase Assay Kit, flow cytometry, and enzyme-linked immunosorbent assays. The protein level was quantified by western blot analysis. The target interaction between miR-92a-3p and mmu_circ_0000037 or Pias1 were predicted by StarbaseV3.0 and validated by dual-luciferase reporter assay and RNA immunoprecipitation assay.
    Mmu_circ_0000037 and Pias1 levels were decreased, whereas miR-92a-3p expression was elevated in caerulein-induced MPC-83 cells. Overexpression of mmu_circ_0000037 protected MPC-83 cells from caerulein-induced the decrease of cell viability, as well as the promotion of amylase activity, apoptosis and inflammation. MiR-92a-3p was targeted by mmu_circ_0000037, and miR-92a-3p overexpression rescued the effect of mmu_circ_0000037 on caerulein-induced MPC-83 cell injury. Pias1 was confirmed as a target of miR-92a-3p and mmu_circ_0000037 regulated the expression of Pias1 by sponging miR-92a-3p.
    Mmu_circ_0000037 relieves caerulein-induced inflammatory injury in MPC-83 cells by targeting miR-92a-3p/Pias1 axis, providing a theoretical basis for the treatment of AP.
    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
    最近,有证据表明核受体相互作用蛋白1(NRIP1)参与急性肺损伤(ALI)的进展,但具体机制尚不清楚。用pcDNA-NRIP1或si-NRIP1转染铜绿假单胞菌(PA)处理的TC-1细胞,我们发现过表达NRIP1抑制细胞活力,促进细胞凋亡和分泌炎症因子,si-NRIP1的转染逆转了这些作用。此外,在线生物信息学分析和免疫共沉淀分析结果表明,NRIP1能与泛素偶联酶E2I(UBE2I)结合,并推广了UBE2I表达式。接下来,PA处理的TC-1细胞用si-NRIP1单独或与pcDNA-UBE2I一起转染,我们观察到用si-NRIP1转染抑制UBE2I表达,促进细胞活力,减少细胞凋亡和炎症因子的分泌,UBE2I过表达可以逆转。此外,UBE2I可以与激活的信号转导和转录激活因子1(PIAS1)的蛋白质抑制剂结合。NRIP1过表达促进UBE2I表达,抑制PIAS1表达,NRIP1促进PIAS1泛素化和UBE2I降解。用si-UBE2I单独或与si-PIAS1一起转染PA处理的TC-1细胞,结果表明si-UBE2I的转染与si-NRIP1的转染具有相同的效果。最后,我们的体内发现表明NRIP1和UBE2I的表达降低,PIAS1表达增加,在NRIP1敲低的小鼠的肺组织中,肺组织炎性浸润减少。总之,我们的研究表明,NRIP1通过促进PIAS1泛素化来加重PA诱导的小鼠肺损伤.
    Recently, evidence has shown that nuclear receptor interacting protein 1 (NRIP1) is involved in acute lung injury (ALI) progression, but the specific mechanism remains unclear. Pseudomonas aeruginosa (PA)-treated TC-1 cells were transfected with pcDNA-NRIP1 or si-NRIP1, and we found that overexpression of NRIP1 inhibited cell viability and promoted cell apoptosis and secretion of inflammatory factors, and transfection of si-NRIP1 reversed these effects. Furthermore, online bioinformatics analysis and co-immunoprecipitation assay results indicated that NRIP1 could bind to Ubiquitin Conjugating Enzyme E2I (UBE2I), and promoted UBE2I expression. Next, the PA-treated TC-1 cells were transfected with si-NRIP1 alone or together with pcDNA-UBE2I, and we observed that transfection with si-NRIP1 inhibited UBE2I expression, promoted cell viability, and reduced cell apoptosis and inflammatory factor secretion, which could be reversed by UBE2I overexpression. Moreover, UBE2I could bind to protein inhibitor of activated signal transducer and activators of transcription 1 (PIAS1). Overexpression of NRIP1 promoted UBE2I expression and inhibited PIAS1 expression, and NRIP1 promoted PIAS1 ubiquitination and degradation by UBE2I. The PA-treated TC-1 cells were transfected with si-UBE2I alone or together with si-PIAS1, and the results indicated that transfection of si-UBE2I had the same effect as transfection of si-NRIP1. Finally, our in vivo findings indicated that the expression of NRIP1 and UBE2I was decreased, and PIAS1 expression was increased, in the lung tissues of mice with NRIP1 knocked-down, and the inflammatory infiltration in the lung tissue was reduced. In conclusion, our study demonstrates that NRIP1 aggravates PA-induced lung injury in mice by promoting PIAS1 ubiquitination.
    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
    慢性丙型肝炎病毒(HCV)感染通常会导致纤维化和慢性肝炎,然后肝硬化和最终肝细胞癌(HCC)。HVC生命周期的过程涉及病毒和宿主细胞蛋白之间的密切相互作用以及脂质代谢。然而,参与这种三方相互作用的分子和机制仍然知之甚少。在这里,我们表明,HCV感染HCC衍生的Huh7.5细胞可促进活化STAT1(PIAS1)蛋白抑制剂的上调。相互,PIAS1调节HCV核心蛋白的表达和HCV诱导的LD积累和HCV复制受损。此外,PIAS1控制HCV促进的隔膜9丝形成和微管聚合。随后,我们发现PIAS1与隔膜9相互作用并控制其在细丝上的组装,从而影响了septin9诱导的脂滴积累。一起来看,这些数据表明,PIAS1调节脂滴的积累,并为HCV如何与宿主蛋白相互作用提供了有意义的见解.
    Chronic hepatitis C virus (HCV) infection often leads to fibrosis and chronic hepatitis, then cirrhosis and ultimately hepatocellular carcinoma (HCC). The processes of the HVC life cycle involve intimate interactions between viral and host cell proteins and lipid metabolism. However, the molecules and mechanisms involved in this tripartite interaction remain poorly understood. Herein, we show that the infection of HCC-derived Huh7.5 cells with HCV promotes upregulation of the protein inhibitor of activated STAT1 (PIAS1). Reciprocally, PIAS1 regulated the expression of HCV core protein and HCV-induced LD accumulation and impaired HCV replication. Furthermore, PIAS1 controlled HCV-promoted septin 9 filament formation and microtubule polymerization. Subsequently, we found that PIAS1 interacted with septin 9 and controlled its assembly on filaments, which thus affected septin 9-induced lipid droplet accumulation. Taken together, these data reveal that PIAS1 regulates the accumulation of lipid droplets and offer a meaningful insight into how HCV interacts with host proteins.
    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
    作为一种保守的翻译后修饰,SUMO化已经显示在染色质相关的生物过程(包括转录)中起重要作用。然而,SUMO化机制如何与染色质相关联尚不清楚。这里,我们提供的证据表明,多个SUMO化机械组件,包括SUMOE1蛋白SAE1和SAE2以及PIAS(激活的STAT蛋白抑制剂)家族SUMOE3连接酶,主要与核基质有关,而不是与染色质有关。我们使用核酸酶消化显示,所有PIAS家族蛋白在不存在染色质的情况下保持核基质缔合。重要的是,我们鉴定了多种组蛋白,包括H3和H2A。Z直接与PIAS1相互作用,并证明这种相互作用需要PIAS1SAP(SAF-A/B,Acinus,和PIAS)域。我们证明PIAS1在SAP域和E3连接酶活性依赖性方式中均促进组蛋白H3和H2B的SUMO化。此外,我们表明PIAS1与热休克诱导的基因结合并抑制其表达,并且该功能还需要SAP结构域。总之,我们的研究首次揭示了核基质是SUMOE1和PIAS家族E3连接酶中最富集的隔室。我们发现PIAS1与组蛋白直接相互作用,这也表明了一种分子机制,即与核基质相关的PIAS1如何能够调节转录和其他染色质相关过程。
    As a conserved posttranslational modification, SUMOylation has been shown to play important roles in chromatin-related biological processes including transcription. However, how the SUMOylation machinery associates with chromatin is not clear. Here, we present evidence that multiple SUMOylation machinery components, including SUMO E1 proteins SAE1 and SAE2 and the PIAS (protein inhibitor of activated STAT) family SUMO E3 ligases, are primarily associated with the nuclear matrix rather than with chromatin. We show using nuclease digestion that all PIAS family proteins maintain nuclear matrix association in the absence of chromatin. Of importance, we identify multiple histones including H3 and H2A.Z as directly interacting with PIAS1 and demonstrate that this interaction requires the PIAS1 SAP (SAF-A/B, Acinus, and PIAS) domain. We demonstrate that PIAS1 promotes SUMOylation of histones H3 and H2B in both a SAP domain- and an E3 ligase activity-dependent manner. Furthermore, we show that PIAS1 binds to heat shock-induced genes and represses their expression and that this function also requires the SAP domain. Altogether, our study reveals for the first time the nuclear matrix as the compartment most enriched in SUMO E1 and PIAS family E3 ligases. Our finding that PIAS1 interacts directly with histone proteins also suggests a molecular mechanism as to how nuclear matrix-associated PIAS1 is able to regulate transcription and other chromatin-related processes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号