Protein Inhibitors of Activated STAT

活化 STAT 的蛋白质抑制剂
  • 文章类型: Journal Article
    严重急性呼吸系统综合症冠状病毒2(SARS-CoV-2)已被证明可以限制宿主干扰素的反应;然而,潜在机制尚不清楚.这里,我们发现SARS-CoV-2感染上调E3泛素连接酶Huwe1,进而促进转录因子Miz1的降解。Miz1的降解阻碍了干扰素α和γ的反应,因此促进病毒复制和阻碍病毒清除。相反,沉默或抑制Huwe1增强了干扰素反应,有效遏制病毒复制。始终如一,过表达Miz1增加了干扰素反应和有限的病毒复制,而沉默Miz1则有相反的效果。靶向Huwe1或过表达Miz1引起转录组改变,其特征在于与增强的抗病毒反应和减少的病毒复制相关的丰富功能。进一步的研究表明,Miz1对特定干扰素信号分子的转录发挥表观遗传控制作用,它们充当共同的上游调节因子,负责在Huwe1或Miz1靶向后观察到的转录组变化。这些发现强调了Huwe1-Miz1轴在控制宿主抗病毒反应中的关键作用,其失调导致在COVID-19期间观察到的干扰素反应受损。
    Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has been demonstrated to limit the host interferon response; however, the underlying mechanism remains unclear. Here, we found that SARS-CoV-2 infection upregulated the E3 ubiquitin ligase Huwe1, which in turn facilitated the degradation of the transcription factor Miz1. The degradation of Miz1 hampered interferon alpha and gamma responses, consequently fostering viral replication and impeding viral clearance. Conversely, silencing or inhibiting Huwe1 enhanced the interferon responses, effectively curbing viral replication. Consistently, overexpressing Miz1 augmented the interferon responses and limited viral replication, whereas silencing Miz1 had the opposite effect. Targeting Huwe1 or overexpressing Miz1 elicited transcriptomic alterations characterized by enriched functions associated with bolstered antiviral response and diminished virus replication. Further study revealed Miz1 exerted epigenetic control over the transcription of specific interferon signaling molecules, which acted as common upstream regulators responsible for the observed transcriptomic changes following Huwe1 or Miz1 targeting. These findings underscore the critical role of the Huwe1-Miz1 axis in governing the host antiviral response, with its dysregulation contributing to the impaired interferon response observed during COVID-19.
    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
    合成致死性为开发靶向癌症疗法提供了有吸引力的策略。例如,具有高水平微卫星不稳定性(MSI-H)的癌细胞依赖于Werner(WRN)解旋酶存活。然而,调节WRN时空动力学的机制仍然知之甚少。这里,我们将单分子示踪(SMT)与WRN抑制剂联合用于检测活癌细胞核内的WRN动力学.WRN抑制捕获染色质上的解旋酶,需要p97/VCP以MSI-H依赖性方式进行提取和蛋白酶体降解。使用表型筛选,我们将PIAS4-RNF4轴鉴定为负责WRN降解的途径。最后,我们显示WRN和SUMO化的共抑制在MSI-H细胞中具有累加毒性作用,并使用MSI-H小鼠异种移植模型证实了WRN抑制的体内活性。这项工作阐明了WRN的调节机制,可以促进新的治疗方式的识别。并强调使用SMT作为药物发现和作用机制研究的工具。
    Synthetic lethality provides an attractive strategy for developing targeted cancer therapies. For example, cancer cells with high levels of microsatellite instability (MSI-H) are dependent on the Werner (WRN) helicase for survival. However, the mechanisms that regulate WRN spatiotemporal dynamics remain poorly understood. Here, we used single-molecule tracking (SMT) in combination with a WRN inhibitor to examine WRN dynamics within the nuclei of living cancer cells. WRN inhibition traps the helicase on chromatin, requiring p97/VCP for extraction and proteasomal degradation in a MSI-H dependent manner. Using a phenotypic screen, we identify the PIAS4-RNF4 axis as the pathway responsible for WRN degradation. Finally, we show that co-inhibition of WRN and SUMOylation has an additive toxic effect in MSI-H cells and confirm the in vivo activity of WRN inhibition using an MSI-H mouse xenograft model. This work elucidates a regulatory mechanism for WRN that may facilitate identification of new therapeutic modalities, and highlights the use of SMT as a tool for drug discovery and mechanism-of-action studies.
    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
    先天免疫信号的病毒破坏是生产性感染的关键决定因素。人巨细胞病毒(HCMV)UL26蛋白可防止感染过程中的抗病毒基因表达,然而,所涉及的机制尚不清楚。我们使用TurboID驱动的邻近蛋白质组学来鉴定感染期间推定的UL26相互作用蛋白以解决这个问题。我们发现UL26与几种免疫调节蛋白形成复合物,包括几个STAT家族成员和各种PIAS蛋白,E3SUMOligases家族。我们的结果表明,UL26可防止感染期间的STAT磷酸化,并拮抗干扰素α(IFNA)或肿瘤坏死因子α(TNFα)诱导的转录激活。此外,我们发现PIAS1的失活使细胞对炎症刺激敏感,导致类似于ΔUL26感染的抗病毒转录环境。Further,PIAS1对于HCMV细胞间的传播很重要,这取决于UL26的存在,这表明UL26-PIAS1相互作用对于调节内在的抗病毒防御至关重要。
    Viral disruption of innate immune signaling is a critical determinant of productive infection. The Human Cytomegalovirus (HCMV) UL26 protein prevents anti-viral gene expression during infection, yet the mechanisms involved are unclear. We used TurboID-driven proximity proteomics to identify putative UL26 interacting proteins during infection to address this issue. We find that UL26 forms a complex with several immuno-regulatory proteins, including several STAT family members and various PIAS proteins, a family of E3 SUMO ligases. Our results indicate that UL26 prevents STAT phosphorylation during infection and antagonizes transcriptional activation induced by either interferon α (IFNA) or tumor necrosis factor α (TNFα). Additionally, we find that the inactivation of PIAS1 sensitizes cells to inflammatory stimulation, resulting in an anti-viral transcriptional environment similar to ΔUL26 infection. Further, PIAS1 is important for HCMV cell-to-cell spread, which depends on the presence of UL26, suggesting that the UL26-PIAS1 interaction is vital for modulating intrinsic anti-viral defense.
    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
    响应于锌损失的锌转运蛋白表达的改变保护心脏细胞免受缺血/再灌注(I/R)损伤。然而,心脏细胞感知锌丢失的潜在分子机制尚不清楚.这里,我们发现锌缺乏诱导活化STAT3(PIAS3)蛋白抑制剂的泛素化和降解,可以通过激活STAT3促进ZIP家族锌转运蛋白基因的表达来减轻心肌I/R损伤。PIAS3内的RING指结构域对于PIAS3退化至关重要,因为PIAS3-dRing(缺失RING结构域)和PIAS3-Mut(锌结合位点突变)在锌缺乏的情况下对降解具有抗性。同时,PIAS3内的环指结构域对于抑制STAT3激活至关重要。此外,PIAS3敲低增加心脏Zn2+水平,减少I/R小鼠心脏的心肌梗死,而野生型PIAS3过表达,但不是PIAS3-Mut,降低心脏Zn2+水平,加剧了心肌梗塞.这些发现阐明了锌感应的独特机制,表明锌缺乏期间锌结合调节蛋白PIAS3的快速降解可以纠正锌代谢障碍并减轻再灌注损伤。
    Alterations in zinc transporter expression in response to zinc loss protect cardiac cells from ischemia/reperfusion (I/R) injury. However, the underlying molecular mechanisms how cardiac cells sense zinc loss remains unclear. Here, we found that zinc deficiency induced ubiquitination and degradation of the protein inhibitor of activated STAT3 (PIAS3), which can alleviate myocardial I/R injury by activating STAT3 to promote the expression of ZIP family zinc transporter genes. The RING finger domain within PIAS3 is vital for PIAS3 degradation, as PIAS3-dRing (missing the RING domain) and PIAS3-Mut (zinc-binding site mutation) were resistant to degradation in the setting of zinc deficiency. Meanwhile, the RING finger domain within PIAS3 is critical for the inhibition of STAT3 activation. Moreover, PIAS3 knockdown increased cardiac Zn2+ levels and reduced myocardial infarction in mouse hearts subjected to I/R, whereas wild-type PIAS3 overexpression, but not PIAS3-Mut, reduced cardiac Zn2+ levels, and exacerbated myocardial infarction. These findings elucidate a unique mechanism of zinc sensing, showing that fast degradation of the zinc-binding regulatory protein PIAS3 during zinc deficiency can correct zinc dyshomeostasis and alleviate reperfusion 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
    E3SUMO连接酶PIAS2在分化型乳头状甲状腺癌中高水平表达,但在间变性甲状腺癌(ATC)中低水平表达。高死亡率的未分化癌。我们在此显示,使用转录的双链RNA指导的RNA干扰(PIAS2b-dsRNAi)消除PIAS2β同种型在体外特异性抑制ATC细胞系和患者原代培养物以及原位患者来源的异种移植物的生长(oPDX)在体内。严重的,PIAS2b-dsRNAi不影响正常或非间变性甲状腺肿瘤培养物的生长(分化癌,良性病变)或细胞系。PIAS2b-dsRNAi还对其他间变性人类癌症(胰腺,肺,和胃)。机械上,PIAS2b是正确的有丝分裂纺锤体和中心体组装所必需的,它是ATC中的剂量敏感蛋白。PIAS2b耗竭促进前期有丝分裂突变。高通量蛋白质组学揭示蛋白酶体(PSMC5)和纺锤体细胞骨架(TUBB3)是有丝分裂起始时PIAS2bSUMO化的直接靶标。这些结果确定PIAS2b-dsRNAi是ATC和其他侵袭性间变性癌的有希望的疗法。
    The E3 SUMO ligase PIAS2 is expressed at high levels in differentiated papillary thyroid carcinomas but at low levels in anaplastic thyroid carcinomas (ATC), an undifferentiated cancer with high mortality. We show here that depletion of the PIAS2 beta isoform with a transcribed double-stranded RNA-directed RNA interference (PIAS2b-dsRNAi) specifically inhibits growth of ATC cell lines and patient primary cultures in vitro and of orthotopic patient-derived xenografts (oPDX) in vivo. Critically, PIAS2b-dsRNAi does not affect growth of normal or non-anaplastic thyroid tumor cultures (differentiated carcinoma, benign lesions) or cell lines. PIAS2b-dsRNAi also has an anti-cancer effect on other anaplastic human cancers (pancreas, lung, and gastric). Mechanistically, PIAS2b is required for proper mitotic spindle and centrosome assembly, and it is a dosage-sensitive protein in ATC. PIAS2b depletion promotes mitotic catastrophe at prophase. High-throughput proteomics reveals the proteasome (PSMC5) and spindle cytoskeleton (TUBB3) to be direct targets of PIAS2b SUMOylation at mitotic initiation. These results identify PIAS2b-dsRNAi as a promising therapy for ATC and other aggressive anaplastic 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
    I型干扰素(IFN)对于抗病毒免疫反应至关重要,和微调I型IFN的产生对于有效清除病毒而不引起有害的免疫病理学至关重要。我们表明,转录因子Miz1通过将组蛋白脱乙酰酶1(HDAC1)募集到Ifna和Ifnb的启动子来表观遗传抑制小鼠肺上皮细胞中编码I型IFN的基因的表达。在甲型流感病毒(IAV)感染期间,Miz1功能的丧失导致这些I型IFN的产生增加,导致在体外和体内提高病毒清除率。IAV感染通过促进cullin-4B(CUL4B)介导的E3泛素连接酶Mule(Mcl-1泛素连接酶E3;也称为Huwe1或Arf-BP1)的泛素化和降解来诱导Miz1积累,以Miz1为目标进行降解。因此,Miz1积累限制了I型IFN的产生并有利于病毒复制。这项研究揭示了Miz1在调节抗病毒防御中的先前未被识别的功能以及流感病毒逃避宿主免疫防御的潜在机制。
    Type I interferons (IFNs) are critical for the antiviral immune response, and fine-tuning type I IFN production is critical to effectively clearing viruses without causing harmful immunopathology. We showed that the transcription factor Miz1 epigenetically repressed the expression of genes encoding type I IFNs in mouse lung epithelial cells by recruiting histone deacetylase 1 (HDAC1) to the promoters of Ifna and Ifnb. Loss of function of Miz1 resulted in augmented production of these type I IFNs during influenza A virus (IAV) infection, leading to improved viral clearance in vitro and in vivo. IAV infection induced Miz1 accumulation by promoting the cullin-4B (CUL4B)-mediated ubiquitylation and degradation of the E3 ubiquitin ligase Mule (Mcl-1 ubiquitin ligase E3; also known as Huwe1 or Arf-BP1), which targets Miz1 for degradation. As a result, Miz1 accumulation limited type I IFN production and favored viral replication. This study reveals a previously unrecognized function of Miz1 in regulating antiviral defense and a potential mechanism for influenza viruses to evade host immune defense.
    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:激活的STATs(PIAS)的蛋白抑制剂具有多效的生物学效应,如蛋白质翻译后修饰,转录共调控和基因编辑。据报道,PIAS家族基因还与癌症中的免疫细胞浸润相关,这突出了它们在肿瘤进展中的未被注意的生物学作用。然而,它们的表达与预后的关系,免疫细胞浸润,肿瘤微环境,泛癌症的免疫疗法很少报道。
    方法:使用多组学数据来研究PIAS家族成员在泛癌症中的表达水平,并通过单因素Cox回归和Kaplan-Meier分析其在不同肿瘤中表达的预后价值。采用相关性分析探讨PIAS基因表达与肿瘤微环境的关系,免疫浸润亚型,干性评分和药物敏感性。此外,我们还使用伤口愈合和transwell实验来验证PIAS家族基因表达对HCC细胞侵袭和转移的生物学效应。
    结果:通过多基因组分析,我们发现PIAS家族基因表达在肿瘤中具有显著异质性,与多种癌症患者的不良预后相关。此外,我们还发现PIAS家族基因的遗传改变不仅在不同类型的人类肿瘤中常见,但也与整个泛癌症的无病生存率(DFS)显着相关。单细胞分析显示PIAS家族基因主要分布在单核细胞/巨噬细胞中。此外,我们还发现它们的表达与肿瘤微环境(包括基质细胞和免疫细胞)和干性评分(DNAss和RNAss)相关。药敏分析表明,PIAS家族基因能够预测化疗和免疫治疗的反应。PIAS家族基因的表达与肿瘤转移密切相关,尤其是PIAS3。高表达PIAS3显著促进肝癌细胞系(HCC-LM3和MHCC97-H)的迁移和侵袭。
    结论:综合考虑,这些发现有助于确定PIAS家族基因是否是潜在的致癌靶基因,对肿瘤免疫治疗的发展有重要贡献。
    BACKGROUND: Protein inhibitor of activated STATs (PIAS) has pleiotropic biological effects, such as protein post-translational modification, transcriptional coregulation and gene editing. It is reported that PIAS family genes are also correlated with immune cells infiltration in cancers that highlights their unnoticed biological role in tumor progression. However, the relationship of their expression with prognosis, immune cell infiltration, tumor microenvironment, and immunotherapy in pan-cancer has been rarely reported.
    METHODS: The multi-omics data were used to investigate the expression level of PIAS family members in pan-cancer, and the prognostic value of their expression in different tumors was analyzed by univariate Cox regression and Kaplan-Meier. Correlation analysis was used to investigate the relationship of PIAS gene expression with tumor microenvironment, immune infiltrating subtypes, stemness score and drug sensitivity. In addition, we also used wound healing and transwell assays to verify the biological effects of PIAS family gene expression on invasion and metastasis of HCC cells.
    RESULTS: We found that PIAS family genes expression is significantly heterogeneous in tumors by multi-genomic analysis, and associated with poor prognosis in patients with multiple types of cancer. Furthermore, we also found that genetic alterations of PIAS family genes were not only common in different types of human tumors, but were also significantly associated with disease-free survival (DFS) across pan-cancer. Single-cell analysis revealed that PIAS family genes were mainly distributed in monocytes/macrophages. Additionally, we also found that their expression was associated with tumor microenvironment (including stromal cells and immune cells) and stemness score (DNAss and RNAss). Drug sensitivity analysis showed that PIAS family genes were able to predict the response to chemotherapy and immunotherapy. PIAS family genes expression is closely related to tumor metastasis, especially PIAS3. High PIAS3 expression significantly promotes the migration and invasion of liver cancer cell lines (HCC-LM3 and MHCC97-H).
    CONCLUSIONS: Taking together, these findings contribute to determine whether the PIAS family genes are a potential oncogenic target gene, which have important contribution for the development of cancer immunotherapy.
    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
    活化的小泛素样修饰剂(SUMO)与缺血性中风后的神经病理过程有关。然而,SUMO化的靶蛋白及其对神经元损伤的贡献仍有待阐明。MLK3(混合谱系激酶3),丝裂原活化蛋白激酶激酶(MAPKKK)家族的成员,是脑缺血后神经元损伤的关键调节因子。这里,我们发现,在全球和局灶性缺血性啮齿动物模型以及氧和葡萄糖剥夺(OGD)的原发性神经元模型中,MLK3的SUMO化增加.在MLK3的Lys401位点处的SUMO1缀合促进了其激活,刺激其下游p38/c-JunN末端激酶(JNK)级联,并导致细胞凋亡。MLK3与SUMO连接酶PIAS3的相互作用,缺血和再灌注后升高。PIAS3的PINIT结构域参与与MLK3的直接相互作用。PIAS3的PINIT结构域的过表达破坏了MLK3-PIAS3的相互作用,抑制MLK3的SUMO化,抑制下游信号,减少细胞凋亡和神经突损伤。在啮齿动物缺血模型中,PINIT域的过度表达减少了脑损伤并减轻了学习缺陷,记忆,和感觉运动功能。我们的发现表明,PIAS3引起的脑缺血诱导的MLK3SUMO化是针对中风后神经元病变和行为障碍的潜在靶标。
    Activated small ubiquitin-like modifiers (SUMOs) have been implicated in neuropathological processes following ischemic stroke. However, the target proteins of SUMOylation and their contribution to neuronal injury remain to be elucidated. MLK3 (mixed-lineage kinase 3), a member of the mitogen-activated protein kinase kinase kinase (MAPKKK) family, is a critical regulator of neuronal lesions following cerebral ischemia. Here, we found that SUMOylation of MLK3 increases in both global and focal ischemic rodent models and primary neuronal models of oxygen and glucose deprivation (OGD). SUMO1 conjugation at the Lys401 site of MLK3 promoted its activation, stimulated its downstream p38/c-Jun N-terminal kinase (JNK) cascades, and led to cell apoptosis. The interaction of MLK3 with PIAS3, a SUMO ligase, was elevated following ischemia and reperfusion. The PINIT domain of PIAS3 was involved in direct interactions with MLK3. Overexpression of the PINIT domain of PIAS3 disrupted the MLK3-PIAS3 interaction, inhibited SUMOylation of MLK3, suppressed downstream signaling, and reduced cell apoptosis and neurite damage. In rodent ischemic models, the overexpression of the PINIT domain reduced brain lesions and alleviated deficits in learning, memory, and sensorimotor functions. Our findings demonstrate that brain ischemia-induced MLK3 SUMOylation by PIAS3 is a potential target against poststroke neuronal lesions and behavioral impairments.
    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
    前列腺癌(PCa)的发病率每年都在上升,随着雄激素受体(AR)成为其生长和进展的关键贡献者。越来越多的证据强调了AR招募辅因子的能力,影响下游基因转录,从而促进PCa细胞的增殖和转移。虽然,涉及AR拮抗剂的临床策略提供了一些缓解,治疗去势抵抗前列腺癌(CRPC)仍然是一个巨大的挑战.因此,小时的需要在于发掘新药或治疗靶点以有效对抗PCa。这篇综述概括了AR的共激活剂和共抑制物所起的关键作用,尤其是PCa中的雄激素受体相关蛋白(ARA)和类固醇受体共激活剂(SRC)。我们的数据揭示了这些辅因子如何复杂地调节组蛋白修饰,细胞循环,SUMOylation,和细胞凋亡通过它们与AR的相互作用。在经过审查的一系列辅因子中,例如ARA70β,ARA24,ARA160,ARA55,ARA54,PIAS1,PIAS3,SRC1,SRC2,SRC3,PCAF,p300/CBP,MED1和CARM1在PCa中表现出上调。相反,其他辅因子,如ARA70α,PIASY,和NCoR/SMRT表现出下调。这种双重性强调了PCa中AR辅因子动力学的复杂性。根据我们的发现,我们认为,操纵辅因子调节以调节AR功能有望成为针对晚期PCa的一种新的治疗途径.面对CRPC的巨大挑战,这种范式转变为寻求有效治疗提供了新的希望。
    Prostate cancer (PCa) is witnessing a concerning rise in incidence annually, with the androgen receptor (AR) emerging as a pivotal contributor to its growth and progression. Mounting evidence underscores the AR\'s ability to recruit cofactors, influencing downstream gene transcription and thereby fueling the proliferation and metastasis of PCa cells. Although, clinical strategies involving AR antagonists provide some relief, managing castration resistant prostate cancer (CRPC) remains a formidable challenge. Thus, the need of the hour lies in unearthing new drugs or therapeutic targets to effectively combat PCa. This review encapsulates the pivotal roles played by coactivators and corepressors of AR, notably androgen receptor-associated protein (ARA) and steroid receptor Coactivators (SRC) in PCa. Our data unveils how these cofactors intricately modulate histone modifications, cell cycling, SUMOylation, and apoptosis through their interactions with AR. Among the array of cofactors scrutinised, such as ARA70β, ARA24, ARA160, ARA55, ARA54, PIAS1, PIAS3, SRC1, SRC2, SRC3, PCAF, p300/CBP, MED1, and CARM1, several exhibit upregulation in PCa. Conversely, other cofactors like ARA70α, PIASy, and NCoR/SMRT demonstrate downregulation. This duality underscores the complexity of AR cofactor dynamics in PCa. Based on our findings, we propose that manipulating cofactor regulation to modulate AR function holds promise as a novel therapeutic avenue against advanced PCa. This paradigm shift offers renewed hope in the quest for effective treatments in the face of CRPC\'s formidable challenges.
    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: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)

公众号