Protein Stability

蛋白质稳定性
  • 文章类型: Journal Article
    SLC7A11是胱氨酸转运蛋白和铁凋亡抑制剂。SLC7A11的稳定性如何在响应环境胱氨酸时协调调节,E3连接酶和去泛素酶(DUB)仍然难以捉摸。这里,我们报道neddylation抑制剂MLN4924通过引起SLC7A11积累来增加胱氨酸的摄取,通过灭活Cullin-RING连接酶-3(CRL-3)。我们将KCTD10鉴定为CRL-3用于SLC7A11泛素化的底物识别亚基,和USP18作为SLC7A11去泛素酶。剥夺胱氨酸后,KCTD10或USP18的蛋白质水平降低或升高,分别,有助于SLC7A11的积累。通过使SLC7A11、KCTD10或USP18不稳定或稳定,反向调节胱氨酸摄取和铁凋亡。生物学,MLN4924与SLC7A11抑制剂咪唑酮伊拉斯汀(IKE)的组合增强了对肿瘤生长的抑制。在人类乳腺肿瘤组织中,SLC7A11水平分别与KCTD10或USP18呈负相关或正相关。总的来说,我们的研究定义了SLC7A11和铁凋亡如何由CRL3KCTD10/E3-USP18/DUB轴协调调节,并提供了合理的药物组合以增强抗癌功效的基本原理。
    SLC7A11 is a cystine transporter and ferroptosis inhibitor. How the stability of SLC7A11 is coordinately regulated in response to environmental cystine by which E3 ligase and deubiquitylase (DUB) remains elusive. Here, we report that neddylation inhibitor MLN4924 increases cystine uptake by causing SLC7A11 accumulation, via inactivating Cullin-RING ligase-3 (CRL-3). We identified KCTD10 as the substrate-recognizing subunit of CRL-3 for SLC7A11 ubiquitylation, and USP18 as SLC7A11 deubiquitylase. Upon cystine deprivation, the protein levels of KCTD10 or USP18 are decreased or increased, respectively, contributing to SLC7A11 accumulation. By destabilizing or stabilizing SLC7A11, KCTD10, or USP18 inversely regulates the cystine uptake and ferroptosis. Biologically, MLN4924 combination with SLC7A11 inhibitor Imidazole Ketone Erastin (IKE) enhanced suppression of tumor growth. In human breast tumor tissues, SLC7A11 levels were negatively or positively correlated with KCTD10 or USP18, respectively. Collectively, our study defines how SLC7A11 and ferroptosis is coordinately regulated by the CRL3KCTD10/E3-USP18/DUB axis, and provides a sound rationale of drug combination to enhance anticancer efficacy.
    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
    长链非编码RNA是癌症生物学中的重要调节因子,并作为肿瘤抑制因子或癌基因发挥作用。它们的失调与肿瘤发生密切相关。LINC00265在肺腺癌中上调,并且是该癌症的预后生物标志物。然而,其在癌症进展中的作用机制仍知之甚少.
    这里,使用肺癌细胞系检查LINC00265在肺腺癌中的调节作用,临床样本,和异种移植。
    我们发现高水平的LINC00265表达与较短的患者总生存率相关,而LINC00265的敲除抑制癌细胞系的增殖和异种移植物中的肿瘤生长。Westernblot和流式细胞术分析表明LINC00265沉默可诱导自噬和凋亡。此外,我们表明LINC00265与转录共阻遏物开关非依赖性3a(SIN3A)相互作用并稳定,它是一种支架蛋白,以依赖环境的方式充当肿瘤抑制因子或癌基因。沉默SIN3A也降低了肺癌细胞的增殖,这与自噬的诱导有关。这些观察结果提高了LINC00265在肺腺癌中促进SIN3A致癌活性的可能性。
    因此,我们的发现将SIN3A确定为LINC00265相关蛋白,并应有助于理解LINC00265介导的肿瘤发生的潜在机制。
    UNASSIGNED: Long non-coding RNAs are important regulators in cancer biology and function either as tumor suppressors or as oncogenes. Their dysregulation has been closely associated with tumorigenesis. LINC00265 is upregulated in lung adenocarcinoma and is a prognostic biomarker of this cancer. However, the mechanism underlying its function in cancer progression remains poorly understood.
    UNASSIGNED: Here, the regulatory role of LINC00265 in lung adenocarcinoma was examined using lung cancer cell lines, clinical samples, and xenografts.
    UNASSIGNED: We found that high levels of LINC00265 expression were associated with shorter overall survival rate of patients, whereas knockdown of LINC00265 inhibited proliferation of cancer cell lines and tumor growth in xenografts. Western blot and flow cytometry analyses indicated that silencing of LINC00265 induced autophagy and apoptosis. Moreover, we showed that LINC00265 interacted with and stabilized the transcriptional co-repressor Switch-independent 3a (SIN3A), which is a scaffold protein functioning either as a tumor repressor or as an oncogene in a context-dependent manner. Silencing of SIN3A also reduced proliferation of lung cancer cells, which was correlated with the induction of autophagy. These observations raise the possibility that LINC00265 functions to promote the oncogenic activity of SIN3A in lung adenocarcinoma.
    UNASSIGNED: Our findings thus identify SIN3A as a LINC00265-associated protein and should help to understand the mechanism underlying LINC00265-mediated oncogenesis.
    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
    肝纤维化是由多种损伤因素引发的持续性损伤修复反应,这导致肝脏组织样本内细胞外基质的异常积累。目前临床上对肝纤维化的治疗是无效的,因此,阐明肝纤维化发生的机制具有重要意义。在这里,研究了lncRNASnhg12在肝纤维化中的功能和相关机制。Snhg12表达在小鼠肝纤维化组织样品中增加,Snhg12基因敲除抑制肝脏病理损伤并下调纤维化相关蛋白的表达水平。机械上,基于生物信息学分析,Snhg12在小鼠肝星状细胞(mHSCs)的早期活化中发挥作用,Snhg12与Igfbp3表达呈正相关。进一步的实验结果表明,Snhg12敲低会阻碍mHSCs的增殖和激活,并下调Igfbp3的蛋白表达。Snhg12可以与IGFBP3相互作用,增强其蛋白稳定性,Igfbp3的过表达通过敲低Snhg12部分逆转了mHSCs增殖和激活的抑制。总之,LncRNASnhg12通过靶向IGFBP3并促进其蛋白稳定性介导肝纤维化,从而促进mHSC增殖和活化。Snhg12已被确定为治疗肝纤维化的潜在靶标。
    Liver fibrosis is a persistent damage repair response triggered by various injury factors, which leads to an abnormal accumulation of extracellular matrix within liver tissue samples. The current clinical treatment of liver fibrosis is currently ineffective; therefore, elucidating the mechanism of liver fibrogenesis is of significant importance. Herein, the function and related mechanisms of lncRNA Snhg12 within hepatic fibrosis were investigated. Snhg12 expression was shown to be increased in mouse hepatic fibrotic tissue samples, and Snhg12 knockdown suppressed hepatic pathological injury and down-regulated the expression levels of fibrosis-associated proteins. Mechanistically, Snhg12 played a role in the early activation of mouse hepatic stellate cells (mHSCs) based on bioinformatics analysis, and Snhg12 was positively correlated with Igfbp3 expression. Further experimental results demonstrated that Snhg12 knockdown impeded mHSCs proliferation and activation and also downregulated the protein expression of Igfbp3. Snhg12 could interact with IGFBP3 and boost its protein stability, and overexpression of Igfbp3 partially reversed the inhibition of mHSCsproliferation and activation by the knockdown of Snhg12. In conclusion, LncRNA Snhg12 mediates liver fibrosis by targeting IGFBP3 and promoting its protein stability, thereby promoting mHSC proliferation and activation. Snhg12 has been identified as an underlying target for treating liver fibrosis.
    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
    ORF9b蛋白,来自SARS-CoV和SARS-CoV-2的核衣壳开放阅读框,通过抑制先天免疫反应,作为病毒免疫逃避至关重要的辅助蛋白。尽管意义重大,其功能背后的精确调控机制仍然难以捉摸。在本研究中,我们揭示了SARS-CoV-2的ORF9b蛋白,包括新兴的突变株,如Delta和Omicron,可以在K67位点进行泛素化,然后通过蛋白酶体途径进行降解,尽管这些菌株之间存在某些突变。此外,我们的研究进一步揭示了外线粒体膜70(TOM70)作为底物受体的转位酶的关键作用,ORF9b与热休克蛋白90α(HSP90α)和Cullin5(CUL5)桥接形成复合物。在这个建筑群中,CUL5引发ORF9b的泛素化和降解,作为宿主抗病毒因子,而HSP90α的作用是稳定它。值得注意的是,用HSP90抑制剂如GA或17-AAG处理加速ORF9b的降解,导致SARS-CoV-2复制的明显抑制。单细胞测序数据显示COVID-19患者肺上皮细胞中HSP90α的上调,提示SARS-CoV-2可能利用HSP90α逃避宿主免疫的潜在机制。我们的研究确定CUL5-TOM70-HSP90α复合物是ORF9b蛋白稳定性的关键调节因子,揭示了复杂的宿主病毒免疫反应动力学,并为临床环境中针对SARS-CoV-2的药物开发提供了有希望的途径。
    The ORF9b protein, derived from the nucleocapsid\'s open-reading frame in both SARS-CoV and SARS-CoV-2, serves as an accessory protein crucial for viral immune evasion by inhibiting the innate immune response. Despite its significance, the precise regulatory mechanisms underlying its function remain elusive. In the present study, we unveil that the ORF9b protein of SARS-CoV-2, including emerging mutant strains like Delta and Omicron, can undergo ubiquitination at the K67 site and subsequent degradation via the proteasome pathway, despite certain mutations present among these strains. Moreover, our investigation further uncovers the pivotal role of the translocase of the outer mitochondrial membrane 70 (TOM70) as a substrate receptor, bridging ORF9b with heat shock protein 90 alpha (HSP90α) and Cullin 5 (CUL5) to form a complex. Within this complex, CUL5 triggers the ubiquitination and degradation of ORF9b, acting as a host antiviral factor, while HSP90α functions to stabilize it. Notably, treatment with HSP90 inhibitors such as GA or 17-AAG accelerates the degradation of ORF9b, leading to a pronounced inhibition of SARS-CoV-2 replication. Single-cell sequencing data revealed an up-regulation of HSP90α in lung epithelial cells from COVID-19 patients, suggesting a potential mechanism by which SARS-CoV-2 may exploit HSP90α to evade the host immunity. Our study identifies the CUL5-TOM70-HSP90α complex as a critical regulator of ORF9b protein stability, shedding light on the intricate host-virus immune response dynamics and offering promising avenues for drug development against SARS-CoV-2 in clinical settings.
    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
    病理性心肌肥厚是心力衰竭和其他心血管疾病的主要危险因素之一。然而,病理性心肌肥厚的潜在机制仍然未知.这里,我们确定了TNFAIP3相互作用蛋白3(TNIP3)是病理性心肌肥大的负调节因子的第一个证据.我们观察到,在接受横向主动脉缩窄(TAC)手术的小鼠心脏和苯肾上腺素(PE)刺激的原代新生大鼠心肌细胞中,TNIP3的显着上调。在Tnip3缺陷小鼠中,TAC手术后心肌肥厚加重。相反,心脏特异性Tnip3转基因(TG)小鼠显示相同表型的显着逆转。因此,TNIP3在体外减轻PE诱导的心肌细胞增大。机械上,RNA测序和相互作用组分析相结合,以鉴定信号转导和转录激活因子1(STAT1)作为潜在靶标,以阐明TNIP3在病理性心脏肥大中的分子机制。通过免疫沉淀和谷胱甘肽S-转移酶测定,我们发现,TNIP3可以直接与STAT1相互作用,并通过抑制K48型泛素化来抑制其在肥大刺激下的降解.值得注意的是,STAT1抑制剂氟达拉滨或STAT1敲低阻断了TNIP3对心肌肥厚的保护作用。我们的研究发现,TNIP3通过促进STAT1的稳定性作为病理性心肌肥大的新抑制剂,这表明TNIP3可能是病理性心肌肥厚和心力衰竭的一个有希望的治疗靶点。
    Pathological cardiac hypertrophy is one of the major risk factors of heart failure and other cardiovascular diseases. However, the mechanisms underlying pathological cardiac hypertrophy remain largely unknown. Here, we identified the first evidence that TNFAIP3 interacting protein 3 (TNIP3) was a negative regulator of pathological cardiac hypertrophy. We observed a significant upregulation of TNIP3 in mouse hearts subjected to transverse aortic constriction (TAC) surgery and in primary neonatal rat cardiomyocytes stimulated by phenylephrine (PE). In Tnip3-deficient mice, cardiac hypertrophy was aggravated after TAC surgery. Conversely, cardiac-specific Tnip3 transgenic (TG) mice showed a notable reversal of the same phenotype. Accordingly, TNIP3 alleviated PE-induced cardiomyocyte enlargement in vitro. Mechanistically, RNA-sequencing and interactome analysis were combined to identify the signal transducer and activator of transcription 1 (STAT1) as a potential target to clarify the molecular mechanism of TNIP3 in pathological cardiac hypertrophy. Via immunoprecipitation and Glutathione S-transferase assay, we found that TNIP3 could interact with STAT1 directly and suppress its degradation by suppressing K48-type ubiquitination in response to hypertrophic stimulation. Remarkably, preservation effect of TNIP3 on cardiac hypertrophy was blocked by STAT1 inhibitor Fludaradbine or STAT1 knockdown. Our study found that TNIP3 serves as a novel suppressor of pathological cardiac hypertrophy by promoting STAT1 stability, which suggests that TNIP3 could be a promising therapeutic target of pathological cardiac hypertrophy and heart failure.
    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
    选择性剪接在增加基因组中表达的mRNA的多样性中起着至关重要的作用。富含丝氨酸/精氨酸的剪接因子3(SRSF3)负责调节其自身mRNA的可变剪接,并确保其表达平衡以维持稳态。此外,SRSF3的外显子跳跃导致产生截短的蛋白质,而不是产生过早终止密码子(PTC)的移码突变.然而,参与SRSF3剪接的精确调控机制尚不清楚.在这项研究中,我们首先建立了共表达全长SRSF3(SRSF3-FL)和SRSF3-PTC的平台,并进一步鉴定了针对SRSF3-FL和截短的SRSF3(SRSF3-TR)蛋白的特异性抗体.接下来,我们发现,外源性过表达SRSF3-FL或SRSF3-PTC未能逆转地高辛的作用,咖啡因,或两者结合在这个分子和它的目标。内质网相关通路,转录因子,发现棕榈酸和磷酸盐等化学物质参与SRSF3表达的调节。在HeLa细胞中,棕榈酸和磷酸盐通过不同的调节机制介导了SRSF3-FL的下调。总之,我们为SRSF3-FL和SRSF3-TR蛋白表达的改变提供了新的见解,以鉴定细胞中SRSF3的功能。
    Alternative splicing plays a crucial role in increasing the diversity of mRNAs expressed in the genome. Serine/arginine-rich splicing factor 3 (SRSF3) is responsible for regulating the alternative splicing of its own mRNA and ensuring that its expression is balanced to maintain homeostasis. Moreover, the exon skipping of SRSF3 leads to the production of a truncated protein instead of a frameshift mutation that generates a premature termination codon (PTC). However, the precise regulatory mechanism involved in the splicing of SRSF3 remains unclear. In this study, we first established a platform for coexpressing full-length SRSF3 (SRSF3-FL) and SRSF3-PTC and further identified a specific antibody against the SRSF3-FL and truncated SRSF3 (SRSF3-TR) proteins. Next, we found that exogenously overexpressing SRSF3-FL or SRSF3-PTC failed to reverse the effects of digoxin, caffeine, or both in combination on this molecule and its targets. Endoplasmic reticulum-related pathways, transcription factors, and chemicals such as palmitic acid and phosphate were found to be involved in the regulation of SRSF3 expression. The downregulation of SRSF3-FL by palmitic acid and phosphate was mediated via different regulatory mechanisms in HeLa cells. In summary, we provide new insights into the altered expression of the SRSF3-FL and SRSF3-TR proteins for the identification of the functions of SRSF3 in cells.
    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
    TBX1基因在22q11.2缺失综合征(22q11.2DS)的发展中起关键作用,与各种表型表现相关的复杂遗传疾病。在这项研究中,我们进行了计算机分析,以鉴定TBX1基因中潜在有害的非同义单核苷酸多态性(nsSNP),并评估其对22q11.2DS的功能和结构影响。采用涉及多种计算工具的综合分析管道来预测nsSNP的致病性。这项研究评估了蛋白质的稳定性,并探索了蛋白质-蛋白质相互作用的潜在变化。结果显示rs751339103(C>A),rs780800634(G>A),rs1936727304(T>C),rs1223320618(G>A),rs1248532217(T>C),rs1294927055(C>T),rs1331240435(A>G,rs1601289406(A>C),rs1936726164(G>A),和rs911796187(G>A)具有影响蛋白质功能和稳定性的高风险潜力。进一步分析了这些nsSNP对翻译后修饰和结构特征的影响,表明它们可能破坏与TBX1及其相互作用伙伴相关的分子途径。这些发现为进一步的实验研究和阐明受22q11.2DS影响的个体的潜在治疗靶标和个性化治疗方法奠定了基础。
    The TBX1 gene plays a critical role in the development of 22q11.2 deletion syndrome (22q11.2DS), a complex genetic disorder associated with various phenotypic manifestations. In this study, we performed in-silico analysis to identify potentially deleterious non-synonymous single nucleotide polymorphisms (nsSNPs) within the TBX1 gene and evaluate their functional and structural impact on 22q11.2DS. A comprehensive analysis pipeline involving multiple computational tools was employed to predict the pathogenicity of nsSNPs. This study assessed protein stability and explored potential alterations in protein-protein interactions. The results revealed the rs751339103(C>A), rs780800634(G>A), rs1936727304(T>C), rs1223320618(G>A), rs1248532217(T>C), rs1294927055 (C>T), rs1331240435 (A>G, rs1601289406 (A>C), rs1936726164 (G>A), and rs911796187(G>A) with a high-risk potential for affecting protein function and stability. These nsSNPs were further analyzed for their impact on post-translational modifications and structural characteristics, indicating their potential disruption of molecular pathways associated with TBX1 and its interacting partners. These findings provide a foundation for further experimental studies and elucidation of potential therapeutic targets and personalized treatment approaches for individuals affected by 22q11.2DS.
    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
    暂无摘要。
    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:越来越多的证据表明去泛素化酶可能有助于肿瘤进展,并且可以作为有希望的治疗靶标。
    方法:通过免疫组织化学和TCGA/GO数据库分析去泛素酶OTUD6B在肺腺癌(LUAD)及其癌旁组织中的过表达。生存分析进一步支持OTUD6B作为LUAD治疗的潜在靶标。我们使用细胞活力测定和TUNEL染色评估了OTUD6B对LUAD细胞生长的影响,迁移,和侵袭实验,探讨OTUD6B对LUAD细胞凋亡和转移的影响。此外,我们在裸鼠体内建立了移植瘤模型来验证我们的发现。最后,使用IP质谱和co-IP实验,我们筛选并证实了RIPK1作为OTUD6B底物在LUAD中的影响。
    结果:OTUD6B在人类LUAD中高度过表达,并预测LUAD患者的预后不良。OTUD6B敲低抑制LUAD细胞的增殖,增强凋亡,抑制LUAD细胞的转移。A549异种移植物显示OTUD6B缺失可以减缓肿瘤生长。此外,OTUD6B可以与RIPK1结合,降低其泛素化水平并增加其蛋白质稳定性。
    结论:我们的结果表明,OTUD6B是LUAD治疗的一个有希望的临床靶点,靶向OTUD6B可能构成一种有效的抗LUAD策略。
    BACKGROUND: There is growing evidence indicating that deubiquitinating enzymes may contribute to tumor progression and can serve as promising therapeutic targets.
    METHODS: The overexpression of deubiquitinase OTUD6B in lung adenocarcinoma (LUAD) and its adjacent tissues was analyzed by immunohistochemistry and TCGA/GO database. Survival analysis further supported OTUD6B as a potential target for LUAD treatment. We assessed the effect of OTUD6B on LUAD cell growth using cell viability assays and conducted TUNEL staining, migration, and invasion experiments to investigate the impact of OTUD6B on the apoptosis and metastasis of LUAD cells. Additionally, we established a transplanted tumor model in nude mice to validate our findings in vivo. Finally, using IP mass spectrometry and co-IP experiments, we screened and confirmed the influence of RIPK1 as a substrate of OTUD6B in LUAD.
    RESULTS: OTUD6B is highly overexpressed in human LUAD and predicts poor prognosis in LUAD patients. OTUD6B knockdown inhibited the proliferation of LUAD cells and enhanced apoptosis and inhibited metastasis in LUAD cells suppressed. A549 xenografts revealed that OTUD6B deletion can slow down tumour growth. Additionally, OTUD6B can bind to RIPK1, reduce its ubiquitination level and increase its protein stability.
    CONCLUSIONS: Our results suggest that OTUD6B is a promising clinical target for LUAD treatment and that targeting OTUD6B may constitute an effective anti-LUAD strategy.
    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
    p53在DNA损伤和氧化应激条件下调节多种信号通路并维持细胞稳态。尽管USP7已被证明通过去泛素化促进p53稳定性,USP7-p53激活机制尚不清楚.这里,我们建议DNA损伤诱导活性氧(ROS)的产生并激活ATM-CHK2,然后CHK2在S168和T231磷酸化USP7。USP7磷酸化是其对p53的去泛素化活性所必需的。USP7还在K119和K131处去泛素化CHK2,增加CHK2稳定性并在CHK2和USP7之间产生正反馈回路。与肿瘤周围组织相比,甲状腺癌和结肠癌组织显示更高的CHK2和磷酸化USP7(S168,T231)水平,这些水平是正相关的。总的来说,我们的结果揭示了一个涉及CHK2-USP7轴的磷酸化-去泛素化正反馈回路,该回路支持p53的稳定和细胞稳态的维持.
    p53 regulates multiple signaling pathways and maintains cell homeostasis under conditions of DNA damage and oxidative stress. Although USP7 has been shown to promote p53 stability via deubiquitination, the USP7-p53 activation mechanism has remained unclear. Here, we propose that DNA damage induces reactive oxygen species (ROS) production and activates ATM-CHK2, and CHK2 then phosphorylates USP7 at S168 and T231. USP7 phosphorylation is essential for its deubiquitination activity toward p53. USP7 also deubiquitinates CHK2 at K119 and K131, increasing CHK2 stability and creating a positive feedback loop between CHK2 and USP7. Compared to peri-tumor tissues, thyroid cancer and colon cancer tissues show higher CHK2 and phosphorylated USP7 (S168, T231) levels, and these levels are positively correlated. Collectively, our results uncover a phosphorylation-deubiquitination positive feedback loop involving the CHK2-USP7 axis that supports the stabilization of p53 and the maintenance of cell homeostasis.
    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