Active Transport, Cell Nucleus

主动转运, 细胞核
  • 文章类型: Journal Article
    异常蛋白质的聚集是神经变性如聚谷氨酰胺(polyQ)和其他重复扩增疾病中的常见病理标志。通过在果蝇肠肠细胞中过表达ataxin3C末端polyQ扩增,我们建立了脊髓小脑性共济失调类型3(SCA3)的肠梗阻模型,并报道了核相关内体(NAEs)的新作用-将polyQ递送至核质.在这个模型中,伴随着显著增加的RAB5阳性NAEs是富含polyQ的大量核质网,异常的核包膜内陷,内质网显著减少,表明功能失调的核质运输和受损的内膜组织。始终如一,Rab5而不是Rab7RNAi进一步降低polyQ相关NAE,抑制内膜解体,缓解疾病模型。有趣的是,自噬蛋白在与polyQ相关的NAEs中富集,并发挥非经典自噬作用,因为自噬分子的遗传操作对NAEs和SCA3毒性表现出不同的影响。即,Atg1或Atg12的下调减轻,而Atg5RNAi加重果蝇肠和复眼的疾病表型。我们的发现,因此,提供新的机制见解,并强调以内体为中心的核质运输和稳态内膜分配在polyQ疾病发病机理中的基本作用。
    Aggregation of aberrant proteins is a common pathological hallmark in neurodegeneration such as polyglutamine (polyQ) and other repeat-expansion diseases. Here through overexpression of ataxin3 C-terminal polyQ expansion in Drosophila gut enterocytes, we generated an intestinal obstruction model of spinocerebellar ataxia type3 (SCA3) and reported a new role of nuclear-associated endosomes (NAEs)-the delivery of polyQ to the nucleoplasm. In this model, accompanied by the prominently increased RAB5-positive NAEs are abundant nucleoplasmic reticulum enriched with polyQ, abnormal nuclear envelope invagination, significantly reduced endoplasmic reticulum, indicating dysfunctional nucleocytoplasmic trafficking and impaired endomembrane organization. Consistently, Rab5 but not Rab7 RNAi further decreased polyQ-related NAEs, inhibited endomembrane disorganization, and alleviated disease model. Interestingly, autophagic proteins were enriched in polyQ-related NAEs and played non-canonical autophagic roles as genetic manipulation of autophagic molecules exhibited differential impacts on NAEs and SCA3 toxicity. Namely, the down-regulation of Atg1 or Atg12 mitigated while Atg5 RNAi aggravated the disease phenotypes both in Drosophila intestines and compound eyes. Our findings, therefore, provide new mechanistic insights and underscore the fundamental roles of endosome-centered nucleocytoplasmic trafficking and homeostatic endomembrane allocation in the pathogenesis of polyQ diseases.
    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
    环状RNA(circularRNAs)在神经发生期间上调。circRNAs在哪里以及如何定位,以及它们在此过程中扮演的角色仍然难以捉摸。比较H9细胞和H9衍生的前脑(FB)神经元之间的核和细胞质circRNAs,我们发现,富含腺苷(A)的circRNAs的一个子集被限制在H9细胞核中,但在分化后输出到细胞溶胶中。circRNAs的这种亚细胞重新定位由poly(A)结合蛋白PABPC1调节。在H9核中,新产生的富含(A)的circRNAs被PABPC1结合并被核篮蛋白TPR捕获以阻止它们的输出。调节circRNAs中富含(A)的基序改变了它们的亚细胞定位,并且在H9胞质中引入富含(A)的circRNAs导致mRNA翻译抑制。此外,神经元分化后减少的核PABPC1能够输出富含(A)的circRNAs,包括circRTN4(2,3),这是神经突生长所必需的。这些发现揭示了circRNAs的亚细胞定位特征,将它们在神经发生过程中的加工和功能联系起来。
    Circular RNAs (circRNAs) are upregulated during neurogenesis. Where and how circRNAs are localized and what roles they play during this process have remained elusive. Comparing the nuclear and cytoplasmic circRNAs between H9 cells and H9-derived forebrain (FB) neurons, we identify that a subset of adenosine (A)-rich circRNAs are restricted in H9 nuclei but exported to cytosols upon differentiation. Such a subcellular relocation of circRNAs is modulated by the poly(A)-binding protein PABPC1. In the H9 nucleus, newly produced (A)-rich circRNAs are bound by PABPC1 and trapped by the nuclear basket protein TPR to prevent their export. Modulating (A)-rich motifs in circRNAs alters their subcellular localization, and introducing (A)-rich circRNAs in H9 cytosols results in mRNA translation suppression. Moreover, decreased nuclear PABPC1 upon neuronal differentiation enables the export of (A)-rich circRNAs, including circRTN4(2,3), which is required for neurite outgrowth. These findings uncover subcellular localization features of circRNAs, linking their processing and function during neurogenesis.
    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
    慢性阻塞性肺疾病(COPD)构成了显着的健康威胁,其特征是主要由肺单核细胞触发的肺部炎症。尽管炎症在COPD中的中心地位,管理这种反应的监管机制仍然难以捉摸,提出了抗炎干预的挑战。在这项研究中,我们评估了exportins在COPD小鼠模型中的表达,揭示小鼠肺中XPO6的显著上调(P=0.0011)。有趣的是,我们观察到来自人和小鼠COPD受试者的肺单核细胞中XPO6的一致上调(P<0.0001)。此外,在人类肺组织中,XPO6表达与TLR2表达呈正相关(P=0)。体外研究表明,XPO6增强TLR2表达,激活MyD88/NF-κB炎症信号通路。这种激活,反过来,促进促炎细胞因子如TNFα的分泌,单核细胞中的IL-6和IL-1β。机械上,XPO6促进TLR2mRNA的核输出,确保其稳定性和随后在单核细胞中的蛋白质表达。总之,我们的发现揭示了COPD肺单核细胞中XPO6的上调通过促进TLR2mRNA的核输出激活MyD88/NF-κB炎症信号通路,从而确定XPO6是COPD抗炎干预的有希望的治疗靶点。
    Chronic obstructive pulmonary disease (COPD) poses a significant health threat characterized by lung inflammation primarily triggered by pulmonary monocytes. Despite the centrality of inflammation in COPD, the regulatory mechanisms governing this response remain elusive, presenting a challenge for anti-inflammatory interventions. In this study, we assessed the expression of exportins in COPD mouse models, revealing a notable upregulation of XPO6 in the mouse lung (P = 0.0011). Intriguingly, we observed a consistent upregulation of XPO6 in pulmonary monocytes from both human and mouse COPD subjects (P < 0.0001). Furthermore, in human lung tissue, XPO6 expression exhibited a positive correlation with TLR2 expression (P = 0). In vitro investigations demonstrated that XPO6 enhances TLR2 expression, activating the MyD88/NF-κB inflammatory signaling pathway. This activation, in turn, promotes the secretion of pro-inflammatory cytokines such as TNFα, IL-6, and IL-1β in monocytes. Mechanistically, XPO6 facilitates the nuclear export of TLR2 mRNA, ensuring its stability and subsequent protein expression in monocytes. In conclusion, our findings unveil that the upregulation of XPO6 in COPD pulmonary monocytes activates the MyD88/NF-κB inflammatory signaling pathway by facilitating the nuclear export of TLR2 mRNA, thereby identifying XPO6 as a promising therapeutic target for anti-inflammatory interventions in COPD.
    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:Exportin-1(XPO1),调节核-细胞质运输的关键蛋白质,在各种癌症中经常过度表达,驱动肿瘤进展和耐药性。这使得XPO1成为有吸引力的治疗靶标。在过去的几十年里,可用的核出口选择性抑制剂的数量一直在增加。只有KPT-330(selinexor)已成功用于治疗血液恶性肿瘤,和KPT-8602(eltanexor)已在临床试验中用于治疗血液肿瘤。然而,在实体肿瘤的临床研究和治疗结果中,尚未对使用核输出选择性抑制剂抑制XPO1表达进行彻底研究.
    方法:我们收集了大量文献来解释XPO1抑制剂在多种实体瘤的临床前和临床研究中的功效。
    结果:在这篇综述中,我们关注XPO1的核输出功能及其抑制剂在实体恶性肿瘤中的临床试验结果.我们总结了XPO1抑制剂的作用机制和治疗潜力。以及不良反应和反应生物标志物。
    结论:XPO1抑制已成为对抗癌症的一种有希望的治疗策略,提供了一种靶向致瘤过程和克服耐药性的新方法。正弦化合物已证明在广泛的实体瘤中有效,正在进行的研究专注于优化它们的使用,识别反应生物标志物,并开发有效的联合疗法。
    结论:Exportin-1(XPO1)在介导核质转运和细胞周期中起关键作用。XPO1功能障碍促进实体瘤的肿瘤发生和耐药性。XPO1抑制剂在实体瘤治疗中的治疗潜力和正在进行的研究。其他研究对于解决安全性问题和确定预测患者对XPO1抑制剂反应的生物标志物至关重要。
    BACKGROUND: Exportin-1 (XPO1), a crucial protein regulating nuclear-cytoplasmic transport, is frequently overexpressed in various cancers, driving tumor progression and drug resistance. This makes XPO1 an attractive therapeutic target. Over the past few decades, the number of available nuclear export-selective inhibitors has been increasing. Only KPT-330 (selinexor) has been successfully used for treating haematological malignancies, and KPT-8602 (eltanexor) has been used for treating haematologic tumours in clinical trials. However, the use of nuclear export-selective inhibitors for the inhibition of XPO1 expression has yet to be thoroughly investigated in clinical studies and therapeutic outcomes for solid tumours.
    METHODS: We collected numerous literatures to explain the efficacy of XPO1 Inhibitors in preclinical and clinical studies of a wide range of solid tumours.
    RESULTS: In this review, we focus on the nuclear export function of XPO1 and results from clinical trials of its inhibitors in solid malignant tumours. We summarized the mechanism of action and therapeutic potential of XPO1 inhibitors, as well as adverse effects and response biomarkers.
    CONCLUSIONS: XPO1 inhibition has emerged as a promising therapeutic strategy in the fight against cancer, offering a novel approach to targeting tumorigenic processes and overcoming drug resistance. SINE compounds have demonstrated efficacy in a wide range of solid tumours, and ongoing research is focused on optimizing their use, identifying response biomarkers, and developing effective combination therapies.
    CONCLUSIONS: Exportin-1 (XPO1) plays a critical role in mediating nucleocytoplasmic transport and cell cycle. XPO1 dysfunction promotes tumourigenesis and drug resistance within solid tumours. The therapeutic potential and ongoing researches on XPO1 inhibitors in the treatment of solid tumours. Additional researches are essential to address safety concerns and identify biomarkers for predicting patient response to XPO1 inhibitors.
    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
    父母经历会影响后代的表型可塑性。在蝗虫中,成年人经历的人口密度调节卵的数量和孵化同步性,导致蝗虫爆发。然而,从父母到后代的信号传递途径仍不清楚。这里,我们发现转录因子Forkheadbox蛋白N1(FOXN1)响应高种群密度并激活蝗虫中的聚嘧啶束结合蛋白1(Ptbp1)。FOXN1-PTBP1作为miR-276的上游调节因子,miR-276是一种控制卵孵化同步性的miRNA。PTBP1以“CU基序”依赖性方式增强前miR-276的核质转运,通过与初级输出蛋白输出蛋白5(XPO5)合作。前miR-276的核输出增强提高了末端卵母细胞中miR-276的表达,其中FOXN1激活Ptbp1并导致卵孵化同步,以响应高人口密度。此外,PTBP1提示的pre-miR-276的核输出在昆虫中是保守的,暗示了一种无处不在的机制来介导跨代效应。
    Parental experiences can affect the phenotypic plasticity of offspring. In locusts, the population density that adults experience regulates the number and hatching synchrony of their eggs, contributing to locust outbreaks. However, the pathway of signal transmission from parents to offspring remains unclear. Here, we find that transcription factor Forkhead box protein N1 (FOXN1) responds to high population density and activates the polypyrimidine tract-binding protein 1 (Ptbp1) in locusts. FOXN1-PTBP1 serves as an upstream regulator of miR-276, a miRNA to control egg-hatching synchrony. PTBP1 boosts the nucleo-cytoplasmic transport of pre-miR-276 in a \"CU motif\"-dependent manner, by collaborating with the primary exportin protein exportin 5 (XPO5). Enhanced nuclear export of pre-miR-276 elevates miR-276 expression in terminal oocytes, where FOXN1 activates Ptbp1 and leads to egg-hatching synchrony in response to high population density. Additionally, PTBP1-prompted nuclear export of pre-miR-276 is conserved in insects, implying a ubiquitous mechanism to mediate transgenerational effects.
    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
    SARS-CoV-2(严重急性呼吸综合征冠状病毒2)的非结构蛋白1(Nsp1)是一种毒力因子,靶向多种细胞途径以抑制宿主基因表达和抗病毒反应。然而,各种Nsp1介导的功能的潜在机制及其对SARS-CoV-2毒力的贡献仍不清楚.Nsp1的靶标是mRNA(信使核糖核酸)输出受体NXF1-NXT1,其介导mRNA从细胞核向细胞质的核输出。基于Nsp1的晶体结构,我们在Nsp1表面上产生了突变体,并鉴定了一个酸性N末端贴片,该贴片对于与NXF1-NXT1的相互作用至关重要。可光活化的Nsp1探针揭示NXF1的RNA识别基序(RRM)结构域作为Nsp1的N-末端结合位点。通过突变Nsp1N末端酸性贴片,我们确定了Nsp1的功能分离突变体,该突变体保留了其翻译抑制功能,但实质上失去了与NXF1的相互作用,并恢复了Nsp1介导的mRNA输出抑制.然后,我们在Nsp1N端酸性贴片上产生了重组(r)SARS-CoV-2突变体,发现该表面是促进NXF1结合和抑制宿主mRNA核输出的关键,病毒复制,和体内致病性。因此,这些发现提供了对Nsp1介导的mRNA输出抑制的机制理解,并确定了该途径在SARS-CoV-2毒力中的重要性。
    The nonstructural protein 1 (Nsp1) of SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2) is a virulence factor that targets multiple cellular pathways to inhibit host gene expression and antiviral response. However, the underlying mechanisms of the various Nsp1-mediated functions and their contributions to SARS-CoV-2 virulence remain unclear. Among the targets of Nsp1 is the mRNA (messenger ribonucleic acid) export receptor NXF1-NXT1, which mediates nuclear export of mRNAs from the nucleus to the cytoplasm. Based on Nsp1 crystal structure, we generated mutants on Nsp1 surfaces and identified an acidic N-terminal patch that is critical for interaction with NXF1-NXT1. Photoactivatable Nsp1 probe reveals the RNA Recognition Motif (RRM) domain of NXF1 as an Nsp1 N-terminal binding site. By mutating the Nsp1 N-terminal acidic patch, we identified a separation-of-function mutant of Nsp1 that retains its translation inhibitory function but substantially loses its interaction with NXF1 and reverts Nsp1-mediated mRNA export inhibition. We then generated a recombinant (r)SARS-CoV-2 mutant on the Nsp1 N-terminal acidic patch and found that this surface is key to promote NXF1 binding and inhibition of host mRNA nuclear export, viral replication, and pathogenicity in vivo. Thus, these findings provide a mechanistic understanding of Nsp1-mediated mRNA export inhibition and establish the importance of this pathway in the virulence of SARS-CoV-2.
    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
    红细胞生成和巨核细胞生成受到信号通路的严格调节。然而,信号通路调节控制红细胞生成和巨核细胞生成的关键转录因子的确切分子机制仍部分了解。在这里,我们确定了热休克同源B(HSCB),以其铁硫簇输送功能而闻名,作为K562人红白血病细胞和脐带血来源的人CD34+CD90+造血干细胞(HSC)红细胞生成过程中GATA1(FOG1)核易位不可缺少的蛋白质,以及在CD34+CD90+HSC的巨核细胞生成过程中。机械上,HSCB可被磷酸肌醇-3-激酶(PI3K)磷酸化,与转化酸性卷曲螺旋蛋白3(TACC3)结合并介导蛋白酶体降解,否则将FOG1滞留在细胞质中,从而促进FOG1核易位。鉴于PI3K在红细胞生成和巨核细胞生成期间都被激活,FOG1是这些过程的关键转录因子,我们的发现阐明了一个重要的,以前未认识到的铁硫簇递送独立于HSCB在红细胞生成和巨核细胞生成中的功能。
    Erythropoiesis and megakaryopoiesis are stringently regulated by signaling pathways. However, the precise molecular mechanisms through which signaling pathways regulate key transcription factors controlling erythropoiesis and megakaryopoiesis remain partially understood. Herein, we identified heat shock cognate B (HSCB), which is well known for its iron-sulfur cluster delivery function, as an indispensable protein for friend of GATA 1 (FOG1) nuclear translocation during erythropoiesis of K562 human erythroleukemia cells and cord-blood-derived human CD34+CD90+hematopoietic stem cells (HSCs), as well as during megakaryopoiesis of the CD34+CD90+HSCs. Mechanistically, HSCB could be phosphorylated by phosphoinositol-3-kinase (PI3K) to bind with and mediate the proteasomal degradation of transforming acidic coiled-coil containing protein 3 (TACC3), which otherwise detained FOG1 in the cytoplasm, thereby facilitating FOG1 nuclear translocation. Given that PI3K is activated during both erythropoiesis and megakaryopoiesis, and that FOG1 is a key transcription factor for these processes, our findings elucidate an important, previously unrecognized iron-sulfur cluster delivery independent function of HSCB in erythropoiesis and megakaryopoiesis.
    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
    严格调节NF-κB途径的激活以防止过度的炎症和免疫应答。在众所周知的负反馈模型中,IκBα依赖性NF-κB终止是活化后期的延迟反应模式,介导活性NF-κB快速终止的机制尚不清楚。这里,我们显示了由CLK2介导的不依赖IκBα的核NF-κB的快速终止,其通过磷酸化核中Ser180处的NF-κB的RelA/p65亚基来负调节活性NF-κB,以限制其通过降解和核输出的转录激活。CLK2的消耗增加了炎性细胞因子的产生,减少病毒复制并增加小鼠的存活率。机械上,CLK2在细胞核中Ser180磷酸化RelA/p65,导致泛素-蛋白酶体介导的降解和胞质再分布。重要的是,CLK2抑制剂促进细胞因子的产生,减少病毒复制,加速鼠牛皮癣.这项研究揭示了NF-κB早期终止的IκBα非依赖性机制,其中磷酸化的Ser180RelA/p65关闭了与转录激活相关的翻译后修饰,最终导致RelA/p65的降解和核输出,以抑制过度的炎症激活。我们的发现表明,RelA/p65在细胞核中Ser180的磷酸化抑制早期NF-κB激活,从而介导NF-κB的负调节。
    Activation of the NF-κB pathway is strictly regulated to prevent excessive inflammatory and immune responses. In a well-known negative feedback model, IκBα-dependent NF-κB termination is a delayed response pattern in the later stage of activation, and the mechanisms mediating the rapid termination of active NF-κB remain unclear. Here, we showed IκBα-independent rapid termination of nuclear NF-κB mediated by CLK2, which negatively regulated active NF-κB by phosphorylating the RelA/p65 subunit of NF-κB at Ser180 in the nucleus to limit its transcriptional activation through degradation and nuclear export. Depletion of CLK2 increased the production of inflammatory cytokines, reduced viral replication and increased the survival of the mice. Mechanistically, CLK2 phosphorylated RelA/p65 at Ser180 in the nucleus, leading to ubiquitin‒proteasome-mediated degradation and cytoplasmic redistribution. Importantly, a CLK2 inhibitor promoted cytokine production, reduced viral replication, and accelerated murine psoriasis. This study revealed an IκBα-independent mechanism of early-stage termination of NF-κB in which phosphorylated Ser180 RelA/p65 turned off posttranslational modifications associated with transcriptional activation, ultimately resulting in the degradation and nuclear export of RelA/p65 to inhibit excessive inflammatory activation. Our findings showed that the phosphorylation of RelA/p65 at Ser180 in the nucleus inhibits early-stage NF-κB activation, thereby mediating the negative regulation of NF-κB.
    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
    癌症转移是乳腺癌(BC)患者死亡的主要原因。肝脏是乳腺癌转移的常见部位,乳腺癌肝转移(BCLMs)患者的5年生存率仅为8.5%左右。CircRNAs参与多种癌症相关的病理行为,它们独特的结构和对RNA降解的抗性使它们成为理想的诊断生物标志物和治疗靶标。因此,研究circRNAs在肿瘤转移中的作用和分子机制具有重要意义。通过circRNA微阵列和qRT-PCR实验将CircLIFR-007鉴定为BC转移中的关键环状RNA。进行细胞功能测定以探索circLIFR-007在乳腺癌细胞中的作用。体内实验验证了circLIFR-007的功能。进行了几种分子测定以研究潜在的机制。我们发现circLIFR-007在乳腺癌肝转移中起负控制因子的作用。CircLIFR-007通过输出hnRNPA1来上调YAP的磷酸化水平,以促进细胞质中hnRNPA1和YAP之间的结合。circirLIFR-007的过表达抑制了肝转移相关蛋白的表达,SREBF1和SNAI1受转录因子YAP调控。功能上,circLIFR-007在体内和体外均抑制乳腺癌细胞的增殖和转移。
    Cancer metastasis is the major cause of death in patients with breast cancer (BC). The liver is a common site of breast cancer metastasis, and the 5-year survival rate of patients with breast cancer liver metastases (BCLMs) is only about 8.5 %. CircRNAs are involved in a variety of cancer-related pathological behaviors, and their unique structure and resistance to RNA degradation enable them to serve as ideal diagnostic biomarkers and therapeutic targets. Therefore, it is important to investigate the role and molecular mechanism of circRNAs in cancer metastasis. CircLIFR-007 was identified as a critical circular RNA in BC metastasis by circRNAs microarray and qRT-PCR experiment. Cell function assays were performed to explore the effect of circLIFR-007 in breast cancer cells. Experiments in vivo validated the function of circLIFR-007. Several molecular assays were performed to investigate the underlying mechanisms. We found that circLIFR-007 acted as a negative controller in breast cancer liver metastasis. CircLIFR-007 upregulates the phosphorylation level of YAP by exporting hnRNPA1 to promote the combination between hnRNPA1 and YAP in the cytoplasm. Overexpression of circLIFR-007 suppressed the expression of liver metastasis-related proteins, SREBF1 and SNAI1, which were regulated by transcription factor YAP. Functionally, circLIFR-007 inhibits the proliferation and metastasis of breast cancer cells both in vivo and in vitro.
    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
    心肌梗塞(MI)是一种潜在的致命疾病,在全球范围内导致大量死亡。增加肌细胞中脂肪酸氧化的策略被认为是加速新陈代谢以满足能量需求的治疗途径。我们进行了这项研究,旨在研究KN-93的作用,它诱导组蛋白去乙酰化酶(HDAC)4穿梭到细胞核,脂肪酸氧化及相关基因的表达。通过给予异丙肾上腺素诱导心肌梗塞的小鼠模型。通过检测心脏损伤标志物来评估心脏损伤。通过检测相关基因的表达来评价脂肪酸氧化水平。利用细胞溶质或细胞核中的免疫荧光和免疫印迹来观察HDAC4的分布。通过免疫共沉淀证实了HDAC4与特异性蛋白(SP)1之间的相互作用。在KN-93处理和HDAC4抑制剂后测试SP1的乙酰化水平。使用耗氧率和免疫印迹实验来确定KN-93对增加脂肪酸氧化的作用是否通过HDAC4和SP1。KN-93的给药在体外和体内均显着减少了心肌梗塞中的心脏损伤并促进了脂肪酸氧化。显示KN-93介导HDAC4的核易位。发现HDAC4与SP1相互作用并减少SP1乙酰化。HDAC4或SP1抑制剂减弱了KN-93对脂肪酸氧化的影响。KN-93促进HDAC4转位到细胞核,从而潜在地增强SPl的脂肪酸氧化。
    Myocardial infarction (MI) is a potentially fatal disease that causes a significant number of deaths worldwide. The strategy of increasing fatty acid oxidation in myocytes is considered a therapeutic avenue to accelerate metabolism to meet energy demands. We conducted the study aiming to investigate the effect of KN-93, which induces histone deacetylase (HDAC)4 shuttling to the nucleus, on fatty acid oxidation and the expression of related genes. A mouse model of myocardial infarction was induced by isoprenaline administration. Heart damage was assessed by the detection of cardiac injury markers. The level of fatty acid oxidation level was evaluated by testing the expression of related genes. Both immunofluorescence and immunoblotting in the cytosol or nucleus were utilized to observe the distribution of HDAC4. The interaction between HDAC4 and specificity protein (SP)1 was confirmed by co-immunoprecipitation. The acetylation level of SP1 was tested after KN-93 treatment and HDAC4 inhibitor. Oxygen consumption rate and immunoblotting experiments were used to determine whether the effect of KN-93 on increasing fatty acid oxidation is through HDAC4 and SP1. Administration of KN-93 significantly reduced cardiac injury in myocardial infarction and promoted fatty acid oxidation both in vitro and in vivo. KN-93 was shown to mediate nuclear translocation of HDAC4. HDAC4 was found to interact with SP1 and reduce SP1 acetylation. HDAC4 or SP1 inhibitors attenuated the effect of KN-93 on fatty acid oxidation. In conclusion, KN-93 promotes HDAC4 translocation to the nucleus, thereby potentially enhancing fatty acid oxidation by SP1.
    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