Nuclear envelope

核包络
  • 文章类型: Journal Article
    中性粒细胞胞外陷阱(NET)形成的诱导剂是异质的,因此,NET的形成没有不可缺少的特定途径或特征分子。但是某些事件,如组蛋白修饰,染色质去凝聚,核包络击穿,和NET版本无处不在。在NET形成过程中,中性粒细胞急剧重排它们的细胞质,颗粒和核含量。然而,NET形成过程中解码每个步骤的确切机制仍然难以捉摸。这里,我们研究了网络形成过程中核包膜击穿的机理。免疫荧光显微镜评估显示,在NET形成过程中,外核膜蛋白nesprin-1逐渐分解,核形态发生改变。由各种诱导物产生的NET的MALDI-TOF分析检测到nesprin-1片段的积累。这表明nesprin-1降解发生在NET发布之前。在钙蛋白酶-1存在下,抑制剂nesprin-1降解在钙驱动的NET形成中降低。显微镜评估证实,层粘连蛋白B受体(LBR)的崩解和肌动蛋白细胞骨架的崩溃发生在NET释放的早期和后期,分别。我们得出结论,钙蛋白酶-1降解nesprin-1,协调核膜的弱化,有助于LBR分解,促进DNA释放,最后,NET形成。
    The inducers of neutrophil extracellular trap (NET) formation are heterogeneous and consequently, there is no specific pathway or signature molecule indispensable for NET formation. But certain events such as histone modification, chromatin decondensation, nuclear envelope breakdown, and NET release are ubiquitous. During NET formation, neutrophils drastically rearrange their cytoplasmic, granular and nuclear content. Yet, the exact mechanism for decoding each step during NET formation still remains elusive. Here, we investigated the mechanism of nuclear envelope breakdown during NET formation. Immunofluorescence microscopic evaluation revealed a gradual disintegration of outer nuclear membrane protein nesprin-1 and alterations in nuclear morphology during NET formation. MALDI-TOF analysis of NETs that had been generated by various inducers detected the accumulation of nesprin-1 fragments. This suggests that nesprin-1 degradation occurs before NET release. In the presence of a calpain-1, inhibitor nesprin-1 degradation was decreased in calcium driven NET formation. Microscopic evaluation confirmed that the disintegration of the lamin B receptor (LBR) and the collapse of the actin cytoskeleton occurs in early and later phases of NET release, respectively. We conclude that the calpain-1 degrades nesprin-1, orchestrates the weakening of the nuclear membrane, contributes to LBR disintegration, and promoting DNA release and finally, NETs formation.
    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:牙周膜干细胞(PDLSCs)是组织工程和临床应用中重要的种子细胞。它们是用于感测各种机械应力的优先受体细胞。Yes相关蛋白(YAP)是公认的机械敏感转录因子。然而,YAP在张力应激(TS)下调节PDLSCs命运的作用及其机制尚不清楚。
    方法:用荧光染色法研究TS对PDLSCs形态和命运的影响,透射电子显微镜,流式细胞术和定量实时聚合酶链反应(qRT-PCR)。然后qRT-PCR,西方印迹,免疫荧光染色和基因敲低实验研究YAP的表达和分布及其与PDLSCs增殖的相关性。随后通过添加细胞骨架抑制剂来探索细胞骨架动力学对YAP核易位的影响。通过qRT-PCR和western印迹证明细胞骨架动力学对LINC复合物表达的影响。腺病毒破坏LINC复合物后,研究了LINC复合物对YAP核易位和PDLSCs增殖的影响.然后进行线粒体相关检测以探索线粒体在YAP核易位中的作用。最后,通过构建Sprague-Dawley大鼠正畸牙齿移动模型对体外结果进行验证。
    结果:TS增强了F-肌动蛋白的聚合和拉伸,上调LINC复合物的表达。这进一步加强了对核弹的拉力,扩大了核孔隙,并促进了YAP的核进入,从而增强增殖相关基因的表达。在这个过程中,线粒体沿着重建的微管运输到细胞核的外围。他们产生ATP以帮助YAP的核易位,并在一定程度上驱动F-肌动蛋白聚合。当LINC复合体被摧毁时,YAP的核易位被抑制,这限制了PDLSCs的增殖,牙周组织改建受阻,阻碍牙齿移动。
    结论:我们的研究证实,适当的TS可以通过机械驱动的F-肌动蛋白/LINC复合物/YAP轴促进PDLSCs增殖和牙周组织重塑,为临床上种子细胞扩增和促进健康有效的牙齿移动提供理论指导。
    BACKGROUND: Periodontal ligament stem cells (PDLSCs) are important seed cells in tissue engineering and clinical applications. They are the priority receptor cells for sensing various mechanical stresses. Yes-associated protein (YAP) is a recognized mechanically sensitive transcription factor. However, the role of YAP in regulating the fate of PDLSCs under tension stress (TS) and its underlying mechanism is still unclear.
    METHODS: The effects of TS on the morphology and fate of PDLSCs were investigated using fluorescence staining, transmission electron microscopy, flow cytometry and quantitative real-time polymerase chain reaction (qRT-PCR). Then qRT-PCR, western blotting, immunofluorescence staining and gene knockdown experiments were performed to investigate the expression and distribution of YAP and its correlation with PDLSCs proliferation. The effects of cytoskeleton dynamics on YAP nuclear translocation were subsequently explored by adding cytoskeleton inhibitors. The effect of cytoskeleton dynamics on the expression of the LINC complex was proved through qRT-PCR and western blotting. After destroying the LINC complex by adenovirus, the effects of the LINC complex on YAP nuclear translocation and PDLSCs proliferation were investigated. Mitochondria-related detections were then performed to explore the role of mitochondria in YAP nuclear translocation. Finally, the in vitro results were verified by constructing orthodontic tooth movement models in Sprague-Dawley rats.
    RESULTS: TS enhanced the polymerization and stretching of F-actin, which upregulated the expression of the LINC complex. This further strengthened the pull on the nuclear envelope, enlarged the nuclear pore, and facilitated YAP\'s nuclear entry, thus enhancing the expression of proliferation-related genes. In this process, mitochondria were transported to the periphery of the nucleus along the reconstructed microtubules. They generated ATP to aid YAP\'s nuclear translocation and drove F-actin polymerization to a certain degree. When the LINC complex was destroyed, the nuclear translocation of YAP was inhibited, which limited PDLSCs proliferation, impeded periodontal tissue remodeling, and hindered tooth movement.
    CONCLUSIONS: Our study confirmed that appropriate TS could promote PDLSCs proliferation and periodontal tissue remodeling through the mechanically driven F-actin/LINC complex/YAP axis, which could provide theoretical guidance for seed cell expansion and for promoting healthy and effective tooth movement in clinical practice.
    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
    丝状肌动蛋白纤维(F-actin)的定量和体积评估由于其相互关联的性质仍然具有挑战性。导致研究人员使用基于阈值或定性的测量方法,可重复性差。在这里,我们介绍了一种新颖的基于机器学习的方法,用于精确量化和重建核相关的F-肌动蛋白。利用卷积神经网络(CNN),我们从3D共聚焦显微镜图像中分割肌动蛋白丝和细胞核,然后通过连接横截面切片上的相交轮廓来重建每根纤维。这允许以可再现的方式测量肌动蛋白丝的总数和单个肌动蛋白丝的长度和体积。关注F-肌动蛋白在支持核细胞骨架连接中的作用,我们定量了顶端F-肌动蛋白,基底F-肌动蛋白,和在核骨架和细胞骨架(LINC)复合物的接头破坏后的间充质干细胞(MSC)的核结构。在间充质干细胞(MSC)中禁用LINC在核膜处产生F-肌动蛋白解体,其特征在于肌动蛋白纤维的长度和体积较短,从而导致较小的细长核形状。我们的发现不仅为机械生物学提供了一种新工具,而且还引入了一种新颖的管道,用于基于F-肌动蛋白的定量测量来开发现实的计算模型。
    Quantitative and volumetric assessment of filamentous actin fibers (F-actin) remains challenging due to their interconnected nature, leading researchers to utilize threshold based or qualitative measurement methods with poor reproducibility. Here we introduce a novel machine learning based methodology for accurate quantification and reconstruction of nuclei-associated F-actin. Utilizing a Convolutional Neural Network (CNN), we segment actin filaments and nuclei from 3D confocal microscopy images and then reconstruct each fiber by connecting intersecting contours on cross-sectional slices. This allowed measurement of the total number of actin filaments and individual actin filament length and volume in a reproducible fashion. Focusing on the role of F-actin in supporting nucleocytoskeletal connectivity, we quantified apical F-actin, basal F-actin, and nuclear architecture in mesenchymal stem cells (MSCs) following the disruption of the Linker of Nucleoskeleton and Cytoskeleton (LINC) Complexes. Disabling LINC in mesenchymal stem cells (MSCs) generated F-actin disorganization at the nuclear envelope characterized by shorter length and volume of actin fibers contributing a less elongated nuclear shape. Our findings not only present a new tool for mechanobiology but introduce a novel pipeline for developing realistic computational models based on quantitative measures of F-actin.
    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
    Cockayne综合征(CS)是一种以小头畸形为特征的过早衰老疾病,生长失败,和神经变性。它是由编码Cockayne综合征B(CSB)和A(CSA)蛋白的ERCC6或ERCC8突变引起的,分别。CSA和CSB在转录偶联核苷酸切除修复中具有明确的作用,负责去除庞大的DNA损伤,包括紫外线照射引起的。这里,我们报道CSA功能障碍导致核膜(NE)完整性缺陷.NE功能障碍是由NE蛋白突变引起的早衰性疾病的特征,比如Hutchinson-Gilford早衰综合征.然而,从未在Cockayne综合征中报道过。我们观察到CSA功能障碍影响了NE处的LEMD2掺入,并增加了肌动蛋白应力纤维,这有助于增强对NE的机械应力。总之,这些导致与cGAS/STING途径激活相关的NE异常。靶向核骨架和细胞骨架复合物的接头足以挽救这些表型。这项工作揭示了由DNA损伤修复蛋白突变引起的早衰综合征中的NE功能障碍,加强NE放松管制和衰老之间的联系。
    Cockayne syndrome (CS) is a premature ageing condition characterized by microcephaly, growth failure, and neurodegeneration. It is caused by mutations in ERCC6 or ERCC8 encoding for Cockayne syndrome B (CSB) and A (CSA) proteins, respectively. CSA and CSB have well-characterized roles in transcription-coupled nucleotide excision repair, responsible for removing bulky DNA lesions, including those caused by UV irradiation. Here, we report that CSA dysfunction causes defects in the nuclear envelope (NE) integrity. NE dysfunction is characteristic of progeroid disorders caused by a mutation in NE proteins, such as Hutchinson-Gilford progeria syndrome. However, it has never been reported in Cockayne syndrome. We observed CSA dysfunction affected LEMD2 incorporation at the NE and increased actin stress fibers that contributed to enhanced mechanical stress to the NE. Altogether, these led to NE abnormalities associated with the activation of the cGAS/STING pathway. Targeting the linker of the nucleoskeleton and cytoskeleton complex was sufficient to rescue these phenotypes. This work reveals NE dysfunction in a progeroid syndrome caused by mutations in a DNA damage repair protein, reinforcing the connection between NE deregulation and ageing.
    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
    氧化损伤引发微核膜破裂和缺陷修复。
    Oxidative damage triggers micronuclear membrane rupture and defective repair.
    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
    含染色体的微核是侵袭性癌症的标志。微核经常经历不可逆的塌陷,将其封闭的染色质暴露于细胞质中。微核破裂催化染色体重排,表观遗传异常,和炎症,然而,保护微核完整性的机制知之甚少。在这项研究中,我们发现线粒体衍生的活性氧(ROS)通过促进带电多囊体蛋白7(CHMP7)的非经典功能来破坏微核,膜修复复合物的支架蛋白,称为转运III(ESCRT-III)所需的内体分选复合物。ROS将CHMP7保留在微核中,同时破坏其与其他ESCRT-III成分的相互作用。ROS诱导的半胱氨酸氧化刺激CHMP7寡聚化并与核膜蛋白LEMD2结合,破坏微核包膜。此外,这种ROS-CHMP7病理轴导致已知由微核破裂引起的染色体破裂。它还在缺氧条件下介导微核不完整,将肿瘤缺氧与驱动癌症进展的下游过程联系起来。
    Chromosome-containing micronuclei are a hallmark of aggressive cancers. Micronuclei frequently undergo irreversible collapse, exposing their enclosed chromatin to the cytosol. Micronuclear rupture catalyzes chromosomal rearrangements, epigenetic abnormalities, and inflammation, yet mechanisms safeguarding micronuclear integrity are poorly understood. In this study, we found that mitochondria-derived reactive oxygen species (ROS) disrupt micronuclei by promoting a noncanonical function of charged multivesicular body protein 7 (CHMP7), a scaffolding protein for the membrane repair complex known as endosomal sorting complex required for transport III (ESCRT-III). ROS retained CHMP7 in micronuclei while disrupting its interaction with other ESCRT-III components. ROS-induced cysteine oxidation stimulated CHMP7 oligomerization and binding to the nuclear membrane protein LEMD2, disrupting micronuclear envelopes. Furthermore, this ROS-CHMP7 pathological axis engendered chromosome shattering known to result from micronuclear rupture. It also mediated micronuclear disintegrity under hypoxic conditions, linking tumor hypoxia with downstream processes driving cancer progression.
    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
    染色体不稳定性(CIN)会产生微核异常的核外结构,从而催化获得癌症中存在的复杂染色体重排。微核的特征是持续的DNA损伤和灾难性的核包膜破裂,将DNA暴露于细胞质中。我们发现自噬受体p62/SQSTM1调节微核稳定性,影响染色体断裂和重排。机械上,微核与线粒体的接近导致氧化驱动的p62同源寡聚化,通过触发自噬降解限制了转运(ESCRT)依赖性微核包膜修复所需的内体分选复合物。我们还发现,p62水平与人类癌细胞系中染色体增生增加以及结直肠肿瘤中CIN增加相关。因此,p62作为微核的调节剂,可能作为高CIN肿瘤的预后标志物。
    Chromosomal instability (CIN) generates micronuclei-aberrant extranuclear structures that catalyze the acquisition of complex chromosomal rearrangements present in cancer. Micronuclei are characterized by persistent DNA damage and catastrophic nuclear envelope collapse, which exposes DNA to the cytoplasm. We found that the autophagic receptor p62/SQSTM1 modulates micronuclear stability, influencing chromosome fragmentation and rearrangements. Mechanistically, proximity of micronuclei to mitochondria led to oxidation-driven homo-oligomerization of p62, limiting endosomal sorting complex required for transport (ESCRT)-dependent micronuclear envelope repair by triggering autophagic degradation. We also found that p62 levels correlate with increased chromothripsis across human cancer cell lines and with increased CIN in colorectal tumors. Thus, p62 acts as a regulator of micronuclei and may serve as a prognostic marker for tumors with high CIN.
    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
    在动物中,有丝分裂涉及核包膜的分解和个体化的分选,浓缩的染色体.在有丝分裂退出期间,新出现的核在单个相互连接的染色体周围重新组装了一个核包膜。核重组的分子机制尚未完全了解。此外,在这个过程中,缺陷的细胞和生理后果在很大程度上是未知的。这里,我们已经描述了果蝇核重组所必需的机制。我们显示Ankle2在重组细胞核时促进BAF和Lamin的PP2A依赖性募集,这种机制的失败会导致有丝分裂后严重的核缺陷。然后,我们利用这种机制的扰动来研究体内组织发育过程中对核重组缺陷的生理反应。部分耗尽Ankle2,BAF,或想象翼盘中的Lamin导致翼发育缺陷并伴有细胞凋亡。我们发现阻断细胞凋亡会强烈增强发育缺陷。阻断p53不会阻止细胞凋亡,但会增强由于细胞周期检查点丢失而导致的缺陷。我们的结果表明,凋亡和p53依赖性反应在保护组织发育以应对零星的核重组缺陷中起着至关重要的作用。
    In animals, mitosis involves the breakdown of the nuclear envelope and the sorting of individualized, condensed chromosomes. During mitotic exit, emerging nuclei reassemble a nuclear envelope around a single mass of interconnecting chromosomes. The molecular mechanisms of nuclear reassembly are incompletely understood. Moreover, the cellular and physiological consequences of defects in this process are largely unexplored. Here, we have characterized a mechanism essential for nuclear reassembly in Drosophila. We show that Ankle2 promotes the PP2A-dependent recruitment of BAF and Lamin at reassembling nuclei, and that failures in this mechanism result in severe nuclear defects after mitosis. We then took advantage of perturbations in this mechanism to investigate the physiological responses to nuclear reassembly defects during tissue development in vivo. Partial depletion of Ankle2, BAF, or Lamin in imaginal wing discs results in wing development defects accompanied by apoptosis. We found that blocking apoptosis strongly enhances developmental defects. Blocking p53 does not prevent apoptosis but enhances defects due to the loss of a cell cycle checkpoint. Our results suggest that apoptotic and p53-dependent responses play a crucial role in safeguarding tissue development in response to sporadic nuclear reassembly defects.
    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
    核膜上的核孔复合物是细胞核与细胞质之间连通的唯一通道,调节各种分子的运输,包括核酸和蛋白质。本工作研究了带负电荷的石墨烯量子点通过核膜的传输动力学,专注于量化它们的运输特征。使用延时共聚焦荧光显微镜在透化的HeLa细胞中进行实验。我们的发现表明,带负电荷的石墨烯量子点表现出快速传输到原子核,在易位过程中涉及两种不同的运输途径。石墨烯量子点的核进出口互补实验验证了输运的双向性,可比的运输率证明了这一点。研究还表明,带负电荷的石墨烯量子点具有良好的保留性能,强调他们作为毒品携带者的潜力。
    The nuclear pore complexes on the nuclear membrane serve as the exclusive gateway for communication between the nucleus and the cytoplasm, regulating the transport of various molecules, including nucleic acids and proteins. The present work investigates the kinetics of the transport of negatively charged graphene quantum dots through nuclear membranes, focusing on quantifying their transport characteristics. Experiments are carried out in permeabilized HeLa cells using time-lapse confocal fluorescence microscopy. Our findings indicate that negatively charged graphene quantum dots exhibit rapid transport to the nuclei, involving two distinct transport pathways in the translocation process. Complementary experiments on the nuclear import and export of graphene quantum dots validate the bi-directionality of transport, as evidenced by comparable transport rates. The study also shows that the negatively charged graphene quantum dots possess favorable retention properties, underscoring their potential as drug carriers.
    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
    亨廷顿病(HD)是由亨廷顿蛋白的多聚谷氨酰胺扩增引起的,导致聚谷氨酰胺聚集体的形成。导致复杂HD病理的毒性机制仅部分了解。这里,我们表明,核多聚谷氨酰胺聚集体会引起核包膜(NE)起泡和破裂,这些破裂通常会被不完全修复。这些破裂与核层的破坏同时发生,并导致层疤痕的形成。扩展显微镜能够解析核聚集体的超微结构,并显示聚谷氨酰胺原纤维在破裂部位粘附到细胞质中,提出了一种不完全修复的机制。此外,我们发现NE修复因子经常积累在核聚集体附近,与停滞的修复一致。这些发现暗示核polyQ聚集体诱导的NE完整性丧失是亨廷顿氏病和其他多聚谷氨酰胺疾病的潜在促成因素。
    Huntington\'s disease (HD) is caused by a polyglutamine expansion of the huntingtin protein, resulting in the formation of polyglutamine aggregates. The mechanisms of toxicity that result in the complex HD pathology remain only partially understood. Here, we show that nuclear polyglutamine aggregates induce nuclear envelope (NE) blebbing and ruptures that are often repaired incompletely. These ruptures coincide with disruptions of the nuclear lamina and lead to lamina scar formation. Expansion microscopy enabled resolving the ultrastructure of nuclear aggregates and revealed polyglutamine fibrils sticking into the cytosol at rupture sites, suggesting a mechanism for incomplete repair. Furthermore, we found that NE repair factors often accumulated near nuclear aggregates, consistent with stalled repair. These findings implicate nuclear polyQ aggregate-induced loss of NE integrity as a potential contributing factor to Huntington\'s disease and other polyglutamine diseases.
    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