nuclear envelope rupture

核包膜破裂
  • 文章类型: Journal Article
    在Lamin相关的扩张型心肌病中,核包膜(NE)破裂是新兴的观察结果,由LaminA/C的功能丧失突变引起的成人发作疾病,核层成分.这里,我们使用Lamin心肌病小鼠模型检验了一个普遍的假设,即NE破裂触发病理性cGAS-STING胞浆DNA传感通路.成年小鼠心肌细胞中LaminA/C的减少导致心肌细胞中普遍的NE破裂,在炎症转录之前,纤维化,和致命的扩张型心肌病.NE破裂后是DNA损伤积累,而不会导致心肌细胞立即死亡。然而,cGAS-STING依赖性炎症信号仍无活性。删除cGas或Sting不能挽救小鼠模型中的心肌病。cGAS-STING活化的缺乏可能是由于在基线时成年心肌细胞中几乎不存在cGAS表达。相反,细胞外基质(ECM)信号被激活,并被预测启动从Lamin减少的心肌细胞到成纤维细胞的促炎通讯。我们的工作提名ECM信号,不是cGAS-STING,作为Lamin心肌病的潜在炎症因素。
    Nuclear envelope (NE) ruptures are emerging observations in Lamin-related dilated cardiomyopathy, an adult-onset disease caused by loss-of-function mutations in Lamin A/C, a nuclear lamina component. Here, we test a prevailing hypothesis that NE ruptures trigger the pathological cGAS-STING cytosolic DNA-sensing pathway using a mouse model of Lamin cardiomyopathy. The reduction of Lamin A/C in cardio-myocyte of adult mice causes pervasive NE ruptures in cardiomyocytes, preceding inflammatory transcription, fibrosis, and fatal dilated cardiomyopathy. NE ruptures are followed by DNA damage accumulation without causing immediate cardiomyocyte death. However, cGAS-STING-dependent inflammatory signaling remains inactive. Deleting cGas or Sting does not rescue cardiomyopathy in the mouse model. The lack of cGAS-STING activation is likely due to the near absence of cGAS expression in adult cardiomyocytes at baseline. Instead, extracellular matrix (ECM) signaling is activated and predicted to initiate pro-inflammatory communication from Lamin-reduced cardiomyocytes to fibroblasts. Our work nominates ECM signaling, not cGAS-STING, as a potential inflammatory contributor in Lamin cardiomyopathy.
    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
    在真核细胞中,核膜(NE)是细胞核和细胞质之间的膜分区,以分隔核内容物。它在促进核功能包括转录中起着重要作用,DNA复制和修复。在哺乳动物细胞中,NE在细胞分裂过程中分解然后重新形成,在中间阶段,它在机械力引起的NE破裂后不久就恢复了。这样,分配效应通过整个细胞周期的动态过程来调节。重建NE结构的失败会触发细胞核和细胞质内容物的混合,导致核功能的灾难性后果。尽管细胞分裂过程中NE重整和间期NE恢复的分子机制的精确细节仍在研究中,在这里,我们主要关注哺乳动物细胞来描述已经确定的关键方面,并讨论它们之间的串扰。
    In eukaryotic cells, the nuclear envelope (NE) is a membrane partition between the nucleus and the cytoplasm to compartmentalize nuclear contents. It plays an important role in facilitating nuclear functions including transcription, DNA replication and repair. In mammalian cells, the NE breaks down and then reforms during cell division, and in interphase it is restored shortly after the NE rupture induced by mechanical force. In this way, the partitioning effect is regulated through dynamic processes throughout the cell cycle. A failure in rebuilding the NE structure triggers the mixing of nuclear and cytoplasmic contents, leading to catastrophic consequences for the nuclear functions. Whereas the precise details of molecular mechanisms for NE reformation during cell division and NE restoration in interphase are still being investigated, here, we mostly focus on mammalian cells to describe key aspects that have been identified and to discuss the crosstalk between them.
    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
    核包膜(NE)的完整性对于维持核的结构稳定性至关重要。在癌细胞中经常观察到NE的破裂,尤其是在机械挑战的背景下,例如物理限制和迁移。然而,自发性NE破裂事件,对细胞没有任何明显的物理挑战,也有描述。这些自发性NE破裂事件的分子机制仍有待探索。这里,我们表明DNA损伤和随后的ATR激活导致NE破裂。DNA损伤后,层粘连蛋白A/C以ATR依赖性方式磷酸化,导致椎板组装的变化,最终,NE破裂。此外,我们表明,具有内在DNA修复缺陷的癌细胞经历频繁的DNA损伤诱导的NE断裂事件,这使得它们对进一步的NE扰动极其敏感。利用这种NE漏洞可以提供一个新的角度来补充传统的,基于DNA损伤的化疗。
    The integrity of the nuclear envelope (NE) is essential for maintaining the structural stability of the nucleus. Rupture of the NE has been frequently observed in cancer cells, especially in the context of mechanical challenges, such as physical confinement and migration. However, spontaneous NE rupture events, without any obvious physical challenges to the cell, have also been described. The molecular mechanism(s) of these spontaneous NE rupture events remain to be explored. Here, we show that DNA damage and subsequent ATR activation leads to NE rupture. Upon DNA damage, lamin A/C is phosphorylated in an ATR-dependent manner, leading to changes in lamina assembly and, ultimately, NE rupture. In addition, we show that cancer cells with intrinsic DNA repair defects undergo frequent events of DNA-damage-induced NE rupture, which renders them extremely sensitive to further NE perturbations. Exploiting this NE vulnerability could provide a new angle to complement traditional, DNA-damage-based chemotherapy.
    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
    微管相关蛋白tau是中枢神经系统神经元的丰富成分。在阿尔茨海默病和其他神经退行性tau病变中,发现tau过度磷酸化并聚集在神经原纤维缠结中。为了更好地了解启动tau发病机制的细胞扰动,我们对增强tau聚集的遗传修饰剂进行了CRISPR-Cas9筛选。这个初步筛选产生了三个基因,BANF1、ANKLE2和PPP2CA,其失活促进tau以磷酸化和不溶性形式的积累。在互补的屏幕中,我们确定了三个额外的基因,LEMD2、LEMD3和CHMP7,当过度表达时,防止tau聚集。由鉴定的基因编码的蛋白质在机械上联系起来,并因其在核包膜的维持和修复中的作用而得到认可。这些结果暗示核包膜完整性的破坏是tau蛋白病变中可能的起始事件,并揭示了治疗干预的靶标。
    The microtubule-associated protein tau is an abundant component of neurons of the central nervous system. In Alzheimer\'s disease and other neurodegenerative tauopathies, tau is found hyperphosphorylated and aggregated in neurofibrillary tangles. To obtain a better understanding of the cellular perturbations that initiate tau pathogenesis, we performed a CRISPR-Cas9 screen for genetic modifiers that enhance tau aggregation. This initial screen yielded three genes, BANF1, ANKLE2, and PPP2CA, whose inactivation promotes the accumulation of tau in a phosphorylated and insoluble form. In a complementary screen, we identified three additional genes, LEMD2, LEMD3, and CHMP7, that, when overexpressed, provide protection against tau aggregation. The proteins encoded by the identified genes are mechanistically linked and recognized for their roles in the maintenance and repair of the nuclear envelope. These results implicate the disruption of nuclear envelope integrity as a possible initiating event in tauopathies and reveal targets for therapeutic intervention.
    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
    核破裂长期以来一直与层板的缺陷或缺陷有关,最近的结果也表明了肌动球蛋白应激的作用,但破裂的关键物理决定因素仍不清楚。这里,lamin-B细丝在低高斯曲率的位置稳定地与核膜相互作用,但在高曲率时稀释以利于破裂,而lamin-A消耗需要高应变率。lamin-B1基因编辑的癌细胞的活细胞成像补充了破裂的固定细胞成像:iPS衍生的早衰患者细胞,跳动的小鸡胚胎心脏内的细胞,癌细胞通过小孔迁移后发生多点破裂。数据拟合从曲面分离的刚性细丝的模型。通过抑制肌球蛋白II和低张应力适度抑制破裂,这减缓了应变率。Lamin-A稀释和破裂概率确实增加到高于核牵拉的阈值速率。质膜蛋白质的曲率感应机制,包括Piezo1,因此可能适用于核膜。摘要声明:即使肌球蛋白应力被抑制,高核曲率也会驱动椎板稀释和核包膜破裂。刚丝通常从高斯曲率高的部位稀释,提供实验的数学拟合。
    Nuclear rupture has long been associated with deficits or defects in lamins, with recent results also indicating a role for actomyosin stress, but key physical determinants of rupture remain unclear. Here, lamin-B filaments stably interact with the nuclear membrane at sites of low Gaussian curvature yet dilute at high curvature to favor rupture, whereas lamin-A depletion requires high strain-rates. Live-cell imaging of lamin-B1 gene-edited cancer cells is complemented by fixed-cell imaging of rupture in: iPS-derived progeria patients cells, cells within beating chick embryo hearts, and cancer cells with multi-site rupture after migration through small pores. Data fit a model of stiff filaments that detach from a curved surface.Rupture is modestly suppressed by inhibiting myosin-II and by hypotonic stress, which slow the strain-rates. Lamin-A dilution and rupture probability indeed increase above a threshold rate of nuclear pulling. Curvature-sensing mechanisms of proteins at plasma membranes, including Piezo1, might thus apply at nuclear membranes.Summary statement: High nuclear curvature drives lamina dilution and nuclear envelope rupture even when myosin stress is inhibited. Stiff filaments generally dilute from sites of high Gaussian curvature, providing mathematical fits of experiments.
    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
    在转移过程中,侵入肿瘤细胞和循环肿瘤细胞(CTC)在通过狭窄孔和细胞挤压的迁移过程中面临多种机械挑战。然而,对机械应力对肿瘤进展和成功侵入新器官的重要性和后果知之甚少。最近,多项研究表明,细胞收缩可导致间期核膜破裂(NER)。这种失去适当的核分区对基因组有深远的影响,是肿瘤进展所需的基因组进化的关键驱动因素。不仅仅是基因组改变的来源,短暂的核包膜塌陷也可以通过涉及先天免疫应答cGAS/STING途径的几种机制支持转移性生长。在这篇综述中,我们将描述细胞挤压在肿瘤发生发展中被低估的作用的重要性。我们将描述肿瘤细胞到达转移部位的复杂性和难度,详细说明了由于NER导致的基因组畸变多样性,并强调了先天免疫途径激活对细胞存活的重要性。细胞适应和核变形可能是许多未知方面转移成功的关键。
    During metastasis, invading tumor cells and circulating tumor cells (CTC) face multiple mechanical challenges during migration through narrow pores and cell squeezing. However, little is known on the importance and consequences of mechanical stress for tumor progression and success in invading a new organ. Recently, several studies have shown that cell constriction can lead to nuclear envelope rupture (NER) during interphase. This loss of proper nuclear compartmentalization has a profound effect on the genome, being a key driver for the genome evolution needed for tumor progression. More than just being a source of genomic alterations, the transient nuclear envelope collapse can also support metastatic growth by several mechanisms involving the innate immune response cGAS/STING pathway. In this review we will describe the importance of the underestimated role of cellular squeezing in the progression of tumorigenesis. We will describe the complexity and difficulty for tumor cells to reach the metastatic site, detail the genomic aberration diversity due to NER, and highlight the importance of the activation of the innate immune pathway on cell survival. Cellular adaptation and nuclear deformation can be the key to the metastasis success in many unsuspected aspects.
    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
    中性粒细胞是最丰富的循环白细胞和先天免疫系统的主要细胞类型之一。中性粒细胞胞外诱捕网(NETs)是核染色质从破裂的核膜和质膜胞外释放的结果。外化染色质是动物在细胞外环境中诱捕和杀死微生物的古老防御武器,从而保护从较低的无脊椎动物到较高的脊椎动物的动物。尽管外化染色质具有作为抗感染药物以防止感染的优势,在高等脊椎动物中,细胞外染色质可能存在问题,因为它们具有适应性免疫系统,可以触发进一步的免疫或自身免疫反应。NET及其相关的核和/或细胞质成分可能会引起无菌性炎症。免疫,和自身免疫反应,导致各种人类疾病。尽管在人类病理生理学中很重要,NET形成(也称为NETosis)的细胞和分子机制尚不清楚。鉴于核染色质形成了NET的骨架,细胞核是细胞核DNA胞外诱捕器的根。因此,核染色质去凝聚,随着核膜和质膜的破裂,是核染色质细胞外释放和NET形成所必需的。到目前为止,大多数文献都集中在某些信号通路上,参与NET形成,但没有解释上述细胞事件和形态变化。这里,我们总结了新的证据并讨论了新的机械理解,用我们的观点,中性粒细胞中的NET形成。
    Neutrophils are the most abundant circulating white blood cells and one of the major cell types of the innate immune system. Neutrophil extracellular traps (NETs) are a result of the extracellular release of nuclear chromatin from the ruptured nuclear envelope and plasma membrane. The externalized chromatin is an ancient defense weapon for animals to entrap and kill microorganisms in the extracellular milieu, thus protecting animals ranging from lower invertebrates to higher vertebrates. Although the externalized chromatin has the advantage of acting as anti-infective to protect against infections, extracellular chromatin might be problematic in higher vertebrate animals as they have an adaptive immune system that can trigger further immune or autoimmune responses. NETs and their associated nuclear and/or cytoplasmic components may induce sterile inflammation, immune, and autoimmune responses, leading to various human diseases. Though important in human pathophysiology, the cellular and molecular mechanisms of NET formation (also called NETosis) are not well understood. Given that nuclear chromatin forms the backbone of NETs, the nucleus is the root of the nuclear DNA extracellular traps. Thus, nuclear chromatin decondensation, along with the rupture of nuclear envelope and plasma membrane, is required for nuclear chromatin extracellular release and NET formation. So far, most of the literature focuses on certain signaling pathways, which are involved in NET formation but without explanation of cellular events and morphological changes described above. Here, we have summarized emerging evidence and discuss new mechanistic understanding, with our perspectives, in NET formation in neutrophils.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    癌症转移,即,肿瘤细胞从原发肿瘤扩散到远处器官,是绝大多数癌症死亡的原因。在这个过程中,癌细胞通过狭窄的间质空间迁移,横截面比细胞小得多。在这种有限的迁移过程中,癌细胞经历广泛的核变形,核包膜破裂,和DNA损伤。导致限制性迁移诱导的DNA损伤的分子机制仍未完全了解。虽然在一些细胞系中,DNA损伤与核包膜破裂密切相关,我们证明,在其他方面,细胞核的机械变形足以造成DNA损伤,即使在没有核膜破裂的情况下。这种变形引起的DNA损伤,与核包膜破裂引起的DNA损伤不同,主要发生在细胞周期的S/G2期,并与复制叉有关。核变形,由于有限的迁移或外部细胞压缩,增加复制压力,可能是通过增加复制叉的失速,为变形诱导的DNA损伤提供了分子机制。因此,我们发现了一种机械诱导DNA损伤的新机制,将细胞核的机械变形与DNA复制应力联系起来。这种机械诱导的DNA损伤不仅会增加转移癌细胞的基因组不稳定性,而且还会导致在发育过程中经历机械压缩的非迁移细胞和组织的DNA损伤。从而促进肿瘤发生和DNA损伤反应激活。
    Cancer metastasis, i.e., the spreading of tumor cells from the primary tumor to distant organs, is responsible for the vast majority of cancer deaths. In the process, cancer cells migrate through narrow interstitial spaces substantially smaller in cross-section than the cell. During such confined migration, cancer cells experience extensive nuclear deformation, nuclear envelope rupture, and DNA damage. The molecular mechanisms responsible for the confined migration-induced DNA damage remain incompletely understood. Although in some cell lines, DNA damage is closely associated with nuclear envelope rupture, we show that, in others, mechanical deformation of the nucleus is sufficient to cause DNA damage, even in the absence of nuclear envelope rupture. This deformation-induced DNA damage, unlike nuclear-envelope-rupture-induced DNA damage, occurs primarily in S/G2 phase of the cell cycle and is associated with replication forks. Nuclear deformation, resulting from either confined migration or external cell compression, increases replication stress, possibly by increasing replication fork stalling, providing a molecular mechanism for the deformation-induced DNA damage. Thus, we have uncovered a new mechanism for mechanically induced DNA damage, linking mechanical deformation of the nucleus to DNA replication stress. This mechanically induced DNA damage could not only increase genomic instability in metastasizing cancer cells but could also cause DNA damage in non-migrating cells and tissues that experience mechanical compression during development, thereby contributing to tumorigenesis and DNA damage response activation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    微核是核外体,主要来自在有丝分裂出口处未掺入初级核中的单端染色体/色素(LC)。与原核不同,大多数微核被核膜(NE)包围,核膜极易发生自发和不可修复的破裂。破裂的微核是染色体样混沌重排和cGAS介导的先天免疫和炎症的触发因素,提出了微核在人类衰老和肿瘤发生中起积极作用的观点。因此,了解微核包膜(mNE)出错的方式变得越来越重要。这里,我们回顾数据,为这个问题提供一个总体框架。我们首先描述有丝分裂后的NE重组和间期的NE修复。同时,我们简要讨论了mNE是如何组织的,以及mNE破裂如何控制微核和微核细胞的命运。作为本次审查的重点,我们重点介绍了目前关于为什么mNE容易破裂和不可修复的知识。为此,我们调查了一系列优雅研究的观察结果,以提供系统的概述。我们得出的结论是,易破裂和不可修复的微核的诞生可能是其细胞内地理起源的累积影响,生物物理特性,和特定的MNE功能。我们建议微核中的DNA损伤和免疫原性从改变的mNE成分逐步增加,mNE破裂,和耐火材料修复。在我们的讨论中,我们注意到尚未解决的mNE脆弱性中的有趣问题。
    Micronuclei are extra-nuclear bodies mainly derived from ana-telophase lagging chromosomes/chromatins (LCs) that are not incorporated into primary nuclei at mitotic exit. Unlike primary nuclei, most micronuclei are enclosed by nuclear envelope (NE) that is highly susceptible to spontaneous and irreparable rupture. Ruptured micronuclei act as triggers of chromothripsis-like chaotic chromosomal rearrangements and cGAS-mediated innate immunity and inflammation, raising the view that micronuclei play active roles in human aging and tumorigenesis. Thus, understanding the ways in which micronuclear envelope (mNE) goes awry acquires increased importance. Here, we review the data to present a general framework for this question. We firstly describe NE reassembly after mitosis and NE repair during interphase. Simultaneously, we briefly discuss how mNE is organized and how mNE rupture controls the fate of micronuclei and micronucleated cells. As a focus of this review, we highlight current knowledge about why mNE is rupture-prone and irreparable. For this, we survey observations from a series of elegant studies to provide a systematic overview. We conclude that the birth of rupture-prone and irreparable micronuclei may be the cumulative effects of their intracellular geographic origins, biophysical properties, and specific mNE features. We propose that DNA damage and immunogenicity in micronuclei increase stepwise from altered mNE components, mNE rupture, and refractory to repair. Throughout our discussion, we note interesting issues in mNE fragility that have yet to be resolved.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    核层对于核壳的结构整合至关重要。核膜破裂和染色质外化是中性粒细胞胞外诱捕网(NETs)形成的标志。NET释放被描述为细胞裂解过程;然而,这个概念最近受到质疑。这里,我们报告说,在NET形成过程中,核层粘连蛋白B不会被破坏性的蛋白水解片段化,而是分解成完整的全长分子。此外,我们证明PKCα的核易位,作为激酶诱导层板蛋白B的磷酸化和分解,导致核包膜破裂。通过PKCα抑制降低层粘连蛋白B磷酸化,遗传缺失,或通过突变层粘连蛋白B上的PKCα共有位点来减弱细胞外陷阱的形成。此外,通过层粘连蛋白B过表达增强核膜可减弱体内NET释放,并降低层粘连蛋白B转基因小鼠UVB照射皮肤中NET相关炎性细胞因子的水平。我们的发现通过表明PKCα介导的层粘连蛋白B磷酸化驱动核包膜破裂以释放中性粒细胞中的染色质,从而促进了对NET形成的机理理解。
    The nuclear lamina is essential for the structural integration of the nuclear envelope. Nuclear envelope rupture and chromatin externalization is a hallmark of the formation of neutrophil extracellular traps (NETs). NET release was described as a cellular lysis process; however, this notion has been questioned recently. Here, we report that during NET formation, nuclear lamin B is not fragmented by destructive proteolysis, but rather disassembled into intact full-length molecules. Furthermore, we demonstrate that nuclear translocation of PKCα, which serves as the kinase to induce lamin B phosphorylation and disassembly, results in nuclear envelope rupture. Decreasing lamin B phosphorylation by PKCα inhibition, genetic deletion, or by mutating the PKCα consensus sites on lamin B attenuates extracellular trap formation. In addition, strengthening the nuclear envelope by lamin B overexpression attenuates NET release in vivo and reduces levels of NET-associated inflammatory cytokines in UVB-irradiated skin of lamin B transgenic mice. Our findings advance the mechanistic understanding of NET formation by showing that PKCα-mediated lamin B phosphorylation drives nuclear envelope rupture for chromatin release in neutrophils.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号