Cellular quiescence

细胞静止
  • 文章类型: Journal Article
    (1)背景:LEO1(左开放阅读框1)蛋白是PAF1C复合物(RNA聚合酶II相关因子1复合物)的保守亚基。PAF1C在转录延伸和RNA加工中具有完善的机制功能。我们之前展示过,在裂殖酵母中,LEO1通过影响染色质中组蛋白H3的转换来控制组蛋白H3K9甲基化水平,并且它对于在细胞静止期间正确调节基因表达至关重要。人成纤维细胞在生长培养基中血清剥夺后进入可逆的静止状态。在这里,我们研究了LEO1在人成纤维细胞中的功能。(2)方法:我们使用CRISPR/Cas9方法在人成纤维细胞中敲除LEO1基因,并通过Westernblot验证LEO1蛋白不可检测。我们用FACS分析和细胞生长测定表征了ΔLEO1敲除细胞的表型。我们使用RNA测序使用刺入对照来测量基因表达和刺入控制的ChIP测序实验来测量组蛋白修饰H3K9me2全基因组。(3)结果:静止细胞中基因表达水平发生改变,然而,在静止的人类细胞中控制染色质和基因表达变化的因素在很大程度上是未知的。与野生型细胞相比,ΔLEO1敲除的成纤维细胞是存活的,但具有降低的代谢活性。与野生型细胞相比,ΔLEO1细胞显示较慢的进入静止状态和不同的形态。在静止的野生型细胞中基因表达通常降低。下调的基因包括参与细胞增殖的基因。少量基因在静止的野生型细胞中上调,包括参与ERK1/ERK2和Wnt信号传导的几个基因。在静止的ΔLEO1细胞中,与野生型细胞相比,许多基因被错误调节。这包括参与钙离子转运和细胞形态发生的基因。最后,掺入控制的ChIP测序实验表明,在静止的ΔLEO1细胞中,组蛋白修饰H3K9me2水平整体增加。(4)结论:因此,LEO1对于正确进入细胞静止很重要,控制H3K9me2水平,和人成纤维细胞中的基因表达。
    (1) Background: The LEO1 (Left open reading frame 1) protein is a conserved subunit of the PAF1C complex (RNA polymerase II-associated factor 1 complex). PAF1C has well-established mechanistic functions in elongation of transcription and RNA processing. We previously showed, in fission yeast, that LEO1 controls histone H3K9 methylation levels by affecting the turnover of histone H3 in chromatin, and that it is essential for the proper regulation of gene expression during cellular quiescence. Human fibroblasts enter a reversible quiescence state upon serum deprivation in the growth media. Here we investigate the function of LEO1 in human fibroblasts. (2) Methods: We knocked out the LEO1 gene using CRISPR/Cas9 methodology in human fibroblasts and verified that the LEO1 protein was undetectable by Western blot. We characterized the phenotype of the ΔLEO1 knockout cells with FACS analysis and cell growth assays. We used RNA-sequencing using spike-in controls to measure gene expression and spike-in controlled ChIP-sequencing experiments to measure the histone modification H3K9me2 genome-wide. (3) Results: Gene expression levels are altered in quiescent cells, however factors controlling chromatin and gene expression changes in quiescent human cells are largely unknown. The ΔLEO1 knockout fibroblasts are viable but have reduced metabolic activity compared to wild-type cells. ΔLEO1 cells showed a slower entry into quiescence and a different morphology compared to wild-type cells. Gene expression was generally reduced in quiescent wild-type cells. The downregulated genes included genes involved in cell proliferation. A small number of genes were upregulated in quiescent wild-type cells including several genes involved in ERK1/ERK2 and Wnt signaling. In quiescent ΔLEO1 cells, many genes were mis-regulated compared to wild-type cells. This included genes involved in Calcium ion transport and cell morphogenesis. Finally, spike-in controlled ChIP-sequencing experiments demonstrated that the histone modification H3K9me2 levels are globally increased in quiescent ΔLEO1 cells. (4) Conclusions: Thus, LEO1 is important for proper entry into cellular quiescence, control of H3K9me2 levels, and gene expression in human fibroblasts.
    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
    细胞静止是单细胞和多细胞真核生物的重要生理状态。停止静止细胞进行增殖并在G0阶段停止细胞周期。使用裂变酵母作为模型生物,我们以前发现,保守的染色质重塑复合物的几个亚基,Ino80C(需要肌醇的核小体重塑因子),是静止生存所必需的。这里,我们证明Ino80C在G0细胞中的基因表达调控中具有关键功能。我们证明了两个Ino80C亚基的空突变体,Iec1和Ies2,推定的亚基Arp42,组蛋白变体H2A的无效突变体。Z,肌醇激酶Asp1的无效突变体在静止期具有非常相似的表型。这些突变体在全基因组范围内显示转录降低,并且特别不能激活149个静止基因,其中许多位于亚端粒区。在归一化ChIP-seq实验中使用尖峰,我们显示H2A的全球减少。静态野生型细胞中的Z水平,而iec1Δ细胞中则没有,并且亚端粒染色体边界元件受到Ino80C的强烈影响。基于这些观察,我们提出了一个Ino80C驱逐H2A的模型。来自静止细胞染色质的Z,从而使亚端粒边界元素失活,导致染色体结构的重组和在静止状态下生存所需的基因的激活。
    Cellular quiescence is an important physiological state both in unicellular and multicellular eukaryotes. Quiescent cells are halted for proliferation and stop the cell cycle at the G0 stage. Using fission yeast as a model organism, we have previously found that several subunits of a conserved chromatin remodeling complex, Ino80C (INOsitol requiring nucleosome remodeling factor), are required for survival in quiescence. Here, we demonstrate that Ino80C has a key function in the regulation of gene expression in G0 cells. We show that null mutants for two Ino80C subunits, Iec1 and Ies2, a putative subunit Arp42, a null mutant for the histone variant H2A.Z, and a null mutant for the Inositol kinase Asp1 have very similar phenotypes in quiescence. These mutants show reduced transcription genome-wide and specifically fail to activate 149 quiescence genes, of which many are localized to the subtelomeric regions. Using spike in normalized ChIP-seq experiments, we show that there is a global reduction of H2A.Z levels in quiescent wild-type cells but not in iec1∆ cells and that a subtelomeric chromosome boundary element is strongly affected by Ino80C. Based on these observations, we propose a model in which Ino80C is evicting H2A.Z from chromatin in quiescent cells, thereby inactivating the subtelomeric boundary element, leading to a reorganization of the chromosome structure and activation of genes required to survive in quiescence.
    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
    营养的可利用性决定着生长和静止,许多动物在饥饿时停止发育。以秀丽隐士L1逮捕为模型,我们证明了基因表达在饥饿中的变化。令人惊讶的是,种系富集基因的相对表达持续数天增加。我们使用生长素诱导的degron系统有条件地降解RNA聚合酶II的大亚基,并分析绝对表达水平。我们发现体细胞转录是生存所必需的,但是种系保持转录静止。成千上万的基因在体细胞中不断转录,尽管它们的绝对丰度下降,这样种系转录本的相对表达增加了极端的转录本稳定性。在饥饿的生殖细胞中异常激活转录会损害生殖,证明转录静止的重要生理功能。这项工作揭示了饥饿期间的替代体细胞和种系基因调控策略,soma维持强大的转录反应以支持生存,种系维持转录静止以支持未来的生殖成功。
    Nutrient availability governs growth and quiescence, and many animals arrest development when starved. Using C. elegans L1 arrest as a model, we show that gene expression changes deep into starvation. Surprisingly, relative expression of germline-enriched genes increases for days. We conditionally degrade the large subunit of RNA polymerase II using the auxin-inducible degron system and analyze absolute expression levels. We find that somatic transcription is required for survival, but the germline maintains transcriptional quiescence. Thousands of genes are continuously transcribed in the soma, though their absolute abundance declines, such that relative expression of germline transcripts increases given extreme transcript stability. Aberrantly activating transcription in starved germ cells compromises reproduction, demonstrating important physiological function of transcriptional quiescence. This work reveals alternative somatic and germline gene-regulatory strategies during starvation, with the soma maintaining a robust transcriptional response to support survival and the germline maintaining transcriptional quiescence to support future reproductive success.
    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
    静止癌细胞是罕见的非潜水细胞,具有逃避化疗并在治疗后恢复细胞分裂的独特能力。尽管癌症复发的风险相关,细胞如何在快速增殖和静止之间可逆地切换仍然是一个长期存在的问题。通过开发一种独特的方法,用于体外细胞亚群中无细胞分选的代谢谱分离,我们揭示了静止细胞的代谢特征,这些代谢特征在很大程度上不受细胞类型和静止诱导刺激的基础差异的影响。与我们基于代谢组的分析一致,我们表明,线粒体脂肪酸β-氧化(FAO)的损害可以诱导静止诱导的细胞凋亡,并阻碍其恢复增殖。我们的研究结果表明,除了介导能量和氧化还原平衡,粮农组织可以在过渡到静止期间防止有毒中间体的积累方面发挥作用。发现要进入的代谢策略,保持,退出静止可以揭示细胞可塑性的基本原理和癌症以外的新的潜在治疗靶点。
    Quiescent cancer cells are rare nondiving cells with the unique ability to evade chemotherapies and resume cell division after treatment. Despite the associated risk of cancer recurrence, how cells can reversibly switch between rapid proliferation and quiescence remains a long-standing open question. By developing a unique methodology for the cell sorting-free separation of metabolic profiles in cell subpopulations in vitro, we unraveled metabolic characteristics of quiescent cells that are largely invariant to basal differences in cell types and quiescence-inducing stimuli. Consistent with our metabolome-based analysis, we show that impairing mitochondrial fatty acid β-oxidation (FAO) can induce apoptosis in quiescence-induced cells and hamper their return to proliferation. Our findings suggest that in addition to mediating energy and redox balance, FAO can play a role in preventing the buildup of toxic intermediates during transitioning to quiescence. Uncovering metabolic strategies to enter, maintain, and exit quiescence can reveal fundamental principles in cell plasticity and new potential therapeutic targets beyond cancer.
    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
    我们提出了一种假设,即在按时间顺序老化的出芽酵母中,将细胞老化与细胞静止联系起来的机制。我们的假设认为这种机制整合了四个不同的过程,所有这些都是在最初含有葡萄糖的培养基中培养的酵母细胞消耗后开始的。在这些培养物中发育的静止细胞可以分离成不同浮力密度的高密度和低密度亚群。所提出的机制的过程1包括G1期的细胞周期停滞,并导致高密度静止细胞的形成。过程2导致高密度静止细胞转化为低密度静止细胞。过程3和4导致低密度或高密度静止细胞的静止性快速或缓慢下降,分别。这里,我们通过评估四种不同的老年保护剂如何影响可能将细胞衰老与细胞静止联系起来的四个过程来检验我们的假设。我们发现,这些老年期保护剂对过程1和2的影响不同,并使过程3和4减速。我们还发现,静止酵母中海藻糖的增加有助于时间老化和静止维持。这些数据共同为细胞衰老和细胞静止之间的机械联系提供了确凿的证据。
    We propose a hypothesis of a mechanism linking cellular aging to cellular quiescence in chronologically aging budding yeast. Our hypothesis posits that this mechanism integrates four different processes, all of which are initiated after yeast cells cultured in a medium initially containing glucose consume it. Quiescent cells that develop in these cultures can be separated into the high- and low-density sub-populations of different buoyant densities. Process 1 of the proposed mechanism consists of a cell-cycle arrest in the G1 phase and leads to the formation of high-density quiescent cells. Process 2 results in converting high-density quiescent cells into low-density quiescent cells. Processes 3 and 4 cause a fast or slow decline in the quiescence of low- or high-density quiescent cells, respectively. Here, we tested our hypothesis by assessing how four different geroprotectors influence the four processes that could link cellular aging to cellular quiescence. We found that these geroprotectors differently affect processes 1 and 2 and decelerate processes 3 and 4. We also found that a rise in trehalose within quiescent yeast contributes to chronological aging and quiescence maintenance. These data collectively provide conclusive evidence for a mechanistic link between cellular aging and cellular quiescence.
    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
    在癌症中,休眠是指一种临床状态,在这种状态下,微观残留疾病变得不增生,并且在很大程度上对化疗难以治疗。休眠最初是在乳腺癌中描述的,在乳腺癌中,疾病可能几十年都没有被发现,最终导致原始恶性肿瘤的复发和临床表现。一个长的潜伏期可以解释为退出细胞增殖(细胞休眠),或保持低水平残留疾病(肿瘤块休眠)的增殖和细胞死亡之间的平衡。对细胞休眠的研究揭示了定义这种状态的特征。它们包括细胞增殖的停滞,细胞代谢改变,以及独特的细胞依赖性和与微环境的相互作用。这些特征可以由来自不同原发疾病部位的休眠细胞共享。这表明它们之间存在共同的特征。高级别浆液性卵巢癌(HGSOC)通过称为球体的细胞聚集体扩散到整个腹腔。这些生长停滞和治疗抗性细胞是疾病复发的重要因素。在这次审查中,我们讨论了卵巢癌细胞在球体和其他癌症疾病部位的休眠特性之间的异同.这揭示了休眠的元素,如细胞周期控制机制和新陈代谢的变化,在大多数形式的细胞休眠中可能是相似的。然而,球体生物学的HGSOC特定方面,包括细胞外基质组织和微环境,必须是疾病部位特异性的。总的来说,我们对当前文献的批判性回顾强调了HGSOC细胞休眠的地方,这可能为理解癌症细胞休眠的广泛原理提供了更易于处理的实验方法.
    In cancer, dormancy refers to a clinical state in which microscopic residual disease becomes non-proliferative and is largely refractory to chemotherapy. Dormancy was first described in breast cancer where disease can remain undetected for decades, ultimately leading to relapse and clinical presentation of the original malignancy. A long latency period can be explained by withdrawal from cell proliferation (cellular dormancy), or a balance between proliferation and cell death that retains low levels of residual disease (tumor mass dormancy). Research into cellular dormancy has revealed features that define this state. They include arrest of cell proliferation, altered cellular metabolism, and unique cell dependencies and interactions with the microenvironment. These characteristics can be shared by dormant cells derived from disparate primary disease sites, suggesting common features exist between them.High-grade serous ovarian cancer (HGSOC) disseminates to locations throughout the abdominal cavity by means of cellular aggregates called spheroids. These growth-arrested and therapy-resistant cells are a strong contributor to disease relapse. In this review, we discuss the similarities and differences between ovarian cancer cells in spheroids and dormant properties reported for other cancer disease sites. This reveals that elements of dormancy, such as cell cycle control mechanisms and changes to metabolism, may be similar across most forms of cellular dormancy. However, HGSOC-specific aspects of spheroid biology, including the extracellular matrix organization and microenvironment, are obligatorily disease site specific. Collectively, our critical review of current literature highlights places where HGSOC cell dormancy may offer a more tractable experimental approach to understand broad principles of cellular dormancy in cancer.
    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
    细胞静止和增殖之间的平衡是组织生理学和稳态的基础。最近的研究表明,静止不是被动和均匀的状态,而是主动维持和异质的状态。这些与静止相关的细胞特征主要在静态培养基下的培养细胞中观察到。然而,体内细胞面临不同的微环境条件,特别是,间质液下的流动通过细胞外基质分布。间质流体流动对细胞和基质应变施加剪切应力,并导致细胞外因子的持续替代。在这项研究中,我们分析了微流体设备中不同流体流速下的单个细胞。我们发现了以前在常规静态介质下确定的静止特征,包括血清信号依赖的静止进入和退出以及时间依赖的静止加深,在连续流体流动下也存在。此外,增加流速驱动细胞较浅的静止,并在生长刺激后更有可能重新进入细胞周期。这种效应是由于流动诱导的物理和生化线索。具体来说,单独增加剪切应力或胞外因子置换,不改变其他参数,导致浅静止。我们表明,我们的实验结果可以通过将细胞外液流连接到Rb-E2f双稳态开关的数学模型来定量解释,该开关调节静止到增殖的过渡。我们的发现揭示了一种以前未被理解的机制,该机制可能是静止细胞在不同生理组织微环境中对组织修复和再生的异质反应的基础。
    The balance between cell quiescence and proliferation is fundamental to tissue physiology and homeostasis. Recent studies have shown that quiescence is not a passive and homogeneous state but actively maintained and heterogeneous. These cellular characteristics associated with quiescence were observed primarily in cultured cells under a static medium. However, cells in vivo face different microenvironmental conditions, particularly, under interstitial fluid flows distributed through extracellular matrices. Interstitial fluid flow exerts shear stress on cells and matrix strain, and results in continuous replacement of extracellular factors. In this study, we analyzed individual cells under varying fluid flow rates in microfluidic devices. We found quiescence characteristics previously identified under conventional static medium, including serum signal-dependant quiescence entry and exit and time-dependant quiescence deepening, are also present under continuous fluid flow. Furthermore, increasing the flow rate drives cells to shallower quiescence and become more likely to reenter the cell cycle upon growth stimulation. This effect is due to flow-induced physical and biochemical cues. Specifically, increasing shear stress or extracellular factor replacement individually, without altering other parameters, results in shallow quiescence. We show our experimental results can be quantitatively explained by a mathematical model connecting extracellular fluid flow to an Rb-E2f bistable switch that regulates the quiescence-to-proliferation transition. Our findings uncover a previously unappreciated mechanism that likely underlies the heterogeneous responses of quiescent cells for tissue repair and regeneration in different physiological tissue microenvironments.
    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
    产生HIV储库的主要方法之一是在感染的活化效应CD4T细胞转变为记忆表型的过程中。QUECEL(静态效应细胞潜伏期)方案有效地模拟该过程并允许产生大量潜伏感染的CD4+T细胞。在极化和膨胀之后,用表达GFP/CD8a的单轮报告病毒感染CD4+T细胞。使用确定的细胞因子混合物,包括TGF-β,将感染的细胞纯化并强制进入静止状态。IL-10和IL-8产生潜伏感染细胞的同质群体。由于可以恢复潜伏感染细胞的同质群体,QUECEL模型具有出色的信噪比,并且在过去5年中进行的许多实验中非常一致和可重复。轻松,效率,和生理条件的准确模拟使QUECEL模型成为研究HIV潜伏期的分子机制的强大和可重复的工具。
    One of the main methods to generate the HIV reservoir is during the transition of infected activated effector CD4 T cells to a memory phenotype. The QUECEL (Quiescent Effector Cell Latency) protocol mimics this process efficiently and allows for production of large numbers of latently infected CD4+ T cells. After polarization and expansion, CD4+ T cells are infected with a single round reporter virus which expressed GFP/CD8a. The infected cells are purified and coerced into quiescence using a defined cocktail of cytokines including TGF-β, IL-10, and IL-8, producing a homogeneous population of latently infected cells. Since homogeneous populations of latently infected cells can be recovered, the QUECEL model has an excellent signal-to-noise ratio, and has been extremely consistent and reproducible in numerous experiments performed during the last 5 years. The ease, efficiency, and accurate mimicking of physiological conditions make the QUECEL model a robust and reproducible tool to study the molecular mechanisms underlying HIV latency.
    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
    在含有0.2%葡萄糖(在热量限制条件下)或2%葡萄糖(在非热量限制条件下)的富含营养的液体培养基中培养的酵母细胞出芽后,将葡萄糖发酵为乙醇,然后消耗乙醇,他们进入固定阶段。他们的时间老化过程开始了。在这一点上,酵母培养物开始积累静止和非静止细胞。这里,我们从热量供应有限的酵母培养物中纯化了静止和非静止细胞的高密度和低密度群体。然后,我们使用基于质谱的定量脂质组学来评估高和低密度细胞脂质的衰老相关变化。我们发现热量限制,保护老年人的饮食干预,在高和低密度静止和非静止细胞群体的大部分时间寿命中,许多脂质类别的浓度会发生变化。具体来说,热量限制减少三酰甘油,增加游离脂肪酸,磷脂升高和心磷脂浓度增加。基于这些发现,我们提出了一个假设模型,用于在出芽酵母的静止和非静止细胞中脂质代谢的热量限制依赖性重组。我们还发现,热量限制会产生这些细胞的脂质组学模式,这与其他两个健壮的老年保护者所建立的模式不同,即Tor1Δ突变和石胆酸。
    After budding yeast cells cultured in a nutrient-rich liquid medium with 0.2% glucose (under caloric restriction conditions) or 2% glucose (under non-caloric restriction conditions), ferment glucose to ethanol and then consume ethanol, they enter the stationary phase. The process of their chronological aging begins. At that point, the yeast culture starts to accumulate quiescent and non-quiescent cells. Here, we purified the high- and low-density populations of quiescent and non-quiescent cells from the yeast cultures limited in calorie supply or not. We then employed mass spectrometry-based quantitative lipidomics to assess the aging-associated changes in high- and low-density cells\' lipidomes. We found that caloric restriction, a geroprotective dietary intervention, alters the concentrations of many lipid classes through most of the chronological lifespan of the high- and low-density populations of quiescent and non-quiescent cells. Specifically, caloric restriction decreased triacylglycerol, increased free fatty acid, elevated phospholipid and amplified cardiolipin concentrations. Based on these findings, we propose a hypothetical model for a caloric restriction-dependent reorganization of lipid metabolism in budding yeast\'s quiescent and non-quiescent cells. We also discovered that caloric restriction creates lipidomic patterns of these cells that differ from those established by two other robust geroprotectors, namely the tor1Δ mutation and lithocholic acid.
    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
    在开发过程中,静止气道基底干细胞是通过细胞周期减慢而衍生于增殖的原始祖细胞。相比之下,基底细胞通过从缓慢循环转变为增殖并随后回到缓慢循环来促进成体组织再生。尽管持续的增殖会导致肿瘤发生,调节这些转变的分子机制仍然未知。使用发育中的鼠气道祖细胞的时间单细胞转录组学和遗传验证实验,我们发现TGF-β信号通过抑制Id2减慢细胞周期,并在发育过程中促进基底细胞的缓慢循环。在成人组织再生中,降低TGF-β信号恢复Id2表达并启动再生。Id2过表达和Tgfbr2敲除增强上皮增殖;然而,持续的Id2表达驱动类似癌前状态的基底细胞增生.一起,TGF-β-Id2轴通常在发育和再生过程中调节基底细胞的增殖转变,它的微调对于正常再生至关重要,同时避免基底细胞增生。
    During development, quiescent airway basal stem cells are derived from proliferative primordial progenitors through the cell-cycle slowdown. In contrast, basal cells contribute to adult tissue regeneration by shifting from slow cycling to proliferating and subsequently back to slow cycling. Although sustained proliferation results in tumorigenesis, the molecular mechanisms regulating these transitions remain unknown. Using temporal single-cell transcriptomics of developing murine airway progenitors and genetic validation experiments, we found that TGF-β signaling decelerated cell cycle by inhibiting Id2 and contributed to slow-cycling basal cell specification during development. In adult tissue regeneration, reduced TGF-β signaling restored Id2 expression and initiated regeneration. Id2 overexpression and Tgfbr2 knockout enhanced epithelial proliferation; however, persistent Id2 expression drove basal cell hyperplasia that resembled a precancerous state. Together, the TGF-β-Id2 axis commonly regulates the proliferation transitions in basal cells during development and regeneration, and its fine-tuning is critical for normal regeneration while avoiding basal cell hyperplasia.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号