genotoxic stress

基因毒性应激
  • 文章类型: Journal Article
    环境变化会引发染色质修饰,导致组蛋白亚基的交换或核小体的重新定位。磷酸化的组蛋白变体H2A。X(γH2A。X)被认为是作为DNA双链断裂(DSB)的确定标记的病灶的形成。然而,H2A的确切作用。X在细胞对基因毒性胁迫的反应和植物激素脱落酸(ABA)的影响仍未完全理解。在这次调查中,我们实施CRISPR/Cas9技术在拟南芥中产生AtHTA3和AtHTA5的功能丧失突变体。athta3和athta5单突变体的表型与野生型Col-0的表型几乎相同。然而,athta3athta5双突变体表现出异常的胚胎发育,增加对DNA损伤的敏感性,对ABA的敏感性更高。RT-qPCR分析表明AtHTA3和AtHTA5负调控ABA信号通路的基本调节因子AtABI3的表达。随后的研究表明,AtABI3通过影响DNA损伤反应基因的表达参与基因毒性应激反应,例如AtBRCA1、AtRAD51和AtWEE1。我们的研究为H2A的作用提供了新的见解。X在拟南芥基因毒性和ABA响应中的作用。
    Environmental variations initiate chromatin modifications, leading to the exchange of histone subunits or the repositioning of nucleosomes. The phosphorylated histone variant H2A.X (γH2A.X) is recognized for the formation of foci that serve as established markers of DNA double-strand breaks (DSBs). Nevertheless, the precise roles of H2A.X in the cellular response to genotoxic stress and the impact of the plant hormone abscisic acid (ABA) remain incompletely understood. In this investigation, we implemented CRISPR/Cas9 technology to produce loss-of-function mutants of AtHTA3 and AtHTA5 in Arabidopsis. The phenotypes of the athta3 and athta5 single mutants were nearly identical to those of the wild-type Col-0. Nevertheless, the athta3 athta5 double mutants exhibited aberrant embryonic development, increased sensitivity to DNA damage, and higher sensitivity to ABA. The RT-qPCR analysis indicates that AtHTA3 and AtHTA5 negatively regulate the expression of AtABI3, a fundamental regulator in the ABA signaling pathway. Subsequent investigation demonstrated that AtABI3 participates in the genotoxic stress response by influencing the expression of DNA damage response genes, such as AtBRCA1, AtRAD51, and AtWEE1. Our research offers new insights into the role of H2A.X in the genotoxic and ABA responses of Arabidopsis.
    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
    克隆造血(CH)是由造血干细胞中的体细胞突变引起的普遍状况。当这些突变发生在“驱动”基因中时,它们可以潜在地赋予受影响的细胞健康优势,导致克隆扩张。虽然大多数突变细胞的克隆扩增通常被认为是无症状的,因为它们不影响整体血细胞数量,CH携带者面临全因死亡和年龄相关疾病的长期风险,包括心血管疾病和血液恶性肿瘤。虽然大量的研究集中在理解CH和这些疾病之间的关联,对探索促进驱动基因克隆扩增的调控因素的关注较少。本文对CH发展背景下环境应激源与遗传风险因素之间的关系进行了综述。更好地了解这些压力源如何影响CH的发展将促进机械研究,并可能导致新的治疗途径来治疗患有这种疾病的个体。
    Clonal hematopoiesis (CH) is a prevalent condition that results from somatic mutations in hematopoietic stem cells. When these mutations occur in \"driver\" genes, they can potentially confer fitness advantages to the affected cells, leading to a clonal expansion. While most clonal expansions of mutant cells are generally considered to be asymptomatic since they do not impact overall blood cell numbers, CH carriers face long-term risks of all-cause mortality and age-associated diseases, including cardiovascular disease and hematological malignancies. While considerable research has focused on understanding the association between CH and these diseases, less attention has been given to exploring the regulatory factors that contribute to the expansion of the driver gene clone. This review focuses on the association between environmental stressors and inherited genetic risk factors in the context of CH development. A better understanding of how these stressors impact CH development will facilitate mechanistic studies and potentially lead to new therapeutic avenues to treat individuals with this condition.
    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
    遗传毒性应激,源于各种环境来源和内源性细胞过程,对基因组稳定性构成持续威胁。细胞已经进化出复杂的机制来检测和修复DNA损伤,协调强大的基因毒性应激反应,以保护基因组的完整性。最近的研究揭示了共同和转录后调控机制在调节细胞对基因毒性应激的反应中的关键作用。在这里,我们重点介绍了最近的进展,这些进展说明了mRNA前加工之间的复杂相互作用,专注于3'端处理,和基因毒性应激反应。
    Genotoxic stress, arising from various environmental sources and endogenous cellular processes, pose a constant threat to genomic stability. Cells have evolved intricate mechanisms to detect and repair DNA damage, orchestrating a robust genotoxic stress response to safeguard the integrity of the genome. Recent research has shed light on the crucial role of co- and post-transcriptional regulatory mechanisms in modulating the cellular response to genotoxic stress. Here we highlight recent advances illustrating the intricate interplay between pre-mRNA processing, with a focus on 3\'-end processing, and genotoxic stress response.
    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: Retraction of Publication
    暂无摘要。
    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
    牛磺酸,一种非蛋白质氨基酸和常用的营养补充剂,可以保护各种组织免受与DNA损伤化疗剂顺铂的作用相关的变性。牛磺酸是否以及如何保护人类卵巢癌(OC)细胞免受顺铂引起的DNA损伤尚不清楚。我们发现OC腹水衍生的细胞比细胞培养模型的OC含有更多的细胞内牛磺酸。在文化中,细胞内牛磺酸浓度升高至OC腹水细胞相关水平抑制了各种OC细胞系和患者来源的类器官的增殖,糖酵解减少,并诱导细胞免受顺铂的保护。牛磺酸细胞保护与顺铂反应的DNA损伤减少有关。RNA测序的组合,反相蛋白质阵列,活细胞显微镜,流式细胞术,生化验证实验为牛磺酸介导的突变型或野生型p53与DNA结合的诱导提供了证据,参与细胞周期负调节的p53效应子的激活(p21),和糖酵解(TIGAR)。矛盾的是,牛磺酸对细胞增殖的抑制与促有丝分裂信号转导的激活有关,包括ERK,mTOR,和主要DNA损伤敏感分子如DNAPK的mRNA表达增加,ATM和ATR。虽然抑制ERK或p53不干扰牛磺酸保护细胞免受顺铂的能力,用Torin2抑制mTOR,一种临床相关的抑制剂,也靶向DNAPK和ATM/ATR,破坏了牛磺酸的细胞保护。我们的研究暗示,细胞内牛磺酸的升高可以抑制细胞生长和代谢,并激活涉及mTOR和DNA损伤感应信号转导的细胞保护机制。
    Taurine, a non-proteogenic amino acid and commonly used nutritional supplement, can protect various tissues from degeneration associated with the action of the DNA-damaging chemotherapeutic agent cisplatin. Whether and how taurine protects human ovarian cancer (OC) cells from DNA damage caused by cisplatin is not well understood. We found that OC ascites-derived cells contained significantly more intracellular taurine than cell culture-modeled OC. In culture, elevation of intracellular taurine concentration to OC ascites-cell-associated levels suppressed proliferation of various OC cell lines and patient-derived organoids, reduced glycolysis, and induced cell protection from cisplatin. Taurine cell protection was associated with decreased DNA damage in response to cisplatin. A combination of RNA sequencing, reverse-phase protein arrays, live-cell microscopy, flow cytometry, and biochemical validation experiments provided evidence for taurine-mediated induction of mutant or wild-type p53 binding to DNA, activation of p53 effectors involved in negative regulation of the cell cycle (p21), and glycolysis (TIGAR). Paradoxically, taurine\'s suppression of cell proliferation was associated with activation of pro-mitogenic signal transduction including ERK, mTOR, and increased mRNA expression of major DNA damage-sensing molecules such as DNAPK, ATM and ATR. While inhibition of ERK or p53 did not interfere with taurine\'s ability to protect cells from cisplatin, suppression of mTOR with Torin2, a clinically relevant inhibitor that also targets DNAPK and ATM/ATR, broke taurine\'s cell protection. Our studies implicate that elevation of intracellular taurine could suppress cell growth and metabolism, and activate cell protective mechanisms involving mTOR and DNA damage-sensing signal transduction.
    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
    面对遗传毒性的压力,真核细胞进化出极其精细的机制。抵抗DNA损伤所带来的威胁的缺陷是罕见疾病Cockayne综合征(CS)的基础,这源于CSA和CSB基因的突变。虽然最初定义为DNA修复蛋白,最近的工作表明,CSA和CSB通过协调DNA修复与转录和细胞分裂,作为基因组应激综合反应的主要调节因子。CSA和CSB通过靶蛋白的泛素化来发挥这种功能,它们是这些过程的效应器/调节器。这篇综述描述了目标底物的泛素化如何成为CSA和CSB参与细胞生命不同方面的共同点,以及它们的突变如何引起复杂的疾病CS。
    To face genotoxic stress, eukaryotic cells evolved extremely refined mechanisms. Defects in counteracting the threat imposed by DNA damage underlie the rare disease Cockayne syndrome (CS), which arises from mutations in the CSA and CSB genes. Although initially defined as DNA repair proteins, recent work shows that CSA and CSB act instead as master regulators of the integrated response to genomic stress by coordinating DNA repair with transcription and cell division. CSA and CSB exert this function through the ubiquitination of target proteins, which are effectors/regulators of these processes. This review describes how the ubiquitination of target substrates is a common denominator by which CSA and CSB participate in different aspects of cellular life and how their mutation gives rise to the complex disease CS.
    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
    应激诱导的启动子相关和反义lncRNAs(si-paancRNAs)起源于通过RNAPII暂停介导的发散反义转录的氧化应激(OS)特异性启动子库。一些研究表明,KDM7A分歧转录基因(KDM7A-DT),编码si-paancRNA,在某些癌症类型中过表达。然而,这种过度表达的机制及其在肿瘤发生和癌症进展中的相应作用知之甚少。我们发现KDM7A-DT表达与高度侵袭性癌症类型和特定的固有确定亚型(例如导管浸润性乳腺癌(BRCA)基底亚型)相关。其调节由亚型特异性环境中的错义TP53突变决定。KDM7A-DT转录几个中等大小的ncRNAs和全长转录本,表现出不同的表达和定位模式。KDM7A-DT的过表达上调非恶性成纤维细胞中TP53蛋白表达和H2AX磷酸化,而在半转化的成纤维细胞中,OS以TP53依赖性方式超诱导KDM7A-DT表达。TP53错义突变的腔内ABRCA变体中的KDM7A-DT敲低和基因表达谱,它被大量表达,表明其在癌症途径中的重要作用。KDM7A-DT的内源性过表达通过TP53BP1介导的途径抑制DNA损伤反应/修复(DDR/R),减少细胞凋亡并促进G2/M检查点的停止。BRCA中更高的KDM7A-DT表达与KDM7A-DT基因座增益/扩增相关,组织学分级较高,非整倍体,缺氧,免疫调节评分,和c-myc途径的激活。较高的KDM7A-DT表达与腔A或基底亚型患者相对较差的生存结果相关。相比之下,它与HER2+ER-或管腔B亚型患者的良好结局相关.KDM7A-DT水平与BRCA中异常表达的关键转录本和蛋白质共同调节,包括通过非同源末端连接和上皮-间质转化途径参与DNA修复的那些。总之,KDM7A-DT及其si-lncRNA表现出一些内在的生物学和临床特征,表明在侵袭性BRCA及其亚型中具有重要作用。KDM7A-DT定义的mRNA和蛋白质子网络为识别临床相关的基于RNA的特征和治疗干预的前瞻性靶标提供了资源。
    Stress-induced promoter-associated and antisense lncRNAs (si-paancRNAs) originate from a reservoir of oxidative stress (OS)-specific promoters via RNAPII pausing-mediated divergent antisense transcription. Several studies have shown that the KDM7A divergent transcript gene (KDM7A-DT), which encodes a si-paancRNA, is overexpressed in some cancer types. However, the mechanisms of this overexpression and its corresponding roles in oncogenesis and cancer progression are poorly understood. We found that KDM7A-DT expression is correlated with highly aggressive cancer types and specific inherently determined subtypes (such as ductal invasive breast carcinoma (BRCA) basal subtype). Its regulation is determined by missense TP53 mutations in a subtype-specific context. KDM7A-DT transcribes several intermediate-sized ncRNAs and a full-length transcript, exhibiting distinct expression and localization patterns. Overexpression of KDM7A-DT upregulates TP53 protein expression and H2AX phosphorylation in nonmalignant fibroblasts, while in semi-transformed fibroblasts, OS superinduces KDM7A-DT expression in a TP53-dependent manner. KDM7A-DT knockdown and gene expression profiling in TP53-missense mutated luminal A BRCA variant, where it is abundantly expressed, indicate its significant role in cancer pathways. Endogenous over-expression of KDM7A-DT inhibits DNA damage response/repair (DDR/R) via the TP53BP1-mediated pathway, reducing apoptosis and promoting G2/M checkpoint arrest. Higher KDM7A-DT expression in BRCA is associated with KDM7A-DT locus gain/amplification, higher histologic grade, aneuploidy, hypoxia, immune modulation scores, and activation of the c-myc pathway. Higher KDM7A-DT expression is associated with relatively poor survival outcomes in patients with luminal A or Basal subtypes. In contrast, it is associated with favorable outcomes in patients with HER2+ER- or luminal B subtypes. KDM7A-DT levels are coregulated with critical transcripts and proteins aberrantly expressed in BRCA, including those involved in DNA repair via non-homologous end joining and epithelial-to-mesenchymal transition pathway. In summary, KDM7A-DT and its si-lncRNA exhibit several intrinsic biological and clinical characteristics that suggest important roles in invasive BRCA and its subtypes. KDM7A-DT-defined mRNA and protein subnetworks offer resources for identifying clinically relevant RNA-based signatures and prospective targets for therapeutic intervention.
    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
    在先天免疫应答期间,炎症控制是关键的。通过模式识别受体(PRR)检测源自病原体或受损宿主细胞的分子来触发这种应答。PRR随后通过不同的途径启动细胞内信号传导,导致i)炎性细胞因子的产生,包括I型干扰素(IFN),和ii)启动促进即时宿主应答以及适应性免疫应答的事件级联。所有人类PYRIN和HIN-200结构域(PYHIN)蛋白家族成员最初被认为是PRRs,尽管这一观点受到了报道的挑战,这些报道揭示了它们对其他细胞机制的影响。在这里相关的,人类PYHIN因子髓样核分化抗原(MNDA)最近被证明直接控制编码调节程序性细胞死亡和炎症的因子的基因的转录。虽然MNDA主要存在于骨髓(中性粒细胞和单核细胞)和淋巴(B细胞)起源的白细胞的细胞核中,它的亚细胞定位已被显示为响应于诱导细胞凋亡的基因毒性因子和细菌成分而被调节,炎症的介质。先前的研究已经注意到MNDA作为某些形式的淋巴瘤的标志物的重要性,并作为以凋亡调节缺陷为特征的造血疾病的临床预后因子。MNDA的异常表达也与细胞因子和其他炎性介质的水平改变有关。完善我们对MNDA和其他PYHIN蛋白表达调控机制的理解,以及增强我们对它们分子功能的定义,可以显着影响许多人类疾病的管理和治疗策略。这里,我们对PYHIN蛋白及其在先天和适应性免疫反应中的作用进行了综述.重点将放在条例上,函数,以及MNDA表达在细胞死亡和炎症过程中控制基因转录和RNA稳定性中的相关性。
    Inflammation control is critical during the innate immune response. Such response is triggered by the detection of molecules originating from pathogens or damaged host cells by pattern-recognition receptors (PRRs). PRRs subsequently initiate intra-cellular signalling through different pathways, resulting in i) the production of inflammatory cytokines, including type I interferon (IFN), and ii) the initiation of a cascade of events that promote both immediate host responses as well as adaptive immune responses. All human PYRIN and HIN-200 domains (PYHIN) protein family members were initially proposed to be PRRs, although this view has been challenged by reports that revealed their impact on other cellular mechanisms. Of relevance here, the human PYHIN factor myeloid nuclear differentiation antigen (MNDA) has recently been shown to directly control the transcription of genes encoding factors that regulate programmed cell death and inflammation. While MNDA is mainly found in the nucleus of leukocytes of both myeloid (neutrophils and monocytes) and lymphoid (B-cell) origin, its subcellular localization has been shown to be modulated in response to genotoxic agents that induce apoptosis and by bacterial constituents, mediators of inflammation. Prior studies have noted the importance of MNDA as a marker for certain forms of lymphoma, and as a clinical prognostic factor for hematopoietic diseases characterized by defective regulation of apoptosis. Abnormal expression of MNDA has also been associated with altered levels of cytokines and other inflammatory mediators. Refining our comprehension of the regulatory mechanisms governing the expression of MNDA and other PYHIN proteins, as well as enhancing our definition of their molecular functions, could significantly influence the management and treatment strategies of numerous human diseases. Here, we review the current state of knowledge regarding PYHIN proteins and their role in innate and adaptive immune responses. Emphasis will be placed on the regulation, function, and relevance of MNDA expression in the control of gene transcription and RNA stability during cell death and inflammation.
    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
    丝裂霉素C(MMC)诱导的遗传毒性应激可被认为是内皮功能障碍和动脉粥样硬化的新触发因素,动脉粥样硬化是全球心血管疾病发病率和死亡率的主要原因。鉴于人类有机体的基因毒性负荷不断增加,基因毒性应激诱导内皮功能障碍的分子通路的解密可以提高我们对基因毒性应激在动脉粥样硬化形成中的作用的认识.这里,我们对暴露于MMC的体外人冠状动脉内皮细胞(HCAECs)和人胸廓内内皮细胞(HITAECs)进行了蛋白质组学分析,以鉴定遗传毒性应激诱导的内皮功能障碍的生化通路和蛋白质.我们表示198和71唯一,在MMC处理的HCAECs和HITAECs中差异表达蛋白(DEPs),分别在HCAECs和HITAECs中仅鉴定出4个DEP。在MMC治疗的HCAECs中,44.5%的DEP上调,55.5%的DEP下调,而在HITAEC,这些百分比是72%和28%,分别。所表示的DEP参与核苷酸和RNA代谢的过程,囊泡介导的转运,翻译后蛋白质修饰,细胞周期控制,小分子的运输,转录和信号转导。获得的结果可以增进我们对动脉粥样硬化形成的基本基础的理解,并有助于将基因毒性应激作为动脉粥样硬化的危险因素。
    Mitomycin C (MMC)-induced genotoxic stress can be considered to be a novel trigger of endothelial dysfunction and atherosclerosis-a leading cause of cardiovascular morbidity and mortality worldwide. Given the increasing genotoxic load on the human organism, the decryption of the molecular pathways underlying genotoxic stress-induced endothelial dysfunction could improve our understanding of the role of genotoxic stress in atherogenesis. Here, we performed a proteomic profiling of human coronary artery endothelial cells (HCAECs) and human internal thoracic endothelial cells (HITAECs) in vitro that were exposed to MMC to identify the biochemical pathways and proteins underlying genotoxic stress-induced endothelial dysfunction. We denoted 198 and 71 unique, differentially expressed proteins (DEPs) in the MMC-treated HCAECs and HITAECs, respectively; only 4 DEPs were identified in both the HCAECs and HITAECs. In the MMC-treated HCAECs, 44.5% of the DEPs were upregulated and 55.5% of the DEPs were downregulated, while in HITAECs, these percentages were 72% and 28%, respectively. The denoted DEPs are involved in the processes of nucleotides and RNA metabolism, vesicle-mediated transport, post-translation protein modification, cell cycle control, the transport of small molecules, transcription and signal transduction. The obtained results could improve our understanding of the fundamental basis of atherogenesis and help in the justification of genotoxic stress as a risk factor for atherosclerosis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Letter
    背景:ATM激酶构成了DNA损伤的主要调控中心,并通过磷酸化MDM2蛋白激活p53反应途径,对p53mRNA二级结构产生亲和力。这种相互作用的破坏阻止了新生p53的激活。MDM2蛋白-p53mRNA与上游DNA损伤传感器ATM激酶相互作用的联系以及p53mRNA在DNA损伤感知机制中的作用,仍然备受期待。
    方法:邻近连接测定(PLA)已被广泛用于揭示蛋白质-mRNA和蛋白质-蛋白质相互作用的亚细胞定位。ELISA和免疫共沉淀证实了体外和细胞中的相互作用。
    结果:这项研究提供了一种新的机制,使p53mRNA与ATM激酶相互作用,并表明L22L同义突变体,已知改变p53mRNA的二级结构,阻止互动。在DNA损伤感知途径中的相关机制作用,这与下游的DNA损伤反应有关,正在探索。DNA损伤(双链DNA断裂激活ATM)后,激活的MDMX蛋白竞争ATM-p53mRNA相互作用,并阻止p53mRNA与NBS1(MRN复合物)的关联。这些数据还揭示了ATM上的结合结构域和磷酸化事件,其调节复合物到细胞质的相互作用和运输。
    结论:提出的模型显示了ATM与p53mRNA的新型相互作用,并描述了DNA损伤传感与下游p53激活途径之间的联系;支持改变二级mRNA结构的同义突变的功能含义上升。
    The ATM kinase constitutes a master regulatory hub of DNA damage and activates the p53 response pathway by phosphorylating the MDM2 protein, which develops an affinity for the p53 mRNA secondary structure. Disruption of this interaction prevents the activation of the nascent p53. The link of the MDM2 protein-p53 mRNA interaction with the upstream DNA damage sensor ATM kinase and the role of the p53 mRNA in the DNA damage sensing mechanism, are still highly anticipated.
    The proximity ligation assay (PLA) has been extensively used to reveal the sub-cellular localisation of the protein-mRNA and protein-protein interactions. ELISA and co-immunoprecipitation confirmed the interactions in vitro and in cells.
    This study provides a novel mechanism whereby the p53 mRNA interacts with the ATM kinase enzyme and shows that the L22L synonymous mutant, known to alter the secondary structure of the p53 mRNA, prevents the interaction. The relevant mechanistic roles in the DNA Damage Sensing pathway, which is linked to downstream DNA damage response, are explored. Following DNA damage (double-stranded DNA breaks activating ATM), activated MDMX protein competes the ATM-p53 mRNA interaction and prevents the association of the p53 mRNA with NBS1 (MRN complex). These data also reveal the binding domains and the phosphorylation events on ATM that regulate the interaction and the trafficking of the complex to the cytoplasm.
    The presented model shows a novel interaction of ATM with the p53 mRNA and describes the link between DNA Damage Sensing with the downstream p53 activation pathways; supporting the rising functional implications of synonymous mutations altering secondary mRNA structures.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号