histones

组蛋白
  • 文章类型: Journal Article
    细胞外组蛋白是涉及多种危重和炎症性疾病的发生和进展的关键损伤相关分子模式,比如败血症,胰腺炎,创伤,急性肝功能衰竭,急性呼吸窘迫综合征,血管炎和关节炎。在过去的十年里,组蛋白介导的炎症的病理生理学机制,内皮功能障碍,凝血激活,已经系统地阐明了疾病中的神经免疫损伤和器官功能障碍。新兴的临床前证据进一步表明,抗组蛋白策略与它们的中和剂(肝素,类肝素,天然血浆蛋白,小阴离子分子和纳米药物,等。)或体外血液净化技术可以显著缓解组蛋白诱导的有害影响,从而改善组蛋白相关的关键和炎症动物模型的结果。然而,目前缺乏对这些组蛋白靶向治疗策略的有效性和安全性的系统评估.在这次审查中,我们首先更新了对组蛋白诱导的炎症过度的潜在分子机制的最新理解,内皮功能障碍,凝血病,和器官功能障碍。然后,我们总结了肝素组蛋白靶向治疗策略的最新进展,抗组蛋白抗体,组蛋白结合蛋白或分子,和临床前研究中的组蛋白亲和血液吸附。最后,还讨论了改善组蛋白靶向治疗策略的临床翻译的挑战和未来前景,以促进更好地管理组蛋白相关疾病患者。
    Extracellular histones are crucial damage-associated molecular patterns involved in the development and progression of multiple critical and inflammatory diseases, such as sepsis, pancreatitis, trauma, acute liver failure, acute respiratory distress syndrome, vasculitis and arthritis. During the past decade, the physiopathologic mechanisms of histone-mediated hyperinflammation, endothelial dysfunction, coagulation activation, neuroimmune injury and organ dysfunction in diseases have been systematically elucidated. Emerging preclinical evidence further shows that anti-histone strategies with either their neutralizers (heparin, heparinoids, nature plasma proteins, small anion molecules and nanomedicines, etc.) or extracorporeal blood purification techniques can significantly alleviate histone-induced deleterious effects, and thus improve the outcomes of histone-related critical and inflammatory animal models. However, a systemic evaluation of the efficacy and safety of these histone-targeting therapeutic strategies is currently lacking. In this review, we first update our latest understanding of the underlying molecular mechanisms of histone-induced hyperinflammation, endothelial dysfunction, coagulopathy, and organ dysfunction. Then, we summarize the latest advances in histone-targeting therapy strategies with heparin, anti-histone antibodies, histone-binding proteins or molecules, and histone-affinity hemoadsorption in pre-clinical studies. Finally, challenges and future perspectives for improving the clinical translation of histone-targeting therapeutic strategies are also discussed to promote better management of patients with histone-related 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
    儿科高级别神经胶质瘤(pHGG)包括广泛的具有不同基因组的神经胶质瘤,表观基因组,和转录组特征。几乎50%的pHG在编码组蛋白3的基因中存在突变,包括具有H3.3-G34突变的亚型。在这种情况下,组蛋白突变通常与TP53和ATRX突变相关,以及PDGFRA和NOTCH2NL扩增。此外,H3.3-G34组蛋白突变诱导免疫相关基因的表观遗传变化,并对微环境发挥调节功能。此外,血脑屏障(BBB)的功能对治疗反应有影响.常规治疗的预后仍然较差,从而引发了额外和替代疗法的研究。有希望的分子靶标包括PDGFRA扩增,BRAF突变,EGFR扩增,NF1损失,和IDH突变。考虑到带有H3.3-G34R突变的pHGs似乎更容易受到免疫疗法(IT)的影响,最近探索了不同的选择,包括免疫检查点抑制剂,抗体介导的IT,还有Car-T细胞.这篇综述旨在总结这组小儿神经胶质瘤中有关癌症生物学和癌症-免疫细胞相互作用的知识。专注于可能的治疗选择。
    Pediatric high-grade glioma (pHGG) encompasses a wide range of gliomas with different genomic, epigenomic, and transcriptomic features. Almost 50% of pHGGs present a mutation in genes coding for histone 3, including the subtype harboring the H3.3-G34 mutation. In this context, histone mutations are frequently associated with mutations in TP53 and ATRX, along with PDGFRA and NOTCH2NL amplifications. Moreover, the H3.3-G34 histone mutation induces epigenetic changes in immune-related genes and exerts modulatory functions on the microenvironment. Also, the functionality of the blood-brain barrier (BBB) has an impact on treatment response. The prognosis remains poor with conventional treatments, thus eliciting the investigation of additional and alternative therapies. Promising molecular targets include PDGFRA amplification, BRAF mutation, EGFR amplification, NF1 loss, and IDH mutation. Considering that pHGGs harboring the H3.3-G34R mutation appear to be more susceptible to immunotherapies (ITs), different options have been recently explored, including immune checkpoint inhibitors, antibody mediated IT, and Car-T cells. This review aims to summarize the knowledge concerning cancer biology and cancer-immune cell interaction in this set of pediatric gliomas, with a focus on possible therapeutic options.
    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
    蛋白质的BET(溴结构域和外结构域)家族,特别是BRD4(含溴结构域蛋白4),在转录调控和表观遗传机制中起着至关重要的作用,影响关键的细胞过程,如增殖,分化,和DNA损伤反应。BRD4这个家族研究最多的成员,与组蛋白和非组蛋白蛋白上的乙酰化赖氨酸结合,从而调节基因表达和影响不同的细胞功能,如细胞周期,肿瘤发生,和对病毒感染的免疫反应。鉴于BRD4参与了这些基本过程,它与各种疾病有关,包括癌症和炎症,使其成为治疗发展的有希望的目标。本文综述了BET家族在基因转录中的作用,DNA损伤反应,和病毒感染,讨论靶向小分子化合物的潜力,并强调BET蛋白作为抗癌治疗的有希望的候选者。
    The BET (bromodomain and extraterminal domain) family of proteins, particularly BRD4 (bromodomain-containing protein 4), plays a crucial role in transcription regulation and epigenetic mechanisms, impacting key cellular processes such as proliferation, differentiation, and the DNA damage response. BRD4, the most studied member of this family, binds to acetylated lysines on both histones and non-histone proteins, thereby regulating gene expression and influencing diverse cellular functions such as the cell cycle, tumorigenesis, and immune responses to viral infections. Given BRD4\'s involvement in these fundamental processes, it is implicated in various diseases, including cancer and inflammation, making it a promising target for therapeutic development. This review comprehensively explores the roles of the BET family in gene transcription, DNA damage response, and viral infection, discussing the potential of targeted small-molecule compounds and highlighting BET proteins as promising candidates for anticancer therapy.
    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
    组蛋白伴侣是染色质动力学不可或缺的部分,促进核小体的组装和拆卸,从而在调节基因表达和维持基因组稳定性中起着至关重要的作用。此外,它们在染色质组装之前防止异常的组蛋白相互作用。组蛋白伴侣功能的破坏可能导致基因组不稳定,这与发病机理有关。这篇综述旨在阐明组蛋白伴侣在癌症病理中的作用,并探讨其作为治疗靶点的潜力。已经发现组蛋白伴侣在各种癌症中失调,随着表达水平的改变,突变,或导致肿瘤发生和癌症进展的异常相互作用。此外,这篇综述旨在强调组蛋白伴侣与致癌因子相互作用的分子机制,强调它们在癌细胞存活和增殖中的作用。组蛋白伴侣的失调与癌症发展显着相关,将它们确立为癌症病理的积极贡献者和治疗干预的可行靶标。这篇综述主张继续研究组蛋白伴侣靶向疗法,这在肿瘤学中具有精准医学的潜力。在理解伴侣功能和相互作用方面的未来进步预计将导致新的癌症治疗。加强患者护理和结果。
    Histone chaperones are integral to chromatin dynamics, facilitating the assembly and disassembly of nucleosomes, thereby playing a crucial role in regulating gene expression and maintaining genomic stability. Moreover, they prevent aberrant histone interactions prior to chromatin assembly. Disruption in histone chaperone function may result in genomic instability, which is implicated in pathogenesis. This review aims to elucidate the role of histone chaperones in cancer pathologies and explore their potential as therapeutic targets. Histone chaperones have been found to be dysregulated in various cancers, with alterations in expression levels, mutations, or aberrant interactions leading to tumorigenesis and cancer progression. In addition, this review intends to highlight the molecular mechanisms of interactions between histone chaperones and oncogenic factors, underscoring their roles in cancer cell survival and proliferation. The dysregulation of histone chaperones is significantly correlated with cancer development, establishing them as active contributors to cancer pathology and viable targets for therapeutic intervention. This review advocates for continued research into histone chaperone-targeted therapies, which hold potential for precision medicine in oncology. Future advancements in understanding chaperone functions and interactions are anticipated to lead to novel cancer treatments, enhancing patient care and outcomes.
    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
    长链非编码RNA(lncRNA)与表观遗传修饰(如染色质/组蛋白甲基化和乙酰化)之间的复杂串扰为肾脏疾病的发病机制和治疗提供了新的视角。lncRNAs,一类超过200个核苷酸的转录本,没有蛋白质编码潜力,现在被认为是通过不同机制影响基因表达的关键调控分子。它们通过招募或阻断负责添加或去除甲基或乙酰基的酶来调节表观遗传修饰。比如DNA,N6-甲基腺苷(m6A)和组蛋白甲基化和乙酰化,随后改变染色质结构和可及性。在急性肾损伤(AKI)等肾脏疾病中,慢性肾脏病(CKD),糖尿病肾病(DN),肾小球肾炎(GN),和肾细胞癌(RCC),DNA/RNA/组蛋白甲基化和乙酰化的异常模式与疾病的发生和进展有关,揭示了与lncRNA动力学的复杂相互作用。最近的研究强调了lncRNAs如何通过影响细胞周期控制中关键基因的表达和功能来影响肾脏病理。纤维化,和炎症反应。这篇综述将分别讨论lncRNAs和表观遗传修饰在肾脏疾病中的作用。特别强调阐明lncRNAs与DNA/RNA/组蛋白甲基化和乙酰化结合的双向调节作用和潜在机制,除了肾脏病变的潜在恶化或肾脏保护作用。了解lncRNAs和表观遗传修饰之间的相互关系不仅会阐明肾脏病理的分子基础,而且还会为治疗干预和生物标志物开发提供新的途径。推进肾病学精准医学。
    The intricate crosstalk between long noncoding RNAs (lncRNAs) and epigenetic modifications such as chromatin/histone methylation and acetylation offer new perspectives on the pathogenesis and treatment of kidney diseases. lncRNAs, a class of transcripts longer than 200 nucleotides with no protein-coding potential, are now recognized as key regulatory molecules influencing gene expression through diverse mechanisms. They modulate the epigenetic modifications by recruiting or blocking enzymes responsible for adding or removing methyl or acetyl groups, such as DNA, N6-methyladenosine (m6A) and histone methylation and acetylation, subsequently altering chromatin structure and accessibility. In kidney diseases such as acute kidney injury (AKI), chronic kidney disease (CKD), diabetic nephropathy (DN), glomerulonephritis (GN), and renal cell carcinoma (RCC), aberrant patterns of DNA/RNA/histone methylation and acetylation have been associated with disease onset and progression, revealing a complex interplay with lncRNA dynamics. Recent studies have highlighted how lncRNAs can impact renal pathology by affecting the expression and function of key genes involved in cell cycle control, fibrosis, and inflammatory responses. This review will separately address the roles of lncRNAs and epigenetic modifications in renal diseases, with a particular emphasis on elucidating the bidirectional regulatory effects and underlying mechanisms of lncRNAs in conjunction with DNA/RNA/histone methylation and acetylation, in addition to the potential exacerbating or renoprotective effects in renal pathologies. Understanding the reciprocal relationships between lncRNAs and epigenetic modifications will not only shed light on the molecular underpinnings of renal pathologies but also present new avenues for therapeutic interventions and biomarker development, advancing precision medicine in nephrology.
    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
    组蛋白赖氨酸乳酸化(Kla)已成为一种独特的表观遗传修饰,通过将乳酰基独特地添加到赖氨酸残基上,与已建立的酰化修饰显着不同。这种修饰不仅改变核小体结构,而且显著影响染色质动力学和基因表达。因此在细胞代谢中起着至关重要的作用,炎症反应,和胚胎发育。组蛋白Kla与各种代谢过程的关联,特别是糖酵解和谷氨酰胺代谢,强调了它在代谢重编程中的关键作用,包括在癌组织中,它有助于肿瘤发生,免疫逃避,和血管生成。此外,组蛋白Kla参与各种疾病的发病机制,特别是几种癌症和神经退行性疾病。组蛋白Kla的鉴定为靶向特定Kla位点的治疗干预开辟了新的途径。在这次审查中,我们总结了组蛋白Kla修饰和其他酰化修饰之间的差异,讨论组蛋白Kla在疾病中的作用和机制,最后描述了现有的药物和潜在的目标。这项研究为将组蛋白Kla与疾病联系起来的机制以及新药物和靶标的发现提供了新的见解。
    Histone lysine lactylation (Kla) has emerged as a distinct epigenetic modification that differs markedly from established acylation modifications through the unique addition of a lactyl group to a lysine residue. Such modifications not only alter nucleosome structure but also significantly impact chromatin dynamics and gene expression, thus playing a crucial role in cellular metabolism, inflammatory responses, and embryonic development. The association of histone Kla with various metabolic processes, particularly glycolysis and glutamine metabolism, underscores its pivotal role in metabolic reprogramming, including in cancerous tissues, where it contributes to tumorigenesis, immune evasion, and angiogenesis. In addition, histone Kla is involved in the pathogenesis of various diseases, particularly several cancers and neurodegenerative diseases. The identification of histone Kla opens new avenues for therapeutic interventions targeting specific Kla sites. In this review, we summarize the differences between histone Kla modifications and other acylation modifications, discuss the mechanisms and roles of histone Kla in disease, and conclude by describing existing drugs and potential targets. This study provides new insights into the mechanisms linking histone Kla to diseases and into the discovery of new drugs and targets.
    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
    室内空气污染正在成为一个日益严重的公共卫生问题,主要是由于家庭燃烧固体燃料和取暖造成的。燃烧这些燃料会产生有害化合物,例如被视为主要健康风险的颗粒物,特别影响呼吸系统疾病的发作和恶化。由于暴露于污染的室内空气会导致DNA损伤,包括DNAsd断裂以及染色体损伤,在本文中,我们的目标是通过回顾使用彗星的科学论文来概述室内空气污染对DNA损伤和基因组稳定性的影响,微核,和γ-H2AX测定。这些方法是人体生物监测和研究各种污染物作用机理的有价值的工具。通过测量DNA和染色体损伤的不同方面,可以很容易地用于评估空气污染物引起的原发性DNA损伤和基因组不稳定性。根据我们的搜索,在选定的研究中(体外,动物模型,和人类生物监测),我们发现,与对照组或未暴露组相比,室内空气污染物导致的DNA链断裂和染色体损伤水平普遍较高.总之,我们的系统回顾揭示了彗星的重要性,微核,和γ-H2AX测定作为评估不同室内空气污染物的DNA和基因组破坏潜力的敏感工具。此外,由于对家庭或公共建筑的室内空气污染水平及其对遗传物质的影响知之甚少,因此有必要进行这方面的研究。未来的研究应集中在调查复杂混合物中室内空气污染物对基因组的可能影响的研究,并将污染物与可能的健康结果联系起来。
    Indoor air pollution is becoming a rising public health problem and is largely resulting from the burning of solid fuels and heating in households. Burning these fuels produces harmful compounds, such as particulate matter regarded as a major health risk, particularly affecting the onset and exacerbation of respiratory diseases. As exposure to polluted indoor air can cause DNA damage including DNA sd breaks as well as chromosomal damage, in this paper, we aim to provide an overview of the impact of indoor air pollution on DNA damage and genome stability by reviewing the scientific papers that have used the comet, micronucleus, and γ-H2AX assays. These methods are valuable tools in human biomonitoring and for studying the mechanisms of action of various pollutants, and are readily used for the assessment of primary DNA damage and genome instability induced by air pollutants by measuring different aspects of DNA and chromosomal damage. Based on our search, in selected studies (in vitro, animal models, and human biomonitoring), we found generally higher levels of DNA strand breaks and chromosomal damage due to indoor air pollutants compared to matched control or unexposed groups. In summary, our systematic review reveals the importance of the comet, micronucleus, and γ-H2AX assays as sensitive tools for the evaluation of DNA and genome damaging potential of different indoor air pollutants. Additionally, research in this particular direction is warranted since little is still known about the level of indoor air pollution in households or public buildings and its impact on genetic material. Future studies should focus on research investigating the possible impact of indoor air pollutants in complex mixtures on the genome and relate pollutants to possible health outcomes.
    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
    作为BET(溴结构域和末端外)蛋白家族的成员,BRD4(含溴结构域蛋白4)是一种染色质相关蛋白,与乙酰化组蛋白相互作用并积极募集调节蛋白,导致基因表达和染色质重塑的调节。BRD4的细胞和表观遗传功能暗示正常发育,纤维化和炎症。BRD4被认为是一个潜在的治疗靶点,因为它经常被过度表达,并且在调节驱动肿瘤细胞增殖的基因表达程序中起着关键作用。生存,迁移和耐药性。为了解决BRD4在癌症中的作用,已经开发了几种特异性靶向BRD4的药物.抑制BRD4在临床前模型中显示出有希望的结果,几种BRD4抑制剂正在进行临床试验,用于治疗各种癌症。头颈部鳞状细胞癌(HNSCC),一组异质性的癌症,仍然是一个健康挑战,发病率高,预后差。HNSCC的常规疗法通常会对患者造成不良影响。因此,以BRD4为目标,代表了使HNSCC对化学和放射疗法敏感的有希望的策略,从而可以降低当前治疗方案的强度并随后减少副作用。然而,需要进一步的研究来充分了解BRD4在HNSCC中的潜在作用机制,以便确定BRD4靶向药物治疗HNSCC患者的最佳剂量和给药方式.
    As a member of BET (bromodomain and extra-terminal) protein family, BRD4 (bromodomain‑containing protein 4) is a chromatin‑associated protein that interacts with acetylated histones and actively recruits regulatory proteins, leading to the modulation of gene expression and chromatin remodeling. The cellular and epigenetic functions of BRD4 implicate normal development, fibrosis and inflammation. BRD4 has been suggested as a potential therapeutic target as it is often overexpressed and plays a critical role in regulating gene expression programs that drive tumor cell proliferation, survival, migration and drug resistance. To address the roles of BRD4 in cancer, several drugs that specifically target BRD4 have been developed. Inhibition of BRD4 has shown promising results in preclinical models, with several BRD4 inhibitors undergoing clinical trials for the treatment of various cancers. Head and neck squamous cell carcinoma (HNSCC), a heterogeneous group of cancers, remains a health challenge with a high incidence rate and poor prognosis. Conventional therapies for HNSCC often cause adverse effects to the patients. Targeting BRD4, therefore, represents a promising strategy to sensitize HNSCC to chemo‑ and radiotherapy allowing de‑intensification of the current therapeutic regime and subsequent reduced side effects. However, further studies are required to fully understand the underlying mechanisms of action of BRD4 in HNSCC in order to determine the optimal dosing and administration of BRD4‑targeted drugs for the treatment of patients with HNSCC.
    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
    与全球气候变化相关的不断升级的天气事件和气温上升对作物生产的威胁越来越大。植物进化了适应机制,包括压力记忆,为了应对非生物胁迫,如热,干旱,和盐度。压力记忆涉及启动,植物记得以前的压力暴露,对随后的应激事件提供增强的反应。压力记忆可以表现为躯体,代际,或者跨代记忆,坚持不同的持续时间。染色质,基因表达的中央调节因子,经历DNA乙酰化等修饰,甲基化,和组蛋白变化对非生物胁迫的反应。组蛋白修改,如H3K4me3和乙酰化,在调节基因表达中起着至关重要的作用。干旱和盐度等非生物胁迫是作物生产的重大挑战,导致产量下降。植物对压力的反应涉及逃避等策略,回避,和宽容,每个对生长阶段的影响不同。土壤盐分通过破坏水势影响植物生长,引起离子毒性,抑制营养吸收。了解植物对这些胁迫的反应需要了解组蛋白介导的修饰,染色质重塑,以及小RNA在应激记忆中的作用。组蛋白介导的修饰,包括乙酰化和甲基化,有助于表观遗传应激记忆,影响植物对环境应激源的适应。染色质重塑在非生物应激反应中起着至关重要的作用,影响应激相关基因的表达。小RNA;miRNA和siRNA,通过指导DNA甲基化和组蛋白修饰参与应激记忆途径。这些表观遗传机制的相互作用有助于植物适应反复出现的胁迫事件并增强其恢复力。总之,揭示植物对非生物胁迫的反应中的表观遗传机制为开发有弹性的农业技术提供了有价值的见解。了解植物如何利用压力记忆,组蛋白修饰,染色质重塑,小RNA对于设计策略以减轻气候变化对作物生产和全球粮食安全的影响至关重要。
    Crop production is increasingly threatened by the escalating weather events and rising temperatures associated with global climate change. Plants have evolved adaptive mechanisms, including stress memory, to cope with abiotic stresses such as heat, drought, and salinity. Stress memory involves priming, where plants remember prior stress exposures, providing enhanced responses to subsequent stress events. Stress memory can manifest as somatic, intergenerational, or transgenerational memory, persisting for different durations. The chromatin, a central regulator of gene expression, undergoes modifications like DNA acetylation, methylation, and histone variations in response to abiotic stress. Histone modifications, such as H3K4me3 and acetylation, play crucial roles in regulating gene expression. Abiotic stresses like drought and salinity are significant challenges to crop production, leading to yield reductions. Plant responses to stress involve strategies like escape, avoidance, and tolerance, each influencing growth stages differently. Soil salinity affects plant growth by disrupting water potential, causing ion toxicity, and inhibiting nutrient uptake. Understanding plant responses to these stresses requires insights into histone-mediated modifications, chromatin remodeling, and the role of small RNAs in stress memory. Histone-mediated modifications, including acetylation and methylation, contribute to epigenetic stress memory, influencing plant adaptation to environmental stressors. Chromatin remodeling play a crucial role in abiotic stress responses, affecting the expression of stress-related genes. Small RNAs; miRNAs and siRNAs, participate in stress memory pathways by guiding DNA methylation and histone modifications. The interplay of these epigenetic mechanisms helps plants adapt to recurring stress events and enhance their resilience. In conclusion, unraveling the epigenetic mechanisms in plant responses to abiotic stresses provides valuable insights for developing resilient agricultural techniques. Understanding how plants utilize stress memory, histone modifications, chromatin remodeling, and small RNAs is crucial for designing strategies to mitigate the impact of climate change on crop production and global food security.
    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: Review
    组蛋白伴侣在维持人体生理过程中起着关键作用。它们以稳定的方式与组蛋白相互作用,确保DNA复制的准确和有效执行,修复和转录。视网膜母细胞瘤结合蛋白(RBBP)4和RBBP7代表一对关键的组蛋白伴侣,它不仅控制组蛋白H3和H4的分子行为,而且还参与几种蛋白质复合物的功能,如多梳抑制复合物2和核小体重塑和脱乙酰酶,从而调节细胞周期,组蛋白修饰,DNA损伤与细胞命运RBBP4/7与一些主要的人类疾病之间有很强的关联,比如癌症,与年龄相关的记忆丧失和传染病。本综述评估了RBBP4/7在调节细胞生物学过程中的分子机制。并重点研究了RBBP4/7表达的变化及其在各种人类疾病中的潜在机制,从而为他们的诊断和治疗提供新的见解。
    Histone chaperones serve a pivotal role in maintaining human physiological processes. They interact with histones in a stable manner, ensuring the accurate and efficient execution of DNA replication, repair and transcription. Retinoblastoma binding protein (RBBP)4 and RBBP7 represent a crucial pair of histone chaperones, which not only govern the molecular behavior of histones H3 and H4, but also participate in the functions of several protein complexes, such as polycomb repressive complex 2 and nucleosome remodeling and deacetylase, thereby regulating the cell cycle, histone modifications, DNA damage and cell fate. A strong association has been indicated between RBBP4/7 and some major human diseases, such as cancer, age‑related memory loss and infectious diseases. The present review assesses the molecular mechanisms of RBBP4/7 in regulating cellular biological processes, and focuses on the variations in RBBP4/7 expression and their potential mechanisms in various human diseases, thus providing new insights for their diagnosis and treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号