Histone deacetylases

组蛋白脱乙酰酶
  • 文章类型: Journal Article
    背景:食管癌仍然是一种具有挑战性的疾病,死亡率高,治疗选择少。鉴于这些困难,表观遗传药物已成为患者护理的潜在替代品。这项研究的目标是评估Panobinostat治疗的效果和生物学后果,HDAC(组蛋白去乙酰化酶)抑制剂已被批准用于多发性骨髓瘤患者的治疗,在正常和恶性来源的食管细胞系中,后者代表了两种主要的组织学亚型:腺癌和鳞状细胞癌。
    结果:Panobinostat治疗抑制生长并阻碍增殖,食管癌细胞的集落形成和侵袭。考虑HDAC组织表达,与肿瘤组织相比,HDAC1在正常食管上皮中显著上调,而HDAC3与非恶性粘膜相比在食管癌中过度表达。在正常组织和肿瘤组织之间没有观察到HDAC2和HDAC8表达的差异。
    结论:Panobinostat暴露可有效损害食管癌细胞的恶性特征。因为HDAC3在食道肿瘤样本中显示过表达,这种表观遗传药物可能是食管癌患者的替代治疗选择.
    BACKGROUND: Oesophageal cancer remains a challenging disease with high mortality rates and few therapeutic options. In view of these difficulties, epigenetic drugs have emerged as potential alternatives for patient care. The goal of this study was to evaluate the effect and biological consequences of Panobinostat treatment, an HDAC (histone deacetylase) inhibitor already approved for treatment of patients with multiple myeloma, in oesophageal cell lines of normal and malignant origin, with the latter being representative of the two main histological subtypes: adenocarcinoma and squamous cell carcinoma.
    RESULTS: Panobinostat treatment inhibited growth and hindered proliferation, colony formation and invasion of oesophageal cancer cells. Considering HDAC tissue expression, HDAC1 was significantly upregulated in normal oesophageal epithelium in comparison with tumour tissue, whereas HDAC3 was overexpressed in oesophageal cancer compared to non-malignant mucosa. No differences between normal and tumour tissue were observed for HDAC2 and HDAC8 expression.
    CONCLUSIONS: Panobinostat exposure effectively impaired malignant features of oesophageal cancer cells. Because HDAC3 was shown to be overexpressed in oesophageal tumour samples, this epigenetic drug may represent an alternative therapeutic option for oesophageal cancer patients.
    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
    活细胞在复杂的机械线索中导航,这些线索影响着它们的行为和命运,来自内部和外部来源。在分子水平上,这些物理刺激转化为细胞反应依赖于机械传感器和传感器的复杂协调,最终影响染色质压实和基因表达。值得注意的是,组蛋白尾部的表观遗传修饰控制着基因调控位点的可及性,从而调节基因表达。在这些修改中,组蛋白乙酰化对机械微环境特别敏感,对细胞活动施加显著控制。然而,由于乙酰化网络的复杂性,组蛋白乙酰化在机械传感和转导中的确切作用仍然难以捉摸。为了解决这个差距,我们的目的是系统地探索组蛋白乙酰化的关键调节因子及其在响应生物力学刺激中的多方面作用。在这次审查中,我们最初引入细胞经历的无处不在的力,然后探索组蛋白乙酰化及其相关辅因子的动态变化,包括HDAC,帽子,和乙酰辅酶A,响应这些生物力学线索。此外,我们深入研究了组蛋白乙酰化和机械传感器/机械传感器之间的复杂相互作用,提供全面的分析。最终,本综述旨在在学术框架内全面了解组蛋白乙酰化与机械力之间的细微差别.
    Living cells navigate a complex landscape of mechanical cues that influence their behavior and fate, originating from both internal and external sources. At the molecular level, the translation of these physical stimuli into cellular responses relies on the intricate coordination of mechanosensors and transducers, ultimately impacting chromatin compaction and gene expression. Notably, epigenetic modifications on histone tails govern the accessibility of gene-regulatory sites, thereby regulating gene expression. Among these modifications, histone acetylation emerges as particularly responsive to the mechanical microenvironment, exerting significant control over cellular activities. However, the precise role of histone acetylation in mechanosensing and transduction remains elusive due to the complexity of the acetylation network. To address this gap, our aim is to systematically explore the key regulators of histone acetylation and their multifaceted roles in response to biomechanical stimuli. In this review, we initially introduce the ubiquitous force experienced by cells and then explore the dynamic alterations in histone acetylation and its associated co-factors, including HDACs, HATs, and acetyl-CoA, in response to these biomechanical cues. Furthermore, we delve into the intricate interactions between histone acetylation and mechanosensors/mechanotransducers, offering a comprehensive analysis. Ultimately, this review aims to provide a holistic understanding of the nuanced interplay between histone acetylation and mechanical forces within an academic framework.
    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
    I类组蛋白脱乙酰酶(HDAC)与多种疾病的发展密切相关。包括癌症,神经退行性疾病,艾滋病毒,和炎症性疾病。考虑到在肿瘤发生中的重要作用,I类HDAC已成为治疗策略的高度理想目标,特别是在抗癌药物开发领域。然而,传统的I类HDAC抑制剂面临着一些挑战,如获得性抗性,固有的毒性,以及抑制HDAC非酶功能的功效有限。为了解决这些问题,新的策略已经出现,包括I类HDAC双作用抑制剂的开发,靶向蛋白质降解(TPD)技术,如PROTACs,分子胶,和HyT降解器,以及共价抑制剂。这篇综述提供了I类HDAC酶和抑制剂的全面概述,通过最初介绍它们的结构和生物学作用。随后,我们专注于I类HDAC调制器的最新进展,包括同工型选择性I类抑制剂,双靶点抑制剂,TPDs,和共价抑制剂,从合理设计原则的角度来看,药效学,药代动力学,和临床进展。最后,我们还提供了挑战,并概述了I类HDAC靶向癌症治疗药物发现领域的未来前景。
    Class I histone deacetylases (HDACs) are closely associated with the development of a diverse array of diseases, including cancer, neurodegenerative disorders, HIV, and inflammatory diseases. Considering the essential roles in tumorigenesis, class I HDACs have emerged as highly desirable targets for therapeutic strategies, particularly in the field of anticancer drug development. However, the conventional class I HDAC inhibitors faced several challenges such as acquired resistance, inherent toxicities, and limited efficacy in inhibiting non-enzymatic functions of HDAC. To address these problems, novel strategies have emerged, including the development of class I HDAC dual-acting inhibitors, targeted protein degradation (TPD) technologies such as PROTACs, molecular glues, and HyT degraders, as well as covalent inhibitors. This review provides a comprehensive overview of class I HDAC enzymes and inhibitors, by initially introducing their structure and biological roles. Subsequently, we focus on the recent advancements of class I HDAC modulators, including isoform-selective class I inhibitors, dual-target inhibitors, TPDs, and covalent inhibitors, from the perspectives of rational design principles, pharmacodynamics, pharmacokinetics, and clinical progress. Finally, we also provide the challenges and outlines future prospects in the realm of class I HDAC-targeted drug discovery for cancer therapeutics.
    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
    弥漫性中线神经胶质瘤(DMG)是中枢神经系统的侵袭性且致命的儿科肿瘤,对治疗具有高度抵抗力。组蛋白H3(H3-K27M)上残基27的赖氨酸替换为甲硫氨酸是DMG中的驱动突变,重塑这些细胞的表观遗传景观以促进肿瘤发生。H3-K27M胶质瘤的特征是组蛋白乙酰化和甲基化途径的失调,以及致癌MYC途径。为了寻找有效的治疗方法,我们研究了组蛋白脱乙酰酶(HDACs)和MYC双重靶向治疗这些肿瘤的潜力.用Sulfopin治疗H3-K27M患者来源的细胞,一种显示在体内阻断MYC驱动的肿瘤的抑制剂,与HDAC抑制剂伏立诺他联合使用,导致细胞活力大幅下降。此外,转录组和表观基因组谱分析揭示了这种药物组合在显著的致癌途径如mTOR的下调中的协同作用。最后,患者来源的原位异种移植模型的体内研究显示,在用该药物组合治疗的小鼠中,肿瘤生长显著减少.这些结果突出了PIN1和HDAC抑制剂的联合治疗作为这些侵袭性肿瘤的有希望的治疗方法。
    弥漫性中线神经胶质瘤(DMGs)是儿童中最具侵袭性和致命性的脑癌。它们通常与组蛋白的变化有关,控制基因活性并赋予染色体结构的蛋白质。大多数患有DMGs的儿童,例如,它们的组蛋白H3蛋白具有相同的异常(称为H3-K27M突变)。这种变化影响了称为甲基和乙酰基的小化学标签如何添加到组蛋白3上,这反过来改变了蛋白质打开和关闭基因的方式。因此,肿瘤开始发展。一种针对DMGs的潜在治疗策略是使用组蛋白去乙酰化酶抑制剂(HDACi),一种有前途的药物,可以抑制从组蛋白中去除乙酰基的酶。患者可以对HDACi产生耐药性,然而,强调需要探索其他方法。一种可能性是用几种药物治疗患者,每个通常针对一个独特的生物过程,有助于癌症的出现。这种联合方法可以有多种好处;药物可能会放大彼此的作用,例如,并且细胞在当时对一种以上的化合物产生抗性的可能性也较小。此外,联合用药中的每种药物都可以较低的剂量使用,以减少副作用并使患者受益。DMG肿瘤细胞通常具有较高的称为MYC的蛋白质活性水平,这可能有助于肿瘤的生长。Algranati,Oren等人。因此,开始测试将HDACi称为Vorinostat与阻断MYC活性的药物(Sulfopin)联合使用是否可以作为这种癌症的有效治疗方法。8名DMG患者的肿瘤样本接受了单独的Sulfopin治疗,或与伏立诺他联合使用的磺福平。暴露于两种药物的细胞不太可能存活,和额外的遗传实验表明,联合治疗导致促进肿瘤发展的途径被阻断。当将磺胺嘧啶和伏立诺他都施用于生长人类DMG肿瘤的小鼠时,动物表现出肿瘤生长的更大减少。DMG的治疗选择通常有限,化疗通常无效,手术不可能。Algranati的作品,Oren等人。提示将HDACi和靶向MYC途径的药物联合使用是一种策略,应进一步研究以确定是否可能临床应用.
    Diffuse midline gliomas (DMGs) are aggressive and fatal pediatric tumors of the central nervous system that are highly resistant to treatments. Lysine to methionine substitution of residue 27 on histone H3 (H3-K27M) is a driver mutation in DMGs, reshaping the epigenetic landscape of these cells to promote tumorigenesis. H3-K27M gliomas are characterized by deregulation of histone acetylation and methylation pathways, as well as the oncogenic MYC pathway. In search of effective treatment, we examined the therapeutic potential of dual targeting of histone deacetylases (HDACs) and MYC in these tumors. Treatment of H3-K27M patient-derived cells with Sulfopin, an inhibitor shown to block MYC-driven tumors in vivo, in combination with the HDAC inhibitor Vorinostat, resulted in substantial decrease in cell viability. Moreover, transcriptome and epigenome profiling revealed synergistic effect of this drug combination in downregulation of prominent oncogenic pathways such as mTOR. Finally, in vivo studies of patient-derived orthotopic xenograft models showed significant tumor growth reduction in mice treated with the drug combination. These results highlight the combined treatment with PIN1 and HDAC inhibitors as a promising therapeutic approach for these aggressive tumors.
    Diffuse midline gliomas (DMGs) are among the most aggressive and fatal brain cancers in children. They are often associated with changes in histones, the proteins that control gene activity and give chromosomes their structure. Most children with DMGs, for example, share the same anomaly in their histone H3 protein (referred to as the H3-K27M mutation). This change affects how small chemical tags called methyl and acetyl groups can be added onto histone 3, which in turn alters the way the protein can switch genes on and off. As a result, tumours start to develop. One potential therapeutic strategy against DMGs is to use histone deacetylase inhibitors (HDACi), a promising type of drugs which inhibits the enzymes that remove acetyl groups from histones. Patients can develop resistance to HDACi, however, highlighting the need to explore other approaches. One possibility is to treat patients with several types of drugs, each usually targeting a distinct biological process that contributes to the emergence of cancer. This combined approach can have multiple benefits; the drugs potentially amplify each other’s effect, for example, and it is also less likely for cells to become resistant to more than one compound at the time. In addition, each drug in the combination can be used in a lower dose to reduce side effects and benefit patients. DMG tumour cells often feature higher activity levels of a protein known as MYC, which can contribute to the growth of the tumour. Algranati, Oren et al. therefore set out to test whether combining an HDACi known as Vorinostat with a drug that blocks MYC activity (Sulfopin) can act as an effective treatment for this cancer. Tumour samples from eight DMG patients were treated with either Sulfopin alone, or Sulfopin in association with Vorinostat. Cells exposed to both drugs were less likely to survive, and additional genetic experiments showed that the combined treatment had resulted in pathways that promote tumour development being blocked. When both Sulfopin and Vorinostat were administered to mice made to grow human DMG tumors, the animals showed a greater reduction in tumor growth. Treatment options for DMG are usually limited, with chemotherapy often being ineffective and surgery impossible. The work by Algranati, Oren et al. suggests that combining HDACi and drugs targeting the MYC pathway is a strategy that should be examined further to determine whether clinical applications are possible.
    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
    暴露于颗粒物(PM)可引起气道炎症并使各种气道疾病恶化。然而,PM引发气道炎症的潜在分子机制尚未完全阐明,缺乏有效的干预措施。我们的研究表明,PM暴露通过METTL3/m6A甲基化/IGF2BP3途径增加了人支气管上皮细胞和小鼠气道上皮中组蛋白脱乙酰酶9(HDAC9)的表达。功能实验表明,HDAC9上调可促进PM诱导的气道炎症和MAPK信号通路的激活。机械上,HDAC9调控双特异性磷酸酶9(DUSP9)启动子区K12(H4K12)组蛋白4乙酰化去乙酰化抑制DUSP9的表达,激活MAPK信号通路,从而促进PM诱导的气道炎症。此外,HDAC9与MEF2A结合以削弱其对PM诱导的气道炎症的抗炎作用。然后,我们开发了一种新型的吸入脂质纳米颗粒系统,用于将HDAC9siRNA递送到气道,为PM引起的气道炎症提供有效的治疗。总的来说,我们阐明了HDAC9在PM诱导的气道炎症中的关键调节机制,并介绍了一种针对HDAC9的吸入治疗方法.这些发现有助于减轻由PM暴露引起的各种气道疾病的负担。
    Exposure to particulate matter (PM) can cause airway inflammation and worsen various airway diseases. However, the underlying molecular mechanism by which PM triggers airway inflammation has not been completely elucidated, and effective interventions are lacking. Our study revealed that PM exposure increased the expression of histone deacetylase 9 (HDAC9) in human bronchial epithelial cells and mouse airway epithelium through the METTL3/m6A methylation/IGF2BP3 pathway. Functional assays showed that HDAC9 upregulation promoted PM-induced airway inflammation and activation of MAPK signaling pathway in vitro and in vivo. Mechanistically, HDAC9 modulated the deacetylation of histone 4 acetylation at K12 (H4K12) in the promoter region of dual specificity phosphatase 9 (DUSP9) to repress the expression of DUSP9 and resulting in the activation of MAPK signaling pathway, thereby promoting PM-induced airway inflammation. Additionally, HDAC9 bound to MEF2A to weaken its anti-inflammatory effect on PM-induced airway inflammation. Then, we developed a novel inhaled lipid nanoparticle system for delivering HDAC9 siRNA to the airway, offering an effective treatment for PM-induced airway inflammation. Collectively, we elucidated the crucial regulatory mechanism of HDAC9 in PM-induced airway inflammation and introduced an inhaled therapeutic approach targeting HDAC9. These findings contribute to alleviating the burden of various airway diseases caused by PM exposure.
    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
    抑制兼性异染色质对于许多生物体的发育过程至关重要。Polycomb抑制复合物2对组蛋白H3赖氨酸27(H3K27)的甲基化是真菌和高等真核生物兼性异染色质的显着特征。尽管这种甲基化通常与沉默有关,压制的详细机制仍未完全理解。我们利用正向遗传学方法来鉴定在粗糙神经孢菌中维持兼性异染色质基因沉默所需的基因,并鉴定了三个以前未表征的对沉默很重要的基因:sds3(NCU01599),rlp1(RPD3L蛋白1;NCU09007),和rlp2(RPD3L蛋白2;NCU02898)。我们发现SDS3,RLP1和RLP2与酿酒酵母Rpd3L复合物的N.crassa同源物相关,并且是抑制H3K27甲基化基因子集所必需的。这些基因的缺失不会导致H3K27甲基化的缺失,但会增加组蛋白H3赖氨酸14在上调基因的乙酰化,这表明RPD3L驱动的脱乙酰是N.crassa中兼性异染色质沉默所需的因素,也许在其他生物体中。
    Repression of facultative heterochromatin is essential for developmental processes in numerous organisms. Methylation of histone H3 lysine 27 (H3K27) by Polycomb repressive complex 2 is a prominent feature of facultative heterochromatin in both fungi and higher eukaryotes. Although this methylation is frequently associated with silencing, the detailed mechanism of repression remains incompletely understood. We utilized a forward genetics approach to identify genes required to maintain silencing at facultative heterochromatin genes in Neurospora crassa and identified three previously uncharacterized genes that are important for silencing: sds3 (NCU01599), rlp1 (RPD3L protein 1; NCU09007), and rlp2 (RPD3L protein 2; NCU02898). We found that SDS3, RLP1, and RLP2 associate with N. crassa homologs of the Saccharomyces cerevisiae Rpd3L complex and are required for repression of a subset of H3K27-methylated genes. Deletion of these genes does not lead to loss of H3K27 methylation but increases acetylation of histone H3 lysine 14 at up-regulated genes, suggesting that RPD3L-driven deacetylation is a factor required for silencing of facultative heterochromatin in N. crassa, and perhaps in other organisms.
    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
    组蛋白脱乙酰酶(HDACs)的失调与癌症的发生和发展密切相关。这里,我们全面分析了所有HDAC家族成员与22种不同肿瘤类型实体瘤的几种临床病理和分子特征之间的关联,主要关注癌症的干性和免疫力。为此,我们使用了公开可用的TCGA数据和几种生物信息学工具(即,GEPIA2,TISIDB,GSCA,Enrichr,GSEA)。我们的分析表明,I类和II类HDAC蛋白与不同的癌症表型相关。转录组分析表明I类HDAC成员,包括HDAC2,与癌症干性呈正相关,而IIA类HDAC蛋白,以HDAC7为代表,显示与实体瘤中癌症干细胞样表型呈负相关。与含有大量HDAC7蛋白的肿瘤相反,HDAC2过表达癌症的转录组特征显著富集了先前确定为干性相关基因的生物学术语.此外,高表达HDAC2的肿瘤被免疫相关过程耗尽,HDAC2表达与肿瘤免疫抑制微环境相关。相反,HDAC7上调与增强的免疫反应显着相关,其次是富集的CD4+和CD8+T细胞浸润。这是第一份全面的报告,展示了特定HDAC家族成员之间强大而通用的关联。癌症去分化,和实体瘤中的抗肿瘤免疫状态。
    Dysregulation of histone deacetylases (HDACs) is closely associated with cancer development and progression. Here, we comprehensively analyzed the association between all HDAC family members and several clinicopathological and molecular traits of solid tumors across 22 distinct tumor types, focusing primarily on cancer stemness and immunity. To this end, we used publicly available TCGA data and several bioinformatic tools (i.e., GEPIA2, TISIDB, GSCA, Enrichr, GSEA). Our analyses revealed that class I and class II HDAC proteins are associated with distinct cancer phenotypes. The transcriptomic profiling indicated that class I HDAC members, including HDAC2, are positively associated with cancer stemness, while class IIA HDAC proteins, represented by HDAC7, show a negative correlation to cancer stem cell-like phenotypes in solid tumors. In contrast to tumors with high amounts of HDAC7 proteins, the transcriptome signatures of HDAC2-overexpressing cancers are significantly enriched with biological terms previously determined as stemness-associated genes. Moreover, high HDAC2-expressing tumors are depleted with immune-related processes, and HDAC2 expression correlates with tumor immunosuppressive microenvironments. On the contrary, HDAC7 upregulation is significantly associated with enhanced immune responses, followed by enriched infiltration of CD4+ and CD8+ T cells. This is the first comprehensive report demonstrating robust and versatile associations between specific HDAC family members, cancer dedifferentiation, and anti-tumor immune statuses in solid tumors.
    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
    III类组蛋白脱乙酰酶SIRT1是研究最广泛的去乙酰化酶。它对广泛的脱乙酰酶抑制剂曲古抑菌素A具有抗性,并且依赖于氧化的烟酰胺腺嘌呤核苷酸(NAD)。SIRT1在许多类型癌症的肿瘤发生中起着至关重要的作用。包括结直肠癌(CRC)。越来越多的证据表明SIRT1是CRC的治疗靶点;然而,SIRT1在CRC中的功能和潜在机制仍有待阐明。在这里,我们提供了一个详细的和更新的审查,以说明SIRT1调节许多在CRC细胞中出错的过程,如细胞凋亡,自噬,扩散,迁移,入侵,转移,氧化应激,对化学放射疗法的抗性,免疫逃避,和代谢重编程。此外,我们将我们的审查与CRC治疗的临床实践紧密联系起来,总结SIRT1抑制剂在CRC治疗中的作用机制及前景。SIRT1抑制剂作为CRC的单一疗法或与化疗联合使用,放射治疗,和免疫疗法进行了全面讨论。从表观遗传调控到其潜在的治疗效果,我们希望对SIRT1在CRC中的作用提供新的见解和全面的理解。
    The class-III histone deacetylase SIRT1 is the most extensively investigated sirtuin deacetylase. It is resistant to the broad deacetylase inhibitor trichostatin A and depends on oxidized nicotinamide adenine nucleotide (NAD+). SIRT1 plays a crucial role in the tumorigenesis of numerous types of cancers, including colorectal cancer (CRC). Accumulating evidence indicates that SIRT1 is a therapeutic target for CRC; however, the function and underlying mechanism of SIRT1 in CRC still need to be elucidated. Herein, we provide a detailed and updated review to illustrate that SIRT1 regulates many processes that go awry in CRC cells, such as apoptosis, autophagy, proliferation, migration, invasion, metastasis, oxidative stress, resistance to chemo-radio therapy, immune evasion, and metabolic reprogramming. Moreover, we closely link our review to the clinical practice of CRC treatment, summarizing the mechanisms and prospects of SIRT1 inhibitors in CRC therapy. SIRT1 inhibitors as monotherapy in CRC or in combination with chemotherapy, radiotherapy, and immune therapies are comprehensively discussed. From epigenetic regulation to its potential therapeutic effect, we hope to offer novel insights and a comprehensive understanding of SIRT1\'s role in CRC.
    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
    目的:大量证据表明组蛋白脱乙酰酶(HDAC)抑制剂可减少骨关节炎(OA)动物模型中的软骨破坏。肿瘤坏死因子(TNF)-α阻断治疗OA可通过减缓关节损伤提供有效的关节保护。探讨依那西普(TNF-α抑制剂)对大鼠OA发生发展的影响及对大鼠伤害性行为和HDACs表达的影响。软骨中的RUNX2和MMP13。
    方法:通过前交叉韧带横断(ACLT)诱导Wistar大鼠OA。ACLT+依那西普(1和5mg/kg)组,在ACLT后连续5周腹膜内给予1或5毫克(mg)依那西普。分析痛觉行为和膝关节宽度的变化。对软骨进行组织学和免疫组织化学评估。
    结果:与单独使用ACLT相比,ACLT+依那西普显著改善了机械异常性疼痛和负重分布。在用依那西普治疗的OA大鼠中,软骨变性和滑膜炎明显低于ACLT大鼠。受OA影响的软骨还显示响应依那西普的HDAC6、7、RUNX-2和MMP-13的表达降低,但HDAC4的表达增加。
    结论:我们的研究表明依那西普治疗(1)减轻了大鼠OA和滑膜炎的发展,(2)减少伤害感受,和(3)调节软骨细胞代谢,可能通过抑制细胞HDAC6和HDAC7,RUNX2和MMP13并增加HDAC4表达。根据新的证据,依那西普可能在OA中具有治疗潜力。
    OBJECTIVE: Mounting evidence suggests that histone deacetylases (HDAC) inhibitors reduce cartilage destruction in animal models of osteoarthritis (OA). Tumor necrosis factor (TNF)-α-blocking treatment for OA may provide effective joint protection by slowing joint damage. To investigate the effects of intraperitoneal administration of etanercept (a TNF-α inhibitor) on OA development in rats and changes in the nociceptive behavior of rats and expression of HDACs, RUNX2, and MMP13 in cartilage.
    METHODS: Induction of OA in Wistar rats was accomplished through anterior cruciate ligament transection (ACLT). One or five milligrams (mg) of etanercept was administered intraperitoneally for 5 consecutive weeks after ACLT to the ACLT + etanercept (1 and 5 mg/kg) groups. Nociceptive behavior and changes in knee joint width were analyzed. Cartilage was evaluated histologically and immunohistochemically.
    RESULTS: ACLT + etanercept significantly improved mechanical allodynia and weight-bearing distribution compared to ACLT alone. In OA rats treated with etanercept, cartilage degeneration and synovitis were significantly less pronounced than those in ACLT rats. OA-affected cartilage also showed reduced expression of HDAC 6, 7, RUNX-2, and MMP-13 in response to etanercept but increased expression of HDAC4.
    CONCLUSIONS: Our study demonstrated that etanercept therapy (1) attenuated the development of OA and synovitis in rats, (2) reduced nociception, and (3) regulated chondrocyte metabolism, possibly by inhibiting cell HDAC6 and HDAC7, RUNX2, and MMP13 and increasing HDAC4 expression. Based on new evidence, etanercept may have therapeutic potential in OA.
    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
    在现代癌症治疗中,阻断多个目标是一种标准方法,并且已经有许多双靶标药物可以通过单个分子实现多重抑制。在这里,我们通过基于STAT3抑制剂E28和HDAC抑制剂MS-275的药效团组合策略,设计并合成了一系列具有信号转导和转录激活因子3(STAT3)和组蛋白去乙酰化酶(HDAC)抑制活性的新型衍生物。其中,化合物24(IC50=8.22±0.27μM)在MCF-7乳腺癌细胞中显示出比临床I类HDAC抑制剂MS-275(IC50=14.65±0.24μM)更好的抗肿瘤活性。此外,通过蛋白质印迹分析验证了化合物24对HDAC和STAT3的双重抑制作用.该研究为进一步探索用单个分子实现的STAT3-HDAC途径抑制剂提供了新的工具化合物。
    In modern cancer therapy, blockage of more than one target is a standard approach, and there are already many dual-target drugs that can achieve multiple inhibition through a single molecule. Herein, we designed and synthesized a series of novel derivatives with signal transducer and activator of transcription 3 (STAT3) and histone deacetylase (HDAC) inhibitory activity through strategy of combining pharmacophore based on the STAT3 inhibitor E28 and HDAC inhibitor MS-275. Among them, compound 24 (IC50 = 8.22 ± 0.27 μM) showed better anti-tumor activity than the clinical Class I HDAC inhibitor MS-275 (IC50 = 14.65 ± 0.24 μM) in MCF-7 breast cancer cells. Furthermore, the dual inhibition to HDAC and STAT3 of compound 24 was validated by western blot analysis. The study provides new tool compounds for further exploration of STAT3-HDAC pathway inhibitor achieved with a single molecule.
    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