histone deacetylase inhibitor

组蛋白去乙酰化酶抑制剂
  • 文章类型: Journal Article
    B细胞淋巴瘤由于其生物学和临床异质性而难以治愈,由于天然的化学抗性。克服癌症诱导的免疫逃避的免疫疗法已成为肿瘤学最新发展的中心。在各种肿瘤中,破坏程序性细胞死亡蛋白1(PD-1)介导的免疫抑制的各种药物的成就强调了这一点。然而,虽然PD-1阻断对许多恶性肿瘤有效,相当比例的癌症,包括B细胞淋巴瘤,对这些治疗策略显示出一定的原发性耐药率。组蛋白去乙酰化酶抑制剂(HDACis)虽然抑制细胞增殖,但仍显示出抗癌活性。诱导分化和触发细胞凋亡。本研究旨在探索B细胞淋巴瘤中结合HDACi(romidepsin)和PD‑1阻断(BMS‑1)的治疗策略,利用构建的B细胞淋巴瘤小鼠模型。两种抑制剂的IC50通过MTT测定证实,它们的抑制作用显示出剂量和时间依赖性。数据表明,romidepsin和BMS-1的联合治疗协同抑制了B细胞淋巴瘤的生长。此外,结果表明,罗米地辛和BMS-1协同触发小鼠B细胞淋巴瘤细胞凋亡。这些药物的协同作用能够激活肿瘤浸润淋巴细胞,特别是CD3+CD4+和CD3+CD8+T细胞。本研究的结果强调了HDAC抑制与PD-1阻断联合作为B细胞淋巴瘤新治疗方法的潜力。强调这两种机制在增强抗肿瘤免疫力方面的协同作用。
    B‑cell lymphoma is difficult to cure because of its biological and clinical heterogeneity, and due to native chemoresistance. Immunotherapies that overcome cancer‑induced immune evasion have been the center of recent developments in oncology. This is emphasized by the accomplishment of various agents that disrupt programmed cell death protein 1 (PD‑1)‑mediated immune suppression in diverse tumors. However, while PD‑1 blockade has been effective in numerous malignancies, a significant proportion of cancers, including B‑cell lymphoma, show certain rates of primary resistance to these therapeutic strategies. Histone deacetylase inhibitors (HDACis) have exhibited anticancer activity though suppressing cell proliferation, inducing differentiation and triggering apoptosis. The present study aimed to explore a therapeutic strategy combining a HDACi (romidepsin) and PD‑1 blockade (BMS‑1) in B‑cell lymphoma, utilizing a constructed mouse model of B‑cell lymphoma. The IC50 of the two inhibitors was confirmed by MTT assay, and their inhibitory effects were revealed to be dose‑ and time‑dependent. The data demonstrated that the combined treatment of romidepsin and BMS‑1 synergistically inhibited the growth of B‑cell lymphoma. Furthermore, it was revealed that romidepsin and BMS‑1 synergistically triggered apoptosis in mouse B‑cell lymphoma. The synergistic effect of these agents was capable of activating tumor‑infiltrating lymphocytes, particularly CD3+CD4+ and CD3+CD8+ T cells. The results of the present study underscore the potential of HDAC inhibition in conjunction with PD‑1 blockade as a novel therapeutic approach for B‑cell lymphoma, highlighting the synergistic effects of these two mechanisms in enhancing antitumor immunity.
    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
    在雌激素受体(ER)阳性乳腺癌的治疗中建立了抗雌激素疗法。然而,为了克服耐药性并改善治疗效果,需要新的策略,例如针对广泛认可的异常表观遗传学.本研究旨在探讨芳香化酶抑制剂依西美坦和组蛋白去乙酰化酶(HDAC)抑制剂和抗氧化剂α-硫辛酸在ER阳性乳腺癌细胞中的联合作用。首先,α-硫辛酸的对映体和外消旋混合物,研究了rac-二氢-硫辛酸对HDAC的抑制作用。我们发现HDAC抑制活性在1-3位数微摩尔范围内,优选HDAC6。Rac-二氢-硫辛酸比rac-α-硫辛酸略强。α-硫辛酸的抗增殖IC50值在3位数微摩尔范围内。值得注意的是,依西美坦和α-硫辛酸的组合在不同的孵育时间(24h至10d)和读数(MTT,活细胞荧光显微镜,半胱天冬酶激活)通过Chou-Talalay方法分析。α-硫辛酸增加线粒体融合和凋亡相关蛋白p21、APAF-1、BIM、FOXO1,并降低抗凋亡蛋白survivin的表达,BCL-2和c-myc。总之,联合使用依西美坦和α-硫辛酸是ER阳性乳腺癌的一种有前景的新型治疗选择.
    Anti-estrogenic therapy is established in the management of estrogen receptor (ER)-positive breast cancer. However, to overcome resistance and improve therapeutic outcome, novel strategies are needed such as targeting widely recognized aberrant epigenetics. The study aims to investigate the combination of the aromatase inhibitor exemestane and the histone deacetylase (HDAC) inhibitor and antioxidant α-lipoic acid in ER-positive breast cancer cells. First, the enantiomers and the racemic mixture of α-lipoic acid, and rac-dihydro-lipoic acid were investigated for HDAC inhibition. We found HDAC inhibitory activity in the 1-3-digit micromolar range with a preference for HDAC6. Rac-dihydro-lipoic acid is slightly more potent than rac-α-lipoic acid. The antiproliferative IC50 value of α-lipoic acid is in the 3-digit micromolar range. Notably, the combination of exemestane and α-lipoic acid resulted in synergistic behavior under various incubation times (24 h to 10 d) and readouts (MTT, live-cell fluorescence microscopy, caspase activation) analyzed by the Chou-Talalay method. α-lipoic acid increases mitochondrial fusion and the expression of apoptosis-related proteins p21, APAF-1, BIM, FOXO1, and decreases expression of anti-apoptotic proteins survivin, BCL-2, and c-myc. In conclusion, combining exemestane with α-lipoic acid is a promising novel treatment option for ER-positive breast 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
    免疫检查点抑制剂(CPIs)已在许多早期和晚期恶性肿瘤中广泛采用。已经提出组蛋白脱乙酰酶抑制剂(HDACis)和烷化剂(AAs)增强CPIs对肿瘤细胞的作用。我们进行了全面的文献综述,以探索CPIs之间的潜在协同活性。AAS,和HDACis。
    在PubMed中使用预定义的搜索字符串识别了临床和非临床研究,这些研究描述了接受CPIs和伴随或序贯(CPI前或后)AA或HDACis的癌症患者的预后。类似地进行关键肿瘤学大会的手动搜索。所有相关文章和摘要都是手动筛选相关性的,根据使用的特定抗癌剂(CPIs,AAS,或HDACis),肿瘤实体,以及治疗是否伴随或序贯。
    总的来说,针对一系列肿瘤类型的227项独特临床研究,实体瘤和血液恶性肿瘤,已确定。审查了关于I期和II期临床研究的109种出版物以及关于III期研究的41种出版物。最常见的肿瘤类型是黑色素瘤,三阴性乳腺癌,非小细胞肺癌,和霍奇金淋巴瘤.随机临床研究确定,所有这些都报告了CPI与AA的组合,与CPI或AA单药治疗相比,联合治疗组的结局更好。同样,CPIs和HDACis的联合治疗证明了有希望的活性。
    CPI与AA或HDACi的序贯或伴随给药可以改善患有多种肿瘤类型的患者的预后。有理由支持对CPI之间协同作用的潜力进行进一步调查,烷化剂和/或HDAC在非临床和临床设置。
    接受癌症治疗的人通常一次会接受一种以上的药物,和癌症药物联合治疗的概念经常被认为是改善患者预后的潜在机会。我们回顾了已发表的临床试验文献和实验室开展的工作,以探索将阻止癌细胞增殖的靶向药物(称为检查点抑制剂)与杀死癌细胞的传统化学疗法相结合是否可能是一种有用的方法。我们在出版物中看到了在化疗的同时使用检查点抑制剂的证据,或在化疗前或后立即给予。最重要的证据来自临床试验,其中直接将接受联合治疗的患者的结果与接受单一治疗的患者的结果进行比较。这些研究表明,与接受单一疗法的患者相比,接受癌症药物组合治疗的患者的预后更好。我们还发现了添加另一类抗癌药物的证据,称为组蛋白脱乙酰酶抑制剂,可能会使肿瘤对检查点抑制剂敏感。这些发现为检查烷化剂和/或组蛋白脱乙酰酶抑制剂与检查点抑制剂的组合提供了理论基础。
    UNASSIGNED: Immune checkpoint inhibitors (CPIs) have been widely adopted in a number of early and advanced malignancies. Histone deacetylase inhibitors (HDACis) and alkylating agents (AAs) have been suggested to potentiate the actions of CPIs on tumor cells. We conducted a comprehensive literature review to explore the potential synergistic activity between CPIs, AAs, and HDACis.
    UNASSIGNED: Clinical and non-clinical studies describing outcomes in patients with cancer receiving CPIs and either concomitant or sequential (pre- or post-CPI) AAs or HDACis were identified in PubMed using pre-defined search strings. Manual searches of key oncology congresses were similarly performed. All relevant articles and abstracts were manually screened for relevance, classified according to the specific anticancer agents used (CPIs, AAs, or HDACis), tumor entity, and whether treatment was concomitant or sequential.
    UNASSIGNED: Overall, 227 unique clinical studies across a range of tumor types, both solid tumors and hematological malignancies, were identified. One hundred and fifty-nine publications on Phase I and II clinical studies together with 41 publications on Phase III studies were examined. The most commonly investigated tumor types were melanoma, triple-negative breast cancer, non-small cell lung cancer, and Hodgkin lymphoma. The randomized clinical studies identified, all of which reported on the combination of a CPI with an AA, demonstrated superior outcomes in the combination arm compared with CPI or AA monotherapy. Similarly, combination therapy with CPIs and HDACis demonstrated promising activity.
    UNASSIGNED: Sequential or concomitant administration of a CPI with an AA or an HDACi may improve outcomes for patients with a range of tumor types. There is a rationale to support further investigation into the potential for synergy between CPIs, alkylating agents and/or HDACis in both the non-clinical and clinical settings.
    People being treated for cancer will often receive more than one drug at a time, and the concept of combining cancer drugs is frequently investigated as a potential opportunity to improve outcomes for patients. We reviewed the published literature for clinical trials and work undertaken in laboratories to explore whether combining targeted agents that stop cancer cells from multiplying (known as checkpoint inhibitors) with traditional chemotherapy that kills cancer cells could be a useful approach. We looked at evidence in publications where checkpoint inhibitors were used at the same time as chemotherapy, or given immediately before or after chemotherapy. The most important evidence came from clinical trials where outcomes for patients receiving combinations of treatment were directly compared with those from patients receiving a single treatment. These studies showed superior outcomes for patients who were treated with a combination of cancer drugs compared with patients receiving monotherapy. We also found evidence that adding another class of cancer drug, called histone deacetylase inhibitors, might sensitize tumors to checkpoint inhibitors. These findings provide a rationale for examining alkylating agents and/or histone deacetylase inhibitors combined with checkpoint inhibitors.
    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(HDAC)催化去除组蛋白和非组蛋白的乙酰化修饰,调节基因表达和其他细胞过程。HDAC抑制剂(HDACi),批准的抗癌剂,成为心脏病的潜在新疗法。在许多心脏疾病的临床前动物模型中观察到HDACi的心脏保护作用。已经开发了遗传小鼠模型来了解每种HDAC在心脏功能中的作用。一些发现是有争议的。这里,我们概述了HDACi和HDAC在生理或病理条件下如何影响心脏功能.我们专注于锌依赖性经典HDAC的体内研究,强调涉及心脏肥大的疾病,心肌梗死(MI),缺血再灌注(I/R)损伤,和心力衰竭。特别是,我们回顾了无偏组学研究如何帮助我们理解HDACi和HDAC心脏效应的潜在机制.
    Histone deacetylases (HDAC) catalyze the removal of acetylation modifications on histones and non-histone proteins, which regulates gene expression and other cellular processes. HDAC inhibitors (HDACi), approved anti-cancer agents, emerge as a potential new therapy for heart diseases. Cardioprotective effects of HDACi are observed in many preclinical animal models of heart diseases. Genetic mouse models have been developed to understand the role of each HDAC in cardiac functions. Some of the findings are controversial. Here, we provide an overview of how HDACi and HDAC impact cardiac functions under physiological or pathological conditions. We focus on in vivo studies of zinc-dependent classical HDACs, emphasizing disease conditions involving cardiac hypertrophy, myocardial infarction (MI), ischemic reperfusion (I/R) injury, and heart failure. In particular, we review how non-biased omics studies can help our understanding of the mechanisms underlying the cardiac effects of HDACi and HDAC.
    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
    EB病毒(EBV)与淋巴瘤和上皮瘤有关,但缺乏专门针对EBV阳性肿瘤的药物。BamHI向右转录本(BART)miRNA在所有EBV阳性肿瘤中表达,抑制裂解感染和宿主细胞凋亡。我们鉴定了辛二酰苯胺异羟肟酸(SAHA),组蛋白脱乙酰酶的抑制剂,作为抑制BART启动子活性和BARTmiRNA转录的试剂。与EBV阴性细胞相比,SAHA治疗对EBV阳性细胞的细胞增殖具有更明显的抑制作用,影响p53野生型和突变型胃上皮细胞。SAHA治疗增强了野生型EBV感染细胞的裂解感染,同时还增强BZLF1缺陷型EBV感染细胞的细胞死亡。它使BART基因表达减少了85%,并增加了BARTmiRNA靶向的促凋亡因子的表达。这些发现表明,SAHA不仅诱导裂解感染,而且通过抑制BARTmiRNA转录和促进凋亡程序导致细胞死亡。
    Epstein-Barr virus (EBV) is linked to lymphoma and epithelioma but lacks drugs specifically targeting EBV-positive tumors. BamHI A Rightward Transcript (BART) miRNAs are expressed in all EBV-positive tumors, suppressing both lytic infection and host cell apoptosis. We identified suberoylanilide hydroxamic acid (SAHA), an inhibitor of histone deacetylase enzymes, as an agent that suppresses BART promoter activity and transcription of BART miRNAs. SAHA treatment demonstrated a more pronounced inhibition of cell proliferation in EBV-positive cells compared to EBV-negative cells, affecting both p53 wild-type and mutant gastric epithelial cells. SAHA treatment enhanced lytic infection in wild-type EBV-infected cells, while also enhancing cell death in BZLF1-deficient EBV-infected cells. It reduced BART gene expression by 85% and increased the expression of proapoptotic factors targeted by BART miRNAs. These findings suggest that SAHA not only induces lytic infection but also leads to cell death by suppressing BART miRNA transcription and promoting the apoptotic program.
    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
    蛋白质错误折叠和错误定位在家族性和偶发性肌萎缩性侧索硬化症(ALS)中很常见。通过诱导热休克蛋白(HSP)以增加伴侣能力来维持蛋白质平衡是治疗ALS的合理治疗策略。然而,上调应激诱导HSP的阈值在神经元中仍然很高,提供治疗障碍。这项研究使用表达ALS变体FUSR521G或SOD1G93A的小鼠模型来跟踪先前在培养的运动神经元中的工作,显示HSP共诱导剂的不同作用,Arimoclomol,和I类组蛋白脱乙酰酶(HDAC)抑制剂对HSP表达的影响取决于所表达的ALS变体。就像培养的神经元一样,无论是转基因的表达还是药物治疗都不诱导皮质中HSPs的表达,FUSR521G小鼠的脊髓或肌肉,表示热休克反应的抑制。尽管如此,Arimoclomol,和RGFP963,恢复了认知测试的表现,并改善了皮质树突脊柱密度。在SOD1G93A小鼠中,多个HSP在后肢骨骼肌中上调,但腰脊髓中没有,HSPB1与星形细胞增多相关。药物治疗改善了收缩力,但减少了肌肉中HSP的增加,而不是促进其表达。数据表明,除了通过Arimoclomol和I类HDAC抑制ALS-FUS小鼠认知功能恢复的热休克反应的放大之外,还表明了对抗ALS认知障碍的潜在益处。额颞叶痴呆和相关疾病。
    Protein misfolding and mislocalization are common to both familial and sporadic forms of amyotrophic lateral sclerosis (ALS). Maintaining proteostasis through induction of heat shock proteins (HSP) to increase chaperoning capacity is a rational therapeutic strategy in the treatment of ALS. However, the threshold for upregulating stress-inducible HSPs remains high in neurons, presenting a therapeutic obstacle. This study used mouse models expressing the ALS variants FUSR521G or SOD1G93A to follow up on previous work in cultured motor neurons showing varied effects of the HSP co-inducer, arimoclomol, and class I histone deacetylase (HDAC) inhibitors on HSP expression depending on the ALS variant being expressed. As in cultured neurons, neither expression of the transgene nor drug treatments induced expression of HSPs in cortex, spinal cord or muscle of FUSR521G mice, indicating suppression of the heat shock response. Nonetheless, arimoclomol, and RGFP963, restored performance on cognitive tests and improved cortical dendritic spine densities. In SOD1G93A mice, multiple HSPs were upregulated in hindlimb skeletal muscle, but not in lumbar spinal cord with the exception of HSPB1 associated with astrocytosis. Drug treatments improved contractile force but reduced the increase in HSPs in muscle rather than facilitating their expression. The data point to mechanisms other than amplification of the heat shock response underlying recovery of cognitive function in ALS-FUS mice by arimoclomol and class I HDAC inhibition and suggest potential benefits in counteracting cognitive impairment in ALS, frontotemporal dementia and related disorders.
    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:组蛋白脱乙酰酶(iHDACs)抑制剂是治疗神经退行性疾病的有前景的药物。我们已经评估了新的iHDACLASSBio-1911在Aβ寡聚体(AβO)毒性模型和星形胶质细胞中的治疗潜力,神经炎症和阿尔茨海默病(AD)的关键参与者。
    方法:通过流式细胞术评估星形胶质细胞表型和突触密度,西方印迹,免疫荧光和qPCR,在体外和小鼠中。使用脑室内输注AβO的小鼠模型通过行为测定来评估认知功能。
    结果:LASSBio-1911调节培养的星形胶质细胞的反应性和突触形成潜能,并改善培养的神经元和小鼠的突触标记。它可以防止AβO触发的小鼠星形胶质细胞反应性,并增强星形胶质细胞的神经保护潜力。LASSBio-1911改善AβO输注小鼠的行为表现并挽救突触和记忆功能。
    结论:这些结果有助于揭示星形胶质细胞在AD中的潜在作用机制,并为使用星形胶质细胞作为AD新药的靶标提供了理论基础。
    OBJECTIVE: Inhibitors of histone deacetylases (iHDACs) are promising drugs for neurodegenerative diseases. We have evaluated the therapeutic potential of the new iHDAC LASSBio-1911 in Aβ oligomer (AβO) toxicity models and astrocytes, key players in neuroinflammation and Alzheimer\'s disease (AD).
    METHODS: Astrocyte phenotype and synapse density were evaluated by flow cytometry, Western blotting, immunofluorescence and qPCR, in vitro and in mice. Cognitive function was evaluated by behavioural assays using a mouse model of intracerebroventricular infusion of AβO.
    RESULTS: LASSBio-1911 modulates reactivity and synaptogenic potential of cultured astrocytes and improves synaptic markers in cultured neurons and in mice. It prevents AβO-triggered astrocytic reactivity in mice and enhances the neuroprotective potential of astrocytes. LASSBio-1911 improves behavioural performance and rescues synaptic and memory function in AβO-infused mice.
    CONCLUSIONS: These results contribute to unveiling the mechanisms underlying astrocyte role in AD and provide the rationale for using astrocytes as targets to new drugs for AD.
    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
    胰腺导管腺癌(PDAC)是胰腺癌的最致命的形式,其特征是治疗抵抗和早期转移。导致存活率低。组蛋白去乙酰化酶(HDAC)抑制剂显示出治疗血液恶性肿瘤的潜力。在PDAC中,HDAC2的过表达与上皮-间质转化(EMT)有关,主要伴随着上皮标志物E-cadherin的下调和转移能力的增加。已知效应细胞因子转化生长因子-β(TGFβ)是PDAC中EMT的主要诱导因子,导致高转移和侵袭潜力。此外,PDAC中HDAC6的过表达与细胞凋亡减少有关。这里,我们已经证明,一种新型HDAC2/6抑制剂不仅显著增加了PANC-1细胞(5.5倍)和3DPDAC共培养球体(2.5倍)中E-cadherin的表达,而且能够逆转TGF-β诱导的E-cadherin表达下调.此外,我们的研究表明,与对照组相比,HDAC抑制剂介导的再分化导致肿瘤细胞侵袭的显著抑制约60%.特别是,我们已经证明HDAC抑制剂诱导细胞凋亡(2倍)和细胞周期停滞.总之,HDAC2/6抑制剂通过上调HDAC2阻断介导的E-cadherin来抑制侵袭,并通过HDAC6抑制诱导细胞周期停滞导致细胞凋亡。这些结果表明,HDAC2/6抑制剂可能代表了治疗PDAC肿瘤发生和转移的新治疗策略。
    Pancreatic ductal adenocarcinoma (PDAC) is the most lethal form of pancreatic cancer characterized by therapy resistance and early metastasis, resulting in a low survival rate. Histone deacetylase (HDAC) inhibitors showed potential for the treatment of hematological malignancies. In PDAC, the overexpression of HDAC 2 is associated with the epithelial-mesenchymal transition (EMT), principally accompanied by the downregulation of the epithelial marker E-cadherin and increased metastatic capacity. The effector cytokine transforming growth factor-β (TGF β) is known to be a major inducer of the EMT in PDAC, leading to high metastatic and invasive potential. In addition, the overexpression of HDAC 6 in PDAC is associated with reduced apoptosis. Here, we have demonstrated that a novel HDAC 2/6 inhibitor not only significantly increased E-cadherin expression in PANC-1 cells (5.5-fold) and in 3D PDAC co-culture spheroids (2.5-fold) but was also able to reverse the TGF-β-induced downregulation of E-cadherin expression. Moreover, our study indicates that the HDAC inhibitor mediated re-differentiation resulting in a significant inhibition of tumor cell invasion by approximately 60% compared to control. In particular, we have shown that the HDAC inhibitor induces both apoptosis (2-fold) and cell cycle arrest. In conclusion, the HDAC 2/6 inhibitor acts by suppressing invasion via upregulating E-cadherin mediated by HDAC 2 blockade and by inducing cell cycle arrest leading to apoptosis via HDAC 6 inhibition. These results suggest that the HDAC 2/6 inhibitor might represent a novel therapeutic strategy for the treatment of PDAC tumorigenesis and metastasis.
    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
    小胶质细胞,中枢神经系统(CNS)中的固有免疫细胞,在维持中枢神经系统稳态中起关键作用。然而,响应脑损伤激活的小胶质细胞产生各种炎症介质,包括一氧化氮(NO)和促炎细胞因子,导致相当大的神经元损伤。诱导型NO合酶(iNOS)产生的NO与超氧化物迅速反应形成剧毒产物,过氧亚硝酸盐.因此,iNOS被认为是脑缺血的推定治疗靶点。这里,我们检查了帕比司他(Pano)的作用,组蛋白脱乙酰酶抑制剂,使用大鼠永生化小胶质细胞HAPI细胞对脂多糖(LPS)诱导的iNOS表达的影响。Pano以剂量依赖的方式抑制LPS诱导的iNOSmRNA表达和NO产生;然而,它对LPS诱导的c-Jun氨基末端激酶(JNK)和p38的激活或核因子-κB(NF-κB)的核易位影响很小。干扰素-β(IFN-β)/信号转导和转录激活因子(STAT)途径对于LPS诱导的巨噬细胞/小胶质细胞中iNOS的表达至关重要。我们还检查了Pano对LPS诱导的IFN-β信号传导的影响。Pano显著抑制LPS诱导的IFN-β表达和随后的STAT1酪氨酸磷酸化。然而,IFN-β的添加恢复了降低的STAT1磷酸化,但没有降低的iNOS表达。此外,Pano抑制了LPS增加的八聚体结合蛋白2和干扰素调节因子9的表达,但是IFN-β的添加也未能恢复这些因子的表达降低。因此,我们得出的结论是,Pano的抑制作用不仅归因于IFN-β/STAT轴的抑制,而且还归因于该轴未涉及的其他因子的下调。
    Microglia, resident immune cells in the central nervous system (CNS), play a critical role in maintaining CNS homeostasis. However, microglia activated in response to brain injury produce various inflammatory mediators, including nitric oxide (NO) and proinflammatory cytokines, leading to considerable neuronal damage. NO generated by inducible NO synthase (iNOS) rapidly reacts with superoxide to form a highly toxic product, peroxynitrite. Therefore, iNOS is considered to be a putative therapeutic target for cerebral ischemia. Here, we examined the effects of panobinostat (Pano), a histone deacetylase inhibitor, on lipopolysaccharide (LPS)-induced iNOS expression using rat immortalized microglia HAPI cells. Pano inhibited LPS-induced expression of iNOS mRNA and NO production in a dose-dependent manner; however, it had little effect on the LPS-induced activation of c-Jun N-terminal kinase (JNK) and p38 or nuclear translocation of nuclear factor-κB (NF-κB). The interferon-β (IFN-β)/signal transducer and activator of transcription (STAT) pathway is essential for LPS-induced iNOS expression in macrophages/microglia. We also examined the effects of Pano on LPS-induced IFN-β signaling. Pano markedly inhibited LPS-induced IFN-β expression and subsequent tyrosine phosphorylation of STAT1. However, the addition of IFN-β restored the decreased STAT1 phosphorylation but not the decreased iNOS expression. In addition, Pano inhibited the LPS-increased expression of octamer binding protein-2 and interferon regulatory factor 9 responsible for iNOS expression, but IFN-β addition also failed to restore the decreased expression of these factors. Thus, we conclude that the inhibitory effects of Pano are due not only to the inhibition of the IFN-β/STAT axis but also to the downregulation of other factors not involved in this axis.
    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
    Schistosomiasis is a neglected zoonotic parasitic disease. Currently, praziquantel is the drug of choice for the treatment of schistosomiasis, and is the only effective chemical for treatment of schistosomiasis japonica. Since its introduction in the 1970s, praziquantel has been used for large-scale chemotherapy of schistosomiasis for over 40 years. However, there have been reports pertaining to the resistance to praziquantel in schistosomes. Therefore, development of novel antischistosomal agents as alternatives of praziquantel, is of great need. Histone deacetylases and histone acetyltransferases have been recently reported to play critical roles in the growth, development and reproduction of schistosomes, and are considered as potential drug targets for the treatment of schistosomiasis. This review summarizes the latest advances of histone deacetylase and histone acetyltransferase inhibitors in the research on antischistosomal drugs, so as to provide insights into research and development of novelantischistosomal agents.
    [摘要] 血吸虫病是一种被忽视的人兽共患寄生虫病。目前, 吡喹酮是治疗血吸虫病的首选药物, 也是治疗日本血吸虫 病的唯一有效药物。自 20 世纪 70 年代问世以来, 吡喹酮已在大规模血吸虫病化疗中应用了 40 余年, 长期大规模使用导 致部分地区血吸虫对其产生了抗性, 因此迫切需要研发新型抗血吸虫药物作为候选替代药物。近期研究表明, 组蛋白去 乙酰化酶和组蛋白乙酰转移酶在血吸虫的生长发育以及繁殖等阶段中均起关键作用, 被认为是治疗血吸虫病的潜在药 物靶点。本文总结了组蛋白去乙酰化酶抑制剂与组蛋白乙酰转移酶抑制剂在抗血吸虫药物研究中的最新进展, 以期为 抗血吸虫新药的研发提供参考。.
    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