Histone Deacetylase 1

组蛋白去乙酰化酶 1
  • 文章类型: Journal Article
    急性髓细胞性白血病(AML)是一种以血液和骨髓浸润为特征的血液恶性肿瘤,缓解率低,复发率高。目前的研究表明,I类HDAC抑制剂可以下调抗凋亡蛋白,导致AML细胞凋亡。在目前的调查中,我们对海洋细胞毒素SantacruzamateA(SCA)进行了结构修饰,一种以其对HDAC的抑制活性而闻名的化合物,导致开发了一系列新型有效的I类HDACs酰肼抑制剂。代表性的基于酰肼的化合物25c表现出作为单一药剂的AML细胞中的细胞凋亡的浓度依赖性诱导。此外,25c与维奈托克联用时表现出协同抗AML作用,用于AML治疗的临床Bcl-2抑制剂。这种组合导致抗凋亡蛋白Mcl-1和Bcl-xL更明显的下调,与单一疗法相比,促凋亡蛋白caspase3和DNA双链断裂生物标志物γ-H2AX的显着上调。这些结果强调了25c作为AML治疗的有希望的先导化合物的潜力。特别是与维奈托克联合使用时。
    Acute myeloid leukemia (AML) is a hematologic malignancy characterized by infiltration of the blood and bone marrow, exhibiting a low remission rate and high recurrence rate. Current research has demonstrated that class I HDAC inhibitors can downregulate anti-apoptotic proteins, leading to apoptosis of AML cells. In the present investigation, we conducted structural modifications of marine cytotoxin Santacruzamate A (SCA), a compound known for its inhibitory activity towards HDACs, resulting in the development of a novel series of potent class I HDACs hydrazide inhibitors. Representative hydrazide-based compound 25c exhibited concentration-dependent induction of apoptosis in AML cells as a single agent. Moreover, 25c exhibited a synergistic anti-AML effect when combined with Venetoclax, a clinical Bcl-2 inhibitor employed in AML therapy. This combination resulted in a more pronounced downregulation of anti-apoptotic proteins Mcl-1 and Bcl-xL, along with a significant upregulation of the pro-apoptotic protein cleaved-caspase3 and the DNA double-strand break biomarker γ-H2AX compared to monotherapy. These results highlighted the potential of 25c as a promising lead compound for AML treatment, particularly when used in combination with Venetoclax.
    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1(HDAC1),一类HDAC酶,对于组蛋白修饰至关重要。目前,它已成为通过癌症表观遗传学设计小分子药物的重要生物学靶标之一。与合成抑制剂一起,不同的天然抑制剂显示出潜在的HDAC1抑制作用。为了深入了解天然抑制剂和HDAC1的分子结构之间的关系,不同的分子建模技术(贝叶斯分类,递归分区,分子对接和分子动力学模拟)已应用于155种具有不同支架的HDAC1自然启发抑制剂的数据集。贝叶斯研究显示了训练集和测试集两者的可接受的ROC值。递归分区研究产生了具有6片叶子的决策树1。Further,进行分子对接研究以生成蛋白质配体复合物,该复合物鉴定了一些潜在的氨基酸残基,例如F205,H28,L271,P29,F150,Y204,用于在天然抑制剂的情况下的结合相互作用。还通过分子动力学模拟研究评估了这些HDAC1-天然抑制剂复合物的稳定性。当前的建模研究试图深入了解调节HDAC1抑制的不同天然化合物之间的不同重要结构指纹。由RamaswamyH.Sarma沟通。
    Histone deacetylase 1 (HDAC1), a class I HDAC enzyme, is crucial for histone modification. Currently, it is emerged as one of the important biological targets for designing small molecule drugs through cancer epigenetics. Along with synthetic inhibitors different natural inhibitors are showing potential HDAC1 inhibitions. In order to gain insights into the relationship between the molecular structures of the natural inhibitors and HDAC1, different molecular modelling techniques (Bayesian classification, recursive partitioning, molecular docking and molecular dynamics simulations) have been applied on a dataset of 155 HDAC1 nature-inspired inhibitors with diverse scaffolds. The Bayesian study showed acceptable ROC values for both the training set and test sets. The Recursive partitioning study produced decision tree 1 with 6 leaves. Further, molecular docking study was processed for generating the protein ligand complex which identified some potential amino acid residues such as F205, H28, L271, P29, F150, Y204 for the binding interactions in case of natural inhibitors. Stability of these HDAC1-natutal inhibitors complexes has been also evaluated by molecular dynamics simulation study. The current modelling study is an attempt to get a deep insight into the different important structural fingerprints among different natural compounds modulating HDAC1 inhibition.Communicated by Ramaswamy H. Sarma.
    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
    妊娠期亚临床甲状腺功能减退症(SCH)是妊娠期甲状腺功能障碍的最常见形式,会影响胎儿神经系统发育,增加出生后神经发育障碍的风险。然而,母体亚临床甲状腺功能减退症对胎儿脑发育和行为表型影响的机制尚不清楚,需要进一步研究。在这项研究中,我们通过在怀孕期间将母猪暴露于含有50ppm丙基硫氧嘧啶(PTU)的饮用水中,构建了母体亚临床甲状腺功能减退症小鼠模型,并通过行为测试发现其后代伴有严重的认知缺陷.机械上,妊娠期SCH导致子代海马中HDAC1/2/3的蛋白表达和活性上调。ChIP分析显示,神经颗粒蛋白(Ng)启动子上的H3K9ac在SCH后代的海马中减少,随着Ng蛋白的显著减少,导致突触可塑性标志物PSD95(突触后密度中的膜相关蛋白)和SYN(突触素,突触前末端的特定标记),突触可塑性受损.此外,MS-275(HDAC1/2/3特异性抑制剂)对SCH后代的给药减轻了后代的突触可塑性受损和认知功能障碍。因此,我们的研究提示,母体亚临床甲状腺功能减退症可能通过HDAC1/2/3-H3K9ac-Ng通路介导后代认知功能障碍.我们的研究有助于理解母体亚临床甲状腺功能减退症介导的后代认知障碍的信号机制。
    Subclinical hypothyroidism (SCH) in pregnancy is the most common form of thyroid dysfunction in pregnancy, which can affect fetal nervous system development and increase the risk of neurodevelopmental disorders after birth. However, the mechanism of the effect of maternal subclinical hypothyroidism on fetal brain development and behavioral phenotypes is still unclear and requires further study. In this study, we constructed a mouse model of maternal subclinical hypothyroidism by exposing dams to drinking water containing 50 ppm propylthiouracil (PTU) during pregnancy and found that its offspring were accompanied by severe cognitive deficits by behavioral testing. Mechanistically, gestational SCH resulted in the upregulation of protein expression and activity of HDAC1/2/3 in the hippocampus of the offspring. ChIP analysis revealed that H3K9ac on the neurogranin (Ng) promoter was reduced in the hippocampus of the offspring of SCH, with a significant reduction in Ng protein, leading to reduced expression levels of synaptic plasticity markers PSD95 (a membrane-associated protein in the postsynaptic density) and SYN (synaptophysin, a specific marker for presynaptic terminals), and impaired synaptic plasticity. In addition, administration of MS-275 (an HDAC1/2/3-specific inhibitor) to SCH offspring alleviated impaired synaptic plasticity and cognitive dysfunction in offspring. Thus, our study suggests that maternal subclinical hypothyroidism may mediate offspring cognitive dysfunction through the HDAC1/2/3-H3K9ac-Ng pathway. Our study contributes to the understanding of the signaling mechanisms underlying maternal subclinical hypothyroidism-mediated cognitive impairment in the offspring.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • DOI:
    文章类型: Journal Article
    PRAME(黑色素瘤优先表达的抗原)的上调与多种癌症的进展有关。包括黑色素瘤.肿瘤抑制因子p53是一种转录调节因子,其响应于应激信号而介导细胞周期停滞和细胞凋亡。这里,我们报道PRAME是一种新的p53抑制靶点。这得到了对携带野生型p53的黑素瘤细胞系和人黑素瘤数据库的分析的支持。PRAME的mRNA表达因p53过表达和使用DNA损伤剂的激活而下调,但通过p53耗竭上调。我们在PRAME的启动子区域中鉴定了p53反应元件(p53RE)。荧光素酶和ChIP分析显示p53抑制PRAME启动子的转录活性,并在依托泊苷处理后与HDAC1一起募集到p53RE。通过测量黑色素瘤细胞中的集落形成和p27表达证明了p53激活介导的PRAME下调的功能意义。这些数据表明p53激活,导致PRAME下调,可能是黑色素瘤细胞的治疗策略。
    Upregulation of PRAME (preferentially expressed antigen of melanoma) has been implicated in the progression of a variety of cancers, including melanoma. The tumor suppressor p53 is a transcriptional regulator that mediates cell cycle arrest and apoptosis in response to stress signals. Here, we report that PRAME is a novel repressive target of p53. This was supported by analysis of melanoma cell lines carrying wild-type p53 and human melanoma databases. mRNA expression of PRAME was downregulated by p53 overexpression and activation using DNA-damaging agents, but upregulated by p53 depletion. We identified a p53-responsive element (p53RE) in the promoter region of PRAME. Luciferase and ChIP assays showed that p53 represses the transcriptional activity of the PRAME promoter and is recruited to the p53RE together with HDAC1 upon etoposide treatment. The functional significance of p53 activationmediated PRAME downregulation was demonstrated by measuring colony formation and p27 expression in melanoma cells. These data suggest that p53 activation, which leads to PRAME downregulation, could be a therapeutic strategy in melanoma cells. [BMB Reports 2024; 57(6): 299-304].
    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
    类风湿性关节炎(RA)是一种全身性免疫介导的疾病,其特征是关节炎症和破坏。这种疾病通常会影响手和脚的小关节,后来发展到涉及更大的关节,如膝盖,肩膀,和臀部。虽然这些联合特定差异的原因尚不清楚,已经报道了与关节位置相关的不同表观遗传模式。在这项研究中,我们评估了RA患者髋关节和膝关节滑膜成纤维细胞样滑膜细胞(FLS)的独特表观遗传景观,重点研究同源盒(HOX)转录因子的表达和调控。这些高度保守的基因在胚胎发育中起关键作用,并且已知在各种成体组织中维持不同的表达模式。我们发现几个HOX基因,尤其是HOXD10,与髋部FLS相比,在膝关节FLS中差异表达。在膝关节和髋关节FLS之间的HOXD10启动子中观察到染色质可及性和组蛋白标记的表观遗传差异。组蛋白修改,特别是组蛋白乙酰化,被鉴定为HOXD10表达的重要调控因子。为了理解HOXD10差异表达的机制,我们用小分子和siRNA抑制组蛋白脱乙酰酶(HDACs)。我们发现HDAC1阻断或缺乏使关节特异性HOXD10表达模式正常化。这些观察表明,表观遗传差异,特别是与HDAC1表达增加相关的组蛋白乙酰化,在关节特异性HOXD10表达中起关键作用。了解这些机制可以提供对RA的区域方面的见解,并可能导致针对疾病过程中特定关节参与模式的治疗策略。
    Rheumatoid arthritis (RA) is a systemic immune-mediated disease characterized by joint inflammation and destruction. The disease typically affects small joints in the hands and feet, later progressing to involve larger joints such as the knees, shoulders, and hips. While the reasons for these joint-specific differences are unclear, distinct epigenetic patterns associated with joint location have been reported. In this study, we evaluated the unique epigenetic landscapes of fibroblast-like synoviocytes (FLS) from hip and knee synovium in RA patients, focusing on the expression and regulation of Homeobox (HOX) transcription factors. These highly conserved genes play a critical role in embryonic development and are known to maintain distinct expression patterns in various adult tissues. We found that several HOX genes, especially HOXD10, were differentially expressed in knee FLS compared with hip FLS. Epigenetic differences in chromatin accessibility and histone marks were observed in HOXD10 promoter between knee and hip FLS. Histone modification, particularly histone acetylation, was identified as an important regulator of HOXD10 expression. To understand the mechanism of differential HOXD10 expression, we inhibited histone deacetylases (HDACs) with small molecules and siRNA. We found that HDAC1 blockade or deficiency normalized the joint-specific HOXD10 expression patterns. These observations suggest that epigenetic differences, specifically histone acetylation related to increased HDAC1 expression, play a crucial role in joint-specific HOXD10 expression. Understanding these mechanisms could provide insights into the regional aspects of RA and potentially lead to therapeutic strategies targeting specific patterns of joint involvement during the course of disease.
    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
    树突状细胞(DC)祖细胞在发育过程中适应其转录程序,生成不同的子集。染色质修饰如何调节这些过程尚不清楚。这里,我们通过基因删除造血祖细胞和CD11c表达细胞中的组蛋白去乙酰化酶1(HDAC1)或HDAC2,研究组蛋白去乙酰化对DCs的影响。虽然HDAC2对DC开发并不重要,HDAC1缺失会损害pro-pDC和成熟pDC的生成,并影响ESAM+cDC2与tDC和pre-cDC2的分化,而cDC1s不变。人类造血细胞中的HDAC1敲低也会损害cDC2的发育,强调它在物种中的关键作用。多组学分析显示HDAC1控制表达,染色质可及性,和cDC2亚群有效发育所需的转录因子IRF4,IRF8和SPIB的组蛋白乙酰化。没有HDAC1,DCs会在免疫上转换,通过增加cDC1成熟和白细胞介素12的产生来加强肿瘤监测,驱动T辅助1介导的免疫和CD8+T细胞募集。我们的研究揭示了组蛋白乙酰化在DC发育和抗肿瘤免疫中的重要性,提示免疫肿瘤学的DC靶向治疗策略。
    Dendritic cell (DC) progenitors adapt their transcriptional program during development, generating different subsets. How chromatin modifications modulate these processes is unclear. Here, we investigate the impact of histone deacetylation on DCs by genetically deleting histone deacetylase 1 (HDAC1) or HDAC2 in hematopoietic progenitors and CD11c-expressing cells. While HDAC2 is not critical for DC development, HDAC1 deletion impairs pro-pDC and mature pDC generation and affects ESAM+cDC2 differentiation from tDCs and pre-cDC2s, whereas cDC1s are unchanged. HDAC1 knockdown in human hematopoietic cells also impairs cDC2 development, highlighting its crucial role across species. Multi-omics analyses reveal that HDAC1 controls expression, chromatin accessibility, and histone acetylation of the transcription factors IRF4, IRF8, and SPIB required for efficient development of cDC2 subsets. Without HDAC1, DCs switch immunologically, enhancing tumor surveillance through increased cDC1 maturation and interleukin-12 production, driving T helper 1-mediated immunity and CD8+ T cell recruitment. Our study reveals the importance of histone acetylation in DC development and anti-tumor immunity, suggesting DC-targeted therapeutic strategies for immuno-oncology.
    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
    背景:肝内胆管癌(ICCA)是一类异质性恶性肿瘤,其特点是复发率高,预后差。异染色质蛋白1α(HP1α)是通过异染色质形成和结构维持参与转录沉默的最重要的非组蛋白染色体蛋白之一。HP1α对ICCA进展的影响尚不清楚。
    方法:通过在两种细胞系和两种ICCA小鼠模型中的实验来检测对ICCA增殖的影响。使用电喷雾电离质谱(ESI-MS)确定HP1α与组蛋白脱乙酰酶1(HDAC1)之间的相互作用,并使用免疫沉淀测定(co-IP)研究结合机制。通过RNA测序(RNA-seq)筛选出目标基因。通过生物信息学方法预测DNA结合蛋白和组蛋白修饰的占用,并通过靶标和标签下的裂解(CUT&Tag)和染色质免疫沉淀(ChIP)进行评估。
    结果:HP1α在肝内胆管癌(ICCA)组织中上调,并通过抑制干扰素途径,以信号转导和转录激活因子1(STAT1)依赖性方式调节ICCA细胞的增殖。机械上,STAT1由HP1α-HDAC1复合物直接和表观遗传地通过启动子结合和不同组蛋白修饰的变化进行转录调节,通过高通量测序验证。广谱HDAC抑制剂(HDACi)通过下调HP1α和靶向异源二聚体激活干扰素途径并抑制ICCA细胞的增殖。发现广谱HDACi加干扰素制备方案可改善体内和体外的抗增殖作用并延迟ICCA的发展,它利用了基础激活以及干扰素途径的直接激活。HP1α参与介导对两种试剂的细胞抗性。
    结论:HP1α-HDAC1复合物通过在转录水平上直接和表观调节STAT1影响干扰素途径的激活。广谱HDACi加干扰素制剂方案抑制ICCA发展,为ICCA治疗提供可行的策略。靶向HP1α-HDAC1-STAT1轴是治疗ICCA的可能策略,尤其是HP1α阳性病例。
    BACKGROUND: Intrahepatic cholangiocarcinoma (ICCA) is a heterogeneous group of malignant tumors characterized by high recurrence rate and poor prognosis. Heterochromatin Protein 1α (HP1α) is one of the most important nonhistone chromosomal proteins involved in transcriptional silencing via heterochromatin formation and structural maintenance. The effect of HP1α on the progression of ICCA remained unclear.
    METHODS: The effect on the proliferation of ICCA was detected by experiments in two cell lines and two ICCA mouse models. The interaction between HP1α and Histone Deacetylase 1 (HDAC1) was determined using Electrospray Ionization Mass Spectrometry (ESI-MS) and the binding mechanism was studied using immunoprecipitation assays (co-IP). The target gene was screened out by RNA sequencing (RNA-seq). The occupation of DNA binding proteins and histone modifications were predicted by bioinformatic methods and evaluated by Cleavage Under Targets and Tagmentation (CUT & Tag) and Chromatin immunoprecipitation (ChIP).
    RESULTS: HP1α was upregulated in intrahepatic cholangiocarcinoma (ICCA) tissues and regulated the proliferation of ICCA cells by inhibiting the interferon pathway in a Signal Transducer and Activator of Transcription 1 (STAT1)-dependent manner. Mechanistically, STAT1 is transcriptionally regulated by the HP1α-HDAC1 complex directly and epigenetically via promoter binding and changes in different histone modifications, as validated by high-throughput sequencing. Broad-spectrum HDAC inhibitor (HDACi) activates the interferon pathway and inhibits the proliferation of ICCA cells by downregulating HP1α and targeting the heterodimer. Broad-spectrum HDACi plus interferon preparation regimen was found to improve the antiproliferative effects and delay ICCA development in vivo and in vitro, which took advantage of basal activation as well as direct activation of the interferon pathway. HP1α participates in mediating the cellular resistance to both agents.
    CONCLUSIONS: HP1α-HDAC1 complex influences interferon pathway activation by directly and epigenetically regulating STAT1 in transcriptional level. The broad-spectrum HDACi plus interferon preparation regimen inhibits ICCA development, providing feasible strategies for ICCA treatment. Targeting the HP1α-HDAC1-STAT1 axis is a possible strategy for treating ICCA, especially HP1α-positive cases.
    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
    雌激素受体阴性[ER(-)]乳腺癌是最具侵袭性的乳腺癌(BC)类型,具有较高的转移和复发率。近年来,饮食预防BC与表观遗传活性植物化学物质受到越来越多的关注,由于其可行性,有效性,和易于实施。在这方面,组合植物化学干预通过同时靶向多个致瘤途径来实现更有效的BC抑制。我们,因此,重点研究了富含萝卜硫素(SFN)的西兰花芽(BSp)和富含维生素A(WA)的Ashwagandha(Ash)组合对雌激素受体阴性[ER(-)]乳腺癌的BC预防的影响使用转基因小鼠。我们的结果表明,与对照治疗相比,组合BSp+Ash治疗显着降低了肿瘤发生率和肿瘤生长(〜75%)以及延迟(〜21%)的肿瘤潜伏期,并且组合BSp+Ash治疗在统计学上更有效地抑制BC与单一BSp或Ash干预相比。在分子水平上,BSp和Ash组合上调肿瘤抑制因子(p53,p57)以及凋亡相关蛋白(BAX,PUMA)和BAX:BCL-2比率。此外,我们的结果表明,由于联合治疗,表观遗传机制HDAC1和DNMT3A在乳腺肿瘤组织中的表达下降。有趣的是,我们报道了BSp和Ash之间的多种协同相互作用,由于BSp和Ash的联合治疗,这些相互作用影响了肿瘤表型和分子表达.我们的RNA-seq分析结果还证明了与多个细胞信号通路相关的基因的全转录组表达重组。由于BSp和Ash联合施用,转录因子活性和表观遗传调控。此外,我们发现,由于组合治疗,肠道微生物组成的改变。总的来说,联合补充BSp和Ash可以通过增强肿瘤抑制来预防ER(-)BC,凋亡诱导和基因表达的全转录组重组可能影响多个细胞信号通路,表观遗传调控与肠道微生物群重塑。
    Estrogen receptor-negative [ER(-)] mammary cancer is the most aggressive type of breast cancer (BC) with higher rate of metastasis and recurrence. In recent years, dietary prevention of BC with epigenetically active phytochemicals has received increased attention due to its feasibility, effectiveness, and ease of implementation. In this regard, combinatorial phytochemical intervention enables more efficacious BC inhibition by simultaneously targeting multiple tumorigenic pathways. We, therefore, focused on investigation of the effect of sulforaphane (SFN)-rich broccoli sprouts (BSp) and withaferin A (WA)-rich Ashwagandha (Ash) combination on BC prevention in estrogen receptor-negative [ER(-)] mammary cancer using transgenic mice. Our results indicated that combinatorial BSp + Ash treatment significantly reduced tumor incidence and tumor growth (~ 75%) as well as delayed (~ 21%) tumor latency when compared to the control treatment and combinatorial BSp + Ash treatment was statistically more effective in suppressing BC compared to single BSp or Ash intervention. At the molecular level, the BSp and Ash combination upregulated tumor suppressors (p53, p57) along with apoptosis associated proteins (BAX, PUMA) and BAX:BCL-2 ratio. Furthermore, our result indicated an expressional decline of epigenetic machinery HDAC1 and DNMT3A in mammary tumor tissue because of combinatorial treatment. Interestingly, we have reported multiple synergistic interactions between BSp and Ash that have impacted both tumor phenotype and molecular expression due to combinatorial BSp and Ash treatment. Our RNA-seq analysis results also demonstrated a transcriptome-wide expressional reshuffling of genes associated with multiple cell-signaling pathways, transcription factor activity and epigenetic regulations due to combined BSp and Ash administration. In addition, we discovered an alteration of gut microbial composition change because of combinatorial treatment. Overall, combinatorial BSp and Ash supplementation can prevent ER(-) BC through enhanced tumor suppression, apoptosis induction and transcriptome-wide reshuffling of gene expression possibly influencing multiple cell signaling pathways, epigenetic regulation and reshaping gut microbiota.
    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
    PA28γ过表达是异常的,并伴随着各种癌症患者的不良预后,这个关键基因在肿瘤微环境中的精确调控机制仍不完全清楚。在这项研究中,以口腔鳞状细胞癌为模型,我们证明了PA28γ在癌症相关成纤维细胞(CAF)中的高表达,其表达与恶性肿瘤临床指标的严重程度显著相关。值得注意的是,我们发现PA28γ+CAFs分泌的IGF2水平升高可以通过激活MAPK/AKT通路以旁分泌方式增强干细胞维持和促进肿瘤细胞侵袭性。机械上,PA28γ通过稳定E2F3蛋白上调IGF2表达,IGF2的转录因子。进一步的机制见解表明,HDAC1主要介导E2F3的去乙酰化和随后的泛素化和降解。值得注意的是,PA28γ与HDAC1相互作用并通过20S蛋白酶体依赖性途径加速其降解。此外,PA28γ+CAFs通过分泌IGF2对肿瘤免疫微环境产生影响。令人兴奋的是,我们的研究提示,靶向PA28γ+CAFs或分泌的IGF2可以提高PD-L1治疗的疗效.因此,我们的研究结果揭示了PA28γ在肿瘤微环境中细胞相互作用中的关键作用,并提出了增强口腔鳞状细胞癌免疫检查点阻断有效性的新策略.
    PA28γ overexpression is aberrant and accompanied by poor patient prognosis in various cancers, the precise regulatory mechanism of this crucial gene in the tumor microenvironment remains incompletely understood. In this study, using oral squamous cell carcinoma as a model, we demonstrated that PA28γ exhibits high expression in cancer-associated fibroblasts (CAFs), and its expression significantly correlates with the severity of clinical indicators of malignancy. Remarkably, we found that elevated levels of secreted IGF2 from PA28γ+ CAFs can enhance stemness maintenance and promote tumor cell aggressiveness through the activation of the MAPK/AKT pathway in a paracrine manner. Mechanistically, PA28γ upregulates IGF2 expression by stabilizing the E2F3 protein, a transcription factor of IGF2. Further mechanistic insights reveal that HDAC1 predominantly mediates the deacetylation and subsequent ubiquitination and degradation of E2F3. Notably, PA28γ interacts with HDAC1 and accelerates its degradation via a 20S proteasome-dependent pathway. Additionally, PA28γ+ CAFs exert an impact on the tumor immune microenvironment by secreting IGF2. Excitingly, our study suggests that targeting PA28γ+ CAFs or secreted IGF2 could increase the efficacy of PD-L1 therapy. Thus, our findings reveal the pivotal role of PA28γ in cell interactions in the tumor microenvironment and propose novel strategies for augmenting the effectiveness of immune checkpoint blockade in oral squamous cell carcinoma.
    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
    恶性疟原虫,导致疟疾的致命原生动物寄生虫,具有与其红细胞内发育周期密切相关的严格调控的基因表达谱。组蛋白乙酰化密码的表观遗传修饰因子已被确定为寄生虫转录组的关键调节因子,但需要进一步研究。在这项研究中,我们绘制了恶性疟原虫组蛋白脱乙酰酶1(PfHDAC1)在整个红细胞无性发育周期中的基因组分布,并发现它对多种发育相关基因具有动态占用。PfHDAC1的过表达导致无性感染周期的连续轮次中寄生虫负荷的逐步增加,并且与多个宿主细胞侵袭因子家族的基因表达增强有关(裂殖子表面蛋白,rhoptry蛋白质,等。)并具有增加的裂殖子侵入效率。随着类特异性抑制剂的使用,我们证明寄生虫中的PfHDAC1活性对于及时的红细胞内发育至关重要.有趣的是,PfHDAC1的过表达导致寄生虫对一线药物双氢青蒿素的敏感性降低。此外,我们发现青蒿素暴露会干扰PfHDAC1丰度和染色质占用,导致与青蒿素反应/抗性有关的基因富集。最后,我们确定双氢青蒿素暴露可以中断PfHDAC1的体外催化脱乙酰酶活性和翻译后磷酸化,这对其基因组功能至关重要。总的来说,我们的结果表明,PfHDAC1是无性寄生虫发育和宿主入侵中关键功能的调节剂,对青蒿素暴露压力有反应,对青蒿素的抗性具有确定性。
    目的:疟疾是一个主要的公共卫生问题,寄生虫恶性疟原虫导致大部分与疟疾相关的死亡率。它通过被感染的蚊子叮咬传播,并导致周期性发烧等症状,发冷,和头痛。然而,如果不及时治疗,它可以迅速发展到更严重和危及生命的形式。世界卫生组织目前建议使用青蒿素联合疗法,多年来一直是黄金标准。不幸的是,东南亚和非洲的某些国家,疟疾发病率很高,报告了耐药感染病例。控制疟疾的主要问题之一是青蒿素耐药性的出现。群体基因组研究已经确定Kelch13基因中的突变是青蒿素抗性的分子标记。然而,此后的几篇报道表明,Kelch13不是主要的介体,而是暗示转录失调是耐药性的主要决定因素。早些时候,我们将PfGCN5确定为应激反应基因的全局调节因子,已知在青蒿素抗性产生中起着核心作用。在这项研究中,我们已经确定了PfHDAC1,一种组蛋白脱乙酰酶作为细胞周期调节因子,在青蒿素抗性产生中发挥重要作用。一起来看,我们的研究确定了在青蒿素抗性产生中起重要作用的关键转录调节因子。
    Plasmodium falciparum, the deadly protozoan parasite responsible for malaria, has a tightly regulated gene expression profile closely linked to its intraerythrocytic development cycle. Epigenetic modifiers of the histone acetylation code have been identified as key regulators of the parasite\'s transcriptome but require further investigation. In this study, we map the genomic distribution of Plasmodium falciparum histone deacetylase 1 (PfHDAC1) across the erythrocytic asexual development cycle and find it has a dynamic occupancy over a wide array of developmentally relevant genes. Overexpression of PfHDAC1 results in a progressive increment in parasite load over consecutive rounds of the asexual infection cycle and is associated with enhanced gene expression of multiple families of host cell invasion factors (merozoite surface proteins, rhoptry proteins, etc.) and with increased merozoite invasion efficiency. With the use of class-specific inhibitors, we demonstrate that PfHDAC1 activity in parasites is crucial for timely intraerythrocytic development. Interestingly, overexpression of PfHDAC1 results in decreased sensitivity to frontline-drug dihydroartemisinin in parasites. Furthermore, we identify that artemisinin exposure can interfere with PfHDAC1 abundance and chromatin occupancy, resulting in enrichment over genes implicated in response/resistance to artemisinin. Finally, we identify that dihydroartemisinin exposure can interrupt the in vitro catalytic deacetylase activity and post-translational phosphorylation of PfHDAC1, aspects that are crucial for its genomic function. Collectively, our results demonstrate PfHDAC1 to be a regulator of critical functions in asexual parasite development and host invasion, which is responsive to artemisinin exposure stress and deterministic of resistance to it.
    OBJECTIVE: Malaria is a major public health problem, with the parasite Plasmodium falciparum causing most of the malaria-associated mortality. It is spread by the bite of infected mosquitoes and results in symptoms such as cyclic fever, chills, and headache. However, if left untreated, it can quickly progress to a more severe and life-threatening form. The World Health Organization currently recommends the use of artemisinin combination therapy, and it has worked as a gold standard for many years. Unfortunately, certain countries in southeast Asia and Africa, burdened with a high prevalence of malaria, have reported cases of drug-resistant infections. One of the major problems in controlling malaria is the emergence of artemisinin resistance. Population genomic studies have identified mutations in the Kelch13 gene as a molecular marker for artemisinin resistance. However, several reports thereafter indicated that Kelch13 is not the main mediator but rather hinted at transcriptional deregulation as a major determinant of drug resistance. Earlier, we identified PfGCN5 as a global regulator of stress-responsive genes, which are known to play a central role in artemisinin resistance generation. In this study, we have identified PfHDAC1, a histone deacetylase as a cell cycle regulator, playing an important role in artemisinin resistance generation. Taken together, our study identified key transcriptional regulators that play an important role in artemisinin resistance generation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号