首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 149 毫秒
1.
在肿瘤发生过程中,组蛋白赖氨酸去甲基化酶(LSD1)的表达失调是一个重要标志。LSD1能够于组蛋白H3的N端与H3K4me2/1和H3K9me2/1相互作用,并使其去甲基化,从而调控多种不同的生理过程。同时,LSD1表达水平变化还与多种基因如p53、DNMT1和EZH2等的表达水平相关联,在胚胎发育、细胞分化和肿瘤增殖转移过程中起重要作用。  相似文献   

2.
赖氨酸特异性组蛋白去甲基化酶1(Lysine specific demethylase 1, LSD1) 的发现, 表明组蛋白的甲基化修饰是一个动态可调节的过程。结构分析显示, LSD1 是一个黄素腺嘌呤二核苷酸(Flavin adenine dinulcleotide, FAD) 依赖性胺氧化酶, 它能够特异性脱去单甲基化和二甲基化组蛋白H3第4位赖氨酸(H3K4) 和H3K9 位点上的甲基基团。功能研究显示, LSD1 定位于细胞核内, 调控着基因转录的激活和抑制, 被誉为细胞深处的基因“开关”, 在胚胎发育和肿瘤发生过程中起着重要的作用。文章主要综述了LSD1 的结构、作用机制及其调控作用研究的新进展。  相似文献   

3.
与其他化学修饰,如乙酰化、磷酸化、泛素化等相似,组蛋白赖氨酸甲基化是一个可以逆转的组蛋白修饰,是一个动态调节的过程。赖氨酸特异性组蛋白去甲基化酶1(lysine specific demethylase 1,LSD1)是一个黄素腺嘌呤二核苷酸(flavin adenine dinulcleotide,FAD)依赖性胺氧化酶,它能够特异性脱去H3K4和H3K9位点上的单甲基化和二甲基化的甲基基团。LSD1参与调控核受体介导的基因转录,并分别维持染色质的活性和非活性状态,被誉为细胞深处的基因"开关"。LSD1的功能失衡可引发多种重要生命现象的改变。主要综述LSD1的结构、作用机制及其在肿瘤发生、胚胎发育、体细胞重编程的调控、细胞分裂和造血等过程中生物学功能的研究新进展。  相似文献   

4.
目的:探讨雷公藤内酯醇(TPL)对多发性骨髓瘤RPMI8226细胞增殖、凋亡和组蛋白H3K4甲基化的影响。方法:以人多发性骨髓瘤细胞株RPMI8226为研究对象,在不同浓度(10、20、40、80、160 nmol/L) TPL中共培养不同时间(24 h、48 h、72 h)后,采用噻唑蓝(MTT)法检测细胞增殖活性;流式细胞术检测细胞凋亡和细胞周期;Western blot法检测组蛋白H3K4me2、H3K4me3的甲基化状态,实时荧光定量RT-PCR分析组蛋白甲基化酶SMYD3和组蛋白去甲基化酶LSD1的表达水平。结果:TPL对RPMI8226细胞有明显的增殖抑制作用,呈剂量和时间依赖性(P<0.05);TPL对RPMI8226细胞有明显诱导凋亡的作用,并且随着TPL作用浓度的增加,细胞凋亡比例逐渐增加(P<0.05);同时TPL还可以诱导RPMI8226细胞周期阻滞于G2/M期;TPL以浓度依赖性降低组蛋白H3K4me2、H3K4me3的甲基化水平(P<0.05,P<0.01),并抑制SMYD3和上调LSD1的表达(P<0.05)。结论:TPL可抑制RPMI8226细胞增殖、引起细胞周期阻滞于G2/M期,并诱导其凋亡;通过抑制组蛋白甲基化酶SMYD3和增强组蛋白去甲基化酶LSD1的表达,降低组蛋白H3K4me3和H3K4me2的甲基化水平,这可能是TPL诱导多发性骨髓瘤细胞凋亡和抗肿瘤作用的机制之一。  相似文献   

5.
PLMT家族成员SET7/9的非组蛋白甲基化作用   总被引:1,自引:0,他引:1  
SET7/9是蛋白赖氨酸甲基化转移酶(protein lysine methyltransferases,PLMTs或PKMTs)家族成员,具有SET结构域。现已发现SET7/9是一种赖氨酸单甲基化转移酶,除了能使组蛋白H3第四位赖氨酸(lysine4 of histone 3,H3K4)单甲基化外,更重要的能使一些转录因子、肿瘤抑制因子、膜相关受体等非组蛋白单甲基化,其甲基化作用主要与蛋白稳定和转录活化有关。该效应受赖氨酸特异性去甲基酶1(lysine specifcdemethylase,LSD1)的抑制。SET7/9与LSD1两者效应的平衡对维持体内活性蛋白质含量、调节基因表达具有重要意义。  相似文献   

6.
目的:检测动脉粥样硬化患者巨噬细胞中组蛋白第9位赖氨酸的二甲基化(histone H3 lysine 9 dimethylation,H3K9Me2)的表达并探究其对炎症因子表达的影响及其机制。方法:选择厦门大学附属东南医院收治的动脉粥样硬化患者(n=20)与健康对照人群(n=22)作为研究对象,采用蛋白免疫印迹方法检测巨噬细胞中H3K9Me2的表达,并通过酶联免疫吸附的方法检测其血清炎症因子表达。随后,通过小干扰RNA(small interfering RNA,siRNA)敲减赖氨酸特异去甲基化酶1A(lysine-specific demethylase 1A,LSD1)后,在氧化低密度脂蛋白(oxidized low-density lipoproteins,ox-LDL)诱导的巨噬细胞中结合白免疫印迹和染色质免疫共沉淀的方法检测细胞整体H3K9Me2的表达及炎症因子启动子区H3K9Me2水平。同时,分析巨噬细胞中上清中炎症因子表达变化。结果:动脉粥样硬化病人巨噬细胞中H3K9Me2水平显著下调,并与血清中炎症因子表达呈负相关。在敲减LSD1后,ox-LDL诱导的巨噬细胞中H3K9Me2整体水平以及炎症因子启动子区H3K9Me2水平下调均被抑制。同时,核因子-κB(NF-κB)结合启动子区介导的炎症因子表达增强也被抑制。结论:巨噬细胞中H3K9Me2的表达与动脉粥样硬化病人血清中炎症因子的表达成负相关,而组蛋白去甲基化酶LSD1很可能在其中参与H3K9Me2水平调控,并通过影响NF-κB与启动子区结合调节炎症因子表达。  相似文献   

7.
组蛋白甲基化是一种重要的表观遗传学修饰,在基因表达调节方面发挥着重要的作用.组蛋白H3赖氨酸27三甲基化(H3K27me3)是一种抑制性组蛋白标记,可被去甲基化酶UTX和JMJD3催化而移去甲基.UTX和JMJD3通过激活HOX基因而参与细胞分化和多能细胞抑制过程.在多种肿瘤中检测到UTX和JMJD3突变或表达下降,同时多种基因启动子区H3K27me3含量增多.UTX和JMJD3均被看作肿瘤抑制基因,其中UTX调节了RB依赖的细胞命运控制,而JMJD3通过激活INK4b-ARF-INK4a位点而参与了癌基因诱导的衰老.组蛋白H3K27去甲基化酶与肿瘤发生的研究使我们对癌症发展过程有了更好的理解,同时也为癌症诊断和治疗提供了新靶点.  相似文献   

8.
SET蛋白家族包含保守的SET结构域,很多家族成员可以利用S-腺苷甲硫氨酸对底物进行甲基化修饰.SET蛋白家族主要对组蛋白进行甲基化修饰,包括组蛋白H3K4、K9、K27、K36以及组蛋白H4K20,调控真核生物中基因的激活和沉默.除组蛋白外,部分SET蛋白家族成员对各种非组蛋白也具有催化酶活性.SET蛋白突变所导致的...  相似文献   

9.
组蛋白翻译后修饰可影响特定基因的表达,从而在多个生理过程中发挥重要作用,是当前生命科学领域的研究热点之一。组蛋白去甲基化酶JMJD1A可催化一甲基化和二甲基化的H3K9(H3K9me1/2)去甲基化,通过解除组蛋白抑制效应而调节基因表达。JMJD1A拥有广泛的生物学功能,参与多种生物学过程包括核受体激活、精子生成、能量代谢、低氧调节和癌症发生等。本文就JMJD1A的特征及功能作一综述。  相似文献   

10.
高文龙  刘红林 《遗传》2007,29(12):1449-1454
组蛋白甲基化是一种重要的组蛋白共价修饰, 在染色质结构和基因表达的调控过程中起着重要的、多样化的作用。DOT1催化核心球体部位的组蛋白H3第79位赖氨酸(H3K79)使其发生甲基化, 是首个被发现的无SET结构域的组蛋白赖氨酸甲基转移酶, 代表了一类新的组蛋白赖氨酸甲基转移酶。DOT1及H3K79甲基化的特点决定了其可能具有重要的、特殊的生物学功能。文章重点综述了DOT1蛋白的结构及特点, DOT1及H3K79甲基化的生物学功能以及组蛋白泛素化修饰对H3K79甲基化的反式调控。  相似文献   

11.
12.
We identify LSD1 (lysine-specific demethylase 1; also known as KDM1A and AOF2) as a key histone modifier that participates in the maintenance of pluripotency through the regulation of bivalent domains, a chromatin environment present at the regulatory regions of developmental genes that contains both H3K4 di/trimethylation and H3K27 trimethylation marks. LSD1 occupies the promoters of a subset of developmental genes that contain bivalent domains and are co-occupied by OCT4 and NANOG in human embryonic stem cells, where it controls the levels of H3K4 methylation through its demethylase activity. Thus, LSD1 has a role in maintaining the silencing of several developmental genes in human embryonic stem cells by regulating the critical balance between H3K4 and H3K27 methylation at their regulatory regions.  相似文献   

13.
Aberrant epigenetic repression of gene expression has been implicated in most cancers, including breast cancer. The nuclear amine oxidase, lysine-specific demethylase 1 (LSD1) has the ability to broadly repress gene expression by removing the activating mono- and di-methylation marks at the lysine 4 residue of histone 3 (H3K4me1 and me2). Additionally, LSD1 is highly expressed in estrogen receptor α negative (ER-) breast cancer cells. Since epigenetic marks are reversible, they make attractive therapeutic targets. Here we examine the effects of polyamine analog inhibitors of LSD1 on gene expression, with the goal of targeting LSD1 as a therapeutic modality in the treatment of breast cancer. Exposure of the ER-negative human breast cancer cells, MDA-MB-231 to the LSD1 inhibitors, 2d or PG11144, significantly increases global H3K4me1 and H3K4me2, and alters gene expression. Array analysis indicated that 98 (75 up and 23 down) and 477 (237 up and 240 down) genes changed expression by at least 1.5-fold or greater after treatment with 2d and PG11144, respectively. The expression of 12 up-regulated genes by 2d and 14 up-regulated genes by PG11144 was validated by quantitative RT-PCR. Quantitative chromatin immunoprecipitation (ChIP) analysis demonstrated that up-regulated gene expression by polyamine analogs is associated with increase of the active histone marks H3K4me1, H3K4me2 and H3K9act, and decrease of the repressive histone marks H3K9me2 and H3K27me3, in the promoter regions of the relevant target genes. These data indicate that the pharmacologic inhibition of LSD1 can effectively alter gene expression and that this therapeutic strategy has potential.  相似文献   

14.
15.
LSD1 is essential for the maintenance of pluripotency of embryonic stem (ES) or embryonic carcinoma/teratocarcinoma (EC) cells. We have previously developed novel LSD1 inhibitors that selectively inhibit ES/EC cells. However, the critical targets of LSD1 remain unclear. Here, we found that LSD1 interacts with histone deacetylase 1 (HDAC1) to regulate the proliferation of ES/EC cells through acetylation of histone H4 at lysine 16 (H4K16), which we show is a critical substrate of HDAC1. The LSD1 demethylase and HDAC1 deacetylase activities were both inactivated if one of them in the complex was chemically inhibited in ES/EC cells or in reconstituted protein complexes. Loss of HDAC1 phenocopied the selective growth-inhibitory effects and increased the levels of H3K4 methylation and H4K16 acetylation of LSD1 inactivation on ES/EC cells. Reduction of acetylated H4K16 by ablation of the acetyltransferase males absent on the first (MOF) is sufficient to rescue the growth inhibition induced by LSD1 inactivation. While LSD1 or HDAC1 inactivation caused the downregulation of Sox2 and Oct4 and induction of differentiation genes, such as FOXA2 or BMP2, depletion of MOF restored the levels of Sox2, Oct4, and FoxA2 in LSD1-deficient cells. Our studies reveal a novel mechanism by which LSD1 acts through the HDAC1- and MOF-mediated regulation of H4K16 acetylation to maintain the pluripotency of ES/EC cells.  相似文献   

16.
《Epigenetics》2013,8(3):129-132
One of the key breakthroughs in the epigenetics/chromatin field in the last several years was the identification of enzymes capable of removing the methyl group from methylated lysines in histone proteins. Lysine-specific demethylase 1 (LSD1) was the first such enzyme identified, which has been shown to demethylate histone H3 on lysine 4 (H3K4) and lysine 9 (H3K9). LSD1 is essential for mammalian development and likely involved in many biological processes. Recent studies show that LSD1 demethylates p53 and Dnmt1 and regulates their cellular functions, indicating that LSD1 fulfills its biological functions by directly acting on both histone and non-histone proteins. LSD1 contains several defined domains and associates with a number of protein complexes. Interacting partners of LSD1 may play key roles in determining/modulating the activity and specificity of LSD1.  相似文献   

17.
18.
Lysine demethylase 1 (LSD1) and Jumonji C domain-containing oxygenase D2C (JMJD2C) participate in regulating the methylation status of histone H3 lysine residues. In some contexts, LSD1 and JMJD2C activity causes enhanced cellular proliferation, which may lead to tumorigenesis. The authors explored the utility of time-resolved fluorescence resonance energy transfer (TR-FRET) immunoassays, which employed peptides consisting of the first 21 amino acids of histone H3 in which lysine 4 (H3K4) or lysine 9 (H3K9) was methylated (me) to quantify LSD1 and JMJD2C activity. The LSD1 assay monitored demethylation of the H3K4me1 peptide using an antibody that recognizes H3K4me1 but not the unmethylated peptide product. The JMJD2C assay measured demethylation of H3K9me3 with an antibody that selectively recognizes H3K9me2. The optimized conditions resulted in robust assays (Z' > 0.7) that required only 3 to 6 nM of enzyme in a reaction volume of 6 to 10 μL. These assays were used to compare the activity of different LSD1 constructs and to determine the apparent K(m) of each JMJD2C substrate. Finally, both assays were used in a high-throughput setting for identifying demethylase inhibitors. Compounds discovered by these TR-FRET methods may lead to powerful tools for ascertaining the roles of demethylases in a cellular context and ultimately for potential cancer treatments.  相似文献   

19.

Introduction

Microsomal prostaglandin E synthase 1 (mPGES-1) catalyzes the terminal step in the biosynthesis of PGE2, a critical mediator in the pathophysiology of osteoarthritis (OA). Histone methylation plays an important role in epigenetic gene regulation. In this study, we investigated the roles of histone H3 lysine 9 (H3K9) methylation in interleukin 1β (IL-1β)-induced mPGES-1 expression in human chondrocytes.

Methods

Chondrocytes were stimulated with IL-1β, and the expression of mPGES-1 mRNA was evaluated using real-time RT-PCR. H3K9 methylation and the recruitment of the histone demethylase lysine-specific demethylase 1 (LSD1) to the mPGES-1 promoter were evaluated using chromatin immunoprecipitation assays. The role of LSD1 was further evaluated using the pharmacological inhibitors tranylcypromine and pargyline and small interfering RNA (siRNA)-mediated gene silencing. The LSD1 level in cartilage was determined by RT-PCR and immunohistochemistry.

Results

The induction of mPGES-1 expression by IL-1β correlated with decreased levels of mono- and dimethylated H3K9 at the mPGES-1 promoter. These changes were concomitant with the recruitment of the histone demethylase LSD1. Treatment with tranylcypromine and pargyline, which are potent inhibitors of LSD1, prevented IL-1β-induced H3K9 demethylation at the mPGES-1 promoter and expression of mPGES-1. Consistently, LSD1 gene silencing with siRNA prevented IL-1β-induced H3K9 demethylation and mPGES-1 expression, suggesting that LSD1 mediates IL-1β-induced mPGES-1 expression via H3K9 demethylation. We show that the level of LSD1 was elevated in OA compared to normal cartilage.

Conclusion

These results indicate that H3K9 demethylation by LSD1 contributes to IL-1β-induced mPGES-1 expression and suggest that this pathway could be a potential target for pharmacological intervention in the treatment of OA and possibly other arthritic conditions.  相似文献   

20.
Lysine methylation is one of the most important histone modifications that modulate chromatin structure. In the present study, the roles of the histone lysine demethylases JMJD2a and LSD1 in CK2 downregulation-mediated senescence were investigated. The ectopic expression of JMJD2a and LSD1 suppressed the induction of senescence-associated β-galactosidase activity and heterochromatin foci formation as well as the reduction of colony-forming and cell migration ability mediated by CK2 knockdown. CK2 downregulation inhibited JMJD2a and LSD1 expression by activating the mammalian target of rapamycin (mTOR)-ribosomal p70 S6 kinase (p70S6K) pathway. In addition, the down-regulation of JMJD2a and LSD1 was involved in activating the p53-p21Cip1/WAF1-SUV39h1-trimethylation of the histone H3 Lys9 (H3K9me3) pathway in CK2-downregulated cells. Further, CK2 downregulation-mediated JMJD2a and LSD1 reduction was found to stimulate the dimethylation of Lys370 on p53 (p53K370me2) and nuclear import of SUV39h1. Therefore, this study indicated that CK2 downregulation reduces JMJD2a and LSD1 expression by activating mTOR, resulting in H3K9me3 induction by increasing the p53K370me2-dependent nuclear import of SUV39h1. These results suggest that CK2 is a potential therapeutic target for age-related diseases.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号