Home>>Signaling Pathways>> Apoptosis>>Methylstat

Methylstat

目录号 : GC67966

Methylstat 是一种有效的组蛋白去甲基化酶 (histone demethylases) 抑制剂。Methylstat 具有抗增殖活性,低细胞毒性。Methylstat 诱导细胞凋亡 (Apoptosis) 和细胞周期停滞在 G0/G1 期。Methylstat 增加 p53 和 p21 蛋白水平的表达。Methylstat 抑制由各种细胞因子诱导的血管生成。Methylstat 可用作化学探针以解决其在血管生成中的作用。

Methylstat Chemical Structure

Cas No.:1310877-95-2

规格 价格 库存 购买数量
10mg
¥2,880.00
现货
25mg
¥6,300.00
现货

电话:400-920-5774 Email: sales@glpbio.cn

Customer Reviews

Based on customer reviews.

Sample solution is provided at 25 µL, 10mM.

产品文档

Quality Control & SDS

View current batch:

产品描述

Methylstat is a potent histone demethylases inhibitor. Methylstat shows anti-proliferative activity with low cytotoxicity. Methylstat induces Apoptosis and cell cycle arrest at G0/G1 phase. Methylstat increases the expression of p53 and p21 protein levels. Methylstat inhibits angiogenesis induced by various cytokines. Methylstat can be used as a chemical probe for addressing its role in angiogenesis[1][2].

Methylstat (0-5 µM; 48, 72 h) shows anti-proliferative activity with no cytotoxicity on HUVECs at 1-2 µM[1].
Methylstat (0, 1, 2 µM; 48 h) induces cell cycle arrest at G0/G1 phase in a dose-dependent manner[1].
Methylstat (0, 1, 2 µM; 48 h) increases the expression of p53 mRNA levels, the H3K27 methylation levels and the accumulation of p53 and p21 protein levels, but suppresses the protein level of cyclinD1[1].
Methylstat (0, 1, 2 µM) shows anti-angiogenic activity induced by VEGF, bFGF and TNF-α in HUVEC cells, and inhibits the f capillary formation during CAM (chick embryo chorioallantoic membrane) development without any sign of thrombosis and hemorrhage[1].
Methylstat (1.1, 2.2 mM for U266 cells, 2.1, 4.2 mM for ARH77 cells; 72 h) induces apoptosis significantly in U266 and ARH77 cells[2].

Cell Cytotoxicity Assay[1]

Cell Line: HUVEC cells
Concentration: 0-5 µM
Incubation Time: 48, 72 h
Result: Did not exhibit cytotoxicity on HUVECs at 1-2 µM.

Cell Viability Assay[1]

Cell Line: HUVEC, HepG2, HeLa, CHANG cells
Concentration: 0-5 µM
Incubation Time: 72 h
Result: Showed anti-proliferative activity with IC50s of 4, 10, 5, 7.5 µM for HUVEC, HepG2, HeLa, CHANG cells, respectively.

Cell Cycle Analysis[1]

Cell Line: HUVEC cells
Concentration: 0, 1, 2 µM
Incubation Time: 48 h
Result: G0/G1 phase increased 16.8% compared to non-treated cells, whereas S andG2/M decreased 5.5% and 6.1% respectively.

Western Blot Analysis[1]

Cell Line: HUVEC cells
Concentration: 0, 1, 2 µM
Incubation Time: 0-48 h
Result: Resulted in accumulation of p53 and p21 protein levels in a time- and dose-dependent manner and increased the H3K27 methylation levels, the but suppressed the protein level of cyclinD1.

Apoptosis Analysis[2]

Cell Line: U266, ARH77 cells
Concentration: 1.1, 2.2 mM for U266 cells, 2.1, 4.2 mM for ARH77 cells
Incubation Time: 72 h
Result: Induced apoptosis in U266, ARH77 cells.

[1]. Yumi Cho, et al. A histone demethylase inhibitor, methylstat, inhibits angiogenesis in vitro and in vivo. RSC Advances, 2014.
[2]. Kac? FN, et al. Synergistic Apoptotic Effects of Bortezomib and Methylstat on Multiple Myeloma Cells. Arch Med Res. 2020 Apr;51(3):187-193.

Chemical Properties

Cas No. 1310877-95-2 SDF Download SDF
分子式 C28H31N3O6 分子量 505.56
溶解度 DMSO : 100 mg/mL (197.80 mM; Need ultrasonic) 储存条件 4°C, protect from light
General tips 请根据产品在不同溶剂中的溶解度选择合适的溶剂配制储备液;一旦配成溶液,请分装保存,避免反复冻融造成的产品失效。
储备液的保存方式和期限:-80°C 储存时,请在 6 个月内使用,-20°C 储存时,请在 1 个月内使用。
为了提高溶解度,请将管子加热至37℃,然后在超声波浴中震荡一段时间。
Shipping Condition 评估样品解决方案:配备蓝冰进行发货。所有其他可用尺寸:配备RT,或根据请求配备蓝冰。

溶解性数据

制备储备液
1 mg 5 mg 10 mg
1 mM 1.978 mL 9.89 mL 19.78 mL
5 mM 0.3956 mL 1.978 mL 3.956 mL
10 mM 0.1978 mL 0.989 mL 1.978 mL
  • 摩尔浓度计算器

  • 稀释计算器

  • 分子量计算器

质量
=
浓度
x
体积
x
分子量
 
 
 
*在配置溶液时,请务必参考产品标签上、MSDS / COA(可在Glpbio的产品页面获得)批次特异的分子量使用本工具。

计算

动物体内配方计算器 (澄清溶液)

第一步:请输入基本实验信息(考虑到实验过程中的损耗,建议多配一只动物的药量)
给药剂量 mg/kg 动物平均体重 g 每只动物给药体积 ul 动物数量
第二步:请输入动物体内配方组成(配方适用于不溶于水的药物;不同批次药物配方比例不同,请联系GLPBIO为您提供正确的澄清溶液配方)
% DMSO % % Tween 80 % saline
计算重置

Research Update

Dynamics and function of DNA methylation in plants

Nat Rev Mol Cell Biol 2018 Aug;19(8):489-506.PMID:29784956DOI:10.1038/s41580-018-0016-z.

DNA methylation is a conserved epigenetic modification that is important for gene regulation and genome stability. Aberrant patterns of DNA methylation can lead to plant developmental abnormalities. A specific DNA methylation state is an outcome of dynamic regulation by de novo methylation, maintenance of methylation and active demethylation, which are catalysed by various enzymes that are targeted by distinct regulatory pathways. In this Review, we discuss DNA methylation in plants, including methylating and demethylating enzymes and regulatory factors, and the coordination of methylation and demethylation activities by a so-called Methylstat mechanism; the functions of DNA methylation in regulating transposon silencing, gene expression and chromosome interactions; the roles of DNA methylation in plant development; and the involvement of DNA methylation in plant responses to biotic and abiotic stress conditions.

Synergistic Apoptotic Effects of Bortezomib and Methylstat on Multiple Myeloma Cells

Arch Med Res 2020 Apr;51(3):187-193.PMID:32111493DOI:10.1016/j.arcmed.2020.01.012.

Background: In this study, we aimed to determine synergistic apoptotic and cytotoxic effects of Methylstat and bortezomib on U266 and ARH77 multiple myeloma (MM) cells. Methods: Cytotoxic effects of the drugs were demonstrated by MTT cell proliferation assay while apoptotic effects were examined by loss of mitochondrial membrane potential (MMP) by JC-1 MMP detection kit, changes in caspase-3 enzyme activity and Annexin-V apoptosis assay by flow cytometry. Expression levels of apoptotic and antiapoptotic genes were examined by qRT-PCR. Results: Our results showed that combination of Methylstat and bortezomib have synergistic antiproliferative effect on MM cells as compared to either agent alone. These results were also confirmed by showing synergistic apoptotic effects determined by increased loss of mitochondrial membrane potential and increased caspase-3 enzyme activity and relocation of phosphotidyleserine on the cell membrane by Annexin-V/PI double staining. Combination of bortezomib with Methylstat arrested cells at the S phase of the cell cycle. Methylstat treatment caused upregulation of FASLG, NGFR, TNF, TNFRS10B and TNFRS1B apoptotic genes and downregulation of AKT1, AVEN, BAG1 BCL2L2 and RELA antiapoptotic genes in a dose and time dependent manner. Conclusion: In conclusion, our data suggested that bortezomib in combination with Methylstat decreased cell proliferation and induced apoptosis significantly in U266 and ARH77 cells. When supported with in vivo analyses, Methylstat might be considered as a potential new agent for the treatment of MM.

Loss of SUV420H2-Dependent Chromatin Compaction Drives Right-Sided Colon Cancer Progression

Gastroenterology 2023 Feb;164(2):214-227.PMID:36402192DOI:10.1053/j.gastro.2022.10.036.

Background & aims: Epigenetic processes regulating gene expression contribute markedly to epithelial cell plasticity in colorectal carcinogenesis. The lysine methyltransferase SUV420H2 comprises an important regulator of epithelial plasticity and is primarily responsible for trimethylation of H4K20 (H4K20me3). Loss of H4K20me3 has been suggested as a hallmark of human cancer due to its interaction with DNMT1. However, the role of Suv4-20h2 in colorectal cancer is unknown. Methods: We examined the alterations in histone modifications in patient-derived colorectal cancer organoids. Patient-derived colorectal cancer organoids and mouse intestinal organoids were genetically manipulated for functional studies in patient-derived xenograft and orthotopic transplantation. Gene expression profiling, micrococcal nuclease assay, and chromatin immunoprecipitation were performed to understand epigenetic regulation of chromatin states and gene expression in patient-derived and mouse intestinal organoids. Results: We found that reduced H4K20me3 levels occurred predominantly in right-sided patient-derived colorectal cancer organoids, which were associated with increased chromatin accessibility. Re-compaction of chromatin by Methylstat, a histone demethylase inhibitor, resulted in reduced growth selectively in subcutaneously grown tumors derived from right-sided cancers. Using mouse intestinal organoids, we confirmed that Suv4-20h2-mediated H4K20me3 is required for maintaining heterochromatin compaction and to prevent R-loop formation. Cross-species comparison of Suv4-20h2-depleted murine organoids with right-sided colorectal cancer organoids revealed a large overlap of gene signatures involved in chromatin silencing, DNA methylation, and stemness/Wnt signaling. Conclusions: Loss of Suv4-20h2-mediated H4K20me3 drives right-sided colorectal tumorigenesis through an epigenetically controlled mechanism of chromatin compaction. Our findings unravel a conceptually novel approach for subtype-specific therapy of this aggressive form of colorectal cancer.

Therapeutic Potentials of Inhibition of Jumonji C Domain-containing Demethylases in Acute Myeloid Leukemia

Turk J Haematol 2020 Feb 20;37(1):5-12.PMID:31833715DOI:10.4274/tjh.galenos.2019.2019.0083.

Objective: Acute myeloid leukemia (AML) is a complex disease affected by both genetic and epigenetic factors. Histone methylation and demethylation are types of epigenetic modification in chromatin remodeling and gene expression. Abnormal expression of histone demethylases is indicated in many types of cancer including AML. Although many commercial drugs are available to treat AML, an absolute cure has not been discovered yet. However, inhibition of demethylases could be a potential cure for AML. Methylstat is a chemical agent that inhibits the Jumonji C domain-containing demethylases. Materials and methods: The cytotoxic and apoptotic effects of Methylstat and doxorubicin on HL-60 cells were detected by MTT cell viability assay, double staining of treated cells with annexin-V/propidium iodide, and caspase-3 activity assay. Mitochondrial activity was analyzed using JC-1 dye. The expression levels of the BCL2 and BCL2L1 anti-apoptotic genes in HL-60 cells were determined using real-time polymerase chain reaction (PCR). Lastly, the cytostatic effect was determined by cell cycle analysis. Results: In our research, cytotoxic, cytostatic, and apoptotic effects of Methylstat on human HL-60 cells were investigated. Cytotoxic and cytostatic analyses revealed that Methylstat decreased cell proliferation in a dose-dependent cytotoxic manner and arrested HL-60 cells in the G2/M and S phases. Methylstat also induced apoptosis through the loss of mitochondrial membrane potential and increases in caspase-3 enzyme activity. The expression levels of BCL2 and BCL2L1 were also decreased according to real-time PCR results. Finally, the combination of Methylstat with doxorubicin resulted in synergistic cytotoxic effects on HL-60 cells. Conclusion: Taken together, these results demonstrate that Methylstat may be a powerful candidate as a drug component of AML treatment protocols.

A model for the aberrant DNA methylomes in aging cells and cancer cells

Biochem Soc Trans 2019 Aug 30;47(4):997-1003.PMID:31320500DOI:10.1042/BST20180218.

DNA methylation at the fifth position of cytosine is a major epigenetic mark conserved in plants and mammals. Genome-wide DNA methylation patterns are dynamically controlled by integrated activities of establishment, maintenance, and removal. In both plants and mammals, a pattern of global DNA hypomethylation coupled with increased methylation levels at some specific genomic regions arises at specific developmental stages and in certain abnormal cells, such as mammalian aging cells and cancer cells as well as some plant epigenetic mutants. Here we provide an overview of this distinct DNA methylation pattern in mammals and plants, and propose that a Methylstat, which is a cis-element responsive to both DNA methylation and active demethylation activities and controlling the transcriptional activity of a key DNA methylation regulator, can help to explain the enigmatic DNA methylation patterns in aging cells and cancer cells.