Home>>Signaling Pathways>> Chromatin/Epigenetics>> Sirtuin>>EX 527 (SEN0014196)

EX 527 (SEN0014196) Sale

(Synonyms: EX-527,SIRT1 Inhibitor III,SEN0014196,EX527, Selisistat) 目录号 : GC10635

A SIRT1 inhibitor

EX 527 (SEN0014196) Chemical Structure

Cas No.:49843-98-3

规格 价格 库存 购买数量
10mM (in 1mL DMSO)
¥347.00
现货
10mg
¥389.00
现货
25mg
¥767.00
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.

105

客户使用产品发表文献 4

产品文档

Quality Control & SDS

View current batch:

实验参考方法

Cell experiment [1]:

Cell lines

T cells

Preparation Method

Purified WT or Sirt-1−/− T cells were labeled with CFSE and cocultured with T-cell-depleted splenocytes as APCs from BALB/c mice for 5 days in the presence of either dimethyl sulfoxide or EX 527 (10 µg/mL). Cells were restimulated with phorbol 12-myristate 13-acetate and ionomycin for cytokine measurement.

Reaction Conditions

10 µg/mL, 5 days

Applications

T cells with allogeneic antigen-presenting cells (APCs) in vitro, and found that EX 527 significantly reduced CD4 T-cell proliferation and IFN-γ production to a comparable level of Sirt-1−/− T cells.

Animal experiment [2]:

Animal models

Male ZDF rats (body weight 300 ± 25 g)

Preparation Method

EX-527 (5 μg/kg, twice weekly) was administered intraperitoneally (i.p.) to HFD-fed rats for ten weeks. The normal diet-fed rats received diets which were devoid of fats. Glucose levels were determined by using a glucometer.

Dosage form

5 μg/kg, i.p.

Applications

Two weeks after HFD-feeding, blood glucose concentration was increased significantly in HFD fed rats than that of normal diet-fed rats. However, fasting blood glucose level was dramatically decreased in the HFD-fed rats following EX 527 treatment.

References:

[1]. Daenthanasanmak A, et al. Targeting Sirt-1 controls GVHD by inhibiting T-cell allo-response and promoting Treg stability in mice. Blood. 2019 Jan 17;133(3):266-279.

[2]. Kundu A, et al. EX-527 Prevents the Progression of High-Fat Diet-Induced Hepatic Steatosis and Fibrosis by Upregulating SIRT4 in Zucker Rats. Cells. 2020 Apr 29;9(5):1101.

产品描述

EX 527, as a SIRT1-selective inhibitor, inhibits Sirt1 ~100-fold more potently than Sirt2 and Sirt3 and has no effect on Sirt5′s deacetylation activit.The IC50 values for Sirt1 and Sirt3 are 0.09 ± 0.03 μM and 22.4 ± 2.7 μM, respectively. Sir2Tm was also efficiently inhibited by Ex-527 with IC50 of 0.9 ± 0.3 μM.[1]

In vitro experiment it shown that cell viability significantly increased and cell death decreased in cancer cells with SIRT1 silencing or?EX 527 (10 μM) treatment compared with the control after exposure to RSL3 or sulfasalazine.[2] In vitro, treatment with 1?μM EX-527 decreased colony formation of ovarian carcinoma cells, with or without overexpression of SIRT172. However, at 600?nM, EX-527 suppressed cell migration and inhibited the occurrence of epithelial–mesenchymal transition (EMT) in chemotherapy resistant oesophageal cancer cells.[4] In vitro experiment it indicated that SIRT 1 inhibition by?EX 527 (10 μM) elevated ROS production. SIRT1 and PGC-1α levels were dramatically decreased.[6]

In vivo efficacy test indicated that pharmacological blockade of Sirt-1 with 2 mg/kg EX 527/mouse/day daily for 3 weeks alleviated GVHD without impairing T-cell-mediated GVL activity.[3] In vivo efficacy study demonstrated that treatment with 5?mg/kg?EX 527 intraperitoneally in C57BL/6J mice abolished the protective effects of melatonin.[4] In vivo, the data indicated that LPS-induced intrapulmonary inflammation and LPS-induced elevation of 4E-BP1 phosphorylation were attenuated by?EX 527 (10?mg/kg, i.p.).[5]

References:
[1]. Gertz M, et al. Ex-527 inhibits Sirtuins by exploiting their unique NAD+-dependent deacetylation mechanism. Proc Natl Acad Sci U S A. 2013 Jul 23;110(30):E2772-81.
[2]. Lee J, et al. Epigenetic reprogramming of epithelial-mesenchymal transition promotes ferroptosis of head and neck cancer. Redox Biol. 2020 Oct;37:101697.
[3]. Daenthanasanmak A, et al. Targeting Sirt-1 controls GVHD by inhibiting T-cell allo-response and promoting Treg stability in mice. Blood. 2019 Jan 17;133(3):266-279.
[4]. Broussy S, et al. Biochemical mechanism and biological effects of the inhibition of silent information regulator 1 (SIRT1) by EX-527 (SEN0014196 or selisistat). J Enzyme Inhib Med Chem. 2020 Dec;35(1):1124-1136.
[5]. Huang J, et al. The SIRT1 inhibitor EX-527 suppresses mTOR activation and alleviates acute lung injury in mice with endotoxiemia. Innate Immun. 2017 Nov;23(8):678-686.
[6]. Waldman M, et al. Regulation of diabetic cardiomyopathy by caloric restriction is mediated by intracellular signaling pathways involving 'SIRT1 and PGC-1α'. Cardiovasc Diabetol. 2018 Aug 2;17(1):111.

EX 527 作为 SIRT1 选择性抑制剂,抑制 Sirt1 的效力比 Sirt2 和 Sirt3 高 100 倍,并且对 Sirt5 的去乙酰化活性没有影响。Sirt1 和 Sirt3 的 IC50 值为 0.09 ± 0.03 μM 和 22.4分别为± 2.7 μM。 Sir2Tm 也被 Ex-527 有效抑制,IC50 为 0.9 ± 0.3 μM。[1]

体外实验表明,与暴露于 RSL3 或柳氮磺胺吡啶后的对照相比,SIRT1 沉默或 EX 527 (10 μM) 处理的癌细胞的细胞活力显着增加,细胞死亡减少。[2] 在体外,用 1μM EX-527 处理可减少卵巢癌细胞的集落形成,有或没有 SIRT172 过表达。然而,在 600nM 时,EX-527 抑制细胞迁移,抑制化疗耐药食管癌细胞上皮-间质转化 (EMT) 的发生。[4] 体外实验表明 SIRT 1 抑制EX 527 (10 μM) 提高了 ROS 的产量。 SIRT1 和 PGC-1α 水平显着降低。[6]

体内药效试验表明,每天 2 mg/kg EX 527/小鼠/天对 Sirt-1 进行药理学阻断,持续 3 周可减轻 GVHD,而不会损害 T 细胞介导的 GVL 活性。[3] 体内功效研究表明,在 C57BL/6J 小鼠中腹膜内注射 5 mg/kg EX 527 可消除褪黑激素的保护作用。[4] 在体内,数据表明 LPS 诱导的肺内EX 527(10mg/kg,i.p.)减弱了炎症和 LPS 诱导的 4E-BP1 磷酸化升高。[5]

Chemical Properties

Cas No. 49843-98-3 SDF
别名 EX-527,SIRT1 Inhibitor III,SEN0014196,EX527, Selisistat
化学名 6-chloro-2,3,4,9-tetrahydro-1H-carbazole-1-carboxamide
Canonical SMILES C1CC(C2=C(C1)C3=C(N2)C=CC(=C3)Cl)C(=O)N
分子式 C13H13ClN2O 分子量 248.71
溶解度 ≥ 12.4 mg/mL in DMSO, ≥ 25.45 mg/mL in EtOH with ultrasonic 储存条件 Store at -20°C
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 4.0207 mL 20.1037 mL 40.2075 mL
5 mM 0.8041 mL 4.0207 mL 8.0415 mL
10 mM 0.4021 mL 2.0104 mL 4.0207 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

Biochemical mechanism and biological effects of the inhibition of silent information regulator 1 (SIRT1) by EX-527 (SEN0014196 or selisistat)

J Enzyme Inhib Med Chem2020 Dec;35(1):1124-1136.PMID: 32366137DOI: 10.1080/14756366.2020.1758691

The human sirtuin silent information regulator 1 (SIRT1) is a NAD+-dependent deacetylase enzyme. It deacetylates many protein substrates, including histones and transcription factors, thereby controlling many physiological and pathological processes. Several synthetic inhibitors and activators of SIRT1 have been developed, and some therapeutic applications have been explored. The indole EX-527 and its derivatives are among the most potent and selective SIRT1 inhibitors. EX-527 has been often used as a pharmacological tool to explore the effect of SIRT1 inhibition in various cell types. Its therapeutic potential has, therefore, been evaluated in animal models for several pathologies, including cancer. It has also been tested in phase II clinical trial for the treatment of Huntington's disease (HD). In this review, we will provide an overview of the literature on EX-527, including its mechanism of inhibition and biological studies.

Protective effect of EX-527 against high-fat diet-induced diabetic nephropathy in Zucker rats

Toxicol Appl Pharmacol2020 Mar 1;390:114899.PMID: 31981641DOI: 10.1016/j.taap.2020.114899

High-fat diet (HFD)-induced obesity is implicated in diabetic nephropathy (DN). EX-527, a selective Sirtuin 1 (SIRT1) inhibitor, has multiple biological functions; however, its protective effect against DN is yet to be properly understood. This study was aimed to explore the protective effect of EX-527 against DN in HFD-induced diabetic Zucker (ZDF) rats. After 21 weeks of continually feeding HFD to the rats, the apparent characteristics of progressive DN were observed, which included an increase in kidney weight (~160%), hyperglycemia, oxidative stress, and inflammatory cytokines, and subsequent renal cell damage. However, the administration of EX-527 for 10 weeks significantly reduced the blood glucose concentration and kidney weight (~59%). Furthermore, EX-527 significantly reduced the serum concentration of transforming growth factor-β1 (49%), interleukin (IL)-1β (52%), and IL-6 in the HFD-fed rats. Overall, the antioxidant activities significantly increased, and oxidative damage to lipids or DNA was suppressed. Particularly, EX-527 significantly reduced blood urea nitrogen (81%), serum creatinine (71%), microalbumin (43%), and urinary excretion of protein-based biomarkers. Histopathological examination revealed expansion of the extracellular mesangial matrix and suppression of glomerulosclerosis following EX-527 administration. EX-527 downregulated the expression of α-SMA (~64%), TGF-β (25%), vimentin, α-tubulin, fibronectin, and collagen-1 in the kidneys of the HFD-fed rats. Additionally, EX-527 substantially reduced claudin-1 and SIRT1 expression, but increased the expression of SIRT3 in the kidneys of the HFD-fed rats. EX-527 also inhibited the growth factor receptors, including EGFR, PDGFR-β, and STAT3, which are responsible for the anti-fibrotic effect of SIRT-1. Therefore, the administration of EX-527 protects against HFD-induced DN.

Hydrogen sulfide-induced GAPDH sulfhydration disrupts the CCAR2-SIRT1 interaction to initiate autophagy

Autophagy2021 Nov;17(11):3511-3529.PMID: 33459133DOI: 10.1080/15548627.2021.1876342

The deacetylase SIRT1 (sirtuin 1) has emerged as a major regulator of nucleocytoplasmic distribution of macroautophagy/autophagy marker MAP1LC3/LC3 (microtubule-associated protein 1 light chain 3). Activation of SIRT1 leads to the deacetylation of LC3 and its translocation from the nucleus into the cytoplasm leading to an increase in the autophagy flux. Notably, hydrogen sulfide (H2S) is a cytoprotective gasotransmitter known to activate SIRT1 and autophagy; however, the underlying mechanism for both remains unknown. Herein, we demonstrate that H2S sulfhydrates the active site cysteine of the glycolytic enzyme GAPDH (glyceraldehyde-3-phosphate dehydrogenase). Sulfhydration of GAPDH leads to its redistribution into the nucleus. Importantly, nuclear localization of GAPDH is critical for H2S-mediated activation of autophagy as H2S does not induce autophagy in cells with GAPDH ablation or cells overexpressing a GAPDH mutant lacking the active site cysteine. Importantly, we observed that nuclear GAPDH interacts with CCAR2/DBC1 (cell cycle activator a nd apoptosis regulator 2) inside the nucleus. CCAR2 interacts with the deacetylase SIRT1 to inhibit its activity. Interaction of GAPDH with CCAR2 disrupts the inhibitory effect of CCAR2 on SIRT1. Activated SIRT1 then deacetylates MAP1LC3B/LC3B (microtubule-associated protein 1 light chain 3 beta) to induce its translocation into the cytoplasm and activate autophagy. Additionally, we demonstrate this pathway's physiological role in autophagy-mediated trafficking of Mycobacterium tuberculosis into lysosomes to restrict intracellular mycobacteria growth. We think that the pathway described here could be involved in H2S-mediated clearance of intracellular pathogens and other health benefits.Abbreviations: ATG5: autophagy related 5; ATG7: autophagy related 7; BECN1: beclin 1, autophagy related; CCAR2/DBC1: cell cycle activator and apoptosis regulator 2; CFU: colony-forming units; DLG4/PSD95: discs large MAGUK scaffold protein 4; EX-527: 6-chloro-2,3,4,9-tetrahydro-1H-carbazole-1-carboxamide; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; H2S: hydrogen sulfide; HEK: human embryonic kidney cells; MAP1LC3B/LC3B: microtubule-associated protein 1 light chain 3 beta; MEF: mouse embryonic fibroblast; Mtb: Mycobacterium tuberculosis; MTOR: mechanistic target of rapamycin kinase; MOI: multiplicity of infection; NO: nitric oxide; PI3K: phosphatidylinositol-4,5-bisphosphate 3-kinase; PLA: proximity ligation assay; PRKAA: protein kinase, AMP-activated, alpha catalytic subunit; SIAH1: siah E3 ubiquitin protein ligase 1A; SIRT1: sirtuin 1; TB: tuberculosis; TP53INP2/DOR: transformation related protein 53 inducible nuclear protein 2; TRP53/TP53: transformation related protein 53.

Sirt1 promotes autophagy and inhibits apoptosis to protect cardiomyocytes from hypoxic stress

Int J Mol Med2019 May;43(5):2033-2043.PMID: 30864731DOI: 10.3892/ijmm.2019.4125

Sirtuin 1 (Sirt1) exerts its cardioprotective effects in various cardiovascular diseases via multiple cellular activities. However, the therapeutic implications of Sirt1 in hypoxic cardiomyocytes and the underlying mechanisms remain elusive. The present study investigated whether Sirt1 regulates autophagy and apoptosis in hypoxic H9C2 cardiomyocytes and in an experimental hypoxic mouse model. Right ventricular outflow tract biopsies were obtained from patients with cyanotic or acyanotic congenital heart diseases. Adenovirus Ad‑Sirt1 was used to activate Sirt1 and Ad‑Sh‑Sirt1 was used to inhibit Sirt1 expression in H9C2 cells, in order to investigate the effect of Sirt1 on cellular autophagy and apoptosis. SRT1720, a pharmacological activator of Sirt1 and EX-527, a Sirt1 antagonist, were administered to mice to explore the role of Sirt1 in hypoxic cardiomyocytes in vivo. The levels of autophagy and apoptosis‑related proteins were evaluated using western blotting. Apoptosis was investigated by TUNEL staining and Annexin V/7‑aminoactinomycin D flow cytometry analysis. Heart tissue samples from cyanotic patients exhibited increased autophagy and apoptosis, as well as elevated Sirt1 levels, compared with the noncyanotic control samples. The data from the western blot analysis revealed that Sirt1 promoted autophagic flux and reduced apoptosis in hypoxic H9C2 cells. In addition, Sirt1 activated AMP‑activated protein kinase (AMPK), and the AMPK inhibitor Compound C abolished the effect of Sirt1 on autophagy activation. Further exploration of the mechanism revealed that Sirt1 protects hypoxic cardiomyocytes from apoptosis, at least in part, through inositol requiring kinase enzyme 1α (IRE1α). Consistent with the in vitro results, treatment with the Sirt1 activator SRT1720 activated AMPK, inhibited IRE1α, enhanced autophagy, and decreased apoptosis in the heart tissues of normoxic mice compared with the hypoxia control group. Opposite changes were observed in hypoxic mice treated with the Sirt1 inhibitor EX-527. These results suggested that Sirt1 promoted autophagy via AMPK activation and reduced hypoxia‑induced apoptosis via the IRE1α pathway, to protect cardiomyocytes from hypoxic stress.

Baicalin Ameliorates Cognitive Impairment and Protects Microglia from LPS-Induced Neuroinflammation via the SIRT1/HMGB1 Pathway

Oxid Med Cell Longev2020 Sep 22;2020:4751349.PMID: 33029280DOI: 10.1155/2020/4751349

Systemic inflammation often induces neuroinflammation and disrupts neural functions, ultimately causing cognitive impairment. Furthermore, neuronal inflammation is the key cause of many neurological conditions. It is particularly important to develop effective neuroprotectants to prevent and control inflammatory brain diseases. Baicalin (BAI) has a wide variety of potent neuroprotective and cognitive enhancement properties in various models of neuronal injury through antioxidation, anti-inflammation, anti-apoptosis, and stimulating neurogenesis. Nevertheless, it remains unclear whether BAI can resolve neuroinflammation and cognitive decline triggered by systemic or distant inflammatory processes. In the present study, intraperitoneal lipopolysaccharide (LPS) administration was used to establish neuroinflammation to evaluate the potential neuroprotective and anti-inflammatory effects of BAI. Here, we report that BAI activated silent information regulator 1 (SIRT1) to deacetylate high-mobility group box 1 (HMGB1) protein in response to acute LPS-induced neuroinflammation and cognitive deficits. Furthermore, we demonstrated the anti-inflammatory and cognitive enhancement effects and the underlying molecular mechanisms of BAI in modulating microglial activation and systemic cytokine production, including tumor necrosis factor- (TNF-) α and interleukin- (IL-) 1β, after LPS exposure in mice and in the microglial cell line, BV2. In the hippocampus, BAI not only reduced reactive microglia and inflammatory cytokine production but also modulated SIRT1/HMGB1 signaling in microglia. Interestingly, pretreatment with SIRT1 inhibitor EX-527 abolished the beneficial effects of BAI against LPS exposure. Specifically, BAI treatment inhibited HMGB1 release via the SIRT1/HMGB1 pathway and reduced the nuclear translocation of HMGB1 in LPS-induced BV2 cells. These effects were reversed in BV2 cells by silencing endogenous SIRT1. Taken together, these findings indicated that BAI reduced microglia-associated neuroinflammation and improved acute neurocognitive deficits in LPS-induced mice via SIRT1-dependent downregulation of HMGB1, suggesting a possible novel protection against acute neurobehavioral deficits, such as delayed neurocognitive recovery after anesthesia and surgery challenges.