Home>>Signaling Pathways>> Others>> Others>>TES-1025

TES-1025 Sale

目录号 : GC31326

TES-1025 is a potent and selective human α-amino-β-carboxymuconate-ε-semialdehyde decarboxylase (ACMSD) inhibitor with an IC50 of 13 nM.

TES-1025 Chemical Structure

Cas No.:1883602-21-8

规格 价格 库存 购买数量
10mM (in 1mL DMSO)
¥2,475.00
现货
5mg
¥2,250.00
现货
10mg
¥4,050.00
现货
25mg
¥8,100.00
现货
50mg
¥12,600.00
现货
100mg
¥18,900.00
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:

实验参考方法

Kinase experiment:

Recombinant hACMSD is expressed in Pichia pastoris and purified. Its enzyme activity is assayed by a coupled spectrophotometric assay. Briefly, in a pre-assay mixture, the ACMS substrate is generated from 10 μM 3-hydroxyanthranilic acid by recombinant 3-hydroxyanthranilate 3,4-dioxigenase from Ralstonia metallidurans. ACMS formation is monitored at 360 nm, and after the reaction is complete, an appropriate amount of ACMSD is added. Activity is calculated from the initial rate of the absorbance decrease subtracted from that of a control reaction mixture in the absence of ACMSD. The effects of the various compounds (e.g., TES-1025) on the enzyme activity are tested by adding the compounds to the assay mixture along with ACMSD. For the IC50 evaluations for each compound, a serial dilution from a stock solution prepared in DMSO is tested, maintaining a DMSO concentration in all the reaction mixtures of 1.0%. One unit is defined as the amount of enzyme that consume 1 μmol of ACMS per minute at 37°C[1].

Animal experiment:

Mice[1]Male CD-1 mice are used.The study is conducted in 3 phases. Phase 1: 18 mice receive an oral administration of TES-1025 at a target dose level of 5 mg/kg. Blood, brain and liver are collected at intervals up to 8 h after dose administration (n=3 animals per each time point). Phase 2: 3 mice receive each an intravenous administration of TES-1025 at a target dose of 0.5 mg/kg. Blood samples are collected from the lateral tail vein at intervals up to 24 h after dose administration. Phase 3: 3 mice receive a single intravenous administration of Elacridar (5 mg/kg) shortly before an oral administration of TES-1025 at a target dose of 5 mg/kg. Blood and brain samples are collected 0.5 h after dose administration. Brain, liver and kidney are collected from all animals of the study[1].

References:

[1]. Pellicciari R, et al. α-Amino-β-carboxymuconate-ε-semialdehyde Decarboxylase (ACMSD) Inhibitors as Novel Modulators of De Novo Nicotinamide Adenine Dinucleotide (NAD+) Biosynthesis. J Med Chem. 2018 Feb 8;61(3):745-759.

产品描述

TES-1025 is a potent and selective human α-amino-β-carboxymuconate-ε-semialdehyde decarboxylase (ACMSD) inhibitor with an IC50 of 13 nM.

[1] Pellicciari R, et al. J Med Chem. 2018 Feb 8;61(3):745-759.

Chemical Properties

Cas No. 1883602-21-8 SDF
Canonical SMILES O=C1C(C#N)=C(C2=CC=CS2)N=C(SCC3=CC=CC(CC(O)=O)=C3)N1
分子式 C18H13N3O3S2 分子量 383.44
溶解度 DMSO : 100 mg/mL (260.80 mM) 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 2.608 mL 13.0398 mL 26.0797 mL
5 mM 0.5216 mL 2.608 mL 5.2159 mL
10 mM 0.2608 mL 1.304 mL 2.608 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

Structural Basis of Human Dimeric α-Amino-β-Carboxymuconate-ε-Semialdehyde Decarboxylase Inhibition With TES-1025

Human α-amino-β-carboxymuconate-ε-semialdehyde decarboxylase (ACMSD) stands at a branch point of the de novo NAD+ synthesis pathway and plays an important role in maintaining NAD+ homeostasis. It has been recently identified as a novel therapeutic target for a wide range of diseases, including inflammatory, metabolic disorders, and aging. So far, in absence of potent and selective enzyme inhibitors, only a crystal structure of the complex of human dimeric ACMSD with pseudo-substrate dipicolinic acid has been resolved. In this study, we report the crystal structure of the complex of human dimeric ACMSD with TES-1025, the first nanomolar inhibitor of this target, which shows a binding conformation different from the previously published predicted binding mode obtained by docking experiments. The inhibitor has a K i value of 0.85 ± 0.22 nM and binds in the catalytic site, interacting with the Zn2+ metal ion and with residues belonging to both chains of the dimer. The results provide new structural information about the mechanism of inhibition exerted by a novel class of compounds on the ACMSD enzyme, a novel therapeutic target for liver and kidney diseases.

Modulation of NAD+ biosynthesis activates SIRT1 and resists cisplatin-induced ototoxicity

Cisplatin, the most widely used platinum-based anticancer drug, often causes progressive and irreversible sensorineural hearing loss in cancer patients. However, the precise mechanism underlying cisplatin-associated ototoxicity is still unclear. Nicotinamide adenine dinucleotide (NAD+), a co-substrate for the sirtuin family and PARPs, has emerged as a potent therapeutic molecular target in various diseases. In our investigates, we observed that NAD+ level was changed in the cochlear explants of mice treated with cisplatin. Supplementation of a specific inhibitor (TES-1025) of α-amino-β-carboxymuconate-ε-semialdehyde decarboxylase (ACMSD), a rate-limiting enzyme of NAD+de novo synthesis pathway, promoted SIRT1 activity, increased mtDNA contents and enhanced AMPK expression, thus significantly reducing hair cells loss and deformation. The protection was blocked by EX527, a specific SIRT1 inhibitor. Meanwhile, the use of NMN, a precursor of NAD+ salvage synthesis pathway, had shown beneficial effect on hair cell under cisplatin administration, effectively suppressing PARP1. In vivo experiments confirmed the hair cell protection of NAD+ modulators in cisplatin treated mice and zebrafish. In conclusion, we demonstrated that modulation of NAD+ biosynthesis via the de novo synthesis pathway and the salvage synthesis pathway could both prevent ototoxicity of cisplatin. These results suggested that direct modulation of cellular NAD+ levels could be a promising therapeutic approach for protection of hearing from cisplatin-induced ototoxicity.

α-Amino-β-carboxymuconate-ε-semialdehyde Decarboxylase (ACMSD) Inhibitors as Novel Modulators of De Novo Nicotinamide Adenine Dinucleotide (NAD+) Biosynthesis

NAD+ has a central function in linking cellular metabolism to major cell-signaling and gene-regulation pathways. Defects in NAD+ homeostasis underpin a wide range of diseases, including cancer, metabolic disorders, and aging. Although the beneficial effects of boosting NAD+ on mitochondrial fitness, metabolism, and lifespan are well established, to date, no therapeutic enhancers of de novo NAD+ biosynthesis have been reported. Herein we report the discovery of 3-[[[5-cyano-1,6-dihydro-6-oxo-4-(2-thienyl)-2-pyrimidinyl]thio]methyl]phenylacetic acid (TES-1025, 22), the first potent and selective inhibitor of human ACMSD (IC50 = 0.013 μM) that increases NAD+ levels in cellular systems. The results of physicochemical-property, ADME, and safety profiling, coupled with in vivo target-engagement studies, support the hypothesis that ACMSD inhibition increases de novo NAD+ biosynthesis and position 22 as a first-class molecule for the evaluation of the therapeutic potential of ACMSD inhibition in treating disorders with perturbed NAD+ supply or homeostasis.

Adult zebrafish as an in vivo drug testing model for ethanol induced acute hepatic injury

Chronic alcohol abuse is common and a leading cause of alcoholic liver disease (ALD). However, a safe and effective therapy for ALD is still elusive. In this study, we evaluated the utility of adult zebrafish as an in vivo model for rapid assessment of drug efficacy in ethanol-induced acute hepatic injury. We exposed adult zebrafish to 0.5 % ethanol for 24, 48, and 72 hours and measured serum alanine aminotransferase (ALT) activities. This treatment resulted in a significant increase in ALT levels at 48 and 72 h of ethanol treatment, compared to untreated control groups. Accompanying this, significant increases in mRNA expression of genes associated with inflammation was observed in the liver during ethanol exposure. To evaluate the effectiveness of drug testing using our zebrafish model for ethanol-induced acute hepatic injury, we investigated the protective function of nicotinamide riboside, a substrate for NAD+, previously shown to be protective in a rodent model of alcoholic liver disease and TES-1025, an inhibitor of α-amino-β-carboxymuconate-ε-semialdehyde decarboxylase (ACMSD), that increases NAD+. We found that both nicotinamide riboside and TES-1025 treatment suppressed ethanol-induced serum ALT levels, post 48 h of ethanol exposure. In a similar manner, riboflavin supplementation also suppressed ethanol-induced serum ALT increase during ethanol exposure. Additionally, both nicotinamide riboside and riboflavin supplementation inhibited the upregulation of mRNA expression of genes associated with inflammation and de novo lipogenesis. In conclusion, we established an adult zebrafish model of ethanol-induced acute hepatic injury that will be valuable for cost-effective in vivo drug screening, which may in the future offer identification of novel therapeutics to mitigate hepatic injury, associated with excessive alcohol consumption.