Home>>Signaling Pathways>> Membrane Transporter/Ion Channel>> Sodium Channel>>RY785

RY785

目录号 : GC44858

A selective inhibitor of Kv2.1 channels

RY785 Chemical Structure

Cas No.:1393748-80-5

规格 价格 库存 购买数量
500μg
¥258.00
现货
1mg
¥489.00
现货
5mg
¥1,479.00
现货
10mg
¥2,549.00
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

RY785 is a selective inhibitor of Kv2.1 voltage-gated potassium channels (IC50 = 0.05 µM) that does not affect Cav2 calcium channels. It displays over 100-fold selectivity for Kv2.1 over Cav1.2 (IC50 = 17 µM).

Chemical Properties

Cas No. 1393748-80-5 SDF
Canonical SMILES COC1=CC=CC(CC(C(NC2=CC(N=C(C3=CSC=N3)N4)=C4C=C2)=O)C)=C1
分子式 C21H20N4O2S 分子量 392.5
溶解度 DMF: 20 mg/ml,DMSO: 15 mg/ml,Ethanol: 15 mg/ml 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 2.5478 mL 12.7389 mL 25.4777 mL
5 mM 0.5096 mL 2.5478 mL 5.0955 mL
10 mM 0.2548 mL 1.2739 mL 2.5478 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

Mechanism of use-dependent Kv2 channel inhibition by RY785

J Gen Physiol 2022 Jun 6;154(6):e202112981.PMID:35435946DOI:10.1085/jgp.202112981.

Understanding the mechanism by which ion channel modulators act is critical for interpretation of their physiological effects and can provide insight into mechanisms of ion channel gating. The small molecule RY785 is a potent and selective inhibitor of Kv2 voltage-gated K+ channels that has a use-dependent onset of inhibition. Here, we investigate the mechanism of RY785 inhibition of rat Kv2.1 (Kcnb1) channels heterologously expressed in CHO-K1 cells. We find that 1 µM RY785 block eliminates Kv2.1 current at all physiologically relevant voltages, inhibiting ≥98% of the Kv2.1 conductance. Both onset of and recovery from RY785 inhibition require voltage sensor activation. Intracellular tetraethylammonium, a classic open-channel blocker, competes with RY785 inhibition. However, channel opening itself does not appear to alter RY785 access. Gating current measurements reveal that RY785 inhibits a component of voltage sensor activation and accelerates voltage sensor deactivation. We propose that voltage sensor activation opens a path into the central cavity of Kv2.1 where RY785 binds and promotes voltage sensor deactivation, trapping itself inside. This gated-access mechanism in conjunction with slow kinetics of unblock supports simple interpretation of RY785 effects: channel activation is required for block by RY785 to equilibrate, after which trapped RY785 will simply decrease the Kv2 conductance density.

A Kv2 inhibitor traps itself in place

J Gen Physiol 2022 Jun 6;154(6):e202213181.PMID:35522189DOI:10.1085/jgp.202213181.

Voltage activation, but not channel opening, is required for RY785 to access the central cavity of Kv2 channels, where it promotes voltage sensor deactivation to trap itself in place.

Identification of novel and selective Kv2 channel inhibitors

Mol Pharmacol 2011 Dec;80(6):959-64.PMID:21948463DOI:10.1124/mol.111.074831.

Identification of selective ion channel inhibitors represents a critical step for understanding the physiological role that these proteins play in native systems. In particular, voltage-gated potassium (K(V)2) channels are widely expressed in tissues such as central nervous system, pancreas, and smooth muscle, but their particular contributions to cell function are not well understood. Although potent and selective peptide inhibitors of K(V)2 channels have been characterized, selective small molecule K(V)2 inhibitors have not been reported. For this purpose, high-throughput automated electrophysiology (IonWorks Quattro; Molecular Devices, Sunnyvale, CA) was used to screen a 200,000-compound mixture (10 compounds per sample) library for inhibitors of K(V)2.1 channels. After deconvolution of 190 active samples, two compounds (A1 and B1) were identified that potently inhibit K(V)2.1 and the other member of the K(V)2 family, K(V)2.2 (IC(50), 0.1-0.2 μM), and that possess good selectivity over K(V)1.2 (IC(50) >10 μM). Modeling studies suggest that these compounds possess a similar three-dimensional conformation. Compounds A1 and B1 are >10-fold selective over Na(V) channels and other K(V) channels and display weak activity (5-9 μM) on Ca(V) channels. The biological activity of compound A1 on native K(V)2 channels was confirmed in electrophysiological recordings of rat insulinoma cells, which are known to express K(V)2 channels. Medicinal chemistry efforts revealed a defined structure-activity relationship and led to the identification of two compounds (RY785 and RY796) without significant Ca(V) channel activity. Taken together, these newly identified channel inhibitors represent important tools for the study of K(V)2 channels in biological systems.