Molecular basis of the urate transporter URAT1 inhibition by gout drugs

Yang Suo, Justin Fedor, Han Zhang, Kalina Tsolova, Xiaoyu Shi, Kedar Sharma, Shweta Kumari, Mario J. Borgnia, Peng Zhan, Wonpil Im, Seok-Yong Lee
{"title":"Molecular basis of the urate transporter URAT1 inhibition by gout drugs","authors":"Yang Suo, Justin Fedor, Han Zhang, Kalina Tsolova, Xiaoyu Shi, Kedar Sharma, Shweta Kumari, Mario J. Borgnia, Peng Zhan, Wonpil Im, Seok-Yong Lee","doi":"10.1101/2024.09.11.612563","DOIUrl":null,"url":null,"abstract":"Hyperuricemia is a condition when uric acid, a waste product of purine metabolism, accumulates in the blood. Untreated hyperuricemia can lead to crystal formation of monosodium urate in the joints, causing a painful inflammatory disease known as gout. These conditions are associated with many other diseases and affect a significant and increasing proportion of the population. The human urate transporter 1 (URAT1) is responsible for the reabsorption of ~90% of uric acid in the kidneys back into the blood, making it a primary target for treating hyperuricemia and gout. Despite decades of research and development, clinically available URAT1 inhibitors have limitations because the molecular basis of URAT1 inhibition by gout drugs remains unknown5. Here we present cryo-electron microscopy structures of URAT1 alone and in complex with three clinically relevant inhibitors: benzbromarone, lesinurad, and the novel compound TD-3. Together with functional experiments and molecular dynamics simulations, we reveal that these inhibitors bind selectively to URAT1 in inward-open states. Furthermore, we discover differences in the inhibitor dependent URAT1 conformations as well as interaction networks, which contribute to drug specificity. Our findings illuminate a general theme for URAT1 inhibition, paving the way for the design of next-generation URAT1 inhibitors in the treatment of gout and hyperuricemia.","PeriodicalId":501048,"journal":{"name":"bioRxiv - Biophysics","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2024-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"bioRxiv - Biophysics","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1101/2024.09.11.612563","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

Abstract

Hyperuricemia is a condition when uric acid, a waste product of purine metabolism, accumulates in the blood. Untreated hyperuricemia can lead to crystal formation of monosodium urate in the joints, causing a painful inflammatory disease known as gout. These conditions are associated with many other diseases and affect a significant and increasing proportion of the population. The human urate transporter 1 (URAT1) is responsible for the reabsorption of ~90% of uric acid in the kidneys back into the blood, making it a primary target for treating hyperuricemia and gout. Despite decades of research and development, clinically available URAT1 inhibitors have limitations because the molecular basis of URAT1 inhibition by gout drugs remains unknown5. Here we present cryo-electron microscopy structures of URAT1 alone and in complex with three clinically relevant inhibitors: benzbromarone, lesinurad, and the novel compound TD-3. Together with functional experiments and molecular dynamics simulations, we reveal that these inhibitors bind selectively to URAT1 in inward-open states. Furthermore, we discover differences in the inhibitor dependent URAT1 conformations as well as interaction networks, which contribute to drug specificity. Our findings illuminate a general theme for URAT1 inhibition, paving the way for the design of next-generation URAT1 inhibitors in the treatment of gout and hyperuricemia.
痛风药物抑制尿酸盐转运体 URAT1 的分子基础
高尿酸血症是指尿酸(一种嘌呤代谢的废物)在血液中积聚。高尿酸血症如不及时治疗,会在关节中形成单钠尿酸盐结晶,引起疼痛性炎症,即痛风。这些疾病与许多其他疾病相关,影响着越来越多的人。人尿酸盐转运体 1(URAT1)负责将肾脏中约 90% 的尿酸重吸收回血液,因此成为治疗高尿酸血症和痛风的主要靶点。尽管经过数十年的研究和开发,临床上可用的URAT1抑制剂仍有局限性,因为痛风药物抑制URAT1的分子基础仍然未知5。在此,我们展示了URAT1单独和与三种临床相关抑制剂(苯溴马隆、来西脲德和新型化合物TD-3)复合物的冷冻电子显微镜结构。通过功能实验和分子动力学模拟,我们发现这些抑制剂在内向开放状态下选择性地与 URAT1 结合。此外,我们还发现了抑制剂依赖的 URAT1 构象以及相互作用网络的差异,这有助于药物的特异性。我们的发现阐明了URAT1抑制的一般主题,为设计治疗痛风和高尿酸血症的下一代URAT1抑制剂铺平了道路。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
自引率
0.00%
发文量
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信