3.9 Article

Zafirlukast Is a Promising Scaffold for Selectively Inhibiting TNFR1 Signaling

期刊

ACS BIO & MED CHEM AU
卷 3, 期 3, 页码 270-282

出版社

AMER CHEMICAL SOC
DOI: 10.1021/acsbiomedchemau.2c00048

关键词

ligand-independent interactions; TNF; TNFR1; NF-kappa B activation; Inflammatory diseases

向作者/读者索取更多资源

In this study, analogues of zafirlukast were synthesized and tested for their efficacy and potency in TNFR1 signaling. Three analogues were identified with significantly improved efficacy and potency, each inducing a conformational change in the receptor. However, despite these improvements, the best candidate only achieved 63% inhibition of NF-kappa B, leaving room for further improvements. Interestingly, the analogues also bound to TNFR2 but did not inhibit its function or cause any conformational changes upon binding.
Tumor necrosis factor (TNF) plays an important role in the pathogenesis of inflammatory and autoimmune diseases such as rheumatoid arthritis and Crohn's disease. The biological effects of TNF are mediated by binding to TNF receptors, TNF receptor 1 (TNFR1), or TNF receptor 2 (TNFR2), and this coupling makes TNFR1-specific inhibition by small-molecule therapies essential to avoid deleterious side effects. Recently, we engineered a time-resolved fluorescence resonance energy transfer biosensor for high-throughput screening of small molecules that modulate TNFR1 conformational states and identified zafirlukast as a compound that inhibits receptor activation, albeit at low potency. Here, we synthesized 16 analogues of zafirlukast and tested their potency and specificity for TNFR1 signaling. Using cell-based functional assays, we identified three analogues with significantly improved efficacy and potency, each of which induces a conformational change in the receptor (as measured by fluorescence resonance energy transfer (FRET) in cells). The best analogue decreased NF-kappa B activation by 2.2-fold, I.Ba efficiency by 3.3-fold, and relative potency by two orders of magnitude. Importantly, we showed that the analogues do not block TNF binding to TNFR1 and that binding to the receptor's extracellular domain is strongly cooperative. Despite these improvements, the best candidate's maximum inhibition of NF-kappa B is only 63%, leaving room for further improvements to the zafirlukast scaffold to achieve full inhibition and prove its potential as a therapeutic lead. Interestingly, while we find that the analogues also bind to TNFR2 in vitro, they do not inhibit TNFR2 function in cells or cause any conformational changes upon binding. Thus, these lead compounds should also be used as reagents to study conformational-dependent activation of TNF receptors.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

3.9
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据