Abstract

Regulated necrosis or necroptosis, mediated by receptor-interacting kinase 1 (RIPK1), RIPK3 and pseudokinase mixed lineage kinase domain-like protein (MLKL), contributes to the pathogenesis of inflammatory, infectious and degenerative diseases. Recently identified necroptosis inhibitors display moderate specificity, suboptimal pharmacokinetics, off-target effects and toxicity, preventing these molecules from reaching the clinic. Here, we developed a cell-based high-throughput screening (HTS) cascade for the identification of small-molecule inhibitors of necroptosis. From the initial library of over 250,000 compounds, the primary screening phase identified 356 compounds that strongly inhibited TNF-α-induced necroptosis, but not apoptosis, in human and murine cell systems, with EC50 < 6.7 μM. From these, 251 compounds were tested for RIPK1 and/or RIPK3 kinase inhibitory activity; some were active and several have novel mechanisms of action. Based on specific chemical descriptors, 110 compounds proceeded into the secondary screening cascade, which then identified seven compounds with maximum ability to reduce MLKL activation, IC50 >100 μM, EC50 2.5–11.5 μM under long-term necroptosis execution in murine fibroblast L929 cells, and full protection from ATP depletion and membrane leakage in human and murine cells. As a proof of concept, compound SN-6109, with binding mode to RIPK1 similar to that of necrostatin-1, confirmed RIPK1 inhibitory activity and appropriate pharmacokinetic properties. SN-6109 was further tested in mice, showing efficacy against TNF-α-induced systemic inflammatory response syndrome. In conclusion, a phenotypic-driven HTS cascade promptly identified robust necroptosis inhibitors with in vivo activity, currently undergoing further medicinal chemistry optimization. Notably, the novel hits highlight the opportunity to identify new molecular mechanisms of action in necroptosis.

Highlights

  • Deregulation of apoptosis represents a key pathologic event in human disease, constituting an appealing therapeutic target[1]

  • AK enzyme release compared with total ATP levels exhibited higher sensitivity in reflecting TNFα-induced cell death, functioning as a reporter of cell lysis and necroptosis[19]

  • A total of 251,328 small-molecule compounds were screened in L929 cells incubated with mTNF-α alone or together with test compounds at 31.7 μM for 8 h

Read more

Summary

Introduction

Deregulation of apoptosis represents a key pathologic event in human disease, constituting an appealing therapeutic target[1]. Despite the benefits of blocking cell death, the scarcity of “druggable” targets in the apoptotic signalling pathway associated with safety concerns has cIAP 1/2, forming the membrane-associated protein complex I. IKKβ, TAB1/2/3/TAK1 and LUBAC are further recruited and activated, the NF-κB and MAPK survival pathways are activated. Receptor-interacting kinase 1 (RIPK1), FADD and caspase-8 form cytosolic complex IIa to activate the caspase cascade and induce apoptosis. When caspase-8 activity and/or survival signalling are inhibited, RIPK1/FADD interacts with RIPK3 and mixed lineage kinase domain-like protein (MLKL) to form the necrosome, or complex IIb. Mechanistically, phosphorylated MLKL triggers cell membrane permeabilization and consequent release of pro-inflammatory intracellular contents to the extracellular space[5]

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call