Abstract

To assess the role of a protein, protein loss phenotypic studies can be used, most commonly through mutagenesis RNAi or CRISPR knockout. Such studies have been critical for the understanding of protein function and the identification of putative therapeutic targets for numerous human disease states. However, these methodological approaches present challenges because they are not easily reversible, and if an essential gene is targeted, an associated loss of cell viability can potentially hinder further studies. Here we present a reversible and conditional live‐cell knockout strategy that is applicable to numerous proteins. This modular protein‐tagging approach regulates target loss at the protein, rather than the genomic, level through the use of HaloPROTAC3, which specifically degrades HaloTag fusion proteins via recruitment of the VHL E3 ligase component. To enable HaloTag‐mediated degradation of endogenous proteins, we provide protocols for HaloTag genomic insertion at the protein N or C terminus via CRISPR/Cas9 and use of HaloTag fluorescent ligands to enrich edited cells via Fluorescence‐Activated Cell Sorting (FACS). Using these approaches, endogenous HaloTag fusion proteins present in various subcellular locations can be degraded by HaloPROTAC3. As detecting the degradation of endogenous targets is challenging, the 11‐amino‐acid peptide tag HiBiT is added to the HaloTag fusion to allows the sensitive luminescence detection of HaloTag fusion levels without the use of antibodies. Lastly, we demonstrate, through comparison of HaloPROTAC3 degradation with that of another fusion tag PROTAC, dTAG‐13, that HaloPROTAC3 has a faster degradation rate and similar extent of degradation. © 2020 The Authors. Basic Protocol 1: CRISPR/Cas9 insertion of HaloTag or HiBiT‐HaloTag Basic Protocol 2: HaloPROTAC3 degradation of endogenous HaloTag fusions

Highlights

  • proteolysis targeting chimeras (PROTACs) result in highly specific removal of target proteins from the cell and can be used to understand protein function and cellular phenotype upon protein loss (Crews, 2018; Deshaies, 2015), providing an excellent alternative to genetic CRISPR knockout or RNA interference (RNAi) strategies (Carthew & Sontheimer, 2009; Jackson & Linsley, 2010; PickarOliver & Gersbach, 2019)

  • PROTAC small molecules are heterobifunctional compounds that induce an interaction between a target protein with an E3 ligase component, most commonly von Hippel–Lindau (VHL) protein or cereblon (CRBN; Schapira, Calabrese, Bullock, & Crews, 2019)

  • To overcome these challenges and provide a more general solution for targeted proteindegradation studies, we developed the strategy of HaloPROTAC3 (Buckley et al, 2015), which degrades the 34-kDa monomeric protein fusion tag HaloTag (Encell et al, 2012; Los et al, 2008; Urh & Rosenberg, 2012)

Read more

Summary

INTRODUCTION

Targeted protein degradation using proteolysis targeting chimeras (PROTACs) is a rapidly growing research area and an exciting new modality of therapeutic treatment (Chamberlain & Hamann, 2019; Ciulli & Farnaby, 2019; Crews, 2018; Cromm & Crews, 2017; Daniels, Riching, & Urh, 2019; Deshaies, 2015; Luh et al, 2020). The HiBiT peptide, which complements with the LgBiT protein to generate NanoBiT luciferase, allows the luminescence detection of the endogenous target protein levels without the need for antibodies (Riching et al, 2018; Schwinn et al, 2018) In this unit we present two basic protocols, the first for CRISPR/Cas insertion of HaloTag or HiBiT-HaloTag, and the second for the degradation of endogenous HaloTag or HiBiT-HaloTag fusion proteins with HaloPROTAC3. Antibodies can be used to detect degradation, optional protocols are provided for luminescence detection of the endogenous HiBiT-HaloTag targets and their respective CRISPR insertion efficiencies and protein levels. Calculate total amounts of tracrRNA and Cas that will be needed for each crRNA reaction before beginning to ensure that proper amounts of each are available

Prepare 50 μM tracrRNA:crRNA duplex by combining the following
Background
Literature Cited
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