Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
ACS Chem Biol ; 14(11): 2430-2440, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31059647

ABSTRACT

Targeted protein degradation has arisen as a powerful strategy for drug discovery allowing the targeting of undruggable proteins for proteasomal degradation. This approach most often employs heterobifunctional degraders consisting of a protein-targeting ligand linked to an E3 ligase recruiter to ubiquitinate and mark proteins of interest for proteasomal degradation. One challenge with this approach, however, is that only a few E3 ligase recruiters currently exist for targeted protein degradation applications, despite the hundreds of known E3 ligases in the human genome. Here, we utilized activity-based protein profiling (ABPP)-based covalent ligand screening approaches to identify cysteine-reactive small-molecules that react with the E3 ubiquitin ligase RNF4 and provide chemical starting points for the design of RNF4-based degraders. The hit covalent ligand from this screen reacted with either of two zinc-coordinating cysteines in the RING domain, C132 and C135, with no effect on RNF4 activity. We further optimized the potency of this hit and incorporated this potential RNF4 recruiter into a bifunctional degrader linked to JQ1, an inhibitor of the BET family of bromodomain proteins. We demonstrate that the resulting compound CCW 28-3 is capable of degrading BRD4 in a proteasome- and RNF4-dependent manner. In this study, we have shown the feasibility of using chemoproteomics-enabled covalent ligand screening platforms to expand the scope of E3 ligase recruiters that can be exploited for targeted protein degradation applications.


Subject(s)
Coordination Complexes/chemistry , Nuclear Proteins/metabolism , Proteolysis/drug effects , Small Molecule Libraries/chemistry , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , Cell Cycle Proteins/metabolism , Coordination Complexes/metabolism , Cysteine/chemistry , Humans , Ligands , Molecular Docking Simulation , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Domains , Small Molecule Libraries/metabolism , Structure-Activity Relationship , Ubiquitination , Zinc/chemistry
2.
Cell Chem Biol ; 24(11): 1368-1376.e4, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-28919038

ABSTRACT

Many natural products that show therapeutic activities are often difficult to synthesize or isolate and have unknown targets, hindering their development as drugs. Identifying druggable hotspots targeted by covalently acting anti-cancer natural products can enable pharmacological interrogation of these sites with more synthetically tractable compounds. Here, we used chemoproteomic platforms to discover that the anti-cancer natural product withaferin A targets C377 on the regulatory subunit PPP2R1A of the tumor-suppressor protein phosphatase 2A (PP2A) complex leading to activation of PP2A activity, inactivation of AKT, and impaired breast cancer cell proliferation. We developed a more synthetically tractable cysteine-reactive covalent ligand, JNS 1-40, that selectively targets C377 of PPP2R1A to impair breast cancer signaling, proliferation, and in vivo tumor growth. Our study highlights the utility of using chemoproteomics to map druggable hotspots targeted by complex natural products and subsequently interrogating these sites with more synthetically tractable covalent ligands for cancer therapy.


Subject(s)
Antineoplastic Agents/metabolism , Biological Products/metabolism , Protein Phosphatase 2/metabolism , Amino Acid Sequence , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Biological Products/chemistry , Biological Products/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cysteine/chemistry , Female , Humans , Ligands , MCF-7 Cells , Protein Phosphatase 2/chemistry , Proteome/drug effects , Proteome/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Withanolides/chemistry , Withanolides/pharmacology
3.
ACS Chem Biol ; 12(6): 1478-1483, 2017 06 16.
Article in English | MEDLINE | ID: mdl-28445029

ABSTRACT

Most of the proteome is considered undruggable, oftentimes hindering translational efforts for drug discovery. Identifying previously unknown druggable hotspots in proteins would enable strategies for pharmacologically interrogating these sites with small molecules. Activity-based protein profiling (ABPP) has arisen as a powerful chemoproteomic strategy that uses reactivity-based chemical probes to map reactive, functional, and ligandable hotspots in complex proteomes, which has enabled inhibitor discovery against various therapeutic protein targets. Here, we report an alkyne-functionalized N-hydroxysuccinimide-ester (NHS-ester) as a versatile reactivity-based probe for mapping the reactivity of a wide range of nucleophilic ligandable hotspots, including lysines, serines, threonines, and tyrosines, encompassing active sites, allosteric sites, post-translational modification sites, protein interaction sites, and previously uncharacterized potential binding sites. Surprisingly, we also show that fragment-based NHS-ester ligands can be made to confer selectivity for specific lysine hotspots on specific targets including Dpyd, Aldh2, and Gstt1. We thus put forth NHS-esters as promising reactivity-based probes and chemical scaffolds for covalent ligand discovery.


Subject(s)
Binding Sites , Esters/chemistry , Ligands , Molecular Probes/chemistry , Proteome/metabolism , Aldehyde Dehydrogenase, Mitochondrial/metabolism , Alkynes/chemistry , Glutathione Transferase/metabolism , Humans , Lysine/metabolism , Succinimides
SELECTION OF CITATIONS
SEARCH DETAIL
...