Your browser doesn't support javascript.
loading
An Activity-Based Oxaziridine Platform for Identifying and Developing Covalent Ligands for Functional Allosteric Methionine Sites: Redox-Dependent Inhibition of Cyclin-Dependent Kinase 4.
Gonzalez-Valero, Angel; Reeves, Audrey G; Page, Annika C S; Moon, Patrick J; Miller, Edward; Coulonval, Katia; Crossley, Steven W M; Xie, Xiao; He, Dan; Musacchio, Patricia Z; Christian, Alec H; McKenna, Jeffrey M; Lewis, Richard A; Fang, Eric; Dovala, Dustin; Lu, Yipin; McGregor, Lynn M; Schirle, Markus; Tallarico, John A; Roger, Pierre P; Toste, F Dean; Chang, Christopher J.
Afiliação
  • Gonzalez-Valero A; Department of Chemistry, University of California, Berkeley, California 94720, United States.
  • Reeves AG; Department of Chemistry, University of California, Berkeley, California 94720, United States.
  • Page ACS; Department of Chemistry, University of California, Berkeley, California 94720, United States.
  • Moon PJ; Department of Chemistry, University of California, Berkeley, California 94720, United States.
  • Miller E; Department of Chemistry, University of California, Berkeley, California 94720, United States.
  • Coulonval K; Faculté de Médecine, Institute of Interdisciplinary Research, Université Libre de Bruxelles, Campus Erasme, Brussels 1070, Belgium.
  • Crossley SWM; Department of Chemistry, University of California, Berkeley, California 94720, United States.
  • Xie X; Department of Chemistry, University of California, Berkeley, California 94720, United States.
  • He D; Department of Chemistry, University of California, Berkeley, California 94720, United States.
  • Musacchio PZ; Department of Chemistry, University of California, Berkeley, California 94720, United States.
  • Christian AH; Department of Chemistry, University of California, Berkeley, California 94720, United States.
  • McKenna JM; Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States.
  • Lewis RA; Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States.
  • Fang E; Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States.
  • Dovala D; Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States.
  • Lu Y; Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States.
  • McGregor LM; Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States.
  • Schirle M; Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States.
  • Tallarico JA; Novartis Institutes for BioMedical Research, Cambridge, Massachusetts 02139, United States.
  • Roger PP; Faculté de Médecine, Institute of Interdisciplinary Research, Université Libre de Bruxelles, Campus Erasme, Brussels 1070, Belgium.
  • Toste FD; Department of Chemistry, University of California, Berkeley, California 94720, United States.
  • Chang CJ; Department of Chemistry, University of California, Berkeley, California 94720, United States.
J Am Chem Soc ; 144(50): 22890-22901, 2022 12 21.
Article em En | MEDLINE | ID: mdl-36484997
ABSTRACT
Activity-based protein profiling (ABPP) is a versatile strategy for identifying and characterizing functional protein sites and compounds for therapeutic development. However, the vast majority of ABPP methods for covalent drug discovery target highly nucleophilic amino acids such as cysteine or lysine. Here, we report a methionine-directed ABPP platform using Redox-Activated Chemical Tagging (ReACT), which leverages a biomimetic oxidative ligation strategy for selective methionine modification. Application of ReACT to oncoprotein cyclin-dependent kinase 4 (CDK4) as a representative high-value drug target identified three new ligandable methionine sites. We then synthesized a methionine-targeting covalent ligand library bearing a diverse array of heterocyclic, heteroatom, and stereochemically rich substituents. ABPP screening of this focused library identified 1oxF11 as a covalent modifier of CDK4 at an allosteric M169 site. This compound inhibited kinase activity in a dose-dependent manner on purified protein and in breast cancer cells. Further investigation of 1oxF11 found prominent cation-π and H-bonding interactions stabilizing the binding of this fragment at the M169 site. Quantitative mass-spectrometry studies validated 1oxF11 ligation of CDK4 in breast cancer cell lysates. Further biochemical analyses revealed cross-talk between M169 oxidation and T172 phosphorylation, where M169 oxidation prevented phosphorylation of the activating T172 site on CDK4 and blocked cell cycle progression. By identifying a new mechanism for allosteric methionine redox regulation on CDK4 and developing a unique modality for its therapeutic intervention, this work showcases a generalizable platform that provides a starting point for engaging in broader chemoproteomics and protein ligand discovery efforts to find and target previously undruggable methionine sites.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Neoplasias da Mama / Metionina Limite: Female / Humans Idioma: En Ano de publicação: 2022 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Neoplasias da Mama / Metionina Limite: Female / Humans Idioma: En Ano de publicação: 2022 Tipo de documento: Article