1
|
Skakuj K, Iglhaut M, Shao Q, Garcia FJ, Huang BY, Brittain SM, Nesvizhskii AI, Schirle M, Nomura DK, Toste FD. Light-Activated Reactivity of Nitrones with Amino Acids and Proteins. Angew Chem Int Ed Engl 2024:e202415976. [PMID: 39509590 DOI: 10.1002/anie.202415976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/02/2024] [Accepted: 11/06/2024] [Indexed: 11/15/2024]
Abstract
Controlled modifications of amino acids are an indispensable tool for advancing fundamental and translational research based on peptides and proteins. Yet, we still lack methods to chemically modify each naturally occurring amino acid sidechain. To help address this gap, we show that N,α-diaryl oxaziridines expand the scope of bioconjugation methods to chemically modify cysteine, methionine, and tryptophan residues with evidence for additional tyrosine labelling in a proteomic context. Conjugation primarily at tryptophan sites can be accessed by selective cleavage of modifications at other sidechains. The N,α-diaryl oxaziridine reagents are accessed through photoisomerization of nitrones, which serve as photocaged reagents, thus providing an additional level of control over reactivity. Initial guiding principles for the design of nitrone reagents are developed by exploring the impact of structure on UV/Vis absorption, photoisomerization, and reactivity. We identify a nitrone structure that maximizes photoisomerization efficiency, the aqueous stability of the oxaziridine, the extent of amino acid modification, and the stability of the resulting amino acid conjugates. We then translate nitrone reagents to modify proteins in aqueous conditions. Finally, we use nitrones to profile reactive residues across the proteome of a mammalian cell line and find that they expand the proteome coverage.
Collapse
Affiliation(s)
- Kacper Skakuj
- Department of Chemistry, University of California, Berkeley, Berkeley, CA-94720, USA
| | - Maximilian Iglhaut
- Department of Chemistry, University of California, Berkeley, Berkeley, CA-94720, USA
| | - Qian Shao
- Department of Chemistry, University of California, Berkeley, Berkeley, CA-94720, USA
| | | | - Bo-Yang Huang
- Department of Chemistry, University of California, Berkeley, Berkeley, CA-94720, USA
| | | | - Alexey I Nesvizhskii
- Department of Pathology, Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Ann Arbor, Michigan, 48109, USA
| | | | - Daniel K Nomura
- Department of Chemistry, University of California, Berkeley, Berkeley, CA-94720, USA
| | - F Dean Toste
- Department of Chemistry, University of California, Berkeley, Berkeley, CA-94720, USA
| |
Collapse
|
2
|
Cheung ST, Kim Y, Cho JH, Brandvold KR, Ghosh B, Del Rosario AM, Bell-Temin H. End-to-End Throughput Chemical Proteomics for Photoaffinity Labeling Target Engagement and Deconvolution. J Proteome Res 2024; 23:4951-4961. [PMID: 39374182 DOI: 10.1021/acs.jproteome.4c00442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
Photoaffinity labeling (PAL) methodologies have proven to be instrumental for the unbiased deconvolution of protein-ligand binding events in physiologically relevant systems. However, like other chemical proteomic workflows, they are limited in many ways by time-intensive sample manipulations and data acquisition techniques. Here, we describe an approach to address this challenge through the innovation of a carboxylate bead-based protein cleanup procedure to remove excess small-molecule contaminants and couple it to plate-based, proteomic sample processing as a semiautomated solution. The analysis of samples via label-free, data-independent acquisition (DIA) techniques led to significant improvements on a workflow time per sample basis over current standard practices. Experiments utilizing three established PAL ligands with known targets, (+)-JQ-1, lenalidomide, and dasatinib, demonstrated the utility of having the flexibility to design experiments with a myriad of variables. Data revealed that this workflow can enable the confident identification and rank ordering of known and putative targets with outstanding protein signal-to-background enrichment sensitivity. This unified end-to-end throughput strategy for processing and analyzing these complex samples could greatly facilitate efficient drug discovery efforts and open up new opportunities in the chemical proteomics field.
Collapse
Affiliation(s)
- Sheldon T Cheung
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Yongkang Kim
- Janssen Research & Development, LLC, 301 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Ji-Hoon Cho
- Janssen Research & Development, LLC, 301 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Kristoffer R Brandvold
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Brahma Ghosh
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Amanda M Del Rosario
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Harris Bell-Temin
- Janssen Research & Development, LLC, 301 Binney Street, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
3
|
Wozniak JM, Li W, Parker CG. Chemical proteomic mapping of reversible small molecule binding sites in native systems. Trends Pharmacol Sci 2024; 45:969-981. [PMID: 39406592 DOI: 10.1016/j.tips.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/16/2024] [Accepted: 09/16/2024] [Indexed: 11/10/2024]
Abstract
The impact of small molecules in human biology are manifold; not only are they critical regulators of physiological processes, but they also serve as probes to investigate biological pathways and leads for therapeutic development. Identifying the protein targets of small molecules, and where they bind, is critical to understanding their functional consequences and potential for pharmacological use. Over the past two decades, chemical proteomics has emerged as a go-to strategy for the comprehensive mapping of small molecule-protein interactions. Recent advancements in this field, particularly innovations of photoaffinity labeling (PAL)-based methods, have enabled the robust identification of small molecule binding sites on protein targets, often in live cells. In this opinion article, we examine these advancements as well as reflect on how their strategic integration with other emerging tools can advance therapeutic development.
Collapse
Affiliation(s)
| | - Weichao Li
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | | |
Collapse
|
4
|
Fu S, Chen Z, Luo Z, Nie M, Fu T, Zhou Y, Yang Q, Zhu F, Ni F. Chem(Pro)2: the atlas of chemoproteomic probes labelling human proteins. Nucleic Acids Res 2024:gkae943. [PMID: 39436046 DOI: 10.1093/nar/gkae943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/25/2024] [Accepted: 10/11/2024] [Indexed: 10/23/2024] Open
Abstract
Chemoproteomic probes (CPPs) have been widely considered as powerful molecular biological tools that enable the highly efficient discovery of both binding proteins and modes of action for the studied compounds. They have been successfully used to validate targets and identify binders. The design of CPP has been considered extremely challenging, which asks for the generalization using a large number of probe data. However, none of the existing databases gives such valuable data of CPPs. Herein, a database entitled 'Chem(Pro)2' was therefore developed to systematically describe the atlas of diverse types of CPPs labelling human protein in living cell/lysate. With the booming application of chemoproteomic technique and artificial intelligence in current chemical biology study, Chem(Pro)2 was expected to facilitate the AI-based learning of interacting pattern among molecules for discovering innovative targets and new drugs. Till now, Chem(Pro)2 has been open to all users without any login requirement at: https://idrblab.org/chemprosquare/.
Collapse
Affiliation(s)
- Songsen Fu
- Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, China
- LeadArt Biotechnologies Ltd., Ningbo 315201, China
| | - Zhen Chen
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Zhiming Luo
- LeadArt Biotechnologies Ltd., Ningbo 315201, China
| | - Meiyun Nie
- LeadArt Biotechnologies Ltd., Ningbo 315201, China
| | - Tingting Fu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Ying Zhou
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Qingxia Yang
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| | - Feng Ni
- Institute of Drug Discovery Technology, Ningbo University, Ningbo 315211, China
- LeadArt Biotechnologies Ltd., Ningbo 315201, China
| |
Collapse
|
5
|
Jackson PA, Kisty E, Pradhan V, Swank C, Bohrer L, Nolan TL, Weerapana E, Lapinsky DJ. Appendage- and Scaffold-Diverse Electrophilic and Photoreactive Probes for Integrated Phenotypic Screening-Target Identification Campaigns via a Minimalist Bifunctional Isocyanide. ACS OMEGA 2024; 9:42557-42570. [PMID: 39431108 PMCID: PMC11483914 DOI: 10.1021/acsomega.4c06879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/16/2024] [Accepted: 09/20/2024] [Indexed: 10/22/2024]
Abstract
One of the grand challenges in chemical biology is identifying a small-molecule modulator for all proteins within a proteome. To expand the variety and number of ligandable proteins for drug discovery, the objective of this study was to synthesize and evaluate the protein target profiles of electrophilic and photoreactive fully functionalized small-molecule probes (FFSMPs) featuring increased scaffold-, appendage-, and protein-reactive functional group (PRFG) diversity. FFSMPs contain: (1) a protein-binding motif, (2) an electrophilic or photoreactive PRFG for target protein capture, and (3) a terminal alkyne for click chemistry-based proteomic applications. These compounds can be directly applied in phenotypic screening programs to identify ligand-protein pairs in cells unbiasedly. Herein, we highlight 17 examples from 34 structurally diverse FFSMPs featuring five electrophiles, three photoreactive groups, and 15 chemical scaffolds. Essential to the synthesis of the FFSMPs was a new minimalist bifunctional isocyanide in an "isocyanide-based multicomponent reaction-Boc deprotection-arming" synthetic sequence. To the best of our knowledge, this is the first report concerning the preparation of appendage- and scaffold-diverse FFSMPs for integrated phenotypic screening-target identification campaigns with the ability to examine either electrophilic or photoreactive PRFGs. In contrast, the status quo for such studies has been appendage-diverse FFSMPs comprised of a single chemical scaffold and a single PRFG, which limits efficient target protein capture and/or chemical space sampling significantly in the quest for discovering new drug targets and/or compounds with novel mechanisms of action. Phenotypic screening of the electrophilic members of our library identified several FFSMPs with potent antiproliferative activity against MCF10CA1a breast cancer cells. One of these FFSMPs (Compound 4a) covalently targeted and potently inhibited protein disulfide isomerase A1 (PDIA1). This study supports the continued use of minimalist bifunctional isocyanides as valuable building blocks for preparing structurally diverse FFSMPs for integrated phenotypic screening-target identification campaigns.
Collapse
Affiliation(s)
- Paul A. Jackson
- Graduate
School of Pharmaceutical Sciences, Duquesne
University, 600 Forbes Avenue, Pittsburgh, Pennsylvania 15282, United States
| | - Eleni Kisty
- Department
of Chemistry, Boston College, Merkert Chemistry
Center, 2609 Beacon Street, Chestnut Hill, Massachusetts 02464, United States
| | - Vandan Pradhan
- Graduate
School of Pharmaceutical Sciences, Duquesne
University, 600 Forbes Avenue, Pittsburgh, Pennsylvania 15282, United States
| | - Christopher Swank
- Graduate
School of Pharmaceutical Sciences, Duquesne
University, 600 Forbes Avenue, Pittsburgh, Pennsylvania 15282, United States
| | - Luke Bohrer
- Graduate
School of Pharmaceutical Sciences, Duquesne
University, 600 Forbes Avenue, Pittsburgh, Pennsylvania 15282, United States
| | - Tammy L. Nolan
- Graduate
School of Pharmaceutical Sciences, Duquesne
University, 600 Forbes Avenue, Pittsburgh, Pennsylvania 15282, United States
| | - Eranthie Weerapana
- Department
of Chemistry, Boston College, Merkert Chemistry
Center, 2609 Beacon Street, Chestnut Hill, Massachusetts 02464, United States
| | - David J. Lapinsky
- Graduate
School of Pharmaceutical Sciences, Duquesne
University, 600 Forbes Avenue, Pittsburgh, Pennsylvania 15282, United States
| |
Collapse
|
6
|
Bon C, Goretzki B, Flamme M, Shelton C, Davis H, Lima F, Garcia F, Brittain S, Brocklehurst CE. Oxadiazolines as Photoreleasable Labels for Drug Target Identification. J Am Chem Soc 2024; 146:26759-26765. [PMID: 39288302 DOI: 10.1021/jacs.4c06936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Photoaffinity labeling is a widely used technique for studying ligand-protein and protein-protein interactions. Traditional photoaffinity labels utilize nonspecific C-H bond insertion reactions mediated by a highly reactive intermediate. Despite being the most widely used photoaffinity labels, diazirines exhibit limited compatibility with downstream organic reactions and suffer from storage stability concerns. This study introduces oxadiazolines as innovative and complementary photoactivatable labels for addition to the toolbox and demonstrates their application in vitro and through in cellulo labeling experiments. Oxadiazolines can be easily synthesized from ketone moieties and cleaved with 302-330 nm light to cleanly liberate a diazo reactive moiety that can covalently modify nucleophilic amino acid residues. Notably, oxadiazolines are compatible with various organic reaction conditions and functional groups, allowing for the exploration of a large chemical space. Several known inhibitors featuring the oxadiazoline functionality were prepared without affecting their binding affinity. Furthermore, we confirmed the ability of oxadiazolines to form covalent bonds with proteins upon UV-irradiation, both in vitro and in cellulo, yielding comparable results to those of the matched diazirine compounds.
Collapse
Affiliation(s)
- Corentin Bon
- Global Discovery Chemistry, Novartis Biomedical Research, Novartis Pharma AG, Basel 4056, Switzerland
| | - Benedikt Goretzki
- Discovery Sciences, Novartis Biomedical Research, Novartis Pharma AG, Basel 4056, Switzerland
| | - Marie Flamme
- Chemical and Analytical Development, Novartis Development, Novartis Pharma AG, Basel 4056, Switzerland
| | - Claude Shelton
- Discovery Sciences, Novartis Biomedical Research, Novartis Pharma AG, Cambridge, Massachusetts 02139, United States
| | - Holly Davis
- Global Discovery Chemistry, Novartis Biomedical Research, Novartis Pharma AG, Basel 4056, Switzerland
| | - Fabio Lima
- Global Discovery Chemistry, Novartis Biomedical Research, Novartis Pharma AG, Basel 4056, Switzerland
| | - Francisco Garcia
- Discovery Sciences, Novartis Biomedical Research, Novartis Pharma AG, Cambridge, Massachusetts 02139, United States
| | - Scott Brittain
- Discovery Sciences, Novartis Biomedical Research, Novartis Pharma AG, Cambridge, Massachusetts 02139, United States
| | - Cara E Brocklehurst
- Global Discovery Chemistry, Novartis Biomedical Research, Novartis Pharma AG, Basel 4056, Switzerland
| |
Collapse
|
7
|
Chen K, Zhang L, Ding Y, Sun Z, Meng J, Luo R, Zhou X, Liu L, Yang S. Activity-based protein profiling in drug/pesticide discovery: Recent advances in target identification of antibacterial compounds. Bioorg Chem 2024; 151:107655. [PMID: 39032407 DOI: 10.1016/j.bioorg.2024.107655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 06/18/2024] [Accepted: 07/14/2024] [Indexed: 07/23/2024]
Abstract
Given the escalating incidence of bacterial diseases and the challenge posed by pathogenic bacterial resistance, it is imperative to identify appropriate methodologies for conducting proteomic investigations on bacteria, and thereby promoting the target-based drug/pesticide discovery. Interestingly, a novel technology termed "activity-based protein profiling" (ABPP) has been developed to identify the target proteins of active molecules. However, few studies have summarized advancements in ABPP for identifying the target proteins in antibacterial-active compounds. In order to accelerate the discovery and development of new drug/agrochemical discovery, we provide a concise overview of ABPP and its recent applications in antibacterial agent discovery. Diversiform cases were cited to demonstrate the potential of ABPP for target identification though highlighting the design strategies and summarizing the reported target protein of antibacterial compounds. Overall, this review is an excellent reference for probe design towards antibacterial compounds, and offers a new perspective of ABPP in bactericide development.
Collapse
Affiliation(s)
- Kunlun Chen
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Ling Zhang
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Yue Ding
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Zhaoju Sun
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Jiao Meng
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Rongshuang Luo
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Xiang Zhou
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China.
| | - Liwei Liu
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China
| | - Song Yang
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China.
| |
Collapse
|
8
|
Wiest A, Kielkowski P. Improved deconvolution of natural products' protein targets using diagnostic ions from chemical proteomics linkers. Beilstein J Org Chem 2024; 20:2323-2341. [PMID: 39290210 PMCID: PMC11406061 DOI: 10.3762/bjoc.20.199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 08/27/2024] [Indexed: 09/19/2024] Open
Abstract
Identification of interactions between proteins and natural products or similar active small molecules is crucial for understanding of their mechanism of action on a molecular level. To search elusive, often labile, and low-abundant conjugates between proteins and active compounds, chemical proteomics introduces a feasible strategy that allows to enrich and detect these conjugates. Recent advances in mass spectrometry techniques and search algorithms provide unprecedented depth of proteome coverage and the possibility to detect desired modified peptides with high sensitivity. The chemical 'linker' connecting an active compound-protein conjugate with a detection tag is the critical component of all chemical proteomic workflows. In this review, we discuss the properties and applications of different chemical proteomics linkers with special focus on their fragmentation releasing diagnostic ions and how these may improve the confidence in identified active compound-peptide conjugates. The application of advanced search options improves the identification rates and may help to identify otherwise difficult to find interactions between active compounds and proteins, which may result from unperturbed conditions, and thus are of high physiological relevance.
Collapse
Affiliation(s)
- Andreas Wiest
- LMU Munich, Department of Chemistry, Butenandtstr. 5-13, 81377 Munich, Germany
| | - Pavel Kielkowski
- LMU Munich, Department of Chemistry, Butenandtstr. 5-13, 81377 Munich, Germany
| |
Collapse
|
9
|
Ábrányi-Balogh P, Bajusz D, Orgován Z, Keeley AB, Petri L, Péczka N, Szalai TV, Pálfy G, Gadanecz M, Grant EK, Imre T, Takács T, Ranđelović I, Baranyi M, Marton A, Schlosser G, Ashraf QF, de Araujo ED, Karancsi T, Buday L, Tóvári J, Perczel A, Bush JT, Keserű GM. Mapping protein binding sites by photoreactive fragment pharmacophores. Commun Chem 2024; 7:168. [PMID: 39085342 PMCID: PMC11292009 DOI: 10.1038/s42004-024-01252-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 07/23/2024] [Indexed: 08/02/2024] Open
Abstract
Fragment screening is a popular strategy of generating viable chemical starting points especially for challenging targets. Although fragments provide a better coverage of chemical space and they have typically higher chance of binding, their weak affinity necessitates highly sensitive biophysical assays. Here, we introduce a screening concept that combines evolutionary optimized fragment pharmacophores with the use of a photoaffinity handle that enables high hit rates by LC-MS-based detection. The sensitivity of our screening protocol was further improved by a target-conjugated photocatalyst. We have designed, synthesized, and screened 100 diazirine-tagged fragments against three benchmark and three therapeutically relevant protein targets of different tractability. Our therapeutic targets included a conventional enzyme, the first bromodomain of BRD4, a protein-protein interaction represented by the oncogenic KRasG12D protein, and the yet unliganded N-terminal domain of the STAT5B transcription factor. We have discovered several fragment hits against all three targets and identified their binding sites via enzymatic digestion, structural studies and modeling. Our results revealed that this protocol outperforms screening traditional fully functionalized and photoaffinity fragments in better exploration of the available binding sites and higher hit rates observed for even difficult targets.
Collapse
Affiliation(s)
- Péter Ábrányi-Balogh
- Medicinal Chemistry Research Group, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- National Drug Research and Development Laboratory, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Budapest, Hungary
| | - Dávid Bajusz
- Medicinal Chemistry Research Group, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- National Drug Research and Development Laboratory, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
| | - Zoltán Orgován
- Medicinal Chemistry Research Group, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- National Drug Research and Development Laboratory, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
| | - Aaron B Keeley
- Medicinal Chemistry Research Group, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
| | - László Petri
- Medicinal Chemistry Research Group, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- National Drug Research and Development Laboratory, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
| | - Nikolett Péczka
- Medicinal Chemistry Research Group, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- National Drug Research and Development Laboratory, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Budapest, Hungary
| | - Tibor Viktor Szalai
- Medicinal Chemistry Research Group, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- National Drug Research and Development Laboratory, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Department of Inorganic and Analytical Chemistry, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Budapest, Hungary
| | - Gyula Pálfy
- Laboratory of Structural Chemistry and Biology & HUN-REN-ELTE Protein Modelling Research Group, Eötvös Loránd University, Budapest, Hungary
| | - Márton Gadanecz
- National Drug Research and Development Laboratory, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Laboratory of Structural Chemistry and Biology & HUN-REN-ELTE Protein Modelling Research Group, Eötvös Loránd University, Budapest, Hungary
- Hevesy György PhD School of Chemistry, Eötvös Loránd University, Budapest, Hungary
| | | | - Tímea Imre
- National Drug Research and Development Laboratory, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- MS Metabolomics Research Group, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
| | - Tamás Takács
- National Drug Research and Development Laboratory, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Signal Transduction and Functional Genomics Research Group, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Doctoral School of Biology, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Ivan Ranđelović
- National Tumor Biology Laboratory and Department of Experimental Pharmacology, National Institute of Oncology, Budapest, Hungary
- KINETO Lab Ltd, Budapest, Hungary
| | - Marcell Baranyi
- KINETO Lab Ltd, Budapest, Hungary
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - András Marton
- Department of Chemical and Environmental Process Engineering, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Budapest, Hungary
- Waters Research Center, Budapest, Hungary
| | - Gitta Schlosser
- MTA-ELTE Lendület Ion Mobility Mass Spectrometry Research Group, Eötvös Loránd University, Budapest, Hungary
| | - Qirat F Ashraf
- Department of Chemical & Physical Sciences, University of Toronto Mississauga, Mississauga, ON, Canada
| | - Elvin D de Araujo
- Centre for Medicinal Chemistry, University of Toronto at Mississauga, Mississauga, ON, Canada
| | - Tamás Karancsi
- Department of Chemical and Environmental Process Engineering, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Budapest, Hungary
- Waters Research Center, Budapest, Hungary
| | - László Buday
- National Drug Research and Development Laboratory, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Signal Transduction and Functional Genomics Research Group, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
| | - József Tóvári
- National Tumor Biology Laboratory and Department of Experimental Pharmacology, National Institute of Oncology, Budapest, Hungary
| | - András Perczel
- National Drug Research and Development Laboratory, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary
- Laboratory of Structural Chemistry and Biology & HUN-REN-ELTE Protein Modelling Research Group, Eötvös Loránd University, Budapest, Hungary
| | | | - György M Keserű
- Medicinal Chemistry Research Group, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary.
- National Drug Research and Development Laboratory, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary.
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Budapest, Hungary.
| |
Collapse
|
10
|
Wozniak JM, Li W, Governa P, Chen LY, Jadhav A, Dongre A, Forli S, Parker CG. Enhanced mapping of small-molecule binding sites in cells. Nat Chem Biol 2024; 20:823-834. [PMID: 38167919 PMCID: PMC11213684 DOI: 10.1038/s41589-023-01514-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 11/29/2023] [Indexed: 01/05/2024]
Abstract
Photoaffinity probes are routinely utilized to identify proteins that interact with small molecules. However, despite this common usage, resolving the specific sites of these interactions remains a challenge. Here we developed a chemoproteomic workflow to determine precise protein binding sites of photoaffinity probes in cells. Deconvolution of features unique to probe-modified peptides, such as their tendency to produce chimeric spectra, facilitated the development of predictive models to confidently determine labeled sites. This yielded an expansive map of small-molecule binding sites on endogenous proteins and enabled the integration with multiplexed quantitation, increasing the throughput and dimensionality of experiments. Finally, using structural information, we characterized diverse binding sites across the proteome, providing direct evidence of their tractability to small molecules. Together, our findings reveal new knowledge for the analysis of photoaffinity probes and provide a robust method for high-resolution mapping of reversible small-molecule interactions en masse in native systems.
Collapse
Affiliation(s)
- Jacob M Wozniak
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Weichao Li
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Paolo Governa
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Li-Yun Chen
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Appaso Jadhav
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Ashok Dongre
- Research and Development, Bristol-Myers Squibb Company, Princeton, NJ, USA
| | - Stefano Forli
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | | |
Collapse
|
11
|
Siriwongsup S, Schmoker AM, Ficarro SB, Marto JA, Kim J. Bioorthogonally activated reactive species for target identification. Chem 2024; 10:1306-1315. [PMID: 38617077 PMCID: PMC11008434 DOI: 10.1016/j.chempr.2024.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
A target identification platform derived from the bioorthogonal activation of reactive species is described. We explore the reactivity of halogenated enamine N-oxides and report that the previously undisclosed α,γ-halogenated enamine N-oxides can be reduced biooorthogonally by diboron reagents to produce highly electrophilic α,β-unsaturated haloiminium ions suitable for labeling a range of amino acid residues on proteins in a 1,2- or 1,4-fashion. Affinity labeling reagents bearing this motif enable ligand-directed protein modification and afford highly sensitive and selective target identification in unbiased chemoproteomics experiments. Target identification is supported in both cell lysate and live cells.
Collapse
Affiliation(s)
- Surached Siriwongsup
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Anna M. Schmoker
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
| | - Scott B. Ficarro
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Blais Proteomics Center, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
- Center for Emergent Drug Targets, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
| | - Jarrod A. Marto
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Blais Proteomics Center, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
- Center for Emergent Drug Targets, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
| | - Justin Kim
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
- Lead Contact
| |
Collapse
|
12
|
Burton NR, Backus KM. Functionalizing tandem mass tags for streamlining click-based quantitative chemoproteomics. Commun Chem 2024; 7:80. [PMID: 38600184 PMCID: PMC11006884 DOI: 10.1038/s42004-024-01162-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 03/27/2024] [Indexed: 04/12/2024] Open
Abstract
Mapping the ligandability or potential druggability of all proteins in the human proteome is a central goal of mass spectrometry-based covalent chemoproteomics. Achieving this ambitious objective requires high throughput and high coverage sample preparation and liquid chromatography-tandem mass spectrometry analysis for hundreds to thousands of reactive compounds and chemical probes. Conducting chemoproteomic screens at this scale benefits from technical innovations that achieve increased sample throughput. Here we realize this vision by establishing the silane-based cleavable linkers for isotopically-labeled proteomics-tandem mass tag (sCIP-TMT) proteomic platform, which is distinguished by early sample pooling that increases sample preparation throughput. sCIP-TMT pairs a custom click-compatible sCIP capture reagent that is readily functionalized in high yield with commercially available TMT reagents. Synthesis and benchmarking of a 10-plex set of sCIP-TMT reveal a substantial decrease in sample preparation time together with high coverage and high accuracy quantification. By screening a focused set of four cysteine-reactive electrophiles, we demonstrate the utility of sCIP-TMT for chemoproteomic target hunting, identifying 789 total liganded cysteines. Distinguished by its compatibility with established enrichment and quantification protocols, we expect sCIP-TMT will readily translate to a wide range of covalent chemoproteomic applications.
Collapse
Affiliation(s)
- Nikolas R Burton
- Department of Biological Chemistry, David Geffen School of Medicine, UCLA, Los Angeles CA, USA
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA
| | - Keriann M Backus
- Department of Biological Chemistry, David Geffen School of Medicine, UCLA, Los Angeles CA, USA.
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA.
- Molecular Biology Institute, UCLA, Los Angeles, CA, USA.
- DOE Institute for Genomics and Proteomics, UCLA, Los Angeles, CA, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA.
| |
Collapse
|
13
|
Csorba N, Ábrányi-Balogh P, Keserű GM. Covalent fragment approaches targeting non-cysteine residues. Trends Pharmacol Sci 2023; 44:802-816. [PMID: 37770315 DOI: 10.1016/j.tips.2023.08.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/30/2023]
Abstract
Covalent fragment approaches combine advantages of covalent binders and fragment-based drug discovery (FBDD) for target identification and validation. Although early applications focused mostly on cysteine labeling, the chemistries of available warheads that target other orthosteric and allosteric protein nucleophiles has recently been extended. The range of different warheads and labeling chemistries provide unique opportunities for screening and optimizing warheads necessary for targeting non-cysteine residues. In this review, we discuss these recently developed amino-acid-specific and promiscuous warheads, as well as emerging labeling chemistries, which includes novel transition metal catalyzed, photoactive, electroactive, and noncatalytic methodologies. We also highlight recent applications of covalent fragments for the development of molecular glues and proteolysis-targeting chimeras (PROTACs), and their utility in chemical proteomics-based target identification and validation.
Collapse
Affiliation(s)
- Noémi Csorba
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, Magyar tudósok krt. 2, 1117, Budapest, Hungary; National Laboratory for Drug Research and Development, Research Centre for Natural Sciences, Magyar tudósok krt. 2, 1117, Budapest, Hungary; Department of Organic Chemistry and Technology, Budapest University of Technology and Economics, Szent Gellért tér 4, 1111 Budapest, Hungary
| | - Péter Ábrányi-Balogh
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, Magyar tudósok krt. 2, 1117, Budapest, Hungary; National Laboratory for Drug Research and Development, Research Centre for Natural Sciences, Magyar tudósok krt. 2, 1117, Budapest, Hungary; Department of Organic Chemistry and Technology, Budapest University of Technology and Economics, Szent Gellért tér 4, 1111 Budapest, Hungary
| | - György M Keserű
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, Magyar tudósok krt. 2, 1117, Budapest, Hungary; National Laboratory for Drug Research and Development, Research Centre for Natural Sciences, Magyar tudósok krt. 2, 1117, Budapest, Hungary; Department of Organic Chemistry and Technology, Budapest University of Technology and Economics, Szent Gellért tér 4, 1111 Budapest, Hungary.
| |
Collapse
|
14
|
Wichroski M, Benci J, Liu SQ, Chupak L, Fang J, Cao C, Wang C, Onorato J, Qiu H, Shan Y, Banas D, Powles R, Locke G, Witt A, Stromko C, Qi H, Zheng X, Martin S, Ding M, Gentles R, Meanwell N, Velaparthi U, Olson R, Wee S, Tenney D, Parker CG, Cravatt BF, Lawrence M, Borzilleri R, Lees E. DGKα/ζ inhibitors combine with PD-1 checkpoint therapy to promote T cell-mediated antitumor immunity. Sci Transl Med 2023; 15:eadh1892. [PMID: 37878674 DOI: 10.1126/scitranslmed.adh1892] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 10/04/2023] [Indexed: 10/27/2023]
Abstract
Programmed cell death protein 1 (PD-1) immune checkpoint blockade therapy has revolutionized cancer treatment. Although PD-1 blockade is effective in a subset of patients with cancer, many fail to respond because of either primary or acquired resistance. Thus, next-generation strategies are needed to expand the depth and breadth of clinical responses. Toward this end, we designed a human primary T cell phenotypic high-throughput screening strategy to identify small molecules with distinct and complementary mechanisms of action to PD-1 checkpoint blockade. Through these efforts, we selected and optimized a chemical series that showed robust potentiation of T cell activation and combinatorial activity with αPD-1 blockade. Target identification was facilitated by chemical proteomic profiling with a lipid-based photoaffinity probe, which displayed enhanced binding to diacylglycerol kinase α (DGKα) in the presence of the active compound, a phenomenon that correlated with the translocation of DGKα to the plasma membrane. We further found that optimized leads within this chemical series were potent and selective inhibitors of both DGKα and DGKζ, lipid kinases that constitute an intracellular T cell checkpoint that blunts T cell signaling through diacylglycerol metabolism. We show that dual DGKα/ζ inhibition amplified suboptimal T cell receptor signaling mediated by low-affinity antigen presentation and low major histocompatibility complex class I expression on tumor cells, both hallmarks of resistance to PD-1 blockade. In addition, DGKα/ζ inhibitors combined with αPD-1 therapy to elicit robust tumor regression in syngeneic mouse tumor models. Together, these findings support targeting DGKα/ζ as a next-generation T cell immune checkpoint strategy.
Collapse
Affiliation(s)
- Michael Wichroski
- Research and Development, Bristol Myers Squibb Company, Cambridge, MA 02142, USA
| | - Joseph Benci
- Research and Development, Bristol Myers Squibb Company, Lawrenceville, NJ 08648, USA
| | - Si-Qi Liu
- Research and Development, Bristol Myers Squibb Company, Cambridge, MA 02142, USA
| | - Louis Chupak
- Research and Development, Bristol Myers Squibb Company, Cambridge, MA 02142, USA
| | - Jie Fang
- Research and Development, Bristol Myers Squibb Company, Lawrenceville, NJ 08648, USA
| | - Carolyn Cao
- Research and Development, Bristol Myers Squibb Company, Lawrenceville, NJ 08648, USA
| | - Cindy Wang
- Research and Development, Bristol Myers Squibb Company, Lawrenceville, NJ 08648, USA
| | - Joelle Onorato
- Research and Development, Bristol Myers Squibb Company, Lawrenceville, NJ 08648, USA
| | - Hongchen Qiu
- Research and Development, Bristol Myers Squibb Company, Cambridge, MA 02142, USA
| | - Yongli Shan
- Research and Development, Bristol Myers Squibb Company, Cambridge, MA 02142, USA
| | - Dana Banas
- Research and Development, Bristol Myers Squibb Company, Lawrenceville, NJ 08648, USA
| | - Ryan Powles
- Research and Development, Bristol Myers Squibb Company, Cambridge, MA 02142, USA
| | - Gregory Locke
- Research and Development, Bristol Myers Squibb Company, Lawrenceville, NJ 08648, USA
| | - Abigail Witt
- Research and Development, Bristol Myers Squibb Company, Cambridge, MA 02142, USA
| | - Caitlyn Stromko
- Research and Development, Bristol Myers Squibb Company, Lawrenceville, NJ 08648, USA
| | - Huilin Qi
- Research and Development, Bristol Myers Squibb Company, Cambridge, MA 02142, USA
| | - Xiaofan Zheng
- Research and Development, Bristol Myers Squibb Company, Cambridge, MA 02142, USA
| | - Scott Martin
- Research and Development, Bristol Myers Squibb Company, Cambridge, MA 02142, USA
| | - Min Ding
- Research and Development, Bristol Myers Squibb Company, Cambridge, MA 02142, USA
| | - Robert Gentles
- Research and Development, Bristol Myers Squibb Company, Cambridge, MA 02142, USA
| | - Nicholas Meanwell
- Research and Development, Bristol Myers Squibb Company, Lawrenceville, NJ 08648, USA
| | - Upender Velaparthi
- Research and Development, Bristol Myers Squibb Company, Lawrenceville, NJ 08648, USA
| | - Richard Olson
- Research and Development, Bristol Myers Squibb Company, Cambridge, MA 02142, USA
| | - Susan Wee
- Research and Development, Bristol Myers Squibb Company, Lawrenceville, NJ 08648, USA
| | - Daniel Tenney
- Research and Development, Bristol Myers Squibb Company, Lawrenceville, NJ 08648, USA
| | | | - Benjamin F Cravatt
- Department of Chemistry, Scripps Research Institute, La Jolla, CA 92037, USA
| | - Michael Lawrence
- Research and Development, Bristol Myers Squibb Company, Lawrenceville, NJ 08648, USA
| | - Robert Borzilleri
- Research and Development, Bristol Myers Squibb Company, Lawrenceville, NJ 08648, USA
| | - Emma Lees
- Research and Development, Bristol Myers Squibb Company, Cambridge, MA 02142, USA
| |
Collapse
|
15
|
Burton NR, Polasky DA, Shikwana F, Ofori S, Yan T, Geiszler DJ, Veiga Leprevost FD, Nesvizhskii AI, Backus KM. Solid-Phase Compatible Silane-Based Cleavable Linker Enables Custom Isobaric Quantitative Chemoproteomics. J Am Chem Soc 2023; 145:21303-21318. [PMID: 37738129 DOI: 10.1021/jacs.3c05797] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/24/2023]
Abstract
Mass spectrometry-based chemoproteomics has emerged as an enabling technology for functional biology and drug discovery. To address limitations of established chemoproteomics workflows, including cumbersome reagent synthesis and low throughput sample preparation, here, we established the silane-based cleavable isotopically labeled proteomics (sCIP) method. The sCIP method is enabled by a high yielding and scalable route to dialkoxydiphenylsilane fluorenylmethyloxycarbonyl (DADPS-Fmoc)-protected amino acid building blocks, which enable the facile synthesis of customizable, isotopically labeled, and chemically cleavable biotin capture reagents. sCIP is compatible with both MS1- and MS2-based quantitation, and the sCIP-MS2 method is distinguished by its click-assembled isobaric tags in which the reporter group is encoded in the sCIP capture reagent and balancer in the pan cysteine-reactive probe. The sCIP-MS2 workflow streamlines sample preparation with early stage isobaric labeling and sample pooling, allowing for high coverage and increased sample throughput via customized low cost six-plex sample multiplexing. When paired with a custom FragPipe data analysis workflow and applied to cysteine-reactive fragment screens, sCIP proteomics revealed established and unprecedented cysteine-ligand pairs, including the discovery that mitochondrial uncoupling agent FCCP acts as a covalent-reversible cysteine-reactive electrophile.
Collapse
Affiliation(s)
- Nikolas R Burton
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095, United States
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Daniel A Polasky
- Department of Pathology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Flowreen Shikwana
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095, United States
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Samuel Ofori
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Tianyang Yan
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095, United States
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Daniel J Geiszler
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109, United States
| | | | - Alexey I Nesvizhskii
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Pathology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Keriann M Backus
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095, United States
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California 90095, United States
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California 90095, United States
- DOE Institute for Genomics and Proteomics, University of California, Los Angeles, Los Angeles, California 90095, United States
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, California 90095, United States
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California 90095, United States
| |
Collapse
|
16
|
Tang J, Li W, Chiu TY, Martínez-Peña F, Luo Z, Chong CT, Wei Q, Gazaniga N, West TJ, See YY, Lairson LL, Parker CG, Baran PS. Synthesis of portimines reveals the basis of their anti-cancer activity. Nature 2023; 622:507-513. [PMID: 37730997 PMCID: PMC10699793 DOI: 10.1038/s41586-023-06535-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 08/11/2023] [Indexed: 09/22/2023]
Abstract
Marine-derived cyclic imine toxins, portimine A and portimine B, have attracted attention because of their chemical structure and notable anti-cancer therapeutic potential1-4. However, access to large quantities of these toxins is currently not feasible, and the molecular mechanism underlying their potent activity remains unknown until now. To address this, a scalable and concise synthesis of portimines is presented, which benefits from the logic used in the two-phase terpenoid synthesis5,6 along with other tactics such as exploiting ring-chain tautomerization and skeletal reorganization to minimize protecting group chemistry through self-protection. Notably, this total synthesis enabled a structural reassignment of portimine B and an in-depth functional evaluation of portimine A, revealing that it induces apoptosis selectively in human cancer cell lines with high potency and is efficacious in vivo in tumour-clearance models. Finally, practical access to the portimines and their analogues simplified the development of photoaffinity analogues, which were used in chemical proteomic experiments to identify a primary target of portimine A as the 60S ribosomal export protein NMD3.
Collapse
Affiliation(s)
- Junchen Tang
- Department of Chemistry, Scripps Research, La Jolla, CA, USA
| | - Weichao Li
- Department of Chemistry, Scripps Research, La Jolla, CA, USA
| | - Tzu-Yuan Chiu
- Department of Chemistry, Scripps Research, La Jolla, CA, USA
| | | | - Zengwei Luo
- Department of Chemistry, Scripps Research, La Jolla, CA, USA
| | | | - Qijia Wei
- Department of Chemistry, Scripps Research, La Jolla, CA, USA
| | | | - Thomas J West
- Department of Chemistry, Scripps Research, La Jolla, CA, USA
| | - Yi Yang See
- Department of Chemistry, Scripps Research, La Jolla, CA, USA
| | - Luke L Lairson
- Department of Chemistry, Scripps Research, La Jolla, CA, USA.
| | | | - Phil S Baran
- Department of Chemistry, Scripps Research, La Jolla, CA, USA.
| |
Collapse
|
17
|
Kawamata Y, Ryu KA, Hermann GN, Sandahl A, Vantourout JC, Olow AK, Adams LTA, Rivera-Chao E, Roberts LR, Gnaim S, Nassir M, Oslund RC, Fadeyi OO, Baran PS. An electroaffinity labelling platform for chemoproteomic-based target identification. Nat Chem 2023; 15:1267-1275. [PMID: 37322100 DOI: 10.1038/s41557-023-01240-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 05/12/2023] [Indexed: 06/17/2023]
Abstract
Target identification involves deconvoluting the protein target of a pharmacologically active, small-molecule ligand, a process that is critical for early drug discovery yet technically challenging. Photoaffinity labelling strategies have become the benchmark for small-molecule target deconvolution, but covalent protein capture requires the use of high-energy ultraviolet light, which can complicate downstream target identification. Thus, there is a strong demand for alternative technologies that allow for controlled activation of chemical probes to covalently label their protein target. Here we introduce an electroaffinity labelling platform that leverages the use of a small, redox-active diazetidinone functional group to enable chemoproteomic-based target identification of pharmacophores within live cell environments. The underlying discovery to enable this platform is that the diazetidinone can be electrochemically oxidized to reveal a reactive intermediate useful for covalent modification of proteins. This work demonstrates the electrochemical platform to be a functional tool for drug-target identification.
Collapse
Affiliation(s)
- Yu Kawamata
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Keun Ah Ryu
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA, USA
| | - Gary N Hermann
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Alexander Sandahl
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | | | - Aleksandra K Olow
- Genetics and Pharmacogenomics, Merck & Co., Inc., San Francisco, CA, USA
| | | | - Eva Rivera-Chao
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Lee R Roberts
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA, USA
| | - Samer Gnaim
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Molhm Nassir
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Rob C Oslund
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA, USA.
- InduPro Therapeutics, Cambridge, MA, USA.
| | - Olugbeminiyi O Fadeyi
- Exploratory Science Center, Merck & Co., Inc., Cambridge, MA, USA.
- InduPro Therapeutics, Cambridge, MA, USA.
| | - Phil S Baran
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
18
|
Zou JX, Chua W, Ser Z, Wang SM, Chiang GSH, Sanmugam K, Tan BY, Sobota RM, Li H. Detection of Bacterial Neutral Ceramidase in Diabetic Foot Ulcers with an Optimized Substrate and Chemoenzymatic Probes. Angew Chem Int Ed Engl 2023; 62:e202307553. [PMID: 37340712 DOI: 10.1002/anie.202307553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 06/22/2023]
Abstract
Ceramidases (CDases) are important in controlling skin barrier integrity by regulating ceramide composition and affording downstream signal molecules. While the functions of epidermal CDases are known, roles of neutral CDases secreted by skin-residing microbes are undefined. Here, we developed a one-step fluorogenic substrate, S-B, for specific detection of bacterial CDase activity and inhibitor screening. We identified a non-hydrolyzable substrate mimic, C6, as the best hit. Based on C6, we designed a photoaffinity probe, JX-1, which efficiently detects bacterial CDases. Using JX-1, we identified endogenous low-abundance PaCDase in a P. aeruginosa monoculture and in a mixed skin bacteria culture. Harnessing both S-B and JX-1, we found that CDase activity positively correlates with the relative abundance of P. aeruginosa and is negatively associated with wound area reduction in clinical diabetic foot ulcer patient samples. Overall, our study demonstrates that bacterial CDases are important regulators of skin ceramides and potentially play a role in wound healing.
Collapse
Affiliation(s)
- Jiao Xia Zou
- Department of Chemistry, National University of Singapore, 3 Science Drive 3, Singapore, 117543, Singapore
| | - Wisely Chua
- Molecular Engineering Lab, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | - Zheng Ser
- Functional Proteomics Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | - Shi Mei Wang
- Functional Proteomics Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | | | | | | | - Radoslaw M Sobota
- Functional Proteomics Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | - Hao Li
- Department of Chemistry, National University of Singapore, 3 Science Drive 3, Singapore, 117543, Singapore
- Molecular Engineering Lab, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| |
Collapse
|
19
|
Faustino AM, Sharma P, Manriquez-Sandoval E, Yadav D, Fried SD. Progress toward Proteome-Wide Photo-Cross-Linking to Enable Residue-Level Visualization of Protein Structures and Networks In Vivo. Anal Chem 2023; 95:10670-10685. [PMID: 37341467 PMCID: PMC11559402 DOI: 10.1021/acs.analchem.3c01369] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2023]
Abstract
Cross-linking mass spectrometry (XL-MS) is emerging as a method at the crossroads of structural and cellular biology, uniquely capable of identifying protein-protein interactions with residue-level resolution and on the proteome-wide scale. With the development of cross-linkers that can form linkages inside cells and easily cleave during fragmentation on the mass spectrometer (MS-cleavable cross-links), it has become increasingly facile to identify contacts between any two proteins in complex samples, including in live cells or tissues. Photo-cross-linkers possess the advantages of high temporal resolution and high reactivity, thereby engaging all residue-types (rather than just lysine); nevertheless, photo-cross-linkers have not enjoyed widespread use and are yet to be employed for proteome-wide studies because their products are challenging to identify. Here, we demonstrate the synthesis and application of two heterobifunctional photo-cross-linkers that feature diazirines and N-hydroxy-succinimidyl carbamate groups, the latter of which unveil doubly fissile MS-cleavable linkages upon acyl transfer to protein targets. Moreover, these cross-linkers demonstrate high water-solubility and cell-permeability. Using these compounds, we demonstrate the feasibility of proteome-wide photo-cross-linking in cellulo. These studies elucidate a small portion of Escherichia coli's interaction network, albeit with residue-level resolution. With further optimization, these methods will enable the detection of protein quinary interaction networks in their native environment at residue-level resolution, and we expect that they will prove useful toward the effort to explore the molecular sociology of the cell.
Collapse
Affiliation(s)
| | - Piyoosh Sharma
- Department of Chemistry, Johns Hopkins University, Baltimore, MD 21218, USA
| | | | - Divya Yadav
- Department of Chemistry, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Stephen D. Fried
- Department of Chemistry, Johns Hopkins University, Baltimore, MD 21218, USA
- Thomas C. Jenkins Department of Biophysics, Johns Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
20
|
Forrest I, Parker CG. Proteome-Wide Fragment-Based Ligand and Target Discovery. Isr J Chem 2023; 63:e202200098. [PMID: 38213795 PMCID: PMC10783656 DOI: 10.1002/ijch.202200098] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Indexed: 02/10/2023]
Abstract
Chemical probes are invaluable tools to investigate biological processes and can serve as lead molecules for the development of new therapies. However, despite their utility, only a fraction of human proteins have selective chemical probes, and more generally, our knowledge of the "chemically-tractable" proteome is limited, leaving many potential therapeutic targets unexploited. To help address these challenges, powerful chemical proteomic approaches have recently been developed to globally survey the ability of proteins to bind small molecules (i. e., ligandability) directly in native systems. In this review, we discuss the utility of such approaches, with a focus on the integration of chemoproteomic methods with fragment-based ligand discovery (FBLD), to facilitate the broad mapping of the ligandable proteome while also providing starting points for progression into lead chemical probes.
Collapse
Affiliation(s)
- Ines Forrest
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Christopher G Parker
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
21
|
Hu L, Liu S, Yao H, Hu Y, Wang Y, Jiang J, Li X, Fu F, Yin Q, Wang H. Identification of a novel heterogeneous nuclear ribonucleoprotein A2B1 (hnRNPA2B1) ligand that disrupts HnRNPA2B1/nucleic acid interactions to inhibit the MDMX-p53 axis in gastric cancer. Pharmacol Res 2023; 189:106696. [PMID: 36791898 DOI: 10.1016/j.phrs.2023.106696] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/01/2023] [Accepted: 02/12/2023] [Indexed: 02/16/2023]
Abstract
Gastric carcinoma is a highly malignant tumor that still lacks effective molecular targets. Heterogeneous nuclear ribonucleoprotein A2B1 (hnRNPA2B1) is an essential oncogenic driver overexpressed in various cancers. The potential role of hnRNPA2B1 in oncotherapy has not been revealed because of the absence of active chemical molecules. In this study, we identified the pseudourea derivative XI-011 as a novel hnRNPA2B1 ligand using chemical proteomics. An interaction study indicated that XI-011 could bind the nucleotide-binding domain to disrupt the recruitment of hnRNPA2B1 to the promoter and untranslated region of the murine double minute X (MDMX) gene, thereby inhibiting its transcription. In addition, chemical targeting of hnRNPA2B1 recovered inactivated p53 and enhanced the therapeutic efficacy of apatinib in vivo. This work presented a novel strategy to restore p53 activity for the treatment of gastric cancers via chemically targeting hnRNPA2B1.
Collapse
Affiliation(s)
- Lei Hu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Shuqi Liu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Hongying Yao
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Yuemiao Hu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
| | - Yingjie Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Jingpeng Jiang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Xiaopeng Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Fenghua Fu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Qikun Yin
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China.
| | - Hongbo Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China.
| |
Collapse
|
22
|
Wilkinson IVL, Bottlinger M, El Harraoui Y, Sieber SA. Profiling the Heme-Binding Proteomes of Bacteria Using Chemical Proteomics. Angew Chem Int Ed Engl 2023; 62:e202212111. [PMID: 36495310 DOI: 10.1002/anie.202212111] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022]
Abstract
Heme is a cofactor with myriad roles and essential to almost all living organisms. Beyond classical gas transport and catalytic functions, heme is increasingly appreciated as a tightly controlled signalling molecule regulating protein expression. However, heme acquisition, biosynthesis and regulation is poorly understood beyond a few model organisms, and the heme-binding proteome has not been fully characterised in bacteria. Yet as heme homeostasis is critical for bacterial survival, heme-binding proteins are promising drug targets. Herein we report a chemical proteomics method for global profiling of heme-binding proteins in live cells for the first time. Employing a panel of heme-based clickable and photoaffinity probes enabled the profiling of 32-54 % of the known heme-binding proteomes in Gram-positive and Gram-negative bacteria. This simple-to-implement profiling strategy could be interchangeably applied to different cell types and systems and fuel future research into heme biology.
Collapse
Affiliation(s)
- Isabel V L Wilkinson
- Centre for Functional Protein Assemblies, Technical University of Munich, Ernst-Otto-Fischer-Straße 8, 85748, Garching, Germany
| | - Max Bottlinger
- Centre for Functional Protein Assemblies, Technical University of Munich, Ernst-Otto-Fischer-Straße 8, 85748, Garching, Germany
| | - Yassmine El Harraoui
- Centre for Functional Protein Assemblies, Technical University of Munich, Ernst-Otto-Fischer-Straße 8, 85748, Garching, Germany
| | - Stephan A Sieber
- Centre for Functional Protein Assemblies, Technical University of Munich, Ernst-Otto-Fischer-Straße 8, 85748, Garching, Germany
| |
Collapse
|
23
|
Chen LY, Wang WW, Wozniak JM, Parker CG. A heterobifunctional molecule system for targeted protein acetylation in cells. Methods Enzymol 2023; 681:287-323. [PMID: 36764762 DOI: 10.1016/bs.mie.2022.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Protein acetylation is a vital biological process that regulates myriad cellular events. Despite its profound effects on protein function, there are limited research tools to dynamically and selectively regulate protein acetylation. To address this, we developed an acetylation tagging system, called AceTAG, to target proteins for chemically induced acetylation directly in live cells. AceTAG uses heterobifunctional molecules composed of a ligand for the lysine acetyltransferase p300/CBP and a FKBP12F36V ligand. Target proteins are genetically tagged with FKBP12F36V and brought in proximity with p300/CBP by AceTAG molecules to subsequently undergo protein-specific acetylation. Targeted acetylation of proteins in cells using AceTAG is selective, rapid, and can be modulated in a dose-dependent fashion, enabling controlled investigations of acetylated protein targets directly in cells. In this protocol, we focus on (1) generation of AceTAG constructs and cell lines, (2) in vitro characterization of AceTAG mediated ternary complex formation and cellular target engagement studies; and (3) in situ characterization of AceTAG induced acetylation of targeted proteins by immunoblotting and quantitative proteomics. The robust procedures described herein should enable the use of AceTAG to explore the roles of acetylation for a variety of protein targets.
Collapse
Affiliation(s)
- Li-Yun Chen
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, United States
| | - Wesley Wei Wang
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, United States
| | - Jacob M Wozniak
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, United States
| | - Christopher G Parker
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, United States.
| |
Collapse
|
24
|
Kozoriz K, Shkel O, Hong KT, Kim DH, Kim YK, Lee JS. Multifunctional Photo-Cross-Linking Probes: From Target Protein Searching to Imaging Applications. Acc Chem Res 2023; 56:25-36. [PMID: 36534922 DOI: 10.1021/acs.accounts.2c00505] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Despite advances in genome sequencing technology, the complete molecular interaction networks reflecting the biological functions of gene products have not been fully elucidated due to the lack of robust molecular interactome profiling techniques. Traditionally, molecular interactions have been investigated in vitro by measuring their affinity. However, such a reductionist approach comes with throughput constraints and does not depict an intact living cell environment. Therefore, molecular interactions in live cells must be captured to minimize false-positive results. The photo-cross-linking technique is a promising tool because the production of a temporally controlled reactive functional group can be induced using light exposure. Photoaffinity labeling is used in biochemistry and medicinal chemistry for bioconjugation, including drug and antibody conjugation, target protein identification of bioactive compounds, and fluorescent labeling of target proteins. This Account summarizes recent advances in multifunctional photo-cross-linkers for drug target identification and bioimaging. In addition to our group's contributions, we reviewed the most notable examples from the last few decades to provide a comprehensive overview of how this field is evolving. Based on cross-linking chemistry, photo-cross-linkers are classified as either (i) reactive intermediate-generating or (ii) electrophile-generating. Reactive intermediates generating photoaffinity tags have been extensively modified to target a molecule of interest using aryl azide, benzophenone, diazirine, diazo, and acyl silanes. These species are highly reactive and can form covalent bonds, irrespective of residue. Their short lifetime is ideal for the instant capture and labeling of biomolecules. Recently, photocaged electrophiles have been investigated to take advantage of their residue selectivity and relatively high yield for adduct formation with tetrazole, nitrobenzyl alcohol, o-nitrophenylethylene, pyrone, and pyrimidone. Multifunctional photo-cross-linkers for two parallel practical applications have been developed using both classes of photoactivatable groups. Unbiased target interactome profiling of small-molecule drugs requires a challenging structure-activity relationship study (SAR) step to retain the nature or biological activity of the lead compound, which led to the design of a multifunctional "minimalist tag" comprising a bio-orthogonal handle, a photoaffinity labeling group, and functional groups to load target molecules. In contrast, fluorogenic photo-cross-linking is advantageous for bioimaging because it does not require an additional bio-orthogonal reaction to introduce a fluorophore to the minimalist tag. Our group has made progress on minimalist tags and fluorogenic photo-cross-linkers through fruitful collaborations with other groups. The current range of photoactivation reactions and applications demonstrate that photoaffinity tags can be improved. We expect exciting days in the rational design of new multifunctional photo-cross-linkers, particularly clinically interesting versions used in photodynamic or photothermal therapy.
Collapse
Affiliation(s)
- Kostiantyn Kozoriz
- Department of Pharmacology, Korea University College of Medicine, 73 Goryeodae-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Olha Shkel
- Convergence Research Center for Brain Science, Korea Institute of Science and Technology (KIST) & Bio-Med Program, KIST-School UST, Hwarang-ro 14 gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Kyung Tae Hong
- Convergence Research Center for Brain Science, Korea Institute of Science and Technology (KIST) & Bio-Med Program, KIST-School UST, Hwarang-ro 14 gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Dong Hoon Kim
- Department of Pharmacology, Korea University College of Medicine, 73 Goryeodae-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Yun Kyung Kim
- Convergence Research Center for Brain Science, Korea Institute of Science and Technology (KIST) & Bio-Med Program, KIST-School UST, Hwarang-ro 14 gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Jun-Seok Lee
- Department of Pharmacology, Korea University College of Medicine, 73 Goryeodae-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| |
Collapse
|
25
|
Thomas RP, Grant EK, Dickinson ER, Zappacosta F, Edwards LJ, Hann MM, House D, Tomkinson NCO, Bush JT. Reactive fragments targeting carboxylate residues employing direct to biology, high-throughput chemistry. RSC Med Chem 2023; 14:671-679. [PMID: 37122547 PMCID: PMC10131605 DOI: 10.1039/d2md00453d] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 01/30/2023] [Indexed: 02/24/2023] Open
Abstract
We present a carboxylate-targeting reactive fragment screening platform using 2-aryl-5-carboxytetrazole (ACT) as the photoreactive functionality. This work will provide a simple accessible method to rapidly discover tool molecules to interrogate important biological targets.
Collapse
Affiliation(s)
- Ross P. Thomas
- GlaxoSmithKline, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
- Department of Pure and Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow, G1 1XL, UK
| | - Emma K. Grant
- GlaxoSmithKline, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
| | | | | | - Lee J. Edwards
- GlaxoSmithKline, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
| | - Michael M. Hann
- GlaxoSmithKline, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
| | - David House
- GlaxoSmithKline, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
| | - Nicholas C. O. Tomkinson
- Department of Pure and Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow, G1 1XL, UK
| | - Jacob T. Bush
- GlaxoSmithKline, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, UK
| |
Collapse
|
26
|
Ge J, Du S, Yao SQ. Bifunctional Lipid-Derived Affinity-Based Probes (A fBPs) for Analysis of Lipid-Protein Interactome. Acc Chem Res 2022; 55:3663-3674. [PMID: 36484537 DOI: 10.1021/acs.accounts.2c00593] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Although lipids are not genetically encoded, they are fundamental building blocks of cell membranes and essential components of cell metabolites. Lipids regulate various biological processes, including energy storage, membrane trafficking, signal transduction, and protein secretion; therefore, their metabolic imbalances cause many diseases. Approximately 47 000 lipid species with diverse structures have been identified, but little is known about their crucial roles in cellular systems. Particularly the structural, metabolic, and signaling functions of lipids often arise from interactions with proteins. Lipids attach to proteins not only by covalent bonds but also through noncovalent interactions, which also influence protein functions and localization. Therefore, it is important to explore this lipid-protein "interactome" to understand its roles in health and disease, which may further provide insight for medicinal development. However, lipid structures are generally quite complicated, rendering the systematic characterization of lipid-protein interactions much more challenging.Chemoproteomics is a well-known chemical biology platform in which small-molecule chemical probes are utilized in combination with high-resolution, quantitative mass spectrometry to study protein-ligand interactions in living cells or organisms, and it has recently been applied to the study of protein-lipid interactions as well. The study of these complicated interactions has been advanced by the development of bifunctional lipid probes, which not only enable probes to form covalent cross-links with lipid-interacting proteins under UV irradiation, but are also capable of enriching these proteins through bioorthogonal reactions.In this Account, we will discuss recent developments in bifunctional lipid-derived, affinity-based probes (AfBP)s that have been developed to investigate lipid-protein interactions in live cell systems. First, we will give a brief introduction of fundamental techniques based on AfBPs which are related to lipid research. Then, we will focus on three aspects, including probes developed on the basis of lipidation, lipid-derived probes with different modification positions (e.g., hydrophobic or hydrophilic parts of a lipid), and, finally, in situ biosynthesis of probes through intrinsic metabolic pathways by using chemically modified building blocks. We will present some case studies to describe these probes' design principles and cellular applications. At the end, we will also highlight key limitations of current approaches so as to provide inspirations for future improvement. The lipid probes that have been constructed are only the tip of the iceberg, and there are still plenty of lipid species that have yet to be explored. We anticipate that AfBP-based chemoproteomics and its further advancement will pave the way for a deep understanding of lipid-protein interactions in the future.
Collapse
Affiliation(s)
- Jingyan Ge
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China
| | - Shubo Du
- School of Bioengineering, Dalian University of Technology, Dalian 116024, People's Republic of China
| | - Shao Q Yao
- Department of Chemistry, National University of Singapore, 4 Science Drive 2, Singapore 117544, Singapore
| |
Collapse
|
27
|
Yu W, Lin Z, Woo CM, Baskin JM. A Chemoproteomics Approach to Profile Phospholipase D-Derived Phosphatidyl Alcohol Interactions. ACS Chem Biol 2022; 17:3276-3283. [PMID: 34908404 DOI: 10.1021/acschembio.1c00584] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Alcohol consumption leads to formation of phosphatidylethanol (PEth) via the transphosphatidylation activity of phospholipase D (PLD) enzymes. Though this non-natural phospholipid routinely serves as a biomarker of chronic alcoholism, its pathophysiological roles remain unknown. We use a minimalist diazirine alkyne alcohol as an ethanol surrogate to generate clickable, photoaffinity lipid reporters of PEth localization and lipid-protein interactions via PLD-mediated transphosphatidylation. We use these tools to visualize phosphatidyl alcohols in a manner compatible with standard permeabilization and immunofluorescence methods. We also use click chemistry tagging, enrichment, and proteomics analysis to define the phosphatidyl alcohol interactome. Our analysis reveals an enrichment of putative interactors at various membrane locations, and we validate one such interaction with the single-pass transmembrane protein basigin/CD147. This study provides a comprehensive view of the molecular interactions of phosphatidyl alcohols with the cellular proteome and points to future work to connect such interactions to potential pathophysiological roles of PEth.
Collapse
Affiliation(s)
- Weizhi Yu
- Department of Chemistry and Chemical Biology and Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York 14853, United States
| | - Zhi Lin
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Christina M Woo
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Jeremy M Baskin
- Department of Chemistry and Chemical Biology and Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
28
|
Hassan AA, Wozniak JM, Vilen Z, Li W, Jadhav A, Parker CG, Huang ML. Chemoproteomic mapping of human milk oligosaccharide (HMO) interactions in cells. RSC Chem Biol 2022; 3:1369-1374. [PMID: 36544572 PMCID: PMC9709932 DOI: 10.1039/d2cb00176d] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 10/09/2022] [Indexed: 12/05/2022] Open
Abstract
Human milk oligosaccharides (HMOs) are a family of unconjugated soluble glycans found in human breast milk that exhibit a myriad of biological activity. While recent studies have uncovered numerous biological functions for HMOs (antimicrobial, anti-inflammatory & probiotic properties), the receptors and protein binding partners involved in these processes are not well characterized. This can be attributed largely in part to the low affinity and transient nature of soluble glycan-protein interactions, precluding the use of traditional characterization techniques to survey binding partners in live cells. Here, we present the use of synthetic photoactivatable HMO probes to capture, enrich and identify HMO protein targets in live cells using mass spectrometry-based chemoproteomics. Following initial validation studies using purified lectins, we profiled the targets of HMO probes in live mouse macrophages. Using this strategy, we mapped hundreds of HMO binding partners across multiple cellular compartments, including many known glycan-binding proteins as well as numerous proteins previously not known to bind glycans. We expect our findings to inform future investigations of the diverse roles of how HMOs may regulate protein function.
Collapse
Affiliation(s)
- Abdullah A Hassan
- Department of Molecular Medicine, Scripps Research, 10550 N Torrey Pines Rd. La Jolla CA 92037 USA
| | - Jacob M Wozniak
- Department of Chemistry, Scripps Research, 10550 N Torrey Pines Rd. La Jolla CA 92037 USA
| | - Zak Vilen
- Department of Molecular Medicine, Scripps Research, 10550 N Torrey Pines Rd. La Jolla CA 92037 USA
- Skaggs Graduate School of Chemical and Biological Sciences, Scripps Research, 10550 N Torrey Pines Rd. La Jolla CA 92037 USA
| | - Weichao Li
- Department of Molecular Medicine, Scripps Research, 10550 N Torrey Pines Rd. La Jolla CA 92037 USA
- Department of Chemistry, Scripps Research, 10550 N Torrey Pines Rd. La Jolla CA 92037 USA
| | - Appaso Jadhav
- Department of Chemistry, Scripps Research, 10550 N Torrey Pines Rd. La Jolla CA 92037 USA
| | - Christopher G Parker
- Department of Chemistry, Scripps Research, 10550 N Torrey Pines Rd. La Jolla CA 92037 USA
- Skaggs Graduate School of Chemical and Biological Sciences, Scripps Research, 10550 N Torrey Pines Rd. La Jolla CA 92037 USA
| | - Mia L Huang
- Department of Molecular Medicine, Scripps Research, 10550 N Torrey Pines Rd. La Jolla CA 92037 USA
- Department of Chemistry, Scripps Research, 10550 N Torrey Pines Rd. La Jolla CA 92037 USA
- Skaggs Graduate School of Chemical and Biological Sciences, Scripps Research, 10550 N Torrey Pines Rd. La Jolla CA 92037 USA
| |
Collapse
|
29
|
West AV, Woo CM. Photoaffinity Labeling Chemistries Used to Map Biomolecular Interactions. Isr J Chem 2022. [DOI: 10.1002/ijch.202200081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Alexander V. West
- Department of Chemistry and Chemical Biology Harvard University 12 Oxford St Cambridge MA USA
| | - Christina M. Woo
- Department of Chemistry and Chemical Biology Harvard University 12 Oxford St Cambridge MA USA
| |
Collapse
|
30
|
West AV, Amako Y, Woo CM. Design and Evaluation of a Cyclobutane Diazirine Alkyne Tag for Photoaffinity Labeling in Cells. J Am Chem Soc 2022; 144:21174-21183. [DOI: 10.1021/jacs.2c08257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Alexander V. West
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Yuka Amako
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Christina M. Woo
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
| |
Collapse
|
31
|
Homan RA, Jadhav AM, Conway LP, Parker CG. A Chemical Proteomic Map of Heme-Protein Interactions. J Am Chem Soc 2022; 144:15013-15019. [PMID: 35960875 PMCID: PMC9811995 DOI: 10.1021/jacs.2c06104] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Heme is an essential cofactor for many human proteins as well as the primary transporter of oxygen in blood. Recent studies have also established heme as a signaling molecule, imparting its effects through binding with protein partners rather than through reactivity of its metal center. However, the comprehensive annotation of such heme-binding proteins in the human proteome remains incomplete. Here, we describe a strategy which utilizes a heme-based photoaffinity probe integrated with quantitative proteomics to map heme-protein interactions across the proteome. In these studies, we identified 350+ unique heme-protein interactions, the vast majority of which were heretofore unknown and consist of targets from diverse functional classes, including transporters, receptors, enzymes, transcription factors, and chaperones. Among these proteins is the immune-related interleukin receptor-associated kinase 1 (IRAK1), where we provide preliminary evidence that heme agonizes its catalytic activity. Our findings should improve the current understanding of heme's regulation as well as its signaling functions and facilitate new insights of its roles in human disease.
Collapse
Affiliation(s)
- Rick A. Homan
- Department of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Appaso M. Jadhav
- Department of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Louis P. Conway
- Department of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Christopher G. Parker
- Department of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
| |
Collapse
|
32
|
Lougee MG, Pagar VV, Kim HJ, Pancoe SX, Chia WK, Mach RH, Garcia BA, Petersson EJ. Harnessing the intrinsic photochemistry of isoxazoles for the development of chemoproteomic crosslinking methods. Chem Commun (Camb) 2022; 58:9116-9119. [PMID: 35880535 PMCID: PMC9922157 DOI: 10.1039/d2cc02263j] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The intrinsic photochemistry of the isoxazole, a common heterocycle in medicinal chemistry, can be applied to offer an alternative to existing strategies using more perturbing, extrinsic photo-crosslinkers. The utility of isoxazole photo-crosslinking is demonstrated in a wide range of biologically relevant experiments, including common proteomics workflows.
Collapse
Affiliation(s)
- Marshall G. Lougee
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Vinayak Vishnu Pagar
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA. .,Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
| | - Hee Jong Kim
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Samantha X. Pancoe
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - W. Kit Chia
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Robert H. Mach
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Benjamin A. Garcia
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - E. James Petersson
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
33
|
Dixit A, Jose GP, Shanbhag C, Tagad N, Kalia J. Metabolic Labeling-Based Chemoproteomics Establishes Choline Metabolites as Protein Function Modulators. ACS Chem Biol 2022; 17:2272-2283. [PMID: 35802552 DOI: 10.1021/acschembio.2c00400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Choline is an essential nutrient for mammalian cells. Our understanding of the cellular functions of choline and its metabolites, independent of their roles as choline lipid metabolism intermediates, remains limited. In addition to fundamental cellular physiology, this knowledge has implications for cancer biology because elevated choline metabolite levels are a hallmark of cancer. Here, we establish a mammalian choline metabolite-interacting proteome by utilizing a photocrosslinkable choline probe. To design this probe, we performed metabolic labeling experiments with structurally diverse choline analogues that resulted in the serendipitous discovery of a choline lipid headgroup remodeling mechanism involving sequential dealkylation and methylation steps. We demonstrate that phosphocholine inhibits the binding of one of the proteins identified, the attractive anticancer target p32, to its endogenous ligands and to the promising p32-targeting anticancer agent, Lyp-1. Our results reveal that choline metabolites play vital roles in cellular physiology by serving as modulators of protein function.
Collapse
Affiliation(s)
- Aditi Dixit
- Department of Chemistry, Indian Institute of Science Education and Research (IISER) Pune, Dr. Homi Bhabha Road, Pashan, Pune 411008, Maharashtra, India.,Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Bhopal, Bhopal Bypass Road, Bhauri, Bhopal 462066, Madhya Pradesh, India
| | - Gregor P Jose
- Department of Chemistry, Indian Institute of Science Education and Research (IISER) Pune, Dr. Homi Bhabha Road, Pashan, Pune 411008, Maharashtra, India
| | - Chitra Shanbhag
- Department of Chemistry, Indian Institute of Science Education and Research (IISER) Pune, Dr. Homi Bhabha Road, Pashan, Pune 411008, Maharashtra, India
| | - Nitin Tagad
- Department of Chemistry, Indian Institute of Science Education and Research (IISER) Pune, Dr. Homi Bhabha Road, Pashan, Pune 411008, Maharashtra, India
| | - Jeet Kalia
- Department of Chemistry, Indian Institute of Science Education and Research (IISER) Pune, Dr. Homi Bhabha Road, Pashan, Pune 411008, Maharashtra, India.,Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Bhopal, Bhopal Bypass Road, Bhauri, Bhopal 462066, Madhya Pradesh, India.,Department of Biology, Indian Institute of Science Education and Research (IISER) Pune, Dr. Homi Bhabha Road, Pashan, Pune 411008, Maharashtra, India.,Department of Chemistry, Indian Institute of Science Education and Research (IISER) Bhopal, Bhopal Bypass Road, Bhauri, Bhopal 462066, Madhya Pradesh, India
| |
Collapse
|
34
|
Yu W, Baskin JM. Photoaffinity labeling approaches to elucidate lipid-protein interactions. Curr Opin Chem Biol 2022; 69:102173. [PMID: 35724595 DOI: 10.1016/j.cbpa.2022.102173] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 05/11/2022] [Accepted: 05/18/2022] [Indexed: 11/30/2022]
Abstract
Lipid-protein interactions serve as the basis for many of the diverse roles of lipids. However, these noncovalent binding events are often weak, transient, or dependent upon environmental cues. Photoaffinity labeling can preserve these interactions under native conditions, enabling their biochemical profiling. Typically, photoaffinity labeling probes contain a diazirine photocrosslinker and a click chemistry handle for enrichment and downstream analysis. In this review, we summarize recent advances in the understanding the mechanisms of diazirine photocrosslinking, and we provide an overview of recent applications of photoaffinity labeling to reveal the interactions of diverse types of lipids with specific members of the proteome.
Collapse
Affiliation(s)
- Weizhi Yu
- Department of Chemistry and Chemical Biology, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, 14853, NY, USA
| | - Jeremy M Baskin
- Department of Chemistry and Chemical Biology, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, 14853, NY, USA.
| |
Collapse
|
35
|
Photoaffinity labeling and bioorthogonal ligation: Two critical tools for designing "Fish Hooks" to scout for target proteins. Bioorg Med Chem 2022; 62:116721. [PMID: 35358862 DOI: 10.1016/j.bmc.2022.116721] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/14/2022] [Accepted: 03/17/2022] [Indexed: 11/21/2022]
Abstract
Small molecules remain an important category of therapeutic agents. Their binding to different proteins can lead to both desired and undesired biological effects. Identification of the proteins that a drug binds to has become an important step in drug development because it can lead to safer and more effective drugs. Parent bioactive molecules can be converted to appropriate probes that allow for visualization and identification of their target proteins. Typically, these probes are designed and synthesized utilizing some or all of five major tools; a photoactivatable group, a reporter tag, a linker, an affinity tag, and a bioorthogonal handle. This review covers two of the most challenging tools, photoactivation and bioorthogonal ligation. We provide a historical and theoretical background along with synthetic routes to prepare them. In addition, the review provides comparative analyses of the available tools that can assist decision making when designing such probes. A survey of most recent literature reports is included as well to identify recent trends in the field.
Collapse
|
36
|
Martín-Acosta P, Meng Q, Klimek J, Reddy AP, David L, Petrie SK, Li BX, Xiao X. A clickable photoaffinity probe of betulinic acid identifies tropomyosin as a target. Acta Pharm Sin B 2022; 12:2406-2416. [PMID: 35646545 PMCID: PMC9136574 DOI: 10.1016/j.apsb.2021.12.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/25/2021] [Accepted: 12/09/2021] [Indexed: 12/30/2022] Open
Abstract
Target identification of bioactive compounds is important for understanding their mechanisms of action and provides critical insights into their therapeutic utility. While it remains a challenge, unbiased chemoproteomics strategy using clickable photoaffinity probes is a useful and validated approach for target identification. One major limitation of this approach is the efficient synthesis of appropriately substituted clickable photoaffinity probes. Herein, we describe an efficient and consistent method to prepare such probes. We further employed this method to prepare a highly stereo-congested probe based on naturally occurring triterpenoid betulinic acid. With this photoaffinity probe, we identified tropomyosin as a novel target for betulinic acid that can account for the unique biological phenotype on cellular cytoskeleton induced by betulinic acid.
Collapse
Affiliation(s)
- Pedro Martín-Acosta
- Program in Chemical Biology, Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA
| | - Qianli Meng
- Program in Chemical Biology, Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA
| | - John Klimek
- Program in Chemical Biology, Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA
| | - Ashok P. Reddy
- Proteomics Shared Resource, Oregon Health & Science University, Portland, OR 97239, USA
| | - Larry David
- Program in Chemical Biology, Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Stefanie Kaech Petrie
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Bingbing X. Li
- Program in Chemical Biology, Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA
| | - Xiangshu Xiao
- Program in Chemical Biology, Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
37
|
Fan J, Shu Q, Li YM, Shi J. Efficient synthesis of terminal-diazirine-based histone peptide probes. Tetrahedron Lett 2022. [DOI: 10.1016/j.tetlet.2022.153878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
38
|
Veale CGL, Talukdar A, Vauzeilles B. ICBS 2021: Looking Toward the Next Decade of Chemical Biology. ACS Chem Biol 2022; 17:728-743. [PMID: 35293726 DOI: 10.1021/acschembio.2c00209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Clinton G. L. Veale
- Department of Chemistry, University of Cape Town, Rondebosch, Cape Town, 7700, South Africa
| | - Arindam Talukdar
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, West Bengal, India
| | - Boris Vauzeilles
- Université Paris-Saclay, CNRS, Institut de Chimie des Substances Naturelles, UPR 2301, 91198, Gif-sur-Yvette, France
| |
Collapse
|
39
|
Page ACS, Scholz SO, Keenan KN, Spradlin JN, Belcher BP, Brittain SM, Tallarico JA, McKenna JM, Schirle M, Nomura DK, Toste FD. Photo-Brook rearrangement of acyl silanes as a strategy for photoaffinity probe design. Chem Sci 2022; 13:3851-3856. [PMID: 35432890 PMCID: PMC8966736 DOI: 10.1039/d2sc00426g] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 03/02/2022] [Indexed: 01/14/2023] Open
Abstract
Photoaffinity labeling (PAL) is a powerful tool for the identification of non-covalent small molecule–protein interactions that are critical to drug discovery and medicinal chemistry, but this approach is limited to only a small subset of robust photocrosslinkers. The identification of new photoreactive motifs capable of covalent target capture is therefore highly desirable. Herein, we report the design, synthesis, and evaluation of a new class of PAL warheads based on the UV-triggered 1,2-photo-Brook rearrangement of acyl silanes, which hitherto have not been explored for PAL workflows. Irradiation of a series of probes in cell lysate revealed an iPr-substituted acyl silane with superior photolabeling and minimal thermal background labeling compared to other substituted acyl silanes. Further, small molecule (+)-JQ1- and rapamycin-derived iPr acyl silanes were shown to selectively label recombinant BRD4-BD1 and FKBP12, respectively, with minimal background. Together, these data highlight the untapped potential of acyl silanes as a novel, tunable scaffold for photoaffinity labeling. Irradiation initiated 1,2-photo Brook rearrangement of acyl silanes generated α-siloxycarbene intermediates that were used for photoaffinity labeling. Optimization of the acyl silane group produced a probe capable of capturing small molecule–protein interactions.![]()
Collapse
Affiliation(s)
- Annika C S Page
- Department of Chemistry, University of California Berkeley California 94720 USA .,Novartis-Berkeley Center for Proteomics and Chemistry Technologies, University of California Berkeley California 94720 USA
| | - Spencer O Scholz
- Department of Chemistry, University of California Berkeley California 94720 USA .,Novartis-Berkeley Center for Proteomics and Chemistry Technologies, University of California Berkeley California 94720 USA
| | - Katherine N Keenan
- Department of Chemistry, University of California Berkeley California 94720 USA .,Novartis-Berkeley Center for Proteomics and Chemistry Technologies, University of California Berkeley California 94720 USA
| | - Jessica N Spradlin
- Department of Chemistry, University of California Berkeley California 94720 USA .,Novartis-Berkeley Center for Proteomics and Chemistry Technologies, University of California Berkeley California 94720 USA.,Innovative Genomics Institute, University of California Berkeley California 94720 USA
| | - Bridget P Belcher
- Department of Chemistry, University of California Berkeley California 94720 USA .,Innovative Genomics Institute, University of California Berkeley California 94720 USA
| | - Scott M Brittain
- Novartis-Berkeley Center for Proteomics and Chemistry Technologies, University of California Berkeley California 94720 USA.,Novartis Institute for BioMedical Research Cambridge Massachusetts 02139 USA
| | - John A Tallarico
- Novartis-Berkeley Center for Proteomics and Chemistry Technologies, University of California Berkeley California 94720 USA.,Novartis Institute for BioMedical Research Cambridge Massachusetts 02139 USA
| | - Jeffrey M McKenna
- Novartis-Berkeley Center for Proteomics and Chemistry Technologies, University of California Berkeley California 94720 USA.,Novartis Institute for BioMedical Research Cambridge Massachusetts 02139 USA
| | - Markus Schirle
- Novartis-Berkeley Center for Proteomics and Chemistry Technologies, University of California Berkeley California 94720 USA.,Novartis Institute for BioMedical Research Cambridge Massachusetts 02139 USA
| | - Daniel K Nomura
- Department of Chemistry, University of California Berkeley California 94720 USA .,Novartis-Berkeley Center for Proteomics and Chemistry Technologies, University of California Berkeley California 94720 USA.,Innovative Genomics Institute, University of California Berkeley California 94720 USA.,Department of Molecular and Cellular Biology, University of California Berkeley California 94720 USA
| | - F Dean Toste
- Department of Chemistry, University of California Berkeley California 94720 USA .,Novartis-Berkeley Center for Proteomics and Chemistry Technologies, University of California Berkeley California 94720 USA
| |
Collapse
|
40
|
Taylor MT. Photochemical protein modification in complex biological environments: recent advances and considerations for future chemical methods development. Biol Chem 2022; 403:413-420. [PMID: 35073619 PMCID: PMC10163948 DOI: 10.1515/hsz-2021-0351] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 01/12/2022] [Indexed: 12/29/2022]
Abstract
Abstract
The development of organic reactions that covalently modify biological matter in complex biological mixtures has become an invaluable asset in drug discovery. Out of the techniques developed to date, optically controlled chemistries are of particular utility owing to both the spatiotemporal control afforded by optical control as well as the impressive array of transformations that are driven by the highly reactive intermediates generated upon excitation. This minireview discusses recent advances in the development of photochemical reactions for use in complex mixtures and highlights key considerations for future photochemical reaction designs.
Collapse
Affiliation(s)
- Michael T Taylor
- Department of Chemistry, University of Wyoming, 1000 E. University Ave., Laramie, WY 82071, USA
| |
Collapse
|
41
|
Hui C, Antonchick AP. Iodonitrene: a direct metal-free electrophilic aminating reagent. Org Chem Front 2022. [DOI: 10.1039/d2qo00739h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Iodonitrene is a new type of reactive electrophilic aminating reagent that opens up opportunities for new discoveries.
Collapse
Affiliation(s)
- Chunngai Hui
- Max Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
- Technical University Dortmund, Faculty of Chemistry and Chemical Biology, Otto-Hahn-Strasse 6, 44221 Dortmund, Germany
| | - Andrey P. Antonchick
- Max Planck Institute of Molecular Physiology, Department of Chemical Biology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany
- Technical University Dortmund, Faculty of Chemistry and Chemical Biology, Otto-Hahn-Strasse 6, 44221 Dortmund, Germany
- Nottingham Trent University, School of Science and Technology, Department of Chemistry and Forensics, Clifton Lane, NG11 8NS Nottingham, UK
| |
Collapse
|
42
|
Tan Y, Fu S, Yang T, Xie Y, Shen G, Yan J, Zhao Y, Ni F. Evaluation of Site-Diversified, Fully Functionalized Diazirine Probes for Chemical Proteomic Applications. Chem Commun (Camb) 2022; 58:9532-9535. [DOI: 10.1039/d2cc03868d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A series of site-diversified, fully functionalized diazirine probes are constructed based on a scaffold shared by several marketed EGFR-targeted drugs. The integrated analysis of protein targets of site-diversified probe toolkit...
Collapse
|
43
|
Graziadei A, Rappsilber J. Leveraging crosslinking mass spectrometry in structural and cell biology. Structure 2021; 30:37-54. [PMID: 34895473 DOI: 10.1016/j.str.2021.11.007] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/11/2021] [Accepted: 11/17/2021] [Indexed: 12/18/2022]
Abstract
Crosslinking mass spectrometry (crosslinking-MS) is a versatile tool providing structural insights into protein conformation and protein-protein interactions. Its medium-resolution residue-residue distance restraints have been used to validate protein structures proposed by other methods and have helped derive models of protein complexes by integrative structural biology approaches. The use of crosslinking-MS in integrative approaches is underpinned by progress in estimating error rates in crosslinking-MS data and in combining these data with other information. The flexible and high-throughput nature of crosslinking-MS has allowed it to complement the ongoing resolution revolution in electron microscopy by providing system-wide residue-residue distance restraints, especially for flexible regions or systems. Here, we review how crosslinking-MS information has been leveraged in structural model validation and integrative modeling. Crosslinking-MS has also been a key technology for cell biology studies and structural systems biology where, in conjunction with cryoelectron tomography, it can provide structural and mechanistic insights directly in situ.
Collapse
Affiliation(s)
- Andrea Graziadei
- Bioanalytics, Institute of Biotechnology, Technische Universität Berlin, 13355 Berlin, Germany
| | - Juri Rappsilber
- Bioanalytics, Institute of Biotechnology, Technische Universität Berlin, 13355 Berlin, Germany; Wellcome Centre for Cell Biology, University of Edinburgh, Max Born Crescent, Edinburgh EH9 3BF, UK.
| |
Collapse
|
44
|
Wang WW, Chen LY, Wozniak JM, Jadhav AM, Anderson H, Malone TE, Parker CG. Targeted Protein Acetylation in Cells Using Heterobifunctional Molecules. J Am Chem Soc 2021; 143:16700-16708. [PMID: 34592107 PMCID: PMC10793965 DOI: 10.1021/jacs.1c07850] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Protein acetylation is a central event in orchestrating diverse cellular processes. However, current strategies to investigate protein acetylation in cells are often nonspecific or lack temporal and magnitude control. Here, we developed an acetylation tagging system, AceTAG, to induce acetylation of targeted proteins. The AceTAG system utilizes bifunctional molecules to direct the lysine acetyltransferase p300/CBP to proteins fused with the small protein tag FKBP12F36V, resulting in their induced acetylation. Using AceTAG, we induced targeted acetylation of a diverse array of proteins in cells, specifically histone H3.3, the NF-κB subunit p65/RelA, and the tumor suppressor p53. We demonstrate that targeted acetylation with the AceTAG system is rapid, selective, reversible and can be controlled in a dose-dependent fashion. AceTAG represents a useful strategy to modulate protein acetylation and should enable the exploration of targeted acetylation in basic biological and therapeutic contexts.
Collapse
Affiliation(s)
- Wesley W Wang
- Department of Chemistry, The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Li-Yun Chen
- Department of Chemistry, The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Jacob M Wozniak
- Department of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Appaso M Jadhav
- Department of Chemistry, The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Hayden Anderson
- Department of Chemistry, The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Taylor E Malone
- Department of Chemistry, The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Christopher G Parker
- Department of Chemistry, The Scripps Research Institute, Jupiter, Florida 33458, United States
- Department of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
| |
Collapse
|
45
|
Burton NR, Kim P, Backus KM. Photoaffinity labelling strategies for mapping the small molecule-protein interactome. Org Biomol Chem 2021; 19:7792-7809. [PMID: 34549230 PMCID: PMC8489259 DOI: 10.1039/d1ob01353j] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Nearly all FDA approved drugs and bioactive small molecules exert their effects by binding to and modulating proteins. Consequently, understanding how small molecules interact with proteins at an molecular level is a central challenge of modern chemical biology and drug development. Complementary to structure-guided approaches, chemoproteomics has emerged as a method capable of high-throughput identification of proteins covalently bound by small molecules. To profile noncovalent interactions, established chemoproteomic workflows typically incorporate photoreactive moieties into small molecule probes, which enable trapping of small molecule-protein interactions (SMPIs). This strategy, termed photoaffinity labelling (PAL), has been utilized to profile an array of small molecule interactions, including for drugs, lipids, metabolites, and cofactors. Herein we describe the discovery of photocrosslinking chemistries, including a comparison of the strengths and limitations of implementation of each chemotype in chemoproteomic workflows. In addition, we highlight key examples where photoaffinity labelling has enabled target deconvolution and interaction site mapping.
Collapse
Affiliation(s)
- Nikolas R Burton
- Department of Chemistry and Biochemistry, College of Arts and Sciences, UCLA, Los Angeles, CA, 90095, USA.
| | - Phillip Kim
- Department of Biological Chemistry, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
| | - Keriann M Backus
- Department of Chemistry and Biochemistry, College of Arts and Sciences, UCLA, Los Angeles, CA, 90095, USA.
- Department of Biological Chemistry, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
| |
Collapse
|
46
|
McCloud RL, Yuan K, Mahoney KE, Bai DL, Shabanowitz J, Ross MM, Hunt DF, Hsu KL. Direct Target Site Identification of a Sulfonyl-Triazole Covalent Kinase Probe by LC-MS Chemical Proteomics. Anal Chem 2021; 93:11946-11955. [PMID: 34431655 DOI: 10.1021/acs.analchem.1c01591] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Chemical proteomics is widely used for the global investigation of protein activity and binding of small molecule ligands. Covalent probe binding and inhibition are assessed using liquid chromatography-tandem mass spectrometry (LC-MS/MS) to gain molecular information on targeted proteins and probe-modified sites. The identification of amino acid sites modified by large complex probes, however, is particularly challenging because of the increased size, hydrophobicity, and charge state of peptides derived from modified proteins. These studies are important for direct evaluation of proteome-wide selectivity of inhibitor scaffolds used to develop targeted covalent inhibitors. Here, we disclose reverse-phase chromatography and MS dissociation conditions tailored for binding site identification using a clickable covalent kinase inhibitor containing a sulfonyl-triazole reactive group (KY-26). We applied this LC-MS/MS strategy to identify tyrosine and lysine sites modified by KY-26 in functional sites of kinases and other ATP-/NAD-binding proteins (>65 in total) in live cells. Our studies revealed key bioanalytical conditions to guide future chemical proteomic workflows for direct target site identification of complex irreversible probes and inhibitors.
Collapse
Affiliation(s)
- Rebecca L McCloud
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, United States
| | - Kun Yuan
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, United States
| | - Keira E Mahoney
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, United States
| | - Dina L Bai
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, United States
| | - Jeffrey Shabanowitz
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, United States
| | - Mark M Ross
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, United States
| | - Donald F Hunt
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, United States
| | - Ku-Lung Hsu
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, United States.,Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia 22908, United States.,University of Virginia Cancer Center, University of Virginia, Charlottesville, Virginia 22903, United States
| |
Collapse
|