1
|
Liu J, Kulkarni A, Gao YQ, Urul DA, Hamelin R, Novotny BÁ, Long MJC, Aye Y. Organ-specific electrophile responsivity mapping in live C. elegans. Cell 2024; 187:7450-7469.e29. [PMID: 39504959 DOI: 10.1016/j.cell.2024.10.014] [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: 06/12/2023] [Revised: 05/30/2024] [Accepted: 10/10/2024] [Indexed: 11/08/2024]
Abstract
Proximity labeling technologies are limited to indexing localized protein residents. Such data-although valuable-cannot inform on small-molecule responsivity of local residents. We here bridge this gap by demonstrating in live C. elegans how electrophile-sensing propensity in specific organs can be quantitatively mapped and ranked. Using this method, >70% of tissue-specific responders exhibit electrophile responsivity, independent of tissue-specific abundance. One responder, cyp-33e1-for which both human and worm orthologs are electrophile responsive-marshals stress-dependent gut functions, despite manifesting uniform abundance across all tissues studied. Cyp-33e1's localized electrophile responsivity operates site specifically, triggering multifaceted responses: electrophile sensing through the catalytic-site cysteine results in partitioning between enzyme inhibition and localized production of a critical metabolite that governs global lipid availability, whereas rapid dual-cysteine site-specific sensing modulates gut homeostasis. Beyond pinpointing chemical actionability within local proteomes, organ-specific electrophile responsivity mapping illuminates otherwise intractable locale-specific metabolite signaling and stress response programs influencing organ-specific decision-making.
Collapse
Affiliation(s)
- Jinmin Liu
- Department of Chemistry, University of Oxford, Oxford OX1 3TA, UK; Swiss Federal Institute of Technology Lausanne, Lausanne 1015, Switzerland
| | - Amogh Kulkarni
- Swiss Federal Institute of Technology Lausanne, Lausanne 1015, Switzerland; Friedrich Schiller University, 07737 Jena, Germany
| | - Yong-Qi Gao
- Swiss Federal Institute of Technology Lausanne, Lausanne 1015, Switzerland
| | - Daniel A Urul
- Swiss Federal Institute of Technology Lausanne, Lausanne 1015, Switzerland; AssayQuant Technologies, Marlboro, MA 01752, USA
| | - Romain Hamelin
- Swiss Federal Institute of Technology Lausanne, Lausanne 1015, Switzerland
| | - Balázs Á Novotny
- Swiss Federal Institute of Technology Lausanne, Lausanne 1015, Switzerland
| | | | - Yimon Aye
- Department of Chemistry, University of Oxford, Oxford OX1 3TA, UK; Swiss Federal Institute of Technology Lausanne, Lausanne 1015, Switzerland.
| |
Collapse
|
2
|
Huang KT, Aye Y. Toward decoding spatiotemporal signaling activities of reactive immunometabolites with precision immuno-chemical biology tools. Commun Chem 2024; 7:195. [PMID: 39223329 PMCID: PMC11369232 DOI: 10.1038/s42004-024-01282-4] [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: 03/10/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
Immune-cell reprogramming driven by mitochondria-derived reactive electrophilic immunometabolites (mt-REMs-e.g., fumarate, itaconate) is an emerging phenomenon of major biomedical importance. Despite their localized production, mt-REMs elicit significantly large local and global footprints within and across cells, through mechanisms involving electrophile signaling. Burgeoning efforts are being put into profiling mt-REMs' potential protein-targets and phenotypic mapping of their multifaceted inflammatory behaviors. Yet, precision indexing of mt-REMs' first-responders with spatiotemporal intelligence and locale-specific function assignments remain elusive. Highlighting the latest advances and overarching challenges, this perspective aims to stimulate thoughts and spur interdisciplinary innovations to address these unmet chemical-biotechnological needs at therapeutic immuno-signaling frontiers.
Collapse
Affiliation(s)
- Kuan-Ting Huang
- Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Yimon Aye
- Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland.
- University of Oxford, Oxford, UK.
| |
Collapse
|
3
|
Long MJC, Aye Y. Formaldehyde regulates one-carbon metabolism and epigenetics. Trends Genet 2024; 40:381-382. [PMID: 38503578 DOI: 10.1016/j.tig.2024.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 03/07/2024] [Indexed: 03/21/2024]
Abstract
Recently, Pham et al. used an array of model systems to uncover a role for the enzyme methionine adenosyltransferase (MAT)-1A, which is mainly expressed in liver, in both sensing formaldehyde and regulating transcriptional responses that protect against it. This provides a new lens for understanding the effects of formaldehyde on gene regulation.
Collapse
Affiliation(s)
| | - Yimon Aye
- Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, 1015, Switzerland.
| |
Collapse
|
4
|
Long MJC, Aye Y. Climbing into their Skin to Understand Contextual Protein-Protein Associations and Localizations: Functional Investigations in Transgenic Live Model Organisms. Chembiochem 2024; 25:e202400005. [PMID: 38511872 DOI: 10.1002/cbic.202400005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/07/2024] [Indexed: 03/22/2024]
Abstract
Borrowing some quotes from Harper Lee's novel "To Kill A Mockingbird" to help frame our manuscript, we discuss methods to profile local proteomes. We initially focus on chemical biology regimens that function in live organisms and use reactive biotin species for this purpose. We then consider ways to add new dimensions to these experimental regimens, principally by releasing less reactive (i. e., more selective) (preter)natural electrophiles. Although electrophile release methods may have lower resolution and label fewer proteins than biotinylation methods, their ability to probe simultaneously protein function and locale raises new and interesting possibilities for the field.
Collapse
Affiliation(s)
| | - Yimon Aye
- Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, 1015, Switzerland
| |
Collapse
|
5
|
Huang KT, Poganik JR, Parvez S, Raja S, Miller B, Long MJC, Fetcho JR, Aye Y. Z-REX: shepherding reactive electrophiles to specific proteins expressed tissue specifically or ubiquitously, and recording the resultant functional electrophile-induced redox responses in larval fish. Nat Protoc 2023; 18:1379-1415. [PMID: 37020146 PMCID: PMC11150335 DOI: 10.1038/s41596-023-00809-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 12/05/2022] [Indexed: 04/07/2023]
Abstract
This Protocol Extension describes the adaptation of an existing Protocol detailing the use of targetable reactive electrophiles and oxidants, an on-demand redox targeting toolset in cultured cells. The adaptation described here is for use of reactive electrophiles and oxidants technologies in live zebrafish embryos (Z-REX). Zebrafish embryos expressing a Halo-tagged protein of interest (POI)-either ubiquitously or tissue specifically-are treated with a HaloTag-specific small-molecule probe housing a photocaged reactive electrophile (either natural electrophiles or synthetic electrophilic drug-like fragments). The reactive electrophile is then photouncaged at a user-defined time, enabling proximity-assisted electrophile-modification of the POI. Functional and phenotypic ramifications of POI-specific modification can then be monitored, by coupling to standard downstream assays, such as click chemistry-based POI-labeling and target-occupancy quantification; immunofluorescence or live imaging; RNA-sequencing and real-time quantitative polymerase chain reaction analyses of downstream-transcript modulations. Transient expression of requisite Halo-POI in zebrafish embryos is achieved by messenger RNA injection. Procedures associated with generation of transgenic zebrafish expressing a tissue-specific Halo-POI are also described. The Z-REX experiments can be completed in <1 week using standard techniques. To successfully execute Z-REX, researchers should have basic skills in fish husbandry, imaging and pathway analysis. Experience with protein or proteome manipulation is useful. This Protocol Extension is aimed at helping chemical biologists study precision redox events in a model organism and fish biologists perform redox chemical biology.
Collapse
Affiliation(s)
- Kuan-Ting Huang
- Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Jesse R Poganik
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Saba Parvez
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, USA
| | - Sruthi Raja
- Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Brian Miller
- Department of Neurobiology and Behavior, Cornell University, Ithaca, NY, USA
| | | | - Joseph R Fetcho
- Department of Neurobiology and Behavior, Cornell University, Ithaca, NY, USA.
| | - Yimon Aye
- Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
6
|
Long MJC, Liu J, Aye Y. Finding a vocation for validation: taking proteomics beyond association and location. RSC Chem Biol 2023; 4:110-120. [PMID: 36794020 PMCID: PMC9906375 DOI: 10.1039/d2cb00214k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 12/01/2022] [Indexed: 12/03/2022] Open
Abstract
First established in the seventies, proteomics, chemoproteomics, and most recently, spatial/proximity-proteomics technologies have empowered researchers with new capabilities to illuminate cellular communication networks that govern sophisticated decision-making processes. With an ever-growing inventory of these advanced proteomics tools, the onus is upon the researchers to understand their individual advantages and limitations, such that we can ensure rigorous implementation and conclusions derived from critical data interpretations backed up by orthogonal series of functional validations. This perspective-based on the authors' experience in applying varied proteomics workflows in complex living models-underlines key book-keeping considerations, comparing and contrasting most-commonly-deployed modern proteomics profiling technologies. We hope this article stimulates thoughts among expert users and equips new-comers with practical knowhow of what has become an indispensable tool in chemical biology, drug discovery, and broader life-science investigations.
Collapse
Affiliation(s)
- Marcus J. C. Long
- University of Lausanne (UNIL)Switzerland,NCCR Chemical Biology, University of Geneva (UNIGE)Switzerland
| | - Jinmin Liu
- Swiss Federal Institute of Technology Lausanne (EPFL) Switzerland .,NCCR Chemical Biology, University of Geneva (UNIGE) Switzerland
| | - Yimon Aye
- Swiss Federal Institute of Technology Lausanne (EPFL) Switzerland .,NCCR Chemical Biology, University of Geneva (UNIGE) Switzerland
| |
Collapse
|
7
|
Dueñas ME, Peltier‐Heap RE, Leveridge M, Annan RS, Büttner FH, Trost M. Advances in high-throughput mass spectrometry in drug discovery. EMBO Mol Med 2023; 15:e14850. [PMID: 36515561 PMCID: PMC9832828 DOI: 10.15252/emmm.202114850] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/03/2022] [Accepted: 10/07/2022] [Indexed: 12/15/2022] Open
Abstract
High-throughput (HT) screening drug discovery, during which thousands or millions of compounds are screened, remains the key methodology for identifying active chemical matter in early drug discovery pipelines. Recent technological developments in mass spectrometry (MS) and automation have revolutionized the application of MS for use in HT screens. These methods allow the targeting of unlabelled biomolecules in HT assays, thereby expanding the breadth of targets for which HT assays can be developed compared to traditional approaches. Moreover, these label-free MS assays are often cheaper, faster, and more physiologically relevant than competing assay technologies. In this review, we will describe current MS techniques used in drug discovery and explain their advantages and disadvantages. We will highlight the power of mass spectrometry in label-free in vitro assays, and its application for setting up multiplexed cellular phenotypic assays, providing an exciting new tool for screening compounds in cell lines, and even primary cells. Finally, we will give an outlook on how technological advances will increase the future use and the capabilities of mass spectrometry in drug discovery.
Collapse
Affiliation(s)
- Maria Emilia Dueñas
- Laboratory for Biomedical Mass Spectrometry, Biosciences InstituteNewcastle UniversityNewcastle‐upon‐TyneUK
| | - Rachel E Peltier‐Heap
- Discovery Analytical, Screening Profiling and Mechanistic Biology, GSK R&DStevenageUK
| | - Melanie Leveridge
- Discovery Analytical, Screening Profiling and Mechanistic Biology, GSK R&DStevenageUK
| | - Roland S Annan
- Discovery Analytical, Screening Profiling and Mechanistic Biology, GSK R&DStevenageUK
| | - Frank H Büttner
- Drug Discovery Sciences, High Throughput BiologyBoehringer Ingelheim Pharma GmbH&CoKGBiberachGermany
| | - Matthias Trost
- Laboratory for Biomedical Mass Spectrometry, Biosciences InstituteNewcastle UniversityNewcastle‐upon‐TyneUK
| |
Collapse
|
8
|
Van Hall-Beauvais A, Poganik JR, Huang KT, Parvez S, Zhao Y, Lin HY, Liu X, Long MJC, Aye Y. Z-REX uncovers a bifurcation in function of Keap1 paralogs. eLife 2022; 11:e83373. [PMID: 36300632 PMCID: PMC9754640 DOI: 10.7554/elife.83373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/12/2022] [Indexed: 11/13/2022] Open
Abstract
Studying electrophile signaling is marred by difficulties in parsing changes in pathway flux attributable to on-target, vis-à-vis off-target, modifications. By combining bolus dosing, knockdown, and Z-REX-a tool investigating on-target/on-pathway electrophile signaling, we document that electrophile labeling of one zebrafish-Keap1-paralog (zKeap1b) stimulates Nrf2- driven antioxidant response (AR) signaling (like the human-ortholog). Conversely, zKeap1a is a dominant-negative regulator of electrophile-promoted Nrf2-signaling, and itself is nonpermissive for electrophile-induced Nrf2-upregulation. This behavior is recapitulated in human cells: (1) zKeap1b-expressing cells are permissive for augmented AR-signaling through reduced zKeap1b-Nrf2 binding following whole-cell electrophile treatment; (2) zKeap1a-expressing cells are non-permissive for AR-upregulation, as zKeap1a-Nrf2 binding capacity remains unaltered upon whole-cell electrophile exposure; (3) 1:1 ZKeap1a:zKeap1b-co-expressing cells show no Nrf2-release from the Keap1-complex following whole-cell electrophile administration, rendering these cells unable to upregulate AR. We identified a zKeap1a-specific point-mutation (C273I) responsible for zKeap1a's behavior during electrophilic stress. Human-Keap1(C273I), of known diminished Nrf2-regulatory capacity, dominantly muted electrophile-induced Nrf2-signaling. These studies highlight divergent and interdependent electrophile signaling behaviors, despite conserved electrophile sensing.
Collapse
Affiliation(s)
| | - Jesse R Poganik
- Swiss Federal Institute of Technology LausanneLausanneSwitzerland
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Kuan-Ting Huang
- Swiss Federal Institute of Technology LausanneLausanneSwitzerland
| | - Saba Parvez
- Department of Pharmacology and Toxicology, College of Pharmacy, University of UtahSalt Lake CityUnited States
| | - Yi Zhao
- Swiss Federal Institute of Technology LausanneLausanneSwitzerland
- BayRay Innovation Center, Shenzhen Bay LaboratoryShenzhenChina
| | - Hong-Yu Lin
- Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen UniversityXiamenChina
| | - Xuyu Liu
- Swiss Federal Institute of Technology LausanneLausanneSwitzerland
- School of Chemistry, The University of SydneySydneyAustralia
- The Heart Research Institute, NewtownNewtownAustralia
| | - Marcus John Curtis Long
- Department of Biochemistry, Faculty of Biology and Medicine, University of LausanneLausanneSwitzerland
| | - Yimon Aye
- Swiss Federal Institute of Technology LausanneLausanneSwitzerland
| |
Collapse
|
9
|
Yang F, Wang C. Site-specific quantitative cysteine profiling with data-independent acquisition-based mass spectrometry. Methods Enzymol 2022; 679:295-322. [PMID: 36682866 DOI: 10.1016/bs.mie.2022.07.037] [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: 01/25/2023]
Abstract
Chemical proteomics methods, such as activity-based protein profiling, have emerged as powerful and versatile tools to annotate the protein functions and targets of bioactive small molecules in complex biological systems. Incorporated with mass spectrometry (MS)-based quantitative proteomics method, changes of protein activities could be captured and investigated with site-specific precision. However, the semi-stochastic nature of data-dependent acquisition and high cost of the isotopic-labeled reagents make it challenging for chemical biology research to systematically and reproducibly analyze a large number of samples in multidimensional analysis and high-throughput screening. In this chapter, we describe an efficient quantitative chemical proteomic strategy, termed DIA-ABPP, with good reproducibility and high quantification accuracy. Cysteinome profiling was used as a proof-of-concept example with the detailed protocol to demonstrate the workflow of the DIA-ABPP method, including dose-dependent analysis of cysteines that are sensitive to modification by a reactive metabolite, screening of a cysteine-reactive fragment library, and profiling of circadian cysteinome fluctuation. This quantitative chemoproteomic strategy would provide an opportunity for in-depth multi-dimensional chemical proteomic profiling and illuminate the function of bioactive small molecules and proteins in complex biological systems.
Collapse
Affiliation(s)
- Fan Yang
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Chu Wang
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
| |
Collapse
|
10
|
Long MJC, Assari M, Aye Y. Hiding in Plain Sight: The Issue of Hidden Variables. ACS Chem Biol 2022; 17:1285-1292. [PMID: 35603432 DOI: 10.1021/acschembio.2c00142] [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
Here we discuss "hidden variables", which are typically introduced during an experiment as a consequence of the application of two independent variables together to create a stimulus. With increased sophistication in modern chemical biology tools and related precision interrogation techniques, hidden variables have become integral to many chemical biologists' routine experiments. For instance, they can appear in the use of light-activatable chemical probes (e.g., μMap, T-REX), or stimulus-induced enzyme activation (e.g., APEX). Unfortunately, control experiments assess only how independent variables affect measured outcomes and not the multiple differences between the two independent variables and the twain. We outline ways to account for potential hidden variables in experimental design and data interpretation as a means to aid developers of new methods, particularly those involving light-driven techniques, chemical activation, or biorthogonal chemistries, to better incorporate well-controlled procedures.
Collapse
Affiliation(s)
- Marcus J. C. Long
- NCCR Chemical Biology and University of Geneva, 1211 Geneva, Switzerland
- University of Lausanne (UNIL), 1110 Epalinges, Switzerland
| | - Mahdi Assari
- Swiss Federal Institute of Technology Lausanne (EPFL), 1015 Lausanne, Switzerland
- NCCR Chemical Biology and University of Geneva, 1211 Geneva, Switzerland
- Department of Chemistry, The University of Chicago, Chicago, Illinois 60637, United States
| | - Yimon Aye
- Swiss Federal Institute of Technology Lausanne (EPFL), 1015 Lausanne, Switzerland
- NCCR Chemical Biology and University of Geneva, 1211 Geneva, Switzerland
| |
Collapse
|
11
|
Long MJC, Miranda Herrera PA, Aye Y. Hitting the Bullseye: Endogenous Electrophiles Show Remarkable Nuance in Signaling Regulation. Chem Res Toxicol 2022; 35:1636-1648. [PMID: 35394758 DOI: 10.1021/acs.chemrestox.2c00006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Our bodies produce a host of electrophilic species that can label specific endogenous proteins in cells. The signaling roles of these molecules are under active debate. However, in our opinion, it is becoming increasingly likely that electrophiles can rewire cellular signaling processes at endogenous levels. Attention is turning more to understanding how nuanced electrophile signaling in cells is. In this Perspective, we describe recent work from our laboratory that has started to inform on different levels of context-specific regulation of proteins by electrophiles. We discuss the relevance of these data to the field and to the broader application of electrophile signaling to precision medicine development, beyond the traditional views of their pleiotropic cytotoxic roles.
Collapse
Affiliation(s)
- Marcus J C Long
- National Centre of Competence in Research Chemical Biology, University of Geneva, 1211 Geneva, Switzerland.,Department of Biochemistry, Faculty of Biology and Medicine, University of Lausanne, 1066 Epalinges, Switzerland
| | - Pierre A Miranda Herrera
- Institute of Chemical Sciences and Engineering, School of Basic Sciences, Swiss Federal Institute of Technology Lausanne, 1015 Lausanne, Switzerland.,National Centre of Competence in Research Chemical Biology, University of Geneva, 1211 Geneva, Switzerland
| | - Yimon Aye
- Institute of Chemical Sciences and Engineering, School of Basic Sciences, Swiss Federal Institute of Technology Lausanne, 1015 Lausanne, Switzerland.,National Centre of Competence in Research Chemical Biology, University of Geneva, 1211 Geneva, Switzerland
| |
Collapse
|
12
|
The emerging role of mass spectrometry-based proteomics in drug discovery. Nat Rev Drug Discov 2022; 21:637-654. [PMID: 35351998 DOI: 10.1038/s41573-022-00409-3] [Citation(s) in RCA: 137] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2022] [Indexed: 12/14/2022]
Abstract
Proteins are the main targets of most drugs; however, system-wide methods to monitor protein activity and function are still underused in drug discovery. Novel biochemical approaches, in combination with recent developments in mass spectrometry-based proteomics instrumentation and data analysis pipelines, have now enabled the dissection of disease phenotypes and their modulation by bioactive molecules at unprecedented resolution and dimensionality. In this Review, we describe proteomics and chemoproteomics approaches for target identification and validation, as well as for identification of safety hazards. We discuss innovative strategies in early-stage drug discovery in which proteomics approaches generate unique insights, such as targeted protein degradation and the use of reactive fragments, and provide guidance for experimental strategies crucial for success.
Collapse
|
13
|
Kemper EK, Zhang Y, Dix MM, Cravatt BF. Global profiling of phosphorylation-dependent changes in cysteine reactivity. Nat Methods 2022; 19:341-352. [PMID: 35228727 PMCID: PMC8920781 DOI: 10.1038/s41592-022-01398-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 01/14/2022] [Indexed: 01/11/2023]
Abstract
Proteomics has revealed that the ~20,000 human genes engender a far greater number of proteins, or proteoforms, that are diversified in large part by post-translational modifications (PTMs). How such PTMs affect protein structure and function is an active area of research but remains technically challenging to assess on a proteome-wide scale. Here, we describe a chemical proteomic method to quantitatively relate serine/threonine phosphorylation to changes in the reactivity of cysteine residues, a parameter that can affect the potential for cysteines to be post-translationally modified or engaged by covalent drugs. Leveraging the extensive high-stoichiometry phosphorylation occurring in mitotic cells, we discover numerous cysteines that exhibit phosphorylation-dependent changes in reactivity on diverse proteins enriched in cell cycle regulatory pathways. The discovery of bidirectional changes in cysteine reactivity often occurring in proximity to serine/threonine phosphorylation events points to the broad impact of phosphorylation on the chemical reactivity of proteins and the future potential to create small-molecule probes that differentially target proteoforms with PTMs.
Collapse
Affiliation(s)
- Esther K Kemper
- The Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA.
| | - Yuanjin Zhang
- The Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Melissa M Dix
- The Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Benjamin F Cravatt
- The Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
14
|
Jung Y, Noda N, Takaya J, Abo M, Toh K, Tajiri K, Cui C, Zhou L, Sato SI, Uesugi M. Discovery of Non-Cysteine-Targeting Covalent Inhibitors by Activity-Based Proteomic Screening with a Cysteine-Reactive Probe. ACS Chem Biol 2022; 17:340-347. [PMID: 35076225 DOI: 10.1021/acschembio.1c00824] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Covalent inhibitors of enzymes are increasingly appreciated as pharmaceutical seeds, yet discovering non-cysteine-targeting inhibitors remains challenging. Herein, we report an intriguing experience during our activity-based proteomic screening of 1601 reactive small molecules, in which we monitored the ability of library molecules to compete with a cysteine-reactive iodoacetamide probe. One epoxide molecule, F8, exhibited unexpected enhancement of the probe reactivity for glyceraldehyde-3-phosphate dehydrogenase (GAPDH), a rate-limiting glycolysis enzyme. In-depth mechanistic analysis suggests that F8 forms a covalent adduct with an aspartic acid in the active site to displace NAD+, a cofactor of the enzyme, with concomitant enhancement of the probe reaction with the catalytic cysteine. The mechanistic underpinning permitted the identification of an optimized aspartate-reactive GAPDH inhibitor. Our findings exemplify that activity-based proteomic screening with a cysteine-reactive probe can be used for discovering covalent inhibitors that react with non-cysteine residues.
Collapse
Affiliation(s)
- Yejin Jung
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
- Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Naotaka Noda
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
- Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Junichiro Takaya
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
- Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Masahiro Abo
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Kohei Toh
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Ken Tajiri
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Changyi Cui
- School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Lu Zhou
- School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Shin-ichi Sato
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Motonari Uesugi
- Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
- School of Pharmacy, Fudan University, Shanghai 201203, China
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Uji, Kyoto 611-0011, Japan
| |
Collapse
|
15
|
Function-guided proximity mapping unveils electrophilic-metabolite sensing by proteins not present in their canonical locales. Proc Natl Acad Sci U S A 2022; 119:2120687119. [PMID: 35082156 PMCID: PMC8812531 DOI: 10.1073/pnas.2120687119] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/14/2021] [Indexed: 11/18/2022] Open
Abstract
Enzyme-assisted posttranslational modifications (PTMs) constitute a major means of signaling across different cellular compartments. However, how nonenzymatic PTMs-despite their direct relevance to covalent drug development-impinge on cross-compartment signaling remains inaccessible as current target-identification (target-ID) technologies offer limited spatiotemporal resolution, and proximity mapping tools are also not guided by specific, biologically-relevant, ligand chemotypes. Here we establish a quantitative and direct profiling platform (Localis-rex) that ranks responsivity of compartmentalized subproteomes to nonenzymatic PTMs. In a setup that contrasts nucleus- vs. cytoplasm-specific responsivity to reactive-metabolite modification (hydroxynonenylation), ∼40% of the top-enriched protein sensors investigated respond in compartments of nonprimary origin or where the canonical activity of the protein sensor is inoperative. CDK9-a primarily nuclear-localized kinase-was hydroxynonenylated only in the cytoplasm. Site-specific CDK9 hydroxynonenylation-which we identified in untreated cells-drives its nuclear translocation, downregulating RNA-polymerase-II activity, through a mechanism distinct from that of commonly used CDK9 inhibitors. Taken together, this work documents an unmet approach to quantitatively profile and decode localized and context-specific signaling/signal-propagation programs orchestrated by reactive covalent ligands.
Collapse
|
16
|
Yang F, Jia G, Guo J, Liu Y, Wang C. Quantitative Chemoproteomic Profiling with Data-Independent Acquisition-Based Mass Spectrometry. J Am Chem Soc 2022; 144:901-911. [PMID: 34986311 DOI: 10.1021/jacs.1c11053] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Activity-based protein profiling (ABPP) has emerged as a powerful and versatile tool to enable annotation of protein functions and discovery of targets of bioactive ligands in complex biological systems. It utilizes chemical probes to covalently label functional sites in proteins so that they can be enriched for mass spectrometry (MS)-based quantitative proteomics analysis. However, the semistochastic nature of data-dependent acquisition and high cost associated with isotopically encoded quantification reagents compromise the power of ABPP in multidimensional analysis and high-throughput screening, when a large number of samples need to be quantified in parallel. Here, we combine the data-independent acquisition (DIA) MS with ABPP to develop an efficient label-free quantitative chemical proteomic method, DIA-ABPP, with good reproducibility and high accuracy for high-throughput quantification. We demonstrated the power of DIA-ABPP for comprehensive profiling of functional cysteineome in three distinct applications, including dose-dependent quantification of cysteines' sensitivity toward a reactive metabolite, screening of ligandable cysteines with a covalent fragment library, and profiling of cysteinome fluctuation in circadian clock cycles. DIA-ABPP will open new opportunities for in-depth and multidimensional profiling of functional proteomes and interactions with bioactive small molecules in complex biological systems.
Collapse
Affiliation(s)
- Fan Yang
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Guogeng Jia
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Jiuzhou Guo
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Yuan Liu
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Chu Wang
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.,Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| |
Collapse
|
17
|
Long MJC, Ly P, Aye Y. A primer on harnessing non-enzymatic post-translational modifications for drug design. RSC Med Chem 2021; 12:1797-1807. [PMID: 34825181 PMCID: PMC8597429 DOI: 10.1039/d1md00157d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 10/08/2021] [Indexed: 11/21/2022] Open
Abstract
Of the manifold concepts in drug discovery and design, covalent drugs have re-emerged as one of the most promising over the past 20-or so years. All such drugs harness the ability of a covalent bond to drive an interaction between a target biomolecule, typically a protein, and a small molecule. Formation of a covalent bond necessarily prolongs target engagement, opening avenues to targeting shallower binding sites, protein complexes, and other difficult to drug manifolds, amongst other virtues. This opinion piece discusses frameworks around which to develop covalent drugs. Our argument, based on results from our research program on natural electrophile signaling, is that targeting specific residues innately involved in native signaling programs are ideally poised to be targeted by covalent drugs. We outline ways to identify electrophile-sensing residues, and discuss how studying ramifications of innate signaling by endogenous molecules can provide a means to predict drug mechanism and function and assess on- versus off-target behaviors.
Collapse
Affiliation(s)
| | - Phillippe Ly
- Swiss Federal Institute of Technology in Lausanne (EPFL) 1015 Lausanne Switzerland
| | - Yimon Aye
- Swiss Federal Institute of Technology in Lausanne (EPFL) 1015 Lausanne Switzerland
| |
Collapse
|
18
|
Long MJC, Huang KT, Aye Y. The not so identical twins: (dis)similarities between reactive electrophile and oxidant sensing and signaling. Chem Soc Rev 2021; 50:12269-12291. [PMID: 34779447 DOI: 10.1039/d1cs00467k] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In this tutorial review, we compare and contrast the chemical mechanisms of electrophile/oxidant sensing, and the molecular mechanisms of signal propagation. We critically analyze biological systems in which these different pathways are believed to be manifest and what the data really mean. Finally, we discuss applications of this knowledge to disease treatment and drug development.
Collapse
Affiliation(s)
| | - Kuan-Ting Huang
- Swiss Federal Institute of Technology in Lausanne (EPFL), 1015 Lausanne, Switzerland.
| | - Yimon Aye
- Swiss Federal Institute of Technology in Lausanne (EPFL), 1015 Lausanne, Switzerland.
| |
Collapse
|
19
|
Catalano C, AL Mughram MH, Guo Y, Kellogg GE. 3D interaction homology: Hydropathic interaction environments of serine and cysteine are strikingly different and their roles adapt in membrane proteins. Curr Res Struct Biol 2021; 3:239-256. [PMID: 34693344 PMCID: PMC8517007 DOI: 10.1016/j.crstbi.2021.09.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/23/2021] [Accepted: 09/23/2021] [Indexed: 12/12/2022] Open
Abstract
Atomic-resolution protein structural models are prerequisites for many downstream activities like structure-function studies or structure-based drug discovery. Unfortunately, this data is often unavailable for some of the most interesting and therapeutically important proteins. Thus, computational tools for building native-like structural models from less-than-ideal experimental data are needed. To this end, interaction homology exploits the character, strength and loci of the sets of interactions that define a structure. Each residue type has its own limited set of backbone angle-dependent interaction motifs, as defined by their environments. In this work, we characterize the interactions of serine, cysteine and S-bridged cysteine in terms of 3D hydropathic environment maps. As a result, we explore several intriguing questions. Are the environments different between the isosteric serine and cysteine residues? Do some environments promote the formation of cystine S-S bonds? With the increasing availability of structural data for water-insoluble membrane proteins, are there environmental differences for these residues between soluble and membrane proteins? The environments surrounding serine and cysteine residues are dramatically different: serine residues are about 50% solvent exposed, while cysteines are only 10% exposed; the latter are more involved in hydrophobic interactions although there are backbone angle-dependent differences. Our analysis suggests that one driving force for -S-S- bond formation is a rather substantial increase in burial and hydrophobic interactions in cystines. Serine and cysteine become less and more, respectively, solvent-exposed in membrane proteins. 3D hydropathic environment maps are an evolving structure analysis tool showing promise as elements in a new protein structure prediction paradigm.
Collapse
Affiliation(s)
- Claudio Catalano
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA, USA
- Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA, USA
| | - Mohammed H. AL Mughram
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA, USA
- Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA, USA
| | - Youzhong Guo
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA, USA
- Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA, USA
| | - Glen E. Kellogg
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA, USA
- Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA, USA
- Center for the Study of Biological Complexity, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
20
|
Poganik JR, Huang KT, Parvez S, Zhao Y, Raja S, Long MJC, Aye Y. Wdr1 and cofilin are necessary mediators of immune-cell-specific apoptosis triggered by Tecfidera. Nat Commun 2021; 12:5736. [PMID: 34593792 PMCID: PMC8484674 DOI: 10.1038/s41467-021-25466-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 08/11/2021] [Indexed: 01/10/2023] Open
Abstract
Despite the emerging importance of reactive electrophilic drugs, deconvolution of their principal targets remains difficult. The lack of genetic tractability/interventions and reliance on secondary validation using other non-specific compounds frequently complicate the earmarking of individual binders as functionally- or phenotypically-sufficient pathway regulators. Using a redox-targeting approach to interrogate how on-target binding of pleiotropic electrophiles translates to a phenotypic output in vivo, we here systematically track the molecular components attributable to innate immune cell toxicity of the electrophilic-drug dimethyl fumarate (Tecfidera®). In a process largely independent of canonical Keap1/Nrf2-signaling, Keap1-specific modification triggers mitochondrial-targeted neutrophil/macrophage apoptosis. On-target Keap1–ligand-engagement is accompanied by dissociation of Wdr1 from Keap1 and subsequent coordination with cofilin, intercepting Bax. This phagocytic-specific cell-killing program is recapitulated by whole-animal administration of dimethyl fumarate, where individual depletions of the players identified above robustly suppress apoptosis. The mechanism-of-action of many electrohilic drugs remains poorly understood. Here, the authors use a redox-targeting approach to elucidate the basis for the innate immune cell toxicity of dimethyl fumarate, showing that it modifies Keap1 to trigger mitochondrial-targeted neutrophil/macrophage apoptosis.
Collapse
Affiliation(s)
- Jesse R Poganik
- Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland.,Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kuan-Ting Huang
- Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Saba Parvez
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT, USA
| | - Yi Zhao
- BayRay Innovation Center, Shenzhen Bay Laboratory (SZBL), Guangdong, China
| | - Sruthi Raja
- Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | | | - Yimon Aye
- Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
21
|
Abstract
CoVID-19 is a multi-symptomatic disease which has made a global impact due to its ability to spread rapidly, and its relatively high mortality rate. Beyond the heroic efforts to develop vaccines, which we do not discuss herein, the response of scientists and clinicians to this complex problem has reflected the need to detect CoVID-19 rapidly, to diagnose patients likely to show adverse symptoms, and to treat severe and critical CoVID-19. Here we aim to encapsulate these varied and sometimes conflicting approaches and the resulting data in terms of chemistry and biology. In the process we highlight emerging concepts, and potential future applications that may arise out of this immense effort.
Collapse
Affiliation(s)
| | - Yimon Aye
- Swiss Federal Institute of Technology in Lausanne (EPFL)1015LausanneSwitzerland
| |
Collapse
|
22
|
Suto N, Kamoshita S, Hosoya S, Sakurai K. Exploration of the Reactivity of Multivalent Electrophiles for Affinity Labeling: Sulfonyl Fluoride as a Highly Efficient and Selective Label. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202104347] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Nanako Suto
- Department of Bioengineering and Life Science Tokyo University of Agriculture and Technology 4-24-16, Naka-cho, Koganei-shi Tokyo 184-8588 Japan
| | - Shione Kamoshita
- Department of Bioengineering and Life Science Tokyo University of Agriculture and Technology 4-24-16, Naka-cho, Koganei-shi Tokyo 184-8588 Japan
| | - Shoichi Hosoya
- Institute of Research Tokyo Medical and Dental University 1-5-45, Yushima, Bunkyo-ku Tokyo 113-8510 Japan
| | - Kaori Sakurai
- Department of Bioengineering and Life Science Tokyo University of Agriculture and Technology 4-24-16, Naka-cho, Koganei-shi Tokyo 184-8588 Japan
| |
Collapse
|
23
|
Zerhouni M, Martin AR, Furstoss N, Gutierrez VS, Jaune E, Tekaya N, Beranger GE, Abbe P, Regazzetti C, Amdouni H, Driowya M, Dubreuil P, Luciano F, Jacquel A, Tulic MK, Cluzeau T, O'Hara BP, Ben-Sahra I, Passeron T, Benhida R, Robert G, Auberger P, Rocchi S. Dual Covalent Inhibition of PKM and IMPDH Targets Metabolism in Cutaneous Metastatic Melanoma. Cancer Res 2021; 81:3806-3821. [PMID: 34099492 DOI: 10.1158/0008-5472.can-20-2114] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 12/08/2020] [Accepted: 06/02/2021] [Indexed: 11/16/2022]
Abstract
Overcoming acquired drug resistance is a primary challenge in cancer treatment. Notably, more than 50% of patients with BRAFV600E cutaneous metastatic melanoma (CMM) eventually develop resistance to BRAF inhibitors. Resistant cells undergo metabolic reprogramming that profoundly influences therapeutic response and promotes tumor progression. Uncovering metabolic vulnerabilities could help suppress CMM tumor growth and overcome drug resistance. Here we identified a drug, HA344, that concomitantly targets two distinct metabolic hubs in cancer cells. HA344 inhibited the final and rate-limiting step of glycolysis through its covalent binding to the pyruvate kinase M2 (PKM2) enzyme, and it concurrently blocked the activity of inosine monophosphate dehydrogenase, the rate-limiting enzyme of de novo guanylate synthesis. As a consequence, HA344 efficiently targeted vemurafenib-sensitive and vemurafenib-resistant CMM cells and impaired CMM xenograft tumor growth in mice. In addition, HA344 acted synergistically with BRAF inhibitors on CMM cell lines in vitro. Thus, the mechanism of action of HA344 provides potential therapeutic avenues for patients with CMM and a broad range of different cancers. SIGNIFICANCE: Glycolytic and purine synthesis pathways are often deregulated in therapy-resistant tumors and can be targeted by the covalent inhibitor described in this study, suggesting its broad application for overcoming resistance in cancer.
Collapse
Affiliation(s)
- Marwa Zerhouni
- Université Côte d'azur, Nice, France
- Inserm U1065, C3M, Team 2, Nice, France
- Inserm U1065, C3M, Team 12, Nice, France
| | - Anthony R Martin
- Université Côte d'azur, Nice, France
- Institut de Chimie de Nice UMR 7272, Nice, France
| | - Nathan Furstoss
- Université Côte d'azur, Nice, France
- Inserm U1065, C3M, Team 2, Nice, France
| | - Vincent S Gutierrez
- Université Côte d'azur, Nice, France
- Institut de Chimie de Nice UMR 7272, Nice, France
| | - Emilie Jaune
- Université Côte d'azur, Nice, France
- Inserm U1065, C3M, Team 12, Nice, France
| | - Nedra Tekaya
- Université Côte d'azur, Nice, France
- Inserm U1065, C3M, Team 12, Nice, France
| | | | - Patricia Abbe
- Université Côte d'azur, Nice, France
- Inserm U1065, C3M, Team 12, Nice, France
| | - Claire Regazzetti
- Université Côte d'azur, Nice, France
- Inserm U1065, C3M, Team 12, Nice, France
| | - Hella Amdouni
- Université Côte d'azur, Nice, France
- Institut de Chimie de Nice UMR 7272, Nice, France
| | - Mohsine Driowya
- Université Côte d'azur, Nice, France
- Institut de Chimie de Nice UMR 7272, Nice, France
| | - Patrice Dubreuil
- CRCM, Team Signalisation, Hématopoïèse et Mécanismes de l'Oncogenèse, Marseille, France
| | - Frédéric Luciano
- Université Côte d'azur, Nice, France
- Inserm U1065, C3M, Team 2, Nice, France
| | - Arnaud Jacquel
- Université Côte d'azur, Nice, France
- Inserm U1065, C3M, Team 2, Nice, France
| | - Meri K Tulic
- Université Côte d'azur, Nice, France
- Inserm U1065, C3M, Team 2, Nice, France
| | - Thomas Cluzeau
- Université Côte d'azur, Nice, France
- Inserm U1065, C3M, Team 2, Nice, France
- CHU de Nice, Nice, France
| | - Brendan P O'Hara
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, Illinois
| | - Issam Ben-Sahra
- Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, Illinois
| | - Thierry Passeron
- Université Côte d'azur, Nice, France
- Inserm U1065, C3M, Team 12, Nice, France
- CHU de Nice, Nice, France
| | | | - Guillaume Robert
- Université Côte d'azur, Nice, France
- Inserm U1065, C3M, Team 2, Nice, France
| | - Patrick Auberger
- Université Côte d'azur, Nice, France.
- Inserm U1065, C3M, Team 2, Nice, France
| | - Stéphane Rocchi
- Université Côte d'azur, Nice, France.
- Inserm U1065, C3M, Team 12, Nice, France
| |
Collapse
|
24
|
Suto N, Kamoshita S, Hosoya S, Sakurai K. Exploration of the Reactivity of Multivalent Electrophiles for Affinity Labeling: Sulfonyl Fluoride as a Highly Efficient and Selective Label. Angew Chem Int Ed Engl 2021; 60:17080-17087. [PMID: 34060195 DOI: 10.1002/anie.202104347] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/16/2021] [Indexed: 12/23/2022]
Abstract
Here we explored the reactivity of a set of multivalent electrophiles cofunctionalized with a carbohydrate ligand on gold nanoparticles to achieve efficient affinity labeling for target protein analysis. Evaluation of the reactivity and selectivity of the electrophiles against three different cognate binding proteins identified arylsulfonyl fluoride as the most efficient protein-reactive group in this study. We demonstrated that multivalent arylsulfonyl fluoride probe 4 at 50 nm concentration achieved selective affinity labeling and enrichment of a model protein PNA in cell lysate, which was more effective than photoaffinity probe 1 with arylazide group. Labeling site analysis by LC-MS/MS revealed that the nanoparticle-immobilized arylsulfonyl fluoride group can target multiple amino acid residues around the ligand binding site of the target proteins. Our study highlights the utility of arylsulfonyl fluoride as a highly effective multivalent affinity label suitable for covalently capturing unknown target proteins.
Collapse
Affiliation(s)
- Nanako Suto
- Department of Bioengineering and Life Science, Tokyo University of Agriculture and Technology, 4-24-16, Naka-cho, Koganei-shi, Tokyo, 184-8588, Japan
| | - Shione Kamoshita
- Department of Bioengineering and Life Science, Tokyo University of Agriculture and Technology, 4-24-16, Naka-cho, Koganei-shi, Tokyo, 184-8588, Japan
| | - Shoichi Hosoya
- Institute of Research, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Kaori Sakurai
- Department of Bioengineering and Life Science, Tokyo University of Agriculture and Technology, 4-24-16, Naka-cho, Koganei-shi, Tokyo, 184-8588, Japan
| |
Collapse
|
25
|
Pogmore JP, Uehling D, Andrews DW. Pharmacological Targeting of Executioner Proteins: Controlling Life and Death. J Med Chem 2021; 64:5276-5290. [PMID: 33939407 DOI: 10.1021/acs.jmedchem.0c02200] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Small-molecule mediated modulation of protein interactions of Bcl-2 (B-cell lymphoma-2) family proteins was clinically validated in 2015 when Venetoclax, a selective inhibitor of the antiapoptotic protein BCL-2, achieved breakthrough status designation by the FDA for treatment of lymphoid malignancies. Since then, substantial progress has been made in identifying inhibitors of other interactions of antiapoptosis proteins. However, targeting their pro-apoptotic counterparts, the "executioners" BAX, BAK, and BOK that both initiate and commit the cell to dying, has lagged behind. However, recent publications demonstrate that these proteins can be positively or negatively regulated using small molecule tool compounds. The results obtained with these molecules suggest that pharmaceutical regulation of apoptosis will have broad implications that extend beyond activating cell death in cancer. We review recent advances in identifying compounds and their utility in the exogenous control of life and death by regulating executioner proteins, with emphasis on the prototype BAX.
Collapse
Affiliation(s)
- Justin P Pogmore
- Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1J7, Canada.,Biological Sciences, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario M4N 3M5, Canada
| | - David Uehling
- Drug Discovery Program, Ontario Institute for Cancer Research, Toronto, Ontario M5G 1M1, Canada
| | - David W Andrews
- Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1J7, Canada.,Biological Sciences, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario M4N 3M5, Canada
| |
Collapse
|
26
|
Reimagining high-throughput profiling of reactive cysteines for cell-based screening of large electrophile libraries. Nat Biotechnol 2021; 39:630-641. [PMID: 33398154 PMCID: PMC8316984 DOI: 10.1038/s41587-020-00778-3] [Citation(s) in RCA: 158] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 11/17/2020] [Indexed: 01/28/2023]
Abstract
Current methods used for measuring amino acid side-chain reactivity lack the throughput needed to screen large chemical libraries for interactions across the proteome. Here we redesigned the workflow for activity-based protein profiling of reactive cysteine residues by using a smaller desthiobiotin-based probe, sample multiplexing, reduced protein starting amounts and software to boost data acquisition in real time on the mass spectrometer. Our method, streamlined cysteine activity-based protein profiling (SLC-ABPP), achieved a 42-fold improvement in sample throughput, corresponding to profiling library members at a depth of >8,000 reactive cysteine sites at 18 min per compound. We applied it to identify proteome-wide targets of covalent inhibitors to mutant Kirsten rat sarcoma (KRAS)G12C and Bruton's tyrosine kinase (BTK). In addition, we created a resource of cysteine reactivity to 285 electrophiles in three human cell lines, which includes >20,000 cysteines from >6,000 proteins per line. The goal of proteome-wide profiling of cysteine reactivity across thousand-member libraries under several cellular contexts is now within reach.
Collapse
|
27
|
Guan I, Williams K, Pan J, Liu X. New Cysteine Covalent Modification Strategies Enable Advancement of Proteome‐wide Selectivity of Kinase Modulators. ASIAN J ORG CHEM 2021. [DOI: 10.1002/ajoc.202100036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Ivy Guan
- School of Chemistry The Heart Research Institute The University of Sydney Sydney New South Wales 2006 Australia
| | - Kayla Williams
- School of Chemistry The University of Sydney Sydney New South Wales 2006 Australia
| | - Jolyn Pan
- Faculty of Science & Engineering The University of Waikato 124 Hillcrest Road, Hillcrest Hamilton 3216 New Zealand
| | - Xuyu Liu
- School of Chemistry The Heart Research Institute The University of Sydney Sydney New South Wales 2006 Australia
| |
Collapse
|
28
|
Long MJC, Kulkarni A, Aye Y. Can Precision Electrophile Signaling Make a Meaningful and Lasting Impression in Drug Design? Chembiochem 2021; 23:e202100051. [PMID: 33826211 DOI: 10.1002/cbic.202100051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/04/2021] [Indexed: 12/14/2022]
Abstract
For several years, drugs with reactive electrophilic appendages have been developed. These units typically confer prolonged residence time of the drugs on their protein targets, and may assist targeting shallow binding sites and/or improving the drug-protein target spectrum. Studies on natural electrophilic molecules have indicated that, in many instances, natural electrophiles use similar mechanisms to alter signaling pathways. However, natural reactive species are also endowed with other important mechanisms to hone signaling properties that are uncommon in drug design. These include ability to be active at low occupancy and elevated inhibitor kinetics. Herein, we discuss how we have begun to harness these properties in inhibitor design.
Collapse
Affiliation(s)
- Marcus J C Long
- University of Lausanne, Department of Biochemistry, Chemin des boveresses 155, Epalinges, 1066, Lausanne, Switzerland
| | - Amogh Kulkarni
- Swiss Federal Institute of Technology in Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Yimon Aye
- Swiss Federal Institute of Technology in Lausanne (EPFL), 1015, Lausanne, Switzerland
| |
Collapse
|
29
|
Long MJC, Rogg C, Aye Y. An Oculus to Profile and Probe Target Engagement In Vivo: How T-REX Was Born and Its Evolution into G-REX. Acc Chem Res 2021; 54:618-631. [PMID: 33228351 DOI: 10.1021/acs.accounts.0c00537] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Here we provide a personal account of innovation and design principles underpinning a method to interrogate precision electrophile signaling that has come to be known as "REX technologies". This Account is framed in the context of trying to improve methods of target mining and understanding of individual target-ligand engagement by a specific natural electrophile and the ramifications of this labeling event in cells and organisms. We start by explaining from a practical standpoint why gleaning such understanding is critical: we are constantly assailed by a battery of electrophilic molecules that exist as a consequence of diet, food preparation, ineluctable endogenous metabolic processes, and potentially disease. The resulting molecules, which are detectable in the body, appear to be able to modify function of specific proteins. Aside from potentially being biologically relevant in their own right, these labeling events are essentially identical to protein-covalent drug interactions. Thus, on what proteins and even in what ways a covalent drug will work can be understood through the eyes of natural electrophiles; extending this logic leads to the postulate that target identification of specific electrophiles can inform on drug design. However, when we entered this field, there was no way to interrogate how a specific labeling event impacted a specific protein in an unperturbed cell. Methods to evaluate stoichiometry of labeling, and even chemospecificity of a specific phenotype were limited. There were further no generally accepted ways to study electrophile signaling that did not hugely disturb physiology.We developed T-REX, a method to study single-protein-specific electrophile engagement, to interrogate how single-protein electrophile labeling shapes pathway flux. Using T-REX, we discovered that labeling of several proteins by a specific electrophile, even at low occupancy, leads to biologically relevant signaling outputs. Further experimentation using T-REX showed that in some instances, single-protein isoforms were electrophile responsive against other isoforms, such as Akt3. Selective electrophile-labeling of Akt3 elicited inhibition of Akt-pathway flux in cells and in zebrafish embryos. Using these data, we rationally designed a molecule to selectively target Akt3. This was a fusion of the naturally derived electrophile and an isoform-nonspecific, reversible Akt inhibitor in phase-II trials, MK-2206. The resulting molecule was a selective inhibitor of Akt3 and was shown to fare better than MK-2206 in breast cancer xenograft mouse models. Recently, we have also developed a means to screen electrophile sensors that is unbiased and uses a precise burst of electrophiles. Using this method, dubbed G-REX, in conjunction with T-REX, we discovered new DNA-damage response upregulation pathways orchestrated by simple natural electrophiles. We thus emphasize how deriving a quantitative understanding of electrophile signaling that is linked to thorough and precise mechanistic studies can open doors to numerous medicinally and biologically relevant insights, from gleaning better understanding of target engagement and target mining to rational design of targeted covalent medicines.
Collapse
Affiliation(s)
- Marcus J. C. Long
- Department of Molecular Biology, University of Geneva, 30 quai Ernest-Ansermet, 1211 Geneva, Switzerland
| | - Chloé Rogg
- Institute of Chemical Sciences and Engineering (ISIC), Swiss Federal Institute of Technology Lausanne (EPFL), Route Cantonale, 1015 Lausanne, Switzerland
| | - Yimon Aye
- Institute of Chemical Sciences and Engineering (ISIC), Swiss Federal Institute of Technology Lausanne (EPFL), Route Cantonale, 1015 Lausanne, Switzerland
| |
Collapse
|
30
|
Meng F, Liang Z, Zhao K, Luo C. Drug design targeting active posttranslational modification protein isoforms. Med Res Rev 2020; 41:1701-1750. [PMID: 33355944 DOI: 10.1002/med.21774] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/29/2020] [Accepted: 12/03/2020] [Indexed: 12/11/2022]
Abstract
Modern drug design aims to discover novel lead compounds with attractable chemical profiles to enable further exploration of the intersection of chemical space and biological space. Identification of small molecules with good ligand efficiency, high activity, and selectivity is crucial toward developing effective and safe drugs. However, the intersection is one of the most challenging tasks in the pharmaceutical industry, as chemical space is almost infinity and continuous, whereas the biological space is very limited and discrete. This bottleneck potentially limits the discovery of molecules with desirable properties for lead optimization. Herein, we present a new direction leveraging posttranslational modification (PTM) protein isoforms target space to inspire drug design termed as "Post-translational Modification Inspired Drug Design (PTMI-DD)." PTMI-DD aims to extend the intersections of chemical space and biological space. We further rationalized and highlighted the importance of PTM protein isoforms and their roles in various diseases and biological functions. We then laid out a few directions to elaborate the PTMI-DD in drug design including discovering covalent binding inhibitors mimicking PTMs, targeting PTM protein isoforms with distinctive binding sites from that of wild-type counterpart, targeting protein-protein interactions involving PTMs, and hijacking protein degeneration by ubiquitination for PTM protein isoforms. These directions will lead to a significant expansion of the biological space and/or increase the tractability of compounds, primarily due to precisely targeting PTM protein isoforms or complexes which are highly relevant to biological functions. Importantly, this new avenue will further enrich the personalized treatment opportunity through precision medicine targeting PTM isoforms.
Collapse
Affiliation(s)
- Fanwang Meng
- Drug Discovery and Design Center, the Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario, Canada
| | - Zhongjie Liang
- Center for Systems Biology, Department of Bioinformatics, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Kehao Zhao
- 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, China
| | - Cheng Luo
- Drug Discovery and Design Center, the Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
31
|
Long MJC, Wang L, Aye Y. Getting the Right Grip? How Understanding Electrophile Selectivity Profiles Could Illuminate Our Understanding of Redox Signaling. Antioxid Redox Signal 2020; 33:1077-1091. [PMID: 31578876 PMCID: PMC7583342 DOI: 10.1089/ars.2019.7894] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Significance: Electrophile signaling is coming into focus as a bona fide cell signaling mechanism. The electrophilic regulation occurs typically through a sensing event (i.e., labeling of a protein) and a signaling event (the labeling event having an effect of the proteins activity, association, etc.). Recent Advances: Herein, we focus on the first step of this process, electrophile sensing. Electrophile sensing is typically a deceptively simple reaction between the thiol of a protein cysteine, of which there are around 200,000 in the human proteome, and a Michael acceptor, of which there are numerous flavors, including enals and enones. Recent data overall paint a picture that despite being a simple chemical reaction, electrophile sensing is a discerning process, showing labeling preferences that are often not in line with reactivity of the electrophile. Critical Issues: With a view to trying to decide what brings about highly electrophile-reactive protein cysteines, and how reactive these sensors may be, we discuss aspects of the thermodynamics and kinetics of covalent/noncovalent binding. Data made available by several laboratories indicate that it is likely that specific proteins exhibit highly stereo- and chemoselective electrophile sensing, which we take as good evidence for recognition between the electrophile and the protein before forming a covalent bond. Future Directions: We propose experiments that could help us gain a better and more quantitative understanding of the mechanisms through which sensing comes about. We further extoll the importance of performing more detailed experiments on labeling and trying to standardize the way we assess protein-specific electrophile sensing.
Collapse
Affiliation(s)
- Marcus J C Long
- 47 Pudding Gate, Bishop Burton, Beverley East Riding of Yorkshire, United Kingdom
| | - Lingxi Wang
- Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Yimon Aye
- Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
32
|
Long MJC. Time to Get Turned on by Chemical Biology. Chembiochem 2020; 22:814-817. [PMID: 33174365 DOI: 10.1002/cbic.202000497] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/07/2020] [Indexed: 11/08/2022]
Abstract
The pressing need for innovation in drug discovery is spurring the emergence of drugs that turn on protein function, as opposed to shutting activity down. Several pharmacophores usher protein target gain-of-function, for instance: PROTACs promote protein target degradation; other drug candidates have been reported to function through dominant-negative inhibition of their target enzyme. Such classes of molecules are typically active at low target occupancy and display numerous advantages relative to canonical inhibitors, whose function is intrinsically tied to achieving, or exceeding a threshold occupancy. However, our ability to generally tap into gain-of-function processes through small molecule interventions is overall in its infancy. Herein, I outline how chemical biology is poised to help us bring this powerful idea to fruition. I further outline means through which gain-of-function events can be identified and harnessed.
Collapse
Affiliation(s)
- Marcus J C Long
- Départment de Biologie Moleculaire; Sciences II, 30 Quai Ernest-Ansermet, 1211, Genève 4, Switzerland
| |
Collapse
|
33
|
Shergalis A, Xue D, Gharbia FZ, Driks H, Shrestha B, Tanweer A, Cromer K, Ljungman M, Neamati N. Characterization of Aminobenzylphenols as Protein Disulfide Isomerase Inhibitors in Glioblastoma Cell Lines. J Med Chem 2020; 63:10263-10286. [PMID: 32830969 PMCID: PMC8103808 DOI: 10.1021/acs.jmedchem.0c00728] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Disulfide bond formation is a critical post-translational modification of newly synthesized polypeptides in the oxidizing environment of the endoplasmic reticulum and is mediated by protein disulfide isomerase (PDIA1). In this study, we report a series of α-aminobenzylphenol analogues as potent PDI inhibitors. The lead compound, AS15, is a covalent nanomolar inhibitor of PDI, and the combination of AS15 analogues with glutathione synthesis inhibitor buthionine sulfoximine (BSO) leads to synergistic cell growth inhibition. Using nascent RNA sequencing, we show that an AS15 analogue triggers the unfolded protein response in glioblastoma cells. A BODIPY-labeled analogue binds proteins including PDIA1, suggesting that the compounds are cell-permeable and reach the intended target. Taken together, these findings demonstrate an extensive biochemical characterization of a novel series of highly potent reactive small molecules that covalently bind to PDI.
Collapse
Affiliation(s)
- Andrea Shergalis
- Department of Medicinal Chemistry, College of Pharmacy, Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Ding Xue
- Department of Medicinal Chemistry, College of Pharmacy, Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Fatma Z. Gharbia
- Department of Medicinal Chemistry, College of Pharmacy, Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Hannah Driks
- Department of Medicinal Chemistry, College of Pharmacy, Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Binita Shrestha
- Department of Medicinal Chemistry, College of Pharmacy, Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Amina Tanweer
- Department of Medicinal Chemistry, College of Pharmacy, Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Kirin Cromer
- Department of Medicinal Chemistry, College of Pharmacy, Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Mats Ljungman
- Department of Radiation Oncology, University of Michigan Medical School and Rogel Cancer Center, School of Public Health, Ann Arbor, Michigan 48109, United States
| | - Nouri Neamati
- Department of Medicinal Chemistry, College of Pharmacy, Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
34
|
Liu X, Long MJC, Hopkins BD, Luo C, Wang L, Aye Y. Precision Targeting of pten-Null Triple-Negative Breast Tumors Guided by Electrophilic Metabolite Sensing. ACS CENTRAL SCIENCE 2020; 6:892-902. [PMID: 32607436 PMCID: PMC7318068 DOI: 10.1021/acscentsci.9b00893] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Indexed: 06/11/2023]
Abstract
Off-target effects continue to impede disease interventions, particularly when targeting a specific protein within a family of similar proteins, such as kinase isoforms that play tumor-subtype-specific roles in cancers. Exploiting the specific electrophilic-metabolite-sensing capability of Akt3, versus moderate or no sensing, respectively, by Akt2 and Akt1, we describe a first-in-class functionally Akt3-selective covalent inhibitor [MK-H(F)NE], wherein the electrophilic core is derived from the native reactive lipid metabolite HNE. Mechanistic profiling and pathway interrogations point to retention of the metabolite's structure-as opposed to implicit electrophilicity-as being essential for biasing isoform preference, which we found translates to tumor-subtype specificity against pten-null triple-negative breast cancers (TNBCs). MK-H(F)NE further enables novel downstream target identification specific to Akt3-function in disease. In TNBC xenografts, MK-H(F)NE fares better than reversible pan-Akt-inhibitors and does not show commonly observed side-effects associated with Akt1-inhibition. Inhibitors derived from native-metabolite sensing are thus an enabling plan-of-action for unmasking kinase-isoform-biased molecular targets and tumor-subtype-specific interventions.
Collapse
Affiliation(s)
- Xuyu Liu
- School
of Chemistry, The University of Sydney, Sydney, New South Wales 2006, Australia
- The
Heart Research Institute, Newtown, New South Wales 2042, Australia
| | - Marcus J. C. Long
- Department
of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York 14850, United States
| | - Benjamin D. Hopkins
- Department
of Genetics and Genomic Sciences, Icahn
School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Chaosheng Luo
- Swiss
Federal Institute of Technology Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Lingxi Wang
- Swiss
Federal Institute of Technology Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Yimon Aye
- Swiss
Federal Institute of Technology Lausanne (EPFL), 1015 Lausanne, Switzerland
| |
Collapse
|
35
|
Poganik JR, Van Hall-Beauvais AK, Long MJC, Disare MT, Zhao Y, Aye Y. The mRNA-Binding Protein HuR Is a Kinetically-Privileged Electrophile Sensor. Helv Chim Acta 2020; 103:e2000041. [PMID: 34113045 PMCID: PMC8188987 DOI: 10.1002/hlca.202000041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 04/11/2020] [Indexed: 12/11/2022]
Abstract
The key mRNA-binding proteins HuR and AUF1 are reported stress sensors in mammals. Intrigued by recent reports of sensitivity of these proteins to the electrophilic lipid prostaglandin A2 and other redox signals, we here examined their sensing abilities to a prototypical redox-linked lipid-derived electrophile, 4-hydroxynonenal (HNE). Leveraging our T-REX electrophile delivery platform, we found that only HuR, and not AUF1, is a kinetically-privileged sensor of HNE in HEK293T cells, and sensing functions through a specific cysteine, C13. Cells depleted of HuR, upon treatment with HNE, manifest unique alterations in cell viability and Nrf2-transcription-factor-driven antioxidant response (AR), which our recent work shows is regulated by HuR at the Nrf2-mRNA level. Mutagenesis studies showed that C13-specific sensing alone is not sufficient to explain HuR-dependent stress responsivities, further highlighting a complex context-dependent layer of Nrf2/AR regulation through HuR.
Collapse
Affiliation(s)
- Jesse R Poganik
- Institute of Chemical Sciences & Engineering (ISIC), Swiss Federal Institute of Technology Lausanne (EPFL), CH-1015 Lausanne
- Department of Chemistry & Chemical Biology, Cornell University, Ithaca, New York, 14853 New York, United States
| | - Alexandra K Van Hall-Beauvais
- Institute of Chemical Sciences & Engineering (ISIC), Swiss Federal Institute of Technology Lausanne (EPFL), CH-1015 Lausanne
| | - Marcus J C Long
- Department of Chemistry & Chemical Biology, Cornell University, Ithaca, New York, 14853 New York, United States
| | - Michael T Disare
- Department of Chemistry & Chemical Biology, Cornell University, Ithaca, New York, 14853 New York, United States
| | - Yi Zhao
- Institute of Chemical Sciences & Engineering (ISIC), Swiss Federal Institute of Technology Lausanne (EPFL), CH-1015 Lausanne
| | - Yimon Aye
- Institute of Chemical Sciences & Engineering (ISIC), Swiss Federal Institute of Technology Lausanne (EPFL), CH-1015 Lausanne
| |
Collapse
|
36
|
Cheng SS, Yang GJ, Wang W, Leung CH, Ma DL. The design and development of covalent protein-protein interaction inhibitors for cancer treatment. J Hematol Oncol 2020; 13:26. [PMID: 32228680 PMCID: PMC7106679 DOI: 10.1186/s13045-020-00850-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 02/20/2020] [Indexed: 12/12/2022] Open
Abstract
Protein-protein interactions (PPIs) are central to a variety of biological processes, and their dysfunction is implicated in the pathogenesis of a range of human diseases, including cancer. Hence, the inhibition of PPIs has attracted significant attention in drug discovery. Covalent inhibitors have been reported to achieve high efficiency through forming covalent bonds with cysteine or other nucleophilic residues in the target protein. Evidence suggests that there is a reduced risk for the development of drug resistance against covalent drugs, which is a major challenge in areas such as oncology and infectious diseases. Recent improvements in structural biology and chemical reactivity have enabled the design and development of potent and selective covalent PPI inhibitors. In this review, we will highlight the design and development of therapeutic agents targeting PPIs for cancer therapy.
Collapse
Affiliation(s)
- Sha-Sha Cheng
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, SAR, China
| | - Guan-Jun Yang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, SAR, China
| | - Wanhe Wang
- Department of Chemistry, Hong Kong Baptist University, Kowloon, 999077, Hong Kong, China.,Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Chung-Hang Leung
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, SAR, China.
| | - Dik-Lung Ma
- Department of Chemistry, Hong Kong Baptist University, Kowloon, 999077, Hong Kong, China.
| |
Collapse
|
37
|
Small-molecule covalent bond formation at tyrosine creates a binding site and inhibits activation of Ral GTPases. Proc Natl Acad Sci U S A 2020; 117:7131-7139. [PMID: 32179690 DOI: 10.1073/pnas.1913654117] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Ral (Ras-like) GTPases are directly activated by oncogenic Ras GTPases. Mutant K-Ras (G12C) has enabled the development of covalent K-Ras inhibitors currently in clinical trials. However, Ral, and the overwhelming majority of mutant oncogenic K-Ras, are devoid of a druggable pocket and lack an accessible cysteine for the development of a covalent inhibitor. Here, we report that covalent bond formation by an aryl sulfonyl fluoride electrophile at a tyrosine residue (Tyr-82) inhibits guanine exchange factor Rgl2-mediated nucleotide exchange of Ral GTPase. A high-resolution 1.18-Å X-ray cocrystal structure shows that the compound binds to a well-defined binding site in RalA as a result of a switch II loop conformational change. The structure, along with additional high-resolution crystal structures of several analogs in complex with RalA, confirm the importance of key hydrogen bond anchors between compound sulfone oxygen atoms and Ral backbone nitrogen atoms. Our discovery of a pocket with features found on known druggable sites and covalent modification of a bystander tyrosine residue present in Ral and Ras GTPases provide a strategy that could lead to therapeutic agent targeting oncogenic Ras mutants that are devoid of a cysteine nucleophile.
Collapse
|
38
|
Silva JRA, Cianni L, Araujo D, Batista PHJ, de Vita D, Rosini F, Leitão A, Lameira J, Montanari CA. Assessment of the Cruzain Cysteine Protease Reversible and Irreversible Covalent Inhibition Mechanism. J Chem Inf Model 2020; 60:1666-1677. [PMID: 32126170 DOI: 10.1021/acs.jcim.9b01138] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Reversible and irreversible covalent ligands are advanced cysteine protease inhibitors in the drug development pipeline. K777 is an irreversible inhibitor of cruzain, a necessary enzyme for the survival of the Trypanosoma cruzi (T. cruzi) parasite, the causative agent of Chagas disease. Despite their importance, irreversible covalent inhibitors are still often avoided due to the risk of adverse effects. Herein, we replaced the K777 vinyl sulfone group with a nitrile moiety to obtain a reversible covalent inhibitor (Neq0682) of cysteine protease. Then, we used advanced experimental and computational techniques to explore details of the inhibition mechanism of cruzain by reversible and irreversible inhibitors. The isothermal titration calorimetry (ITC) analysis shows that inhibition of cruzain by an irreversible inhibitor is thermodynamically more favorable than by a reversible one. The hybrid Quantum Mechanics/Molecular Mechanics (QM/MM) and Molecular Dynamics (MD) simulations were used to explore the mechanism of the reaction inhibition of cruzain by K777 and Neq0682. The calculated free energy profiles show that the Cys25 nucleophilic attack and His162 proton transfer occur in a single step for a reversible inhibitor and two steps for an irreversible covalent inhibitor. The hybrid QM/MM calculated free energies for the inhibition reaction correspond to -26.7 and -5.9 kcal mol-1 for K777 and Neq0682 at the MP2/MM level, respectively. These results indicate that the ΔG of the reaction is very negative for the process involving K777, consequently, the covalent adduct cannot revert to a noncovalent protein-ligand complex, and its binding tends to be irreversible. Overall, the present study provides insights into a covalent inhibition mechanism of cysteine proteases.
Collapse
Affiliation(s)
- José Rogério A Silva
- Laboratório de Planejamento e Desenvolvimento de Fármacos. Instituto de Ciências Exatas e Naturais, Universidade Federal do Pará, Rua Augusto Corrêa 01, CEP 66075-110, Belém, Pará, Brazil
| | - Lorenzo Cianni
- Medicinal Chemistry Group, Institute of Chemistry of São Carlos, University of São Paulo, Avenue Trabalhador Sancarlense 400, 23566-590, São Carlos, São Paulo, Brazil
| | - Deborah Araujo
- Medicinal Chemistry Group, Institute of Chemistry of São Carlos, University of São Paulo, Avenue Trabalhador Sancarlense 400, 23566-590, São Carlos, São Paulo, Brazil
| | - Pedro Henrique Jatai Batista
- Medicinal Chemistry Group, Institute of Chemistry of São Carlos, University of São Paulo, Avenue Trabalhador Sancarlense 400, 23566-590, São Carlos, São Paulo, Brazil
| | - Daniela de Vita
- Medicinal Chemistry Group, Institute of Chemistry of São Carlos, University of São Paulo, Avenue Trabalhador Sancarlense 400, 23566-590, São Carlos, São Paulo, Brazil
| | - Fabiana Rosini
- Medicinal Chemistry Group, Institute of Chemistry of São Carlos, University of São Paulo, Avenue Trabalhador Sancarlense 400, 23566-590, São Carlos, São Paulo, Brazil
| | - Andrei Leitão
- Medicinal Chemistry Group, Institute of Chemistry of São Carlos, University of São Paulo, Avenue Trabalhador Sancarlense 400, 23566-590, São Carlos, São Paulo, Brazil
| | - Jerônimo Lameira
- Laboratório de Planejamento e Desenvolvimento de Fármacos. Instituto de Ciências Exatas e Naturais, Universidade Federal do Pará, Rua Augusto Corrêa 01, CEP 66075-110, Belém, Pará, Brazil
| | - Carlos A Montanari
- Medicinal Chemistry Group, Institute of Chemistry of São Carlos, University of São Paulo, Avenue Trabalhador Sancarlense 400, 23566-590, São Carlos, São Paulo, Brazil
| |
Collapse
|
39
|
Potential Applications of NRF2 Modulators in Cancer Therapy. Antioxidants (Basel) 2020; 9:antiox9030193. [PMID: 32106613 PMCID: PMC7139512 DOI: 10.3390/antiox9030193] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 02/21/2020] [Accepted: 02/21/2020] [Indexed: 01/17/2023] Open
Abstract
The nuclear factor erythroid 2-related factor 2 (NRF2)-Kelch-like ECH-associated protein 1 (KEAP1) regulatory pathway plays an essential role in protecting cells and tissues from oxidative, electrophilic, and xenobiotic stress. By controlling the transactivation of over 500 cytoprotective genes, the NRF2 transcription factor has been implicated in the physiopathology of several human diseases, including cancer. In this respect, accumulating evidence indicates that NRF2 can act as a double-edged sword, being able to mediate tumor suppressive or pro-oncogenic functions, depending on the specific biological context of its activation. Thus, a better understanding of the mechanisms that control NRF2 functions and the most appropriate context of its activation is a prerequisite for the development of effective therapeutic strategies based on NRF2 modulation. In line of principle, the controlled activation of NRF2 might reduce the risk of cancer initiation and development in normal cells by scavenging reactive-oxygen species (ROS) and by preventing genomic instability through decreased DNA damage. In contrast however, already transformed cells with constitutive or prolonged activation of NRF2 signaling might represent a major clinical hurdle and exhibit an aggressive phenotype characterized by therapy resistance and unfavorable prognosis, requiring the use of NRF2 inhibitors. In this review, we will focus on the dual roles of the NRF2-KEAP1 pathway in cancer promotion and inhibition, describing the mechanisms of its activation and potential therapeutic strategies based on the use of context-specific modulation of NRF2.
Collapse
|
40
|
Poganik JR, Aye Y. Electrophile Signaling and Emerging Immuno- and Neuro-modulatory Electrophilic Pharmaceuticals. Front Aging Neurosci 2020; 12:1. [PMID: 32116644 PMCID: PMC7019031 DOI: 10.3389/fnagi.2020.00001] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 01/07/2020] [Indexed: 12/11/2022] Open
Abstract
With a lipid-rich environment and elevated oxygen consumption, the central nervous system (CNS) is subject to intricate regulation by lipid-derived electrophiles (LDEs). Investigations into oxidative damage and chronic LDE generation in neural disorders have spurred the development of tools that can detect and catalog the gamut of LDE-adducted proteins. Despite these advances, deconstructing the precise consequences of individual protein-specific LDE modifications remained largely impossible until recently. In this perspective, we first overview emerging toolsets that can decode electrophile-signaling events in a protein/context-specific manner, and how the accumulating mechanistic insights brought about by these tools have begun to offer new means to modulate pathways relevant to multiple sclerosis (MS). By surveying the latest data surrounding the blockbuster MS drug dimethyl fumarate that functions through LDE-signaling-like mechanisms, we further provide a vision for how chemical biology tools probing electrophile signaling may be leveraged toward novel interventions in CNS disease.
Collapse
Affiliation(s)
- Jesse R Poganik
- Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Yimon Aye
- Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
41
|
Wang X, Wang X, Pu X, Pu W, Wang Y, Liu Y, Gong Y, Jin X, Peng Y, Dai L. An Unbiased Immunoaffinity-Based Strategy for Profiling Covalent Drug Targets In Vivo. Anal Chem 2019; 91:15818-15825. [PMID: 31743002 DOI: 10.1021/acs.analchem.9b04118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Activity-based chemical proteomics approaches used for identifying cellular targets of drugs are mainly dependent on the availability of probes derived from drugs. However, all chemical probes are structurally different from the drugs themselves and cannot fully mimic the real actions of drugs in cells. Here we present a concise and unbiased immunoaffinity-based strategy for identifying covalent drug targets in vivo. By using the specific antibody, we not only confirm the well-known ibrutinib-binding target BTK, but also identify some previously undescribed strongly binding proteins, such as CKAP4 in human cell lines and TAP1 in mouse organs. The observed target profiles between species may partially explain why certain drug candidates are very effective in mice but not in humans. This approach avoids the chemical modification of drugs, eliminates the nonspecific bindings of chemical probes, and allows to unbiasedly decode the underlying mechanisms of action of covalent drugs.
Collapse
Affiliation(s)
- Xinyuan Wang
- Department of General Practice and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital , Sichuan University, and Collaborative Innovation Center of Biotherapy , Chengdu 610041 , China
| | - Xiuxuan Wang
- Department of General Practice and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital , Sichuan University, and Collaborative Innovation Center of Biotherapy , Chengdu 610041 , China
| | - Xinghua Pu
- Department of General Practice and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital , Sichuan University, and Collaborative Innovation Center of Biotherapy , Chengdu 610041 , China
| | - Wenchen Pu
- Department of General Practice and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital , Sichuan University, and Collaborative Innovation Center of Biotherapy , Chengdu 610041 , China
| | - Yuqi Wang
- Department of General Practice and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital , Sichuan University, and Collaborative Innovation Center of Biotherapy , Chengdu 610041 , China
| | - Yu Liu
- Department of General Practice and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital , Sichuan University, and Collaborative Innovation Center of Biotherapy , Chengdu 610041 , China
| | - Yanqiu Gong
- Department of General Practice and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital , Sichuan University, and Collaborative Innovation Center of Biotherapy , Chengdu 610041 , China
| | - Xiuxiu Jin
- Department of General Practice and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital , Sichuan University, and Collaborative Innovation Center of Biotherapy , Chengdu 610041 , China
| | - Yong Peng
- Department of General Practice and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital , Sichuan University, and Collaborative Innovation Center of Biotherapy , Chengdu 610041 , China
| | - Lunzhi Dai
- Department of General Practice and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital , Sichuan University, and Collaborative Innovation Center of Biotherapy , Chengdu 610041 , China
| |
Collapse
|
42
|
Kulkarni A, Soni I, Kelkar DS, Dharmaraja AT, Sankar RK, Beniwal G, Rajendran A, Tamhankar S, Chopra S, Kamat SS, Chakrapani H. Chemoproteomics of an Indole-Based Quinone Epoxide Identifies Druggable Vulnerabilities in Vancomycin-Resistant Staphylococcus aureus. J Med Chem 2019; 62:6785-6795. [PMID: 31241934 PMCID: PMC6660313 DOI: 10.1021/acs.jmedchem.9b00774] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
![]()
The alarming global
rise in fatalities from multidrug-resistant Staphylococcus
aureus (S. aureus)
infections has underscored a need to develop new therapies to address
this epidemic. Chemoproteomics is valuable in identifying targets
for new drugs in different human diseases including bacterial infections.
Targeting functional cysteines is particularly attractive, as they
serve critical catalytic functions that enable bacterial survival.
Here, we report an indole-based quinone epoxide scaffold with a unique
boat-like conformation that allows steric control in modulating thiol
reactivity. We extensively characterize a lead compound (4a), which potently inhibits clinically derived vancomycin-resistant S. aureus. Leveraging diverse chemoproteomic platforms,
we identify and biochemically validate important transcriptional factors
as potent targets of 4a. Interestingly, each identified
transcriptional factor has a conserved catalytic cysteine residue
that confers antibiotic tolerance to these bacteria. Thus, the chemical
tools and biological targets that we describe here prospect new therapeutic
paradigms in combatting S. aureus infections.
Collapse
Affiliation(s)
| | - Isha Soni
- Division of Microbiology , CSIR-Central Drug Research Institute , Sector 10, Janakipuram Extension, Sitapur Road , Lucknow 226021 , Uttar Pradesh , India
| | | | | | | | | | | | | | - Sidharth Chopra
- Division of Microbiology , CSIR-Central Drug Research Institute , Sector 10, Janakipuram Extension, Sitapur Road , Lucknow 226021 , Uttar Pradesh , India
| | | | | |
Collapse
|
43
|
Abstract
Targeted covalent modification is assuming consolidated importance in drug discovery. In this context, the electrophilic tuning of redox-dependent cell signaling is attracting major interest, as it opens prospect for treating numerous pathologic conditions. Herein, we discuss the rationale and the issues of electrophile-based approaches, focusing on the transcriptional Nrf2-Keap1 pathway as a test case. We also highlight relevant medicinal chemistry strategies researchers have devised to meet the ambitious goal, dwelling on the investigational and therapeutic potential of modulating redox-signaling networks through regulatory cysteine switches.
Collapse
|
44
|
Long MJ, Liu X, Aye Y. Genie in a bottle: controlled release helps tame natural polypharmacology? Curr Opin Chem Biol 2019; 51:48-56. [PMID: 30913473 DOI: 10.1016/j.cbpa.2019.02.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 02/02/2019] [Accepted: 02/12/2019] [Indexed: 02/06/2023]
Abstract
Ability to faithfully report drug-target interactions constitutes a major critical parameter in preclinical/clinical settings. Yet the assessment of target engagement remains challenging, particularly for promiscuous and/or polypharmacologic ligands. Drawing from our improved insights into native electrophile signaling and emerging technologies that profile and interrogate these non-enzyme-assisted signaling subsystems, we posit that 'trained' polypharmocologic covalent inhibitors can be designed. Accumulating evidence indicates that electrophile-modified states at fractional occupancy can alter cell fate. Thus, by understanding sensing preferences and ligandable regions favored by the natural electrophilic signals at individual protein-ligand resolution, we can better evaluate target engagement and develop a function-guided understanding of polypharmacology.
Collapse
Affiliation(s)
- Marcus Jc Long
- 47 Pudding Gate, Bishop Burton, Beverley East Riding of Yorkshire, HU17 8QH, UK
| | - Xuyu Liu
- École Polytechnique Fédérale de Lausanne, Institute of Chemical Sciences and Engineering, 1015, Lausanne, Switzerland
| | - Yimon Aye
- École Polytechnique Fédérale de Lausanne, Institute of Chemical Sciences and Engineering, 1015, Lausanne, Switzerland.
| |
Collapse
|
45
|
Long MJC, Liu X, Aye Y. Chemical Biology Gateways to Mapping Location, Association, and Pathway Responsivity. Front Chem 2019; 7:125. [PMID: 30949469 PMCID: PMC6437114 DOI: 10.3389/fchem.2019.00125] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 02/18/2019] [Indexed: 12/14/2022] Open
Abstract
Here we discuss, how by applying chemical concepts to biological problems, methods have been developed to map spatiotemporal regulation of proteins and small-molecule modulation of proteome signaling responses. We outline why chemical-biology platforms are ideal for such purposes. We further discuss strengths and weaknesses of chemical-biology protocols, contrasting them against classical genetic and biochemical approaches. We make these evaluations based on three parameters: occupancy; functional information; and spatial restriction. We demonstrate how the specific choice of chemical reagent and experimental set-up unite to resolve biological problems. Potential improvements/extensions as well as specific controls that in our opinion are often overlooked or employed incorrectly are also considered. Finally, we discuss some of the latest emerging methods to illuminate how chemical-biology innovations provide a gateway toward information hitherto inaccessible by conventional genetic/biochemical means. Finally, we also caution against solely relying on chemical-biology strategies and urge the field to undertake orthogonal validations to ensure robustness of results.
Collapse
Affiliation(s)
| | - Xuyu Liu
- École Polytechnique Fédérale de Lausanne, Institute of Chemical Sciences and Engineering, Lausanne, Switzerland
| | - Yimon Aye
- École Polytechnique Fédérale de Lausanne, Institute of Chemical Sciences and Engineering, Lausanne, Switzerland
| |
Collapse
|
46
|
Simaan H, Lev S, Horwitz BA. Oxidant-Sensing Pathways in the Responses of Fungal Pathogens to Chemical Stress Signals. Front Microbiol 2019; 10:567. [PMID: 30941117 PMCID: PMC6433817 DOI: 10.3389/fmicb.2019.00567] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 03/05/2019] [Indexed: 12/04/2022] Open
Abstract
Host defenses expose fungal pathogens to oxidants and antimicrobial chemicals. The fungal cell employs conserved eukaryotic signaling pathways and dedicated transcription factors to program its response to these stresses. The oxidant-sensitive transcription factor of yeast, YAP1, and its orthologs in filamentous fungi, are central to tolerance to oxidative stress. The C-terminal domain of YAP1 contains cysteine residues that, under oxidizing conditions, form an intramolecular disulfide bridge locking the molecule in a conformation where the nuclear export sequence is masked. YAP1 accumulates in the nucleus, promoting transcription of genes that provide the cell with the ability to counteract oxidative stress. Chemicals including xenobiotics and plant signals can also promote YAP1 nuclearization in yeast and filamentous fungi. This could happen via direct or indirect oxidative stress, or by a different biochemical pathway. Plant phenolics are known antioxidants, yet they have been shown to elicit cellular responses that would usually be triggered to counter oxidant stress. Here we will discuss the evidence that YAP1 and MAPK pathways respond to phenolic compounds. Following this and other examples, we explore here how oxidative-stress sensing networks of fungi might have evolved to detect chemical stressors. Furthermore, we draw functional parallels between fungal YAP1 and mammalian Keap1-Nrf2 signaling systems.
Collapse
Affiliation(s)
- Hiba Simaan
- Faculty of Biology, Technion - Israel Institute of Technology, Haifa, Israel
| | - Sophie Lev
- Centre for Infectious Diseases and Microbiology, The Westmead Institute for Medical Research, Westmead, NSW, Australia
| | - Benjamin A Horwitz
- Faculty of Biology, Technion - Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
47
|
Poganik JR, Long MJC, Aye Y. Interrogating Precision Electrophile Signaling. Trends Biochem Sci 2019; 44:380-381. [PMID: 30765181 DOI: 10.1016/j.tibs.2019.01.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 01/15/2019] [Indexed: 12/17/2022]
Abstract
Understanding the targets and signaling roles of reactive electrophilic species (RES) at a specific cellular space and time has long been hampered by the reliance of the field on the bulk administration of excess RES from outside of cells and/or animals. Uncontrolled bolus methods provide limited understanding of target engagement for these individual nonenzymatic RES-modification events. REX technologies [targetable reactive electrophiles and oxidants (T-REX) and its genome-wide variant (G-REX)] were developed as a gateway to address these limitations. These protocols offer a new ability to both profile kinetically privileged sensors (KPSs) of RES at a systems level (G-REX™ profiling) and monitor signaling responses at the sensor protein-of-interest (POI)-specific level (T-REX™ delivery) with high spatiotemporal resolution. REX technologies are compatible with several model systems and are built on a HaloTag-targetable small-molecule photocaged precursor to a native RES.
Collapse
Affiliation(s)
- Jesse R Poganik
- École Polytechnique Fédérale de Lausanne, Institute of Chemical Sciences and Engineering, 1015 Lausanne, Switzerland; Cornell University, Ithaca, NY 14850, USA
| | | | - Yimon Aye
- École Polytechnique Fédérale de Lausanne, Institute of Chemical Sciences and Engineering, 1015 Lausanne, Switzerland.
| |
Collapse
|
48
|
Proteomics and Beyond: Cell Decision-Making Shaped by Reactive Electrophiles. Trends Biochem Sci 2018; 44:75-89. [PMID: 30327250 DOI: 10.1016/j.tibs.2018.09.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 08/21/2018] [Accepted: 09/19/2018] [Indexed: 12/11/2022]
Abstract
Revolutionary proteomic strategies have enabled rapid profiling of the cellular targets of electrophilic small molecules. However, precise means to directly interrogate how these individual electrophilic modifications at low occupancy functionally reshape signaling networks have until recently been largely limited. We highlight here new methods that transcend proteomic platforms to forge a quantitative link between protein target-selective engagement and downstream signaling. We focus on recent progress in the study of non-enzyme-assisted signaling mechanisms and crosstalk choreographed by native reactive electrophilic species (RES). Using this as a model, we offer a long-term vision of how these toolsets together with fundamental biochemical knowledge of precision electrophile signaling may be harnessed to assist covalent ligand-target matching and ultimately amend disease-specific signaling dysfunction.
Collapse
|
49
|
Abstract
The concept of cell signaling in the context of nonenzyme-assisted protein modifications by reactive electrophilic and oxidative species, broadly known as redox signaling, is a uniquely complex topic that has been approached from numerous different and multidisciplinary angles. Our Review reflects on five aspects critical for understanding how nature harnesses these noncanonical post-translational modifications to coordinate distinct cellular activities: (1) specific players and their generation, (2) physicochemical properties, (3) mechanisms of action, (4) methods of interrogation, and (5) functional roles in health and disease. Emphasis is primarily placed on the latest progress in the field, but several aspects of classical work likely forgotten/lost are also recollected. For researchers with interests in getting into the field, our Review is anticipated to function as a primer. For the expert, we aim to stimulate thought and discussion about fundamentals of redox signaling mechanisms and nuances of specificity/selectivity and timing in this sophisticated yet fascinating arena at the crossroads of chemistry and biology.
Collapse
Affiliation(s)
- Saba Parvez
- Department of Pharmacology and Toxicology, College of
Pharmacy, University of Utah, Salt Lake City, Utah, 84112, USA
- Department of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York, 14853, USA
| | - Marcus J. C. Long
- Department of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York, 14853, USA
| | - Jesse R. Poganik
- Ecole Polytechnique Fédérale de Lausanne,
Institute of Chemical Sciences and Engineering, 1015, Lausanne, Switzerland
- Department of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York, 14853, USA
| | - Yimon Aye
- Ecole Polytechnique Fédérale de Lausanne,
Institute of Chemical Sciences and Engineering, 1015, Lausanne, Switzerland
- Department of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York, 14853, USA
- Department of Biochemistry, Weill Cornell Medicine, New
York, New York, 10065, USA
| |
Collapse
|
50
|
Li T, Maltais R, Poirier D, Lin SX. Combined Biophysical Chemistry Reveals a New Covalent Inhibitor with a Low-Reactivity Alkyl Halide. J Phys Chem Lett 2018; 9:5275-5280. [PMID: 30148957 DOI: 10.1021/acs.jpclett.8b02225] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
17β-Hydroxysteroid dehydrogenase type 1 (17β-HSD1) plays a pivotal role in the progression of estrogen-related diseases because of its involvement in the biosynthesis of estradiol (E2), constituting a valuable therapeutic target for endocrine treatment. In the present study, we successfully cocrystallized the enzyme with the reversible inhibitor 2-methoxy-16β-( m-carbamoylbenzyl)-E2 (2-MeO-CC-156) as well as the enzyme with the irreversible inhibitor 3-(2-bromoethyl)-16β-( m-carbamoylbenzyl)-17β-hydroxy-1,3,5(10)-estratriene (PBRM). The structures of ternary complexes of 17β-HSD1-2-MeO-CC-156-NADP+ and 17β-HSD1-PBRM-NADP+ comparatively show the formation of a covalent bond between His221 and the bromoethyl side chain of the inhibitor in the PBRM structure. A dynamic process including beneficial molecular interactions that favor the specific binding of a low-reactivity inhibitor and subsequent N-alkylation event through the participation of His221 in the enzyme catalytic site clearly demonstrates the covalent bond formation. This finding opens the door to a new design of alkyl halide-based specific covalent inhibitors as potential therapeutic agents for different enzymes, contributing to the development of highly efficient inhibitors.
Collapse
Affiliation(s)
- Tang Li
- CHU de Québec - Research Center , 2705 Boulevard Laurier , Québec , QC G1V 4G2 , Canada
- Faculty of Medicine , Université Laval , Québec , QC G1V 0A6 , Canada
| | - René Maltais
- CHU de Québec - Research Center , 2705 Boulevard Laurier , Québec , QC G1V 4G2 , Canada
| | - Donald Poirier
- CHU de Québec - Research Center , 2705 Boulevard Laurier , Québec , QC G1V 4G2 , Canada
- Faculty of Medicine , Université Laval , Québec , QC G1V 0A6 , Canada
| | - Sheng-Xiang Lin
- CHU de Québec - Research Center , 2705 Boulevard Laurier , Québec , QC G1V 4G2 , Canada
- Faculty of Medicine , Université Laval , Québec , QC G1V 0A6 , Canada
| |
Collapse
|