1
|
Song J. Applications of the Cellular Thermal Shift Assay to Drug Discovery in Natural Products: A Review. Int J Mol Sci 2025; 26:3940. [PMID: 40362180 PMCID: PMC12072176 DOI: 10.3390/ijms26093940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/21/2025] [Accepted: 04/21/2025] [Indexed: 05/15/2025] Open
Abstract
Natural products play a crucial role in drug discovery because of their structural diversity and biological activity. However, identifying their molecular targets remains a challenge. Traditional target identification approaches such as affinity-based protein profiling and activity-based protein profiling are limited by the need for chemical modification or reactive groups in natural products. The emergence of label-free techniques offers a powerful alternative for studying drug-target engagement in a physiological context. In particular, the cellular thermal shift assay (CETSA) exploits ligand-induced protein stabilization-a phenomenon where ligand binding enhances a protein's thermal stability by reducing conformational flexibility-to assess drug binding without requiring chemical modifications. CETSA's integration with advanced mass spectrometry and high-throughput platforms has dramatically expanded proteome coverage and sensitivity, enabling the simultaneous quantification of thousands of proteins and the identification of low-abundance targets in native cellular environments. This review highlights the application of key CETSA-based methods to target identification in natural products including Western blot-based CETSA, isothermal dose-response CETSA, mass spectrometry-based CETSA, and high-throughput CETSA. Case studies are presented that demonstrate their effectiveness in uncovering the mechanisms of action of different drugs. The current limitations of CETSA-based strategies are also explored, and future improvements to optimize their potential for drug discovery are discussed. Integrating CETSA with complementary approaches can enhance the target identification accuracy and efficiency for natural products and ultimately advance development of therapeutic applications.
Collapse
Affiliation(s)
- Jayoung Song
- Department of Pharmacology, College of Medicine, Dankook University, Cheonan 31116, Republic of Korea
| |
Collapse
|
2
|
Cacace E, Tietgen M, Steinhauer M, Mateus A, Schultze TG, Eckermann M, Galardini M, Varik V, Koumoutsi A, Parzeller JJ, Corona F, Orakov A, Knopp M, Brauer-Nikonow A, Bork P, Romao CV, Zimmermann M, Cloetens P, Savitski MM, Typas A, Göttig S. Uncovering nitroxoline activity spectrum, mode of action and resistance across Gram-negative bacteria. Nat Commun 2025; 16:3783. [PMID: 40263263 PMCID: PMC12015411 DOI: 10.1038/s41467-025-58730-5] [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: 06/17/2024] [Accepted: 03/27/2025] [Indexed: 04/24/2025] Open
Abstract
Nitroxoline is a bacteriostatic quinoline antibiotic, known to form complexes with metals. Its clinical indications are limited to uncomplicated urinary tract infections, with a susceptibility breakpoint only available for Escherichia coli. Here, we test > 1000 clinical isolates and demonstrate a much broader activity spectrum and species-specific bactericidal activity, including Gram-negative bacteria for which therapeutic options are limited due to multidrug resistance. By combining genetic and proteomic approaches with direct measurement of intracellular metals, we show that nitroxoline acts as a metallophore, inducing copper and zinc intoxication in bacterial cells. The compound displays additional effects on bacterial physiology, including alteration of outer membrane integrity, which underpins nitroxoline's synergies with large-scaffold antibiotics and resensitization of colistin-resistant Enterobacteriaceae in vitro and in vivo. Furthermore, we identify conserved resistance mechanisms across bacterial species, often leading to nitroxoline efflux.
Collapse
Affiliation(s)
- Elisabetta Cacace
- Goethe University Frankfurt, University Hospital, Institute for Medical Microbiology and Infection Control, Frankfurt, Germany
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany
| | - Manuela Tietgen
- Goethe University Frankfurt, University Hospital, Institute for Medical Microbiology and Infection Control, Frankfurt, Germany
| | - Meike Steinhauer
- Goethe University Frankfurt, University Hospital, Institute for Medical Microbiology and Infection Control, Frankfurt, Germany
| | - André Mateus
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany
| | - Tilman G Schultze
- Goethe University Frankfurt, University Hospital, Institute for Medical Microbiology and Infection Control, Frankfurt, Germany
| | - Marina Eckermann
- European Synchrotron Radiation Facility (ESRF), Grenoble, France
- Institute of Applied Physics, University of Bern, Bern, Switzerland
| | - Marco Galardini
- Institute for Molecular Bacteriology, TWINCORE Centre for Experimental and Clinical Infection Research, a joint venture between the Hannover Medical School (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School (MHH), Hannover, Germany
| | - Vallo Varik
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany
| | - Alexandra Koumoutsi
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany
| | - Jordan J Parzeller
- Goethe University Frankfurt, University Hospital, Institute for Medical Microbiology and Infection Control, Frankfurt, Germany
| | - Federico Corona
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany
| | - Askarbek Orakov
- European Molecular Biology Laboratory, Structural and Computational Biology Unit, Heidelberg, Germany
| | - Michael Knopp
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany
| | - Amber Brauer-Nikonow
- European Molecular Biology Laboratory, Structural and Computational Biology Unit, Heidelberg, Germany
| | - Peer Bork
- European Molecular Biology Laboratory, Structural and Computational Biology Unit, Heidelberg, Germany
| | - Celia V Romao
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Michael Zimmermann
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School (MHH), Hannover, Germany
| | - Peter Cloetens
- European Synchrotron Radiation Facility (ESRF), Grenoble, France
| | - Mikhail M Savitski
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany
| | - Athanasios Typas
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany
| | - Stephan Göttig
- Goethe University Frankfurt, University Hospital, Institute for Medical Microbiology and Infection Control, Frankfurt, Germany.
| |
Collapse
|
3
|
Deschner F, Mostert D, Daniel JM, Voltz A, Schneider DC, Khangholi N, Bartel J, Pessanha de Carvalho L, Brauer M, Gorelik TE, Kleeberg C, Risch T, Haeckl FPJ, Herraiz Benítez L, Andreas A, Kany AM, Jézéquel G, Hofer W, Müsken M, Held J, Bischoff M, Seemann R, Brötz-Oesterhelt H, Schneider T, Sieber S, Müller R, Herrmann J. Natural products chlorotonils exert a complex antibacterial mechanism and address multiple targets. Cell Chem Biol 2025; 32:586-602.e15. [PMID: 40203831 DOI: 10.1016/j.chembiol.2025.03.005] [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: 08/21/2024] [Revised: 12/12/2024] [Accepted: 03/18/2025] [Indexed: 04/11/2025]
Abstract
Antimicrobial resistance is a threat to human health rendering current first-line antibiotics ineffective. New agents overcoming resistance mechanisms are urgently needed to guarantee successful treatment of human disease in the future. Chlorotonils, a natural product class with yet unknown mode of action, were shown to have broad-spectrum activity against multi-resistant Gram-positive bacteria and the malaria parasite Plasmodium falciparum, with promising activity and safety in murine infection models. Here, we report that chlorotonils can target the cell membrane, cell wall, and protein biosynthesis. They can be characterized by a rapid onset of action via interference with ion homeostasis leading to membrane depolarization, however, without inducing severe barrier failure or cellular lysis. Further characterization confirmed binding of chlorotonils to bacterial membrane lipids eventually leading to uncontrolled potassium transport. Additionally, we identified functional inhibition of the peptidoglycan biosynthesis protein YbjG and methionine aminopeptidase MetAP as secondary targets of chlorotonils.
Collapse
Affiliation(s)
- Felix Deschner
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany; German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany
| | - Dietrich Mostert
- Center for Functional Protein Assemblies, TUM School of Natural Sciences, Technical University of Munich, 85748 Garching, Germany
| | - Jan-Martin Daniel
- German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany; Institute for Pharmaceutical Microbiology, University of Bonn, University Hospital Bonn, 53127 Bonn, Germany
| | - Alexander Voltz
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany; German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany
| | - Dana Carina Schneider
- German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany; Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Department of Microbial Bioactive Compounds, University of Tübingen, 72074 Tübingen, Germany
| | - Navid Khangholi
- Experimental Physics and Center for Biophysics, Saarland University, 66123 Saarbrücken, Germany
| | - Jürgen Bartel
- Department of Microbial Proteomics, Institute of Microbiology, University of Greifswald, 17489 Greifswald, Germany
| | | | - Madita Brauer
- Department of Microbial Physiology and Molecular Biology, Institute of Microbiology, University of Greifswald, 17489 Greifswald, Germany
| | - Tatiana E Gorelik
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; Institute of Inorganic and Analytical Chemistry, Goethe University Frankfurt, 60438 Frankfurt am Main, Germany; Ernst Ruska-Centre for Microscopy and Spectroscopy with Electrons, Forschungszentrum Jülich GmbH, Jülich 52428, Germany
| | - Christian Kleeberg
- Institute for Inorganic and Analytical Chemistry, Technical University of Braunschweig, 38106 Braunschweig, Germany
| | - Timo Risch
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany; German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany
| | - F P Jake Haeckl
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany
| | - Laura Herraiz Benítez
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany
| | - Anastasia Andreas
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany
| | - Andreas Martin Kany
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany; German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany
| | - Gwenaëlle Jézéquel
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany
| | - Walter Hofer
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany
| | - Mathias Müsken
- Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany
| | - Jana Held
- German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany; Institute of Tropical Medicine, Eberhard Karls University Tübingen, 72074 Tübingen, Germany
| | - Markus Bischoff
- Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; Institute for Medical Microbiology and Hygiene, Saarland University, 66421 Homburg, Germany
| | - Ralf Seemann
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Department of Microbial Bioactive Compounds, University of Tübingen, 72074 Tübingen, Germany
| | - Heike Brötz-Oesterhelt
- German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany; Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Department of Microbial Bioactive Compounds, University of Tübingen, 72074 Tübingen, Germany; Cluster or Excellence "Controlling Microbes to Fight Infections", Tübingen, Germany
| | - Tanja Schneider
- German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany; Institute for Pharmaceutical Microbiology, University of Bonn, University Hospital Bonn, 53127 Bonn, Germany
| | - Stephan Sieber
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Center for Functional Protein Assemblies, TUM School of Natural Sciences, Technical University of Munich, 85748 Garching, Germany
| | - Rolf Müller
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany; German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany
| | - Jennifer Herrmann
- Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany; Helmholtz Centre for Infection Research (HZI), 38124 Braunschweig, Germany; Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany; German Centre for Infection Research (DZIF), partner sites: Bonn-Cologne, Hannover-Braunschweig, and Tübingen, 38124 Braunschweig, Germany.
| |
Collapse
|
4
|
Hoffmanns L, Svedberg D, Mateus A. Protein O-glycosylation in the Bacteroidota phylum. FEBS Open Bio 2025. [PMID: 40231347 DOI: 10.1002/2211-5463.70041] [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: 02/21/2025] [Revised: 03/26/2025] [Accepted: 04/08/2025] [Indexed: 04/16/2025] Open
Abstract
Glycans play crucial roles in bacteria, such as providing structural integrity or enabling interactions with the ecosystem. They can be linked to lipids, peptides, or proteins. In proteins, they modify either asparagine (N-glycosylation) or serine or threonine (O-glycosylation). Species of the Bacteroidota phylum, a major component of the human microbiome and marine and soil ecosystems, have a unique type of O-glycosylation that modifies multiple noncytoplasmic proteins containing a specific amino acid sequence. Only a small number of species have currently been characterized, but within one species, generally all proteins are modified with the same glycan structure. Most species share a common inner part but differ in the sugar composition and branching of the outer part of their glycan. This suggests that the biosynthesis of the glycan occurs in two separate steps. Both the inner core and the outer glycan are likely assembled from nucleotide-activated monosaccharides on undecaprenyl phosphate on the cytoplasmic side of the inner membrane, prior to being flipped to the periplasm and transferred to the protein. A genomic locus responsible for the biosynthesis of the outer glycan has been identified, containing some conserved genes across species. Despite substantial progress in the characterization of this O-glycosylation system, its function, the overall diversity of glycan structures across the phylum, and the complete biosynthetic pathway remain mostly unknown. Due to the importance of this group of species for the human gut microbiome, elucidating these aspects can open up strategies to modulate the composition of the microbiome community toward a healthy state.
Collapse
Affiliation(s)
| | | | - André Mateus
- Department of Chemistry, Umeå University, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
- Umeå Center for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| |
Collapse
|
5
|
Lavallée-Adam M, Pelletier A, Diedrich JK, Pinto AFM, Martínez-Bartolomé S, Petrascheck M, Moresco JJ, Yates JR. TargetSeeker-MS: A Bayesian Inference Approach for Drug-Target Discovery Using Protein Fractionation Coupled to Mass Spectrometry. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2025; 36:664-679. [PMID: 40067882 PMCID: PMC11968059 DOI: 10.1021/jasms.4c00269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
To understand the mechanism of action of a drug and assess its clinical usefulness and viability, it is imperative that its affinity for its putative targets is determined. When coupled to mass spectrometry (MS), energetics-based protein separation (EBPS) techniques, such as a thermal shift assay, have shown great potential to identify the targets of a drug on a proteome scale. Nevertheless, the computational analyses assessing the confidence of drug-target predictions made by these methods have remained tightly tied to the protocol under which the data were produced. To identify drug targets in data sets produced using different EBPS-MS techniques, we have developed a novel flexible Bayesian inference approach named TargetSeeker-MS. We showed that TargetSeeker-MS identifies known and novel drug targets in Caenorhabditis elegans and HEK 293 samples treated with the fungicide benomyl. We also demonstrated that TargetSeeker-MS' drug-target identifications are reproducible in C. elegans samples that were processed using two different EBPS techniques (thermal shift assay and a differential precipitation of proteins, named DiffPOP). In addition, we validated a novel benomyl target by measuring its altered enzymatic activity upon drug treatment in vitro. TargetSeeker-MS, which is available as a web server (https://targetseeker.scripps.edu/), allows for the rapid, versatile, and confident identification of targets of a drug on a proteome scale, thereby providing a better understanding of its mechanisms and facilitating the evaluation of its clinical viability.
Collapse
Affiliation(s)
- Mathieu Lavallée-Adam
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd., La Jolla, California, 92037, USA
- (Current affiliation) Department of Biochemistry, Microbiology and Immunology and Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Alexander Pelletier
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd., La Jolla, California, 92037, USA
| | - Jolene K. Diedrich
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd., La Jolla, California, 92037, USA
- The Mass Spectrometry Core for Proteomics and Metabolomics, The Salk Institute for Biological Studies, 10010 N Torrey Pines Rd., La Jolla, California, 92037, USA
| | - Antonio F. M. Pinto
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd., La Jolla, California, 92037, USA
- The Mass Spectrometry Core for Proteomics and Metabolomics, The Salk Institute for Biological Studies, 10010 N Torrey Pines Rd., La Jolla, California, 92037, USA
| | - Salvador Martínez-Bartolomé
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd., La Jolla, California, 92037, USA
| | - Michael Petrascheck
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd., La Jolla, California, 92037, USA
| | - James J. Moresco
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd., La Jolla, California, 92037, USA
- The Mass Spectrometry Core for Proteomics and Metabolomics, The Salk Institute for Biological Studies, 10010 N Torrey Pines Rd., La Jolla, California, 92037, USA
- (Current affiliation) Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - John R. Yates
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd., La Jolla, California, 92037, USA
- The Mass Spectrometry Core for Proteomics and Metabolomics, The Salk Institute for Biological Studies, 10010 N Torrey Pines Rd., La Jolla, California, 92037, USA
| |
Collapse
|
6
|
Matilla MA, Gavira JA, Monteagudo-Cascales E, Zhulin IB, Krell T. Structural and functional diversity of sensor domains in bacterial transmembrane receptors. Trends Microbiol 2025:S0966-842X(25)00076-9. [PMID: 40121131 DOI: 10.1016/j.tim.2025.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/25/2025]
Abstract
The ability of bacteria to adapt to changing environmental conditions largely depends on transmembrane receptors that sense signal molecules and generate responses such as chemotaxis, changes in gene expression, or alterations in second-messenger levels. Although these receptors differ significantly in function, they share a common mode of activation that involves signal molecule interaction with sensor domains. A major challenge in microbiology lies in the limited knowledge of ligands that stimulate receptors. Here, we review recent advances in this field, including the occurrence of multi-modular sensor domains, the identification of co-component signal transduction systems, evidence for sensor domain evolution from transporters, and the use of binding pocket sequence motifs to identify sensor domain ligands.
Collapse
Affiliation(s)
- Miguel A Matilla
- Department of Biotechnology and Environmental Protection, Estación Experimental del Zaidín, Consejo Superior de Investigaciones Científicas, Prof. Albareda 1, Granada, 18008, Spain
| | - José A Gavira
- Laboratory of Crystallographic Studies, CSIC-UGR, Avenida de las Palmeras 4, Armilla, 18100, Spain
| | - Elizabet Monteagudo-Cascales
- Department of Biotechnology and Environmental Protection, Estación Experimental del Zaidín, Consejo Superior de Investigaciones Científicas, Prof. Albareda 1, Granada, 18008, Spain
| | - Igor B Zhulin
- Department of Microbiology and Translational Data Analytics Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Tino Krell
- Department of Biotechnology and Environmental Protection, Estación Experimental del Zaidín, Consejo Superior de Investigaciones Científicas, Prof. Albareda 1, Granada, 18008, Spain.
| |
Collapse
|
7
|
Ramos M, Jernigan RL, Kilinc M. ESMStabP: A Regression Model for Protein Thermostability Prediction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.18.638450. [PMID: 40027616 PMCID: PMC11870573 DOI: 10.1101/2025.02.18.638450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Accurately predicting protein thermostability is crucial for numerous applications in biotechnology, pharmaceuticals, and food science. Experimental methods for determining protein melting temperatures are often time-consuming and costly, driving the need for efficient computational alternatives. In this paper, we introduce ESMStabP, an enhanced regression model for predicting protein thermostability. To improve model performance and generalizability, we assembled a significantly larger dataset by combining and cleaning datasets previously utilized in other thermostability models. Building on DeepStabP, ESMStabP incorporates significant improvements, using embeddings from the ESM2 protein language model and thermophilic classifications. The predictions from ESMStabP outperform DeepStabP and other existing predictors, achieving an R2 of 0.95 and a Pearson correlation coefficient (PCC) of 0.97. Despite these improvements, challenges such as dataset availability. This work underscores the critical role of specific layer identification for model development and outlines potential directions for future advancements in protein stability predictions.
Collapse
Affiliation(s)
- Marcus Ramos
- Iowa State University, Ames, IA 50011, United States
| | | | - Mesih Kilinc
- Iowa State University, Ames, IA 50011, United States
| |
Collapse
|
8
|
Khan A, Liu Y, Gad M, Kenny TC, Birsoy K. Solute carriers: The gatekeepers of metabolism. Cell 2025; 188:869-884. [PMID: 39983672 PMCID: PMC11875512 DOI: 10.1016/j.cell.2025.01.015] [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: 08/01/2024] [Revised: 10/23/2024] [Accepted: 01/07/2025] [Indexed: 02/23/2025]
Abstract
Solute carrier (SLC) proteins play critical roles in maintaining cellular and organismal homeostasis by transporting small molecules and ions. Despite a growing body of research over the past decade, physiological substrates and functions of many SLCs remain elusive. This perspective outlines key challenges in studying SLC biology and proposes an evidence-based framework for defining SLC substrates. To accelerate the deorphanization process, we explore systematic technologies, including human genetics, biochemistry, and computational and structural approaches. Finally, we suggest directions to better understand SLC functions beyond substrate identification in physiology and disease.
Collapse
Affiliation(s)
- Artem Khan
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, NY, USA
| | - Yuyang Liu
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, NY, USA
| | - Mark Gad
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, NY, USA; Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Timothy C Kenny
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, NY, USA
| | - Kıvanç Birsoy
- Laboratory of Metabolic Regulation and Genetics, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
9
|
Doron-Mandel E, Bokor BJ, Ma Y, Street LA, Tang LC, Abdou AA, Shah NH, Rosenberger G, Jovanovic M. SEC-MX: an approach to systematically study the interplay between protein assembly states and phosphorylation. Nat Commun 2025; 16:1176. [PMID: 39885126 PMCID: PMC11782603 DOI: 10.1038/s41467-025-56303-0] [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: 05/16/2024] [Accepted: 01/13/2025] [Indexed: 02/01/2025] Open
Abstract
A protein's molecular interactions and post-translational modifications (PTMs), such as phosphorylation, can be co-dependent and reciprocally co-regulate each other. Although this interplay is central for many biological processes, a systematic method to simultaneously study assembly states and PTMs from the same sample is critically missing. Here, we introduce SEC-MX (Size Exclusion Chromatography fractions MultipleXed), a global quantitative method combining Size Exclusion Chromatography and PTM-enrichment for simultaneous characterization of PTMs and assembly states. SEC-MX enhances throughput, allows phosphopeptide enrichment, and facilitates quantitative differential comparisons between biological conditions. Conducting SEC-MX on HEK293 and HCT116 cells, we generate a proof-of-concept dataset, mapping thousands of phosphopeptides and their assembly states. Our analysis reveals intricate relationships between phosphorylation events and assembly states and generates testable hypotheses for follow-up studies. Overall, we establish SEC-MX as a valuable tool for exploring protein functions and regulation beyond abundance changes.
Collapse
Affiliation(s)
- Ella Doron-Mandel
- Department of Biological Sciences, Columbia University, New York, NY, USA.
| | - Benjamin J Bokor
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Yanzhe Ma
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Lena A Street
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Lauren C Tang
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Ahmed A Abdou
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Neel H Shah
- Department of Chemistry, Columbia University, New York, NY, USA
| | | | - Marko Jovanovic
- Department of Biological Sciences, Columbia University, New York, NY, USA.
| |
Collapse
|
10
|
Khan MM, Galea G, Jung J, Zukowska J, Lauer D, Tuechler N, Halavatyi A, Tischer C, Haberkant P, Stein F, Jung F, Landry JJM, Khan AM, Oorschot V, Becher I, Neumann B, Muley T, Winter H, Duerr J, Mall MA, Grassi A, de la Cueva E, Benes V, Gote-Schniering J, Savitski M, Pepperkok R. Dextromethorphan inhibits collagen and collagen-like cargo secretion to ameliorate lung fibrosis. Sci Transl Med 2024; 16:eadj3087. [PMID: 39693409 DOI: 10.1126/scitranslmed.adj3087] [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: 06/19/2023] [Revised: 04/25/2024] [Accepted: 11/01/2024] [Indexed: 12/20/2024]
Abstract
Excessive deposition of fibrillar collagen in the interstitial extracellular matrix (ECM) of human lung tissue causes fibrosis, which can ultimately lead to organ failure. Despite our understanding of the molecular mechanisms underlying the disease, no cure for pulmonary fibrosis has yet been found. We screened a drug library and found that dextromethorphan (DXM), a cough expectorant, reduced the amount of excess fibrillar collagen deposited in the ECM in cultured primary human lung fibroblasts, a bleomycin mouse model, and a cultured human precision-cut lung slice model of lung fibrosis. The reduced extracellular fibrillar collagen upon DXM treatment was due to reversible trafficking inhibition of collagen type I (COL1) in the endoplasmic reticulum (ER) in TANGO1- and HSP47-positive structures. Mass spectrometric analysis showed that DXM promoted hyperhydroxylation of proline and lysine residues on various collagens (COL1, COL3, COL4, COL5, COL7, and COL12) and latent transforming growth factor-β-binding protein (LTBP1 and LTBP2) peptides, coinciding with their secretion block. Additionally, proteome profiling of DXM-treated cells showed increased thermal stability of prolyl-hydroxylases P3H2, P3H3, P3H4, P4HA1, and P4HA2, suggesting a change in their activity. Transcriptome analysis of profibrotic stimulated primary human lung fibroblasts and human ex vivo lung slices after DXM treatment showed activation of an antifibrotic program through regulation of multiple pathways, including the MMP-ADAMTS axis, WNT signaling, and fibroblast-to-myofibroblast differentiation. Together, these data obtained from in vitro, in vivo, and ex vivo models of lung fibrogenesis show that DXM has the potential to limit fibrosis through inhibition of COL1 membrane trafficking in the ER.
Collapse
Affiliation(s)
- Muzamil M Khan
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), 69120 Heidelberg, Germany
| | - George Galea
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Juan Jung
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Joanna Zukowska
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - David Lauer
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
- Department of Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Nadine Tuechler
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory and Heidelberg University, 69117 Heidelberg, Germany
- Institute for Computational Biomedicine (ICB), Faculty of Medicine, Heidelberg University and Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Aliaksandr Halavatyi
- Advanced Light Microscopy Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Christian Tischer
- Advanced Light Microscopy Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Per Haberkant
- Proteomics Core Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Frank Stein
- Proteomics Core Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Ferris Jung
- Genomics Core Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Jonathan J M Landry
- Genomics Core Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Arif M Khan
- Centre for Bioimage Analysis, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Viola Oorschot
- Electron Microscopy Core Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Isabelle Becher
- Genome Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Beate Neumann
- Advanced Light Microscopy Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Thomas Muley
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), 69120 Heidelberg, Germany
- Translational Research Unit/Lung Biobank, Thoraxklinik, University Hospital Heidelberg, 69117 Heidelberg, Germany
| | - Hauke Winter
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), 69120 Heidelberg, Germany
- Translational Research Unit/Lung Biobank, Thoraxklinik, University Hospital Heidelberg, 69117 Heidelberg, Germany
| | - Julia Duerr
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin, 13353 Berlin, Germany
- German Center for Lung Research (DZL), Associated Partner Site, 13353 Berlin, Germany
| | - Marcus A Mall
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin, 13353 Berlin, Germany
- German Center for Lung Research (DZL), Associated Partner Site, 13353 Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, 10178 Berlin, Germany
| | - Alessandro Grassi
- Laboratory Animal Resources, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Ernesto de la Cueva
- Laboratory Animal Resources, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Vladimir Benes
- Genomics Core Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Janine Gote-Schniering
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Department of Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Department of Rheumatology and Immunology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
| | - Mikhail Savitski
- Proteomics Core Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
- Genome Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Rainer Pepperkok
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), 69120 Heidelberg, Germany
- Advanced Light Microscopy Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| |
Collapse
|
11
|
Bizzarri L, Steinbrunn D, Quennesson T, Lacour A, Bianchino GI, Bravo P, Chaignon P, Lohse J, Mäser P, Seemann M, Calenbergh SV, Hirsch AKH, Hahne H. Studying Target-Engagement of Anti-Infectives by Solvent-Induced Protein Precipitation and Quantitative Mass Spectrometry. ACS Infect Dis 2024; 10:4087-4102. [PMID: 39566904 DOI: 10.1021/acsinfecdis.4c00417] [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: 11/22/2024]
Abstract
Antimicrobial resistance (AMR) poses a serious threat to global health. The rapid emergence of resistance contrasts with the slow pace of antimicrobial development, emphasizing the urgent need for innovative drug discovery approaches. This study addresses a critical bottleneck in early drug development by introducing integral solvent-induced protein precipitation (iSPP) to rapidly assess the target-engagement of lead compounds in extracts of pathogenic microorganisms under close-to-physiological conditions. iSPP measures the change in protein stability against solvent-induced precipitation in the presence of ligands. The iSPP method for bacteria builds upon established SPP procedures and features optimized denaturation gradients and minimized sample input amounts. The effectiveness of the iSPP workflow was initially demonstrated through a multidrug target-engagement study. Using quantitative mass spectrometry (LC-MS/MS), we successfully identified known drug targets of seven different antibiotics in cell extracts of four AMR-related pathogens: the three Gram-negative bacteria Escherichia coli, Klebsiella pneumoniae, Pseudomonas aeruginosa and the Gram-positive bacterium Staphylococcus aureus. The iSPP method was ultimately applied to demonstrate target-engagement of compounds derived from target-based drug discovery. We employed five small molecules targeting three enzymes in the 2-C-methyl-d-erythritol 4-phosphate (MEP) pathway─a promising focus for anti-infective drug development. The study showcases iSPP adaptability and efficiency in identifying anti-infective drug targets, advancing early-stage drug discovery against AMR.
Collapse
Affiliation(s)
- Lorenzo Bizzarri
- OmicScouts GmbH, Lise-Meitner-Straße 30, Freising D-85354, Germany
- Department of Pharmacy, Saarland University, Campus E8.1, Saarbrücken D-66123, Germany
| | - Dominik Steinbrunn
- OmicScouts GmbH, Lise-Meitner-Straße 30, Freising D-85354, Germany
- TUM School of Natural Sciences, Department of Bioscience, Technical University of Munich, Center for Functional Protein Assemblies (CPA), Garching bei München D-85748, Germany
| | - Thibaut Quennesson
- Laboratory for Medicinal Chemistry (Campus Heymans), Ghent University, Ottergemsesteenweg 460, Gent B-9000, Belgium
| | - Antoine Lacour
- Department of Pharmacy, Saarland University, Campus E8.1, Saarbrücken D-66123, Germany
- Helmholtz Institute for Pharmaceutical Research (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, Campus E8.1, Saarbrücken D-66123, Germany
| | - Gabriella Ines Bianchino
- Equipe Chimie Biologique et Applications Thérapeutiques, Institut de Chimie de Strasbourg, UMR 7177, Université de Strasbourg/CNRS, 4, Rue Blaise Pascal, Strasbourg F-67070, France
| | - Patricia Bravo
- Swiss Tropical and Public Health Institute (Swiss TPH), Kreuzstrasse 2, Allschwil CH-4123, Switzerland
- University of Basel, Petersgraben 1, Basel CH-4001, Switzerland
| | - Philippe Chaignon
- Equipe Chimie Biologique et Applications Thérapeutiques, Institut de Chimie de Strasbourg, UMR 7177, Université de Strasbourg/CNRS, 4, Rue Blaise Pascal, Strasbourg F-67070, France
| | - Jonas Lohse
- OmicScouts GmbH, Lise-Meitner-Straße 30, Freising D-85354, Germany
| | - Pascal Mäser
- Swiss Tropical and Public Health Institute (Swiss TPH), Kreuzstrasse 2, Allschwil CH-4123, Switzerland
- University of Basel, Petersgraben 1, Basel CH-4001, Switzerland
| | - Myriam Seemann
- Equipe Chimie Biologique et Applications Thérapeutiques, Institut de Chimie de Strasbourg, UMR 7177, Université de Strasbourg/CNRS, 4, Rue Blaise Pascal, Strasbourg F-67070, France
| | - Serge Van Calenbergh
- Laboratory for Medicinal Chemistry (Campus Heymans), Ghent University, Ottergemsesteenweg 460, Gent B-9000, Belgium
| | - Anna K H Hirsch
- Department of Pharmacy, Saarland University, Campus E8.1, Saarbrücken D-66123, Germany
- Helmholtz Institute for Pharmaceutical Research (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, Campus E8.1, Saarbrücken D-66123, Germany
| | - Hannes Hahne
- OmicScouts GmbH, Lise-Meitner-Straße 30, Freising D-85354, Germany
| |
Collapse
|
12
|
Meng Z, Saei AA, Lyu H, Gaetani M, Zubarev RA. One-Pot Time-Induced Proteome Integral Solubility Alteration Assay for Automated and Sensitive Drug-Target Identification. Anal Chem 2024; 96:18917-18921. [PMID: 39567183 PMCID: PMC11618734 DOI: 10.1021/acs.analchem.4c05127] [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: 09/23/2024] [Revised: 10/31/2024] [Accepted: 11/15/2024] [Indexed: 11/22/2024]
Abstract
The proteome integral solubility alteration (PISA) assay is widely used for identifying drug targets, but it is labor-intensive and time-consuming and requires a substantial amount of biological sample. Aiming at enabling automation and greatly reducing the sample amount, we developed one-pot time-induced (OPTI)-PISA. Here, we demonstrate OPTI-PISA performance on identifying targets of multiple drugs in cell lysate and scaling down the sample amount to sub-microgram levels, making the PISA method suitable for NanoProteomics. OPTI-PISA can be implemented using only the standard equipment of a proteomics lab.
Collapse
Affiliation(s)
- Zhaowei Meng
- Division
of Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
- Chemical
Proteomics Unit, Science for Life Laboratory
(SciLifeLab), 17165 Stockholm, Sweden
- Chemical
Proteomics, Swedish National Infrastructure
for Biological Mass Spectrometry (BioMS), 17177 Stockholm, Sweden
| | - Amir Ata Saei
- Department
of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Hezheng Lyu
- Division
of Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
- Biomotif
AB, 18212 Danderyd, Sweden
| | - Massimiliano Gaetani
- Division
of Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
- Chemical
Proteomics Unit, Science for Life Laboratory
(SciLifeLab), 17165 Stockholm, Sweden
- Chemical
Proteomics, Swedish National Infrastructure
for Biological Mass Spectrometry (BioMS), 17177 Stockholm, Sweden
| | - Roman A. Zubarev
- Division
of Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
- Chemical
Proteomics Unit, Science for Life Laboratory
(SciLifeLab), 17165 Stockholm, Sweden
- Chemical
Proteomics, Swedish National Infrastructure
for Biological Mass Spectrometry (BioMS), 17177 Stockholm, Sweden
- Department
of Pharmaceutical and Toxicological Chemistry, Medical Institute, Peoples’ Friendship University of Russia named
after Patrice Lumumba (RUDN University), 6 Miklukho-Maklaya St., Moscow 117198, Russian Federation
| |
Collapse
|
13
|
Searle BC. Characterizing protein-protein interactions with thermal proteome profiling. Curr Opin Struct Biol 2024; 89:102946. [PMID: 39481280 PMCID: PMC11602378 DOI: 10.1016/j.sbi.2024.102946] [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: 05/02/2024] [Revised: 09/30/2024] [Accepted: 10/04/2024] [Indexed: 11/02/2024]
Abstract
Thermal proteome profiling (TPP) is an innovative technique that uses the principle of protein thermal stability to identify potential protein interaction partners. Employing quantitative mass spectrometry, TPP measures protein stability across the proteome, offering a comprehensive snapshot of protein interactions in a single experiment. When studying protein-protein interactions (PPI), TPP leverages changes in apparent protein melting temperatures to identify transient and weak interactions that most traditional PPI detection methodologies struggle to measure. This review discusses current TPP methodologies, the challenges of interpreting the resulting complex datasets, and opportunities to deepen and improve PPI networks. By advancing our grasp of intricate protein interactions, TPP promises to illuminate the molecular basis of diseases and drive the discovery of novel therapeutic targets.
Collapse
Affiliation(s)
- Brian C Searle
- Department of Biomedical Informatics, The Ohio State University Medical Center, Columbus, OH, 43210, USA; Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, 43210, USA; Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
14
|
He SJ, Li J, Zhou JC, Yang ZY, Liu X, Ge YW. Chemical proteomics accelerates the target discovery of natural products. Biochem Pharmacol 2024; 230:116609. [PMID: 39510194 DOI: 10.1016/j.bcp.2024.116609] [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: 08/10/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/15/2024]
Abstract
More than half of the global novel drugs are directly or indirectly derived from natural products (NPs) because of their better selectivity towards proteins. Traditional medicines perform multiple bioactivities through various NPs binding to drug targets, which highlights the opportunities of target discovery for drug development. However, detecting the binding relationship between NPs and targets remains challenging. Chemical proteomics, an interdisciplinary field of chemistry, proteomics, biology, and bioinformatics, has emerged as a potential approach for uncovering drug-target interactions. This review summarizes the principles and characteristics of the current widely applied chemical proteomic technologies, while delving into their latest applications in the target discovery of natural medicine. These endeavours demonstrate the potential of chemical proteomics for target discovery to supply dependable methodologies for the target elucidation of NPs.
Collapse
Affiliation(s)
- Shu-Jie He
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Innovative Team of Research on Effective Substances of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jun Li
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Innovative Team of Research on Effective Substances of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jie-Chun Zhou
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Innovative Team of Research on Effective Substances of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhi-You Yang
- College of Food Science and Technology, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Institute of Nutrition and Marine Drugs, Guangdong Ocean University, Zhanjiang, China
| | - Xi Liu
- School of Medical Information Engineering, Guangdong Pharmaceutical University, Guangzhou, China.
| | - Yue-Wei Ge
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China; Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM, Innovative Team of Research on Effective Substances of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China.
| |
Collapse
|
15
|
Locke TM, Fields R, Gizinski H, Otto GM, MacEwen MJS, Rusnac DV, He P, Shechner DM, McGann CD, Berg MD, Villen J, Sancak Y, Schweppe DK. High-throughput identification of calcium-regulated proteins across diverse proteomes. Cell Rep 2024; 43:114879. [PMID: 39425928 PMCID: PMC11921809 DOI: 10.1016/j.celrep.2024.114879] [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: 02/21/2024] [Revised: 08/27/2024] [Accepted: 09/30/2024] [Indexed: 10/21/2024] Open
Abstract
Calcium ions play important roles in nearly every biological process, yet whole-proteome analysis of calcium effectors has been hindered by a lack of high-throughput, unbiased, and quantitative methods to identify protein-calcium engagement. To address this, we adapted protein thermostability assays in budding yeast, human cells, and mouse mitochondria. Based on calcium-dependent thermostability, we identified 2,884 putative calcium-regulated proteins across human, mouse, and yeast proteomes. These data revealed calcium engagement of signaling hubs and cellular processes, including metabolic enzymes and the spliceosome. Cross-species comparison of calcium-protein engagement and mutagenesis experiments identified residue-specific cation engagement, even within well-known EF-hand domains. Additionally, we found that the dienoyl-coenzyme A (CoA) reductase DECR1 binds calcium at physiologically relevant concentrations with substrate-specific affinity, suggesting direct calcium regulation of mitochondrial fatty acid oxidation. These discovery-based proteomic analyses of calcium effectors establish a key resource to dissect cation engagement and its mechanistic effects across multiple species and diverse biological processes.
Collapse
Affiliation(s)
- Timothy M Locke
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Rose Fields
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA; Brotman Baty Institute for Precision Medicine, Seattle, WA, USA; Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Hayden Gizinski
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - George M Otto
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Melissa J S MacEwen
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Domnita-Valeria Rusnac
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA; Howard Hughes Medical Institute, Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Peixian He
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - David M Shechner
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Chris D McGann
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA; Brotman Baty Institute for Precision Medicine, Seattle, WA, USA; Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Matthew D Berg
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Judit Villen
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Yasemin Sancak
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA.
| | - Devin K Schweppe
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA; Brotman Baty Institute for Precision Medicine, Seattle, WA, USA; Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
16
|
Rider MH, Vertommen D, Johanns M. How mass spectrometry can be exploited to study AMPK. Essays Biochem 2024; 68:283-294. [PMID: 39056150 DOI: 10.1042/ebc20240009] [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: 05/29/2024] [Revised: 07/12/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024]
Abstract
AMP-activated protein kinase (AMPK) is a key regulator of metabolism and a recognised target for the treatment of metabolic diseases such as Type 2 diabetes (T2D). Here, we review how mass spectrometry (MS) can be used to study short-term control by AMPK via protein phosphorylation and long-term control due to changes in protein expression. We discuss how MS can quantify AMPK subunit levels in tissues from different species. We propose hydrogen-deuterium exchange (HDX)-MS to investigate molecular mechanisms of AMPK activation and thermoproteomic profiling (TPP) to assess off-target effects of pharmacological AMPK activators/inhibitors. Lastly, because large MS data sets are generated, we consider different approaches that can be used for their interpretation.
Collapse
Affiliation(s)
- Mark H Rider
- Protein Phosphorylation (PHOS) laboratory, Université catholique de Louvain and de Duve Institute, Avenue Hippocrate 75, B-1200 Brussels, Belgium
| | - Didier Vertommen
- Protein Phosphorylation (PHOS) laboratory, Université catholique de Louvain and de Duve Institute, Avenue Hippocrate 75, B-1200 Brussels, Belgium
| | - Manuel Johanns
- Protein Phosphorylation (PHOS) laboratory, Université catholique de Louvain and de Duve Institute, Avenue Hippocrate 75, B-1200 Brussels, Belgium
| |
Collapse
|
17
|
Qian L, Sun R, Aebersold R, Bühlmann P, Sander C, Guo T. AI-empowered perturbation proteomics for complex biological systems. CELL GENOMICS 2024; 4:100691. [PMID: 39488205 PMCID: PMC11605689 DOI: 10.1016/j.xgen.2024.100691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 09/02/2024] [Accepted: 10/06/2024] [Indexed: 11/04/2024]
Abstract
The insufficient availability of comprehensive protein-level perturbation data is impeding the widespread adoption of systems biology. In this perspective, we introduce the rationale, essentiality, and practicality of perturbation proteomics. Biological systems are perturbed with diverse biological, chemical, and/or physical factors, followed by proteomic measurements at various levels, including changes in protein expression and turnover, post-translational modifications, protein interactions, transport, and localization, along with phenotypic data. Computational models, employing traditional machine learning or deep learning, identify or predict perturbation responses, mechanisms of action, and protein functions, aiding in therapy selection, compound design, and efficient experiment design. We propose to outline a generic PMMP (perturbation, measurement, modeling to prediction) pipeline and build foundation models or other suitable mathematical models based on large-scale perturbation proteomic data. Finally, we contrast modeling between artificially and naturally perturbed systems and highlight the importance of perturbation proteomics for advancing our understanding and predictive modeling of biological systems.
Collapse
Affiliation(s)
- Liujia Qian
- School of Medicine, Westlake University, Hangzhou, Zhejiang Province, China; Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province, China; Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, Zhejiang Province, China
| | - Rui Sun
- School of Medicine, Westlake University, Hangzhou, Zhejiang Province, China; Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province, China; Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, Zhejiang Province, China
| | - Ruedi Aebersold
- Department of Biology, Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland
| | | | - Chris Sander
- Harvard Medical School, Boston, MA, USA; Broad Institute of Harvard and MIT, Boston, MA, USA; Ludwig Center at Harvard, Boston, MA, USA.
| | - Tiannan Guo
- School of Medicine, Westlake University, Hangzhou, Zhejiang Province, China; Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province, China; Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
18
|
Van Vranken JG, Li J, Mintseris J, Wei TY, Sniezek CM, Gadzuk-Shea M, Gygi SP, Schweppe DK. Large-scale characterization of drug mechanism of action using proteome-wide thermal shift assays. eLife 2024; 13:RP95595. [PMID: 39526730 PMCID: PMC11554310 DOI: 10.7554/elife.95595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
In response to an ever-increasing demand of new small molecules therapeutics, numerous chemical and genetic tools have been developed to interrogate compound mechanism of action. Owing to its ability to approximate compound-dependent changes in thermal stability, the proteome-wide thermal shift assay has emerged as a powerful tool in this arsenal. The most recent iterations have drastically improved the overall efficiency of these assays, providing an opportunity to screen compounds at a previously unprecedented rate. Taking advantage of this advance, we quantified more than one million thermal stability measurements in response to multiple classes of therapeutic and tool compounds (96 compounds in living cells and 70 compounds in lysates). When interrogating the dataset as a whole, approximately 80% of compounds (with quantifiable targets) caused a significant change in the thermal stability of an annotated target. There was also a wealth of evidence portending off-target engagement despite the extensive use of the compounds in the laboratory and/or clinic. Finally, the combined application of cell- and lysate-based assays, aided in the classification of primary (direct ligand binding) and secondary (indirect) changes in thermal stability. Overall, this study highlights the value of these assays in the drug development process by affording an unbiased and reliable assessment of compound mechanism of action.
Collapse
Affiliation(s)
| | - Jiaming Li
- Department of Cell Biology, Harvard Medical SchoolBostonUnited States
| | - Julian Mintseris
- Department of Cell Biology, Harvard Medical SchoolBostonUnited States
| | - Ting-Yu Wei
- Department of Cell Biology, Harvard Medical SchoolBostonUnited States
| | | | - Meagan Gadzuk-Shea
- Department of Genome Sciences, University of WashingtonSeattleUnited States
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical SchoolBostonUnited States
| | - Devin K Schweppe
- Department of Genome Sciences, University of WashingtonSeattleUnited States
| |
Collapse
|
19
|
Aliashkevich A, Guest T, Alvarez L, Gilmore MC, Rea D, Amstutz J, Mateus A, Schiffthaler B, Ruiz I, Typas A, Savitski MM, Brown PJB, Cava F. LD-transpeptidation is crucial for fitness and polar growth in Agrobacterium tumefaciens. PLoS Genet 2024; 20:e1011449. [PMID: 39432536 PMCID: PMC11527210 DOI: 10.1371/journal.pgen.1011449] [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: 06/26/2024] [Revised: 10/31/2024] [Accepted: 10/04/2024] [Indexed: 10/23/2024] Open
Abstract
Peptidoglycan (PG), a mesh-like structure which is the primary component of the bacterial cell wall, is crucial to maintain cell integrity and shape. While most bacteria rely on penicillin binding proteins (PBPs) for crosslinking, some species also employ LD-transpeptidases (LDTs). Unlike PBPs, the essentiality and biological functions of LDTs remain largely unclear. The Hyphomicrobiales order of the Alphaproteobacteria, known for their polar growth, have PG which is unusually rich in LD-crosslinks, suggesting that LDTs may play a more significant role in PG synthesis in these bacteria. Here, we investigated LDTs in the plant pathogen Agrobacterium tumefaciens and found that LD-transpeptidation, resulting from at least one of 14 putative LDTs present in this bacterium, is essential for its survival. Notably, a mutant lacking a distinctive group of 7 LDTs which are broadly conserved among the Hyphomicrobiales exhibited reduced LD-crosslinking and tethering of PG to outer membrane β-barrel proteins. Consequently, this mutant suffered severe fitness loss and cell shape rounding, underscoring the critical role played by these Hyphomicrobiales-specific LDTs in maintaining cell wall integrity and promoting elongation. Tn-sequencing screens further revealed non-redundant functions for A. tumefaciens LDTs. Specifically, Hyphomicrobiales-specific LDTs exhibited synthetic genetic interactions with division and cell cycle proteins, and a single LDT from another group. Additionally, our findings demonstrate that strains lacking all LDTs except one displayed distinctive phenotypic profiles and genetic interactions. Collectively, our work emphasizes the critical role of LD-crosslinking in A. tumefaciens cell wall integrity and growth and provides insights into the functional specialization of these crosslinking activities.
Collapse
Affiliation(s)
- Alena Aliashkevich
- Department of Molecular Biology and Laboratory for Molecular Infection Medicine Sweden, Umeå Centre for Microbial Research, SciLifeLab, Umeå University, Umeå, Sweden
| | - Thomas Guest
- Department of Molecular Biology and Laboratory for Molecular Infection Medicine Sweden, Umeå Centre for Microbial Research, SciLifeLab, Umeå University, Umeå, Sweden
| | - Laura Alvarez
- Department of Molecular Biology and Laboratory for Molecular Infection Medicine Sweden, Umeå Centre for Microbial Research, SciLifeLab, Umeå University, Umeå, Sweden
| | - Michael C. Gilmore
- Department of Molecular Biology and Laboratory for Molecular Infection Medicine Sweden, Umeå Centre for Microbial Research, SciLifeLab, Umeå University, Umeå, Sweden
| | - Daniel Rea
- Department of Molecular Biology and Laboratory for Molecular Infection Medicine Sweden, Umeå Centre for Microbial Research, SciLifeLab, Umeå University, Umeå, Sweden
| | - Jennifer Amstutz
- Division of Biological Sciences, University of Missouri-Columbia, Columbia, Missouri, United States of America
| | - André Mateus
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Bastian Schiffthaler
- Department of Molecular Biology and Laboratory for Molecular Infection Medicine Sweden, Umeå Centre for Microbial Research, SciLifeLab, Umeå University, Umeå, Sweden
| | - Iñigo Ruiz
- Department of Molecular Biology and Laboratory for Molecular Infection Medicine Sweden, Umeå Centre for Microbial Research, SciLifeLab, Umeå University, Umeå, Sweden
| | - Athanasios Typas
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Mikhail M. Savitski
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Pamela J. B. Brown
- Division of Biological Sciences, University of Missouri-Columbia, Columbia, Missouri, United States of America
| | - Felipe Cava
- Department of Molecular Biology and Laboratory for Molecular Infection Medicine Sweden, Umeå Centre for Microbial Research, SciLifeLab, Umeå University, Umeå, Sweden
| |
Collapse
|
20
|
Tóth D, Tengölics R, Aarabi F, Karlsson A, Vidal-Meireles A, Kovács L, Kuntam S, Körmöczi T, Fernie AR, Hudson EP, Papp B, Tóth SZ. Chloroplastic ascorbate modifies plant metabolism and may act as a metabolite signal regardless of oxidative stress. PLANT PHYSIOLOGY 2024; 196:1691-1711. [PMID: 39106412 PMCID: PMC11444284 DOI: 10.1093/plphys/kiae409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 06/06/2024] [Accepted: 07/01/2024] [Indexed: 08/09/2024]
Abstract
Ascorbate (Asc) is a major plant metabolite that plays crucial roles in various processes, from reactive oxygen scavenging to epigenetic regulation. However, to what extent and how Asc modulates metabolism is largely unknown. We investigated the consequences of chloroplastic and total cellular Asc deficiencies by studying chloroplastic Asc transporter mutant lines lacking PHOSPHATE TRANSPORTER 4; 4 and the Asc-deficient vtc2-4 mutant of Arabidopsis (Arabidopsis thaliana). Under regular growth conditions, both Asc deficiencies caused minor alterations in photosynthesis, with no apparent signs of oxidative damage. In contrast, metabolomics analysis revealed global and largely overlapping alterations in the metabolome profiles of both Asc-deficient mutants, suggesting that chloroplastic Asc modulates plant metabolism. We observed significant alterations in amino acid metabolism, particularly in arginine metabolism, activation of nucleotide salvage pathways, and changes in secondary metabolism. In addition, proteome-wide analysis of thermostability revealed that Asc may interact with enzymes involved in arginine metabolism, the Calvin-Benson cycle, and several photosynthetic electron transport components. Overall, our results suggest that, independent of oxidative stress, chloroplastic Asc modulates the activity of diverse metabolic pathways in vascular plants and may act as an internal metabolite signal.
Collapse
Affiliation(s)
- Dávid Tóth
- Laboratory for Molecular Photobioenergetics, HUN-REN Biological Research Centre, Institute of Plant Biology, Temesvári krt. 62, Szeged H-6726, Hungary
- Doctoral School of Biology, University of Szeged, Közép fasor 52, Szeged H-6722, Hungary
| | - Roland Tengölics
- HCEMM-BRC Metabolic Systems Biology Lab, Temesvári krt. 62, Szeged H-6726, Hungary
- Synthetic and Systems Biology Unit, HUN-REN Biological Research Centre, Institute of Biochemistry, Temesvári krt. 62, Szeged H-6726, Hungary
- Metabolomics Lab, Core Facilities, HUN-REN Biological Research Centre, Temesvári krt. 62, Szeged H-6726, Hungary
| | - Fayezeh Aarabi
- Max Planck Institute of Molecular Plant Physiology, Am Mühlenberg 1, Potsdam-Golm D-14476, Germany
| | - Anna Karlsson
- Science for Life Laboratory, School of Engineering Science in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, PO Box 1031, Solna 171 21, Sweden
| | - André Vidal-Meireles
- Laboratory for Molecular Photobioenergetics, HUN-REN Biological Research Centre, Institute of Plant Biology, Temesvári krt. 62, Szeged H-6726, Hungary
| | - László Kovács
- Laboratory for Molecular Photobioenergetics, HUN-REN Biological Research Centre, Institute of Plant Biology, Temesvári krt. 62, Szeged H-6726, Hungary
| | - Soujanya Kuntam
- Laboratory for Molecular Photobioenergetics, HUN-REN Biological Research Centre, Institute of Plant Biology, Temesvári krt. 62, Szeged H-6726, Hungary
| | - Tímea Körmöczi
- HCEMM-BRC Metabolic Systems Biology Lab, Temesvári krt. 62, Szeged H-6726, Hungary
| | - Alisdair R Fernie
- Max Planck Institute of Molecular Plant Physiology, Am Mühlenberg 1, Potsdam-Golm D-14476, Germany
| | - Elton P Hudson
- Science for Life Laboratory, School of Engineering Science in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, PO Box 1031, Solna 171 21, Sweden
| | - Balázs Papp
- HCEMM-BRC Metabolic Systems Biology Lab, Temesvári krt. 62, Szeged H-6726, Hungary
- Synthetic and Systems Biology Unit, HUN-REN Biological Research Centre, Institute of Biochemistry, Temesvári krt. 62, Szeged H-6726, Hungary
- National Laboratory for Health Security, HUN-REN Biological Research Centre, Temesvári krt. 62, Szeged H-6726, Hungary
| | - Szilvia Z Tóth
- Laboratory for Molecular Photobioenergetics, HUN-REN Biological Research Centre, Institute of Plant Biology, Temesvári krt. 62, Szeged H-6726, Hungary
| |
Collapse
|
21
|
Lu H, Zhu Z, Fields L, Zhang H, Li L. Mass Spectrometry Structural Proteomics Enabled by Limited Proteolysis and Cross-Linking. MASS SPECTROMETRY REVIEWS 2024. [PMID: 39300771 DOI: 10.1002/mas.21908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 08/31/2024] [Accepted: 09/02/2024] [Indexed: 09/22/2024]
Abstract
The exploration of protein structure and function stands at the forefront of life science and represents an ever-expanding focus in the development of proteomics. As mass spectrometry (MS) offers readout of protein conformational changes at both the protein and peptide levels, MS-based structural proteomics is making significant strides in the realms of structural and molecular biology, complementing traditional structural biology techniques. This review focuses on two powerful MS-based techniques for peptide-level readout, namely limited proteolysis-mass spectrometry (LiP-MS) and cross-linking mass spectrometry (XL-MS). First, we discuss the principles, features, and different workflows of these two methods. Subsequently, we delve into the bioinformatics strategies and software tools used for interpreting data associated with these protein conformation readouts and how the data can be integrated with other computational tools. Furthermore, we provide a comprehensive summary of the noteworthy applications of LiP-MS and XL-MS in diverse areas including neurodegenerative diseases, interactome studies, membrane proteins, and artificial intelligence-based structural analysis. Finally, we discuss the factors that modulate protein conformational changes. We also highlight the remaining challenges in understanding the intricacies of protein conformational changes by LiP-MS and XL-MS technologies.
Collapse
Affiliation(s)
- Haiyan Lu
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Zexin Zhu
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Lauren Fields
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Hua Zhang
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Lingjun Li
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Lachman Institute for Pharmaceutical Development, School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
22
|
Kim D, Nita-Lazar A. Progress in mass spectrometry approaches to profiling protein-protein interactions in the studies of the innate immune system. JOURNAL OF PROTEINS AND PROTEOMICS 2024; 15:545-559. [PMID: 39380887 PMCID: PMC11460538 DOI: 10.1007/s42485-024-00156-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 06/04/2024] [Accepted: 06/24/2024] [Indexed: 10/10/2024]
Abstract
Understanding protein-protein interactions (PPIs) is pivotal for deciphering the intricacies of biological processes. Dysregulation of PPIs underlies a spectrum of diseases, including cancer, neurodegenerative disorders, and autoimmune conditions, highlighting the imperative of investigating these interactions for therapeutic advancements. This review delves into the realm of mass spectrometry-based techniques for elucidating PPIs and their profound implications in biological research. Mass spectrometry in the PPI research field not only facilitates the evaluation of protein-protein interaction modulators but also discovers unclear molecular mechanisms and sheds light on both on- and off-target effects, thus aiding in drug development. Our discussion navigates through six pivotal techniques: affinity purification mass spectrometry (AP-MS), proximity labeling mass spectrometry (PL-MS), cross-linking mass spectrometry (XL-MS), size exclusion chromatography coupled with mass spectrometry (SEC-MS), limited proteolysis-coupled mass spectrometry (LiP-MS), and thermal proteome profiling (TPP).
Collapse
Affiliation(s)
- Doeun Kim
- Functional Cellular Networks Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-1892, USA
| | - Aleksandra Nita-Lazar
- Functional Cellular Networks Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-1892, USA
| |
Collapse
|
23
|
Le Sueur C, Rattray M, Savitski M. GPMelt: A hierarchical Gaussian process framework to explore the dark meltome of thermal proteome profiling experiments. PLoS Comput Biol 2024; 20:e1011632. [PMID: 39331673 PMCID: PMC11463780 DOI: 10.1371/journal.pcbi.1011632] [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: 10/25/2023] [Revised: 10/09/2024] [Accepted: 08/23/2024] [Indexed: 09/29/2024] Open
Abstract
Thermal proteome profiling (TPP) is a proteome wide technology that enables unbiased detection of protein drug interactions as well as changes in post-translational state of proteins between different biological conditions. Statistical analysis of temperature range TPP (TPP-TR) datasets relies on comparing protein melting curves, describing the amount of non-denatured proteins as a function of temperature, between different conditions (e.g. presence or absence of a drug). However, state-of-the-art models are restricted to sigmoidal melting behaviours while unconventional melting curves, representing up to 50% of TPP-TR datasets, have recently been shown to carry important biological information. We present a novel statistical framework, based on hierarchical Gaussian process models and named GPMelt, to make TPP-TR datasets analysis unbiased with respect to the melting profiles of proteins. GPMelt scales to multiple conditions, and extension of the model to deeper hierarchies (i.e. with additional sub-levels) allows to deal with complex TPP-TR protocols. Collectively, our statistical framework extends the analysis of TPP-TR datasets for both protein and peptide level melting curves, offering access to thousands of previously excluded melting curves and thus substantially increasing the coverage and the ability of TPP to uncover new biology.
Collapse
Affiliation(s)
- Cecile Le Sueur
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Magnus Rattray
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Mikhail Savitski
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| |
Collapse
|
24
|
Zhu C, Zhang M, Gong S, Du J, Ma L, Liu Y, Li Y, Yu J, Liu N. Identification of Matrine as a Kirsten rats Arcomaviral oncogene homolog inhibitor alleviating chemotherapy-induced neuropathic pain. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155841. [PMID: 38971025 DOI: 10.1016/j.phymed.2024.155841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 06/13/2024] [Accepted: 06/20/2024] [Indexed: 07/08/2024]
Abstract
BACKGROUND Chemotherapy-induced peripheral neuropathy (CIPN) represents a prevailing and severe clinical concern, characterized by limited availability of clinically effective treatment strategies. Current evidence endorses matrine's potential as a neuroprotective and analgesic agent for CIPN. Nevertheless, the precise targets and mechanisms of action of matrine remain insufficiently explored, impeding comprehensive pharmacological investigation and clinical application. OBJECTIVE This study endeavors to elucidate the analgesic and neuroprotective effects of matrine in mice with vincristine-induced neuropathic pain. A focal point is the identification of matrine's specific target and the underlying molecular mechanisms governing its analgesic and neuroprotective actions. METHODS To discern matrine's analgesic effects in CIPN mice, we conducted behavioral experiments encompassing the Von Frey filament test and Hargreaves Test. Furthermore, we conducted electrophysiological and histopathological assessments involving HE staining, Nissl staining, and Fluoro-Jade B staining to evaluate matrine's effects on neuroprotection within dorsal root ganglia and the spinal cord of CIPN mice. Sequentially, thermal shift assay, GTP hydrolysis assay, and nucleotide exchange assay were executed to validate matrine's inhibitory effects on KRAS. Molecular docking and site-directed mutagenesis experiments were implemented to identify the precise binding pocket of matrine on KRAS. Lastly, matrine's inhibitory effects on downstream signaling pathways of KRAS were confirmed through experiments conducted at animal model. RESULTS Matrine exhibited a notable increase in mechanical withdrawal threshold and thermal withdrawal latency in vincristine-treated mice. This compound substantially ameliorated the neurofunctional blockade associated with sensory and motor functions induced by vincristine. Moreover, matrine mitigated pathological damage within DRG and the L4-L5 spinal cord regions. The study's MST experiments indicated matrine's substantial elevation of KRAS's melting temperature. The GTP hydrolysis and nucleotide exchange assays revealed concentration-dependent inhibition of KRAS activity by matrine. Molecular docking provided insight into the binding mode of matrine with KRAS, while site-directed mutagenesis verified the specific binding site of matrine on KRAS. Lastly, matrine's inhibition of downstream Raf/Erk1/2 and PI3K/Akt/mTOR signaling pathways of KRAS was confirmed in VCR mice. CONCLUSION Compared to previous studies, our research has identified matrine as a natural inhibitor of the elusive protein KRAS, often considered "undruggable." Furthermore, this study has revealed that matrine exerts its therapeutic effects on chemotherapy-induced peripheral neuropathy (CIPN) by inhibiting KRAS activation, subsequently suppressing downstream signaling pathways such as Raf/Erk1/2 and PI3K/Akt/mTOR. This investigation signifies the discovery of a novel target for matrine, thus expanding the potential scope of its involvement in KRAS-related biological functions and diseases. These findings hold the promise of providing a crucial experimental foundation for forthcoming drug development initiatives centered around matrine, thereby advancing the field of pharmaceutical research.
Collapse
Affiliation(s)
- Chunhao Zhu
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, PR China; Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China
| | - Mengting Zhang
- People's Hospital of Ningxia Hui Autonomous Region, Yinchuan 750004, PR China
| | - Shuaishuai Gong
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, PR China
| | - Juan Du
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, PR China
| | - Lin Ma
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, PR China
| | - Yue Liu
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, PR China
| | - Yuxiang Li
- School of nursing, Ningxia Medical University, Yinchuan 750004, Ningxia, PR China
| | - Jianqiang Yu
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, PR China; Ningxia Key Laboratory of Drug Development and Generic Drug Research, Ningxia Medical University, Yinchuan 750004, PR China.
| | - Ning Liu
- School of Pharmacy, Ningxia Medical University, Yinchuan 750004, PR China; Ningxia Key Laboratory of Drug Development and Generic Drug Research, Ningxia Medical University, Yinchuan 750004, PR China.
| |
Collapse
|
25
|
Jiang Y, Rex DA, Schuster D, Neely BA, Rosano GL, Volkmar N, Momenzadeh A, Peters-Clarke TM, Egbert SB, Kreimer S, Doud EH, Crook OM, Yadav AK, Vanuopadath M, Hegeman AD, Mayta M, Duboff AG, Riley NM, Moritz RL, Meyer JG. Comprehensive Overview of Bottom-Up Proteomics Using Mass Spectrometry. ACS MEASUREMENT SCIENCE AU 2024; 4:338-417. [PMID: 39193565 PMCID: PMC11348894 DOI: 10.1021/acsmeasuresciau.3c00068] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 05/03/2024] [Accepted: 05/03/2024] [Indexed: 08/29/2024]
Abstract
Proteomics is the large scale study of protein structure and function from biological systems through protein identification and quantification. "Shotgun proteomics" or "bottom-up proteomics" is the prevailing strategy, in which proteins are hydrolyzed into peptides that are analyzed by mass spectrometry. Proteomics studies can be applied to diverse studies ranging from simple protein identification to studies of proteoforms, protein-protein interactions, protein structural alterations, absolute and relative protein quantification, post-translational modifications, and protein stability. To enable this range of different experiments, there are diverse strategies for proteome analysis. The nuances of how proteomic workflows differ may be challenging to understand for new practitioners. Here, we provide a comprehensive overview of different proteomics methods. We cover from biochemistry basics and protein extraction to biological interpretation and orthogonal validation. We expect this Review will serve as a handbook for researchers who are new to the field of bottom-up proteomics.
Collapse
Affiliation(s)
- Yuming Jiang
- Department
of Computational Biomedicine, Cedars Sinai
Medical Center, Los Angeles, California 90048, United States
- Smidt Heart
Institute, Cedars Sinai Medical Center, Los Angeles, California 90048, United States
- Advanced
Clinical Biosystems Research Institute, Cedars Sinai Medical Center, Los
Angeles, California 90048, United States
| | - Devasahayam Arokia
Balaya Rex
- Center for
Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, India
| | - Dina Schuster
- Department
of Biology, Institute of Molecular Systems
Biology, ETH Zurich, Zurich 8093, Switzerland
- Department
of Biology, Institute of Molecular Biology
and Biophysics, ETH Zurich, Zurich 8093, Switzerland
- Laboratory
of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institute, Villigen 5232, Switzerland
| | - Benjamin A. Neely
- Chemical
Sciences Division, National Institute of
Standards and Technology, NIST, Charleston, South Carolina 29412, United States
| | - Germán L. Rosano
- Mass
Spectrometry
Unit, Institute of Molecular and Cellular
Biology of Rosario, Rosario, 2000 Argentina
| | - Norbert Volkmar
- Department
of Biology, Institute of Molecular Systems
Biology, ETH Zurich, Zurich 8093, Switzerland
| | - Amanda Momenzadeh
- Department
of Computational Biomedicine, Cedars Sinai
Medical Center, Los Angeles, California 90048, United States
- Smidt Heart
Institute, Cedars Sinai Medical Center, Los Angeles, California 90048, United States
- Advanced
Clinical Biosystems Research Institute, Cedars Sinai Medical Center, Los
Angeles, California 90048, United States
| | - Trenton M. Peters-Clarke
- Department
of Pharmaceutical Chemistry, University
of California—San Francisco, San Francisco, California, 94158, United States
| | - Susan B. Egbert
- Department
of Chemistry, University of Manitoba, Winnipeg, Manitoba, R3T 2N2 Canada
| | - Simion Kreimer
- Smidt Heart
Institute, Cedars Sinai Medical Center, Los Angeles, California 90048, United States
- Advanced
Clinical Biosystems Research Institute, Cedars Sinai Medical Center, Los
Angeles, California 90048, United States
| | - Emma H. Doud
- Center
for Proteome Analysis, Indiana University
School of Medicine, Indianapolis, Indiana, 46202-3082, United States
| | - Oliver M. Crook
- Oxford
Protein Informatics Group, Department of Statistics, University of Oxford, Oxford OX1 3LB, United
Kingdom
| | - Amit Kumar Yadav
- Translational
Health Science and Technology Institute, NCR Biotech Science Cluster 3rd Milestone Faridabad-Gurgaon
Expressway, Faridabad, Haryana 121001, India
| | | | - Adrian D. Hegeman
- Departments
of Horticultural Science and Plant and Microbial Biology, University of Minnesota, Twin Cities, Minnesota 55108, United States
| | - Martín
L. Mayta
- School
of Medicine and Health Sciences, Center for Health Sciences Research, Universidad Adventista del Plata, Libertador San Martin 3103, Argentina
- Molecular
Biology Department, School of Pharmacy and Biochemistry, Universidad Nacional de Rosario, Rosario 2000, Argentina
| | - Anna G. Duboff
- Department
of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Nicholas M. Riley
- Department
of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Robert L. Moritz
- Institute
for Systems biology, Seattle, Washington 98109, United States
| | - Jesse G. Meyer
- Department
of Computational Biomedicine, Cedars Sinai
Medical Center, Los Angeles, California 90048, United States
- Smidt Heart
Institute, Cedars Sinai Medical Center, Los Angeles, California 90048, United States
- Advanced
Clinical Biosystems Research Institute, Cedars Sinai Medical Center, Los
Angeles, California 90048, United States
| |
Collapse
|
26
|
Emery-Corbin SJ, Yousef JM, Adhikari S, Sumardy F, Nhu D, van Delft MF, Lessene G, Dziekan J, Webb AI, Dagley LF. Improved drug target deconvolution with PISA-DIA using an extended, overlapping temperature gradient. Proteomics 2024; 24:e2300644. [PMID: 38766901 DOI: 10.1002/pmic.202300644] [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: 12/19/2023] [Revised: 05/03/2024] [Accepted: 05/07/2024] [Indexed: 05/22/2024]
Abstract
Thermal proteome profiling (TPP) is a powerful tool for drug target deconvolution. Recently, data-independent acquisition mass spectrometry (DIA-MS) approaches have demonstrated significant improvements to depth and missingness in proteome data, but traditional TPP (a.k.a. CEllular Thermal Shift Assay "CETSA") workflows typically employ multiplexing reagents reliant on data-dependent acquisition (DDA). Herein, we introduce a new experimental design for the Proteome Integral Solubility Alteration via label-free DIA approach (PISA-DIA). We highlight the proteome coverage and sensitivity achieved by using multiple overlapping thermal gradients alongside DIA-MS, which maximizes efficiencies in PISA sample concatenation and safeguards against missing protein targets that exist at high melting temperatures. We demonstrate our extended PISA-DIA design has superior proteome coverage as compared to using tandem-mass tags (TMT) necessitating DDA-MS analysis. Importantly, we demonstrate our PISA-DIA approach has the quantitative and statistical rigor using A-1331852, a specific inhibitor of BCL-xL. Due to the high melt temperature of this protein target, we utilized our extended multiple gradient PISA-DIA workflow to identify BCL-xL. We assert our novel overlapping gradient PISA-DIA-MS approach is ideal for unbiased drug target deconvolution, spanning a large temperature range whilst minimizing target dropout between gradients, increasing the likelihood of resolving the protein targets of novel compounds.
Collapse
Affiliation(s)
- Samantha J Emery-Corbin
- Advanced Technology and Biology Division, the Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Jumana M Yousef
- Advanced Technology and Biology Division, the Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Subash Adhikari
- Advanced Technology and Biology Division, the Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Fransisca Sumardy
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
- ACRF Chemical Biology Division, the Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Duong Nhu
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
- ACRF Chemical Biology Division, the Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Mark F van Delft
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
- Blood Cells and Blood Cancer Division, the Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Guillaume Lessene
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
- ACRF Chemical Biology Division, the Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, Victoria, Australia
| | - Jerzy Dziekan
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
- Infection and Immunity Division, the Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Andrew I Webb
- Advanced Technology and Biology Division, the Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Laura F Dagley
- Advanced Technology and Biology Division, the Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
27
|
Shanley HT, Taki AC, Nguyen N, Wang T, Byrne JJ, Ang CS, Leeming MG, Williamson N, Chang BCH, Jabbar A, Sleebs BE, Gasser RB. Comparative structure activity and target exploration of 1,2-diphenylethynes in Haemonchus contortus and Caenorhabditis elegans. Int J Parasitol Drugs Drug Resist 2024; 25:100534. [PMID: 38554597 PMCID: PMC10992699 DOI: 10.1016/j.ijpddr.2024.100534] [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: 02/01/2024] [Revised: 03/14/2024] [Accepted: 03/17/2024] [Indexed: 04/01/2024]
Abstract
Infections and diseases caused by parasitic nematodes have a major adverse impact on the health and productivity of animals and humans worldwide. The control of these parasites often relies heavily on the treatment with commercially available chemical compounds (anthelmintics). However, the excessive or uncontrolled use of these compounds in livestock animals has led to major challenges linked to drug resistance in nematodes. Therefore, there is a need to develop new anthelmintics with novel mechanism(s) of action. Recently, we identified a small molecule, designated UMW-9729, with nematocidal activity against the free-living model organism Caenorhabditis elegans. Here, we evaluated UMW-9729's potential as an anthelmintic in a structure-activity relationship (SAR) study in C. elegans and the highly pathogenic, blood-feeding Haemonchus contortus (barber's pole worm), and explored the compound-target relationship using thermal proteome profiling (TPP). First, we synthesised and tested 25 analogues of UMW-9729 for their nematocidal activity in both H. contortus (larvae and adults) and C. elegans (young adults), establishing a preliminary nematocidal pharmacophore for both species. We identified several compounds with marked activity against either H. contortus or C. elegans which had greater efficacy than UMW-9729, and found a significant divergence in compound bioactivity between these two nematode species. We also identified a UMW-9729 analogue, designated 25, that moderately inhibited the motility of adult female H. contortus in vitro. Subsequently, we inferred three H. contortus proteins (HCON_00134350, HCON_00021470 and HCON_00099760) and five C. elegans proteins (F30A10.9, F15B9.8, B0361.6, DNC-4 and UNC-11) that interacted directly with UMW-9729; however, no conserved protein target was shared between the two nematode species. Future work aims to extend the SAR investigation in these and other parasitic nematode species, and validate individual proteins identified here as possible targets of UMW-9729. Overall, the present study evaluates this anthelmintic candidate and highlights some challenges associated with early anthelmintic investigation.
Collapse
Affiliation(s)
- Harrison T Shanley
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria, 3010, Australia; Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia
| | - Aya C Taki
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Nghi Nguyen
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia
| | - Tao Wang
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Joseph J Byrne
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Ching-Seng Ang
- Melbourne Mass Spectrometry and Proteomics Facility, The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Michael G Leeming
- Melbourne Mass Spectrometry and Proteomics Facility, The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Nicholas Williamson
- Melbourne Mass Spectrometry and Proteomics Facility, The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Bill C H Chang
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Abdul Jabbar
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Brad E Sleebs
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria, 3010, Australia; Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia.
| | - Robin B Gasser
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria, 3010, Australia.
| |
Collapse
|
28
|
Geißler A, Junca H, Kany AM, Daumann LJ, Hirsch AKH, Pieper DH, Sieber SA. Isocyanides inhibit bacterial pathogens by covalent targeting of essential metabolic enzymes. Chem Sci 2024; 15:11946-11955. [PMID: 39092115 PMCID: PMC11290450 DOI: 10.1039/d4sc01940g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 06/23/2024] [Indexed: 08/04/2024] Open
Abstract
Isonitrile natural products, also known as isocyanides, demonstrate potent antimicrobial activities, yet our understanding of their molecular targets remains limited. Here, we focus on the so far neglected group of monoisonitriles to gain further insights into their antimicrobial mode of action (MoA). Screening a focused monoisonitrile library revealed a potent S. aureus growth inhibitor with a different MoA compared to previously described isonitrile antibiotics. Chemical proteomics via competitive cysteine reactivity profiling, uncovered covalent modifications of two essential metabolic enzymes involved in the fatty acid biosynthetic process (FabF) and the hexosamine pathway (GlmS) at their active site cysteines. In-depth studies with the recombinant enzymes demonstrated concentration-dependent labeling, covalent binding to the catalytic site and corresponding functional inhibition by the isocyanide. Thermal proteome profiling and full proteome studies of compound-treated S. aureus further highlighted the destabilization and dysregulation of proteins related to the targeted pathways. Cytotoxicity and the inhibition of cytochrome P450 enzymes require optimization of the hit molecule prior to therapeutic application. The here described novel, covalent isocyanide MoA highlights the versatility of the functional group, making it a useful tool and out-of-the-box starting point for the development of innovative antibiotics.
Collapse
Affiliation(s)
- Alexandra Geißler
- Center for Functional Protein Assemblies, Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich Ernst-Otto-Fischer-Straße 8 85748 Garching Germany
| | - Howard Junca
- Microbial Interactions and Processes Research Group, Helmholtz Centre for Infection Research Inhoffenstraße 7 38124 Braunschweig Germany
| | - Andreas M Kany
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI) Campus E8.1 66123 Saarbrücken Germany
- Deutsches Zentrum für Infektionsforschung (DZIF) e.V. 38124 Braunschweig Germany
| | - Lena J Daumann
- Chair of Bioinorganic Chemistry, Heinrich-Heine-Universität Düsseldorf Universitätsstraße 1 40225 Düsseldorf Germany
| | - Anna K H Hirsch
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI) Campus E8.1 66123 Saarbrücken Germany
- Saarland University, Department of Pharmacy 66123 Saarbrücken Germany
- Deutsches Zentrum für Infektionsforschung (DZIF) e.V. 38124 Braunschweig Germany
| | - Dietmar H Pieper
- Microbial Interactions and Processes Research Group, Helmholtz Centre for Infection Research Inhoffenstraße 7 38124 Braunschweig Germany
| | - Stephan A Sieber
- Center for Functional Protein Assemblies, Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich Ernst-Otto-Fischer-Straße 8 85748 Garching Germany
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI) Campus E8.1 66123 Saarbrücken Germany
| |
Collapse
|
29
|
Yang H, Ibrahim MM, Zhang S, Sun Y, Chang J, Qi H, Yang S. Targeting post-stroke neuroinflammation with Salvianolic acid A: molecular mechanisms and preclinical evidence. Front Immunol 2024; 15:1433590. [PMID: 39139557 PMCID: PMC11319147 DOI: 10.3389/fimmu.2024.1433590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 07/16/2024] [Indexed: 08/15/2024] Open
Abstract
Salvianolic acid A (SalA), a bioactive compound extracted from Salvia miltiorrhiza, has garnered considerable interest for its potential in ameliorating the post-stroke neuroinflammation. This review delineates the possible molecular underpinnings of anti-inflammatory and neuroprotective roles of SalA, offering a comprehensive analysis of its therapeutic efficacy in preclinical studies of ischemic stroke. We explore the intricate interplay between post-stroke neuroinflammation and the modulatory effects of SalA on pro-inflammatory cytokines, inflammatory signaling pathways, the peripheral immune cell infiltration through blood-brain barrier disruption, and endothelial cell function. The pharmacokinetic profiles of SalA in the context of stroke, characterized by enhanced cerebral penetration post-ischemia, makes it particularly suitable as a therapeutic agent. Preliminary clinical findings have demonstrated that salvianolic acids (SA) has a positive impact on cerebral perfusion and neurological deficits in stroke patients, warranting further investigation. This review emphasizes SalA as a potential anti-inflammatory agent for the advancement of innovative therapeutic approaches in the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Hongchun Yang
- Department of Neurosurgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Muhammad Mustapha Ibrahim
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Siyu Zhang
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
- Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yao Sun
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Junlei Chang
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Hui Qi
- Department of Neurosurgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Shilun Yang
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
30
|
Gassaway BM, Huttlin EL, Huntsman EM, Yaron-Barir TM, Johnson JL, Kurmi K, Cantley LC, Paulo JA, Ringel AE, Gygi SP, Haigis MC. Profiling Proteins and Phosphorylation Sites During T Cell Activation Using an Integrated Thermal Shift Assay. Mol Cell Proteomics 2024; 23:100801. [PMID: 38880243 PMCID: PMC11298636 DOI: 10.1016/j.mcpro.2024.100801] [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: 11/03/2023] [Revised: 05/24/2024] [Accepted: 06/13/2024] [Indexed: 06/18/2024] Open
Abstract
T cell activation is a complex biological process of naive cells maturing into effector cells. Proteomic and phospho-proteomic approaches have provided critical insights into this process, yet it is not always clear how changes in individual proteins or phosphorylation sites have functional significance. Here, we developed the Phosphorylation Integrated Thermal Shift Assay (PITSA) that combines the measurement of protein or phosphorylation site abundance and thermal stability into a single tandem mass tags experiment and apply this method to study T cell activation. We quantified the abundance and thermal stability of over 7500 proteins and 5000 phosphorylation sites and identified significant differences in chromatin-related, TCR signaling, DNA repair, and proliferative phosphoproteins. PITSA may be applied to a wide range of biological contexts to generate hypotheses as to which proteins or phosphorylation sites are functionally regulated in a given system as well as the mechanisms by which this regulation may occur.
Collapse
Affiliation(s)
- Brandon M Gassaway
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA; Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah, USA
| | - Edward L Huttlin
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Emily M Huntsman
- Meyer Cancer Center and Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Tomer M Yaron-Barir
- Meyer Cancer Center and Department of Medicine, Weill Cornell Medicine, New York, New York, USA; Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Jared L Johnson
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA; Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Kiran Kurmi
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Lewis C Cantley
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA; Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Alison E Ringel
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA; Ragon Institute of Mass General, MIT, and Harvard, Cambridge, Massachusetts, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA; Koch Institute for Integrative Cancer Research, Cambridge, Massachusetts, USA.
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA.
| | - Marcia C Haigis
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
31
|
Ruan ML, Ni WX, Chu JCH, Lam TL, Law KC, Zhang Y, Yang G, He Y, Zhang C, Fung YME, Liu T, Huang T, Lok CN, Chan SLF, Che CM. Iridium(III) carbene complexes as potent girdin inhibitors against metastatic cancers. Proc Natl Acad Sci U S A 2024; 121:e2316615121. [PMID: 38861602 PMCID: PMC11194514 DOI: 10.1073/pnas.2316615121] [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: 10/06/2023] [Accepted: 04/27/2024] [Indexed: 06/13/2024] Open
Abstract
Many cancer-driving protein targets remain undruggable due to a lack of binding molecular scaffolds. In this regard, octahedral metal complexes with unique and versatile three-dimensional structures have rarely been explored as inhibitors of undruggable protein targets. Here, we describe antitumor iridium(III) pyridinium-N-heterocyclic carbene complex 1a, which profoundly reduces the viability of lung and breast cancer cells as well as cancer patient-derived organoids at low micromolar concentrations. Compound 1a effectively inhibits the growth of non-small-cell lung cancer and triple-negative breast cancer xenograft tumors, impedes the metastatic spread of breast cancer cells, and can be modified into an antibody-drug conjugate payload to achieve precise tumor delivery in mice. Identified by thermal proteome profiling, an important molecular target of 1a in cellulo is Girdin, a multifunctional adaptor protein that is overexpressed in cancer cells and unequivocally serves as a signaling hub for multiple pivotal oncogenic pathways. However, specific small-molecule inhibitors of Girdin have not yet been developed. Notably, 1a exhibits high binding affinity to Girdin with a Kd of 1.3 μM and targets the Girdin-linked EGFR/AKT/mTOR/STAT3 cancer-driving pathway, inhibiting cancer cell proliferation and metastatic activity. Our study reveals a potent Girdin-targeting anticancer compound and demonstrates that octahedral metal complexes constitute an untapped library of small-molecule inhibitors that can fit into the ligand-binding pockets of key oncoproteins.
Collapse
Affiliation(s)
- Mei-Ling Ruan
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Hong Kong Science Park, Shatin, Hong Kong, China
- State Key Laboratory of Synthetic Chemistry and Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong, China
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan430079, China
| | - Wen-Xiu Ni
- Department of Medicinal Chemistry, Shantou University Medical College, Shantou515041, Guangdong, China
- Chemistry and Chemical Engineering of Guangdong Laboratory, Shantou515041, Guangdong, China
| | - Jacky C. H. Chu
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Hong Kong Science Park, Shatin, Hong Kong, China
- State Key Laboratory of Synthetic Chemistry and Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Tsz-Lung Lam
- State Key Laboratory of Synthetic Chemistry and Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Kwok-Chung Law
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Hong Kong Science Park, Shatin, Hong Kong, China
- State Key Laboratory of Synthetic Chemistry and Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Yiwei Zhang
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Hong Kong Science Park, Shatin, Hong Kong, China
- State Key Laboratory of Synthetic Chemistry and Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Guanya Yang
- AI And Life Sciences Institute (Hong Kong) Limited, Hong Kong Science Park, Shatin, Hong Kong, China
| | - Ying He
- AI And Life Sciences Institute (Hong Kong) Limited, Hong Kong Science Park, Shatin, Hong Kong, China
| | - Chunlei Zhang
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Hong Kong Science Park, Shatin, Hong Kong, China
- State Key Laboratory of Synthetic Chemistry and Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Yi Man Eva Fung
- State Key Laboratory of Synthetic Chemistry and Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Tao Liu
- Department of Medicinal Chemistry, Shantou University Medical College, Shantou515041, Guangdong, China
- Chemistry and Chemical Engineering of Guangdong Laboratory, Shantou515041, Guangdong, China
| | - Tao Huang
- Department of Medicinal Chemistry, Shantou University Medical College, Shantou515041, Guangdong, China
- Chemistry and Chemical Engineering of Guangdong Laboratory, Shantou515041, Guangdong, China
| | - Chun-Nam Lok
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Hong Kong Science Park, Shatin, Hong Kong, China
- State Key Laboratory of Synthetic Chemistry and Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Sharon Lai-Fung Chan
- Department of Applied Biology and Chemical Biology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | - Chi-Ming Che
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Hong Kong Science Park, Shatin, Hong Kong, China
- State Key Laboratory of Synthetic Chemistry and Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| |
Collapse
|
32
|
Feng H, Liu G, Li L, Ren X, Jiang Y, Hou W, Liu R, Liu K, Liu H, Huang H. Quantitative Proteomics Reveal the Role of Matrine in Regulating Lipid Metabolism. ACS OMEGA 2024; 9:24308-24320. [PMID: 38882153 PMCID: PMC11170650 DOI: 10.1021/acsomega.3c09983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 04/11/2024] [Accepted: 04/23/2024] [Indexed: 06/18/2024]
Abstract
Hyperlipidemia (HLP) is a prevalent systemic metabolic disorder characterized by disrupted lipid metabolism. Statin drugs have long been the primary choice for managing lipid levels, but intolerance issues have prompted the search for alternative treatments. Matrine, a compound derived from the traditional Chinese medicine Kushen, exhibits anti-inflammatory and lipid-lowering properties. Nevertheless, the mechanism by which matrine modulates lipid metabolism remains poorly understood. Here, we investigated the molecular mechanisms underlying matrine's regulation of lipid metabolism. Employing quantitative proteomics, we discovered that matrine increases the expression of LDL receptor (LDLR) in HepG2 and A549 cells, with subsequent experiments validating its role in enhancing LDL uptake. Notably, in hyperlipidemic hamsters, matrine effectively lowered lipid levels without affecting body weight, which highlights LDLR as a critical target for matrine's impact on HLP. Moreover, matrine's potential inhibitory effects on tumor cell LDL uptake hint at broader applications in cancer research. Additionally, thermal proteome profiling analysis identified lipid metabolism-related proteins that may interact with matrine. Together, our study reveals matrine's capacity to upregulate LDLR expression and highlights its potential in treating HLP. These findings offer insights into matrine's mechanism of action and open new avenues for drug research and lipid metabolism regulation.
Collapse
Affiliation(s)
- Huixu Feng
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210203, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Guobin Liu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210203, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Luhan Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xuelian Ren
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yue Jiang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Mechanical Engineering and Automation, Northeastern University, Shenyang 110819, China
| | - Wanting Hou
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Ruilong Liu
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Kun Liu
- School of Mechanical Engineering and Automation, Northeastern University, Shenyang 110819, China
| | - Hong Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - He Huang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210203, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China
| |
Collapse
|
33
|
Stincone P, Naimi A, Saviola AJ, Reher R, Petras D. Decoding the molecular interplay in the central dogma: An overview of mass spectrometry-based methods to investigate protein-metabolite interactions. Proteomics 2024; 24:e2200533. [PMID: 37929699 DOI: 10.1002/pmic.202200533] [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: 07/07/2023] [Revised: 10/15/2023] [Accepted: 10/23/2023] [Indexed: 11/07/2023]
Abstract
With the emergence of next-generation nucleotide sequencing and mass spectrometry-based proteomics and metabolomics tools, we have comprehensive and scalable methods to analyze the genes, transcripts, proteins, and metabolites of a multitude of biological systems. Despite the fascinating new molecular insights at the genome, transcriptome, proteome and metabolome scale, we are still far from fully understanding cellular organization, cell cycles and biology at the molecular level. Significant advances in sensitivity and depth for both sequencing as well as mass spectrometry-based methods allow the analysis at the single cell and single molecule level. At the same time, new tools are emerging that enable the investigation of molecular interactions throughout the central dogma of molecular biology. In this review, we provide an overview of established and recently developed mass spectrometry-based tools to probe metabolite-protein interactions-from individual interaction pairs to interactions at the proteome-metabolome scale.
Collapse
Affiliation(s)
- Paolo Stincone
- University of Tuebingen, CMFI Cluster of Excellence, Interfaculty Institute of Microbiology and Infection Medicine, Tuebingen, Germany
- University of Tuebingen, Center for Plant Molecular Biology, Tuebingen, Germany
| | - Amira Naimi
- University of Marburg, Institute of Pharmaceutical Biology and Biotechnology, Marburg, Germany
| | | | - Raphael Reher
- University of Marburg, Institute of Pharmaceutical Biology and Biotechnology, Marburg, Germany
| | - Daniel Petras
- University of Tuebingen, CMFI Cluster of Excellence, Interfaculty Institute of Microbiology and Infection Medicine, Tuebingen, Germany
- University of California Riverside, Department of Biochemistry, Riverside, USA
| |
Collapse
|
34
|
Smith CF, Modahl CM, Ceja Galindo D, Larson KY, Maroney SP, Bahrabadi L, Brandehoff NP, Perry BW, McCabe MC, Petras D, Lomonte B, Calvete JJ, Castoe TA, Mackessy SP, Hansen KC, Saviola AJ. Assessing Target Specificity of the Small Molecule Inhibitor MARIMASTAT to Snake Venom Toxins: A Novel Application of Thermal Proteome Profiling. Mol Cell Proteomics 2024; 23:100779. [PMID: 38679388 PMCID: PMC11154231 DOI: 10.1016/j.mcpro.2024.100779] [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: 11/12/2023] [Revised: 04/09/2024] [Accepted: 04/25/2024] [Indexed: 05/01/2024] Open
Abstract
New treatments that circumvent the pitfalls of traditional antivenom therapies are critical to address the problem of snakebite globally. Numerous snake venom toxin inhibitors have shown promising cross-species neutralization of medically significant venom toxins in vivo and in vitro. The development of high-throughput approaches for the screening of such inhibitors could accelerate their identification, testing, and implementation and thus holds exciting potential for improving the treatments and outcomes of snakebite envenomation worldwide. Energetics-based proteomic approaches, including thermal proteome profiling and proteome integral solubility alteration (PISA) assays, represent "deep proteomics" methods for high throughput, proteome-wide identification of drug targets and ligands. In the following study, we apply thermal proteome profiling and PISA methods to characterize the interactions between venom toxin proteoforms in Crotalus atrox (Western Diamondback Rattlesnake) and the snake venom metalloprotease (SVMP) inhibitor marimastat. We investigate its venom proteome-wide effects and characterize its interactions with specific SVMP proteoforms, as well as its potential targeting of non-SVMP venom toxin families. We also compare the performance of PISA thermal window and soluble supernatant with insoluble precipitate using two inhibitor concentrations, providing the first demonstration of the utility of a sensitive high-throughput PISA-based approach to assess the direct targets of small molecule inhibitors for snake venom.
Collapse
Affiliation(s)
- Cara F Smith
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, Colorado, USA
| | - Cassandra M Modahl
- Centre for Snakebite Research and Interventions, Liverpool School of Tropical Medicine, Liverpool, UK
| | - David Ceja Galindo
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, Colorado, USA
| | - Keira Y Larson
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, Colorado, USA
| | - Sean P Maroney
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, Colorado, USA
| | - Lilyrose Bahrabadi
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, Colorado, USA
| | - Nicklaus P Brandehoff
- Rocky Mountain Poison and Drug Center, Denver Health and Hospital Authority, Denver, Colorado, USA
| | - Blair W Perry
- School of Biological Sciences, Washington State University, Pullman, Washington, USA
| | - Maxwell C McCabe
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, Colorado, USA
| | - Daniel Petras
- CMFI Cluster of Excellence, University of Tuebingen, Tuebingen, Germany; Department of Biochemistry, University of California Riverside, Riverside, California, USA
| | - Bruno Lomonte
- Instituto Clodomiro Picado, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
| | - Juan J Calvete
- Evolutionary and Translational Venomics Laboratory, Consejo Superior de Investigaciones Científicas (CSIC), Valencia, Spain
| | - Todd A Castoe
- Department of Biology, The University of Texas Arlington, Texas, USA
| | - Stephen P Mackessy
- Department of Biological Sciences, University of Northern Colorado, Greeley, Colorado, USA
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, Colorado, USA
| | - Anthony J Saviola
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, Colorado, USA.
| |
Collapse
|
35
|
Tan X, Han Y, Zhai S, Dong H, Zhang T, Zhang K. An Integrated Analytical Approach for Screening Functional Post-Translational Modification Sites in Metabolic Enzymes. ACS OMEGA 2024; 9:19003-19008. [PMID: 38708225 PMCID: PMC11064186 DOI: 10.1021/acsomega.3c09514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 04/07/2024] [Accepted: 04/10/2024] [Indexed: 05/07/2024]
Abstract
Post-translational modifications (PTMs) are pivotal in the orchestration of diverse physiological and pathological processes. Despite this, the identification of functional PTM sites within the vast amount of data remains challenging. Conventionally, those PTM sites are discerned through labor-intensive and time-consuming experiments. Here, we developed an integrated analytical approach for the identification of functional PTM sites on metabolic enzymes via a screening process. Through gene ontology (GO) analysis, we identified 269 enzymes with lysine 2-hydroxyisobutyrylation (Khib) from our proteomics data set of Escherichia coli. The first round of screening was performed based on the enzyme structures/predicted structures using the TM-score engineer, a tool designed to evaluate the impact of PTM on the protein structure. Subsequently, we examined the influence of Khib on the enzyme-substrate interactions through both static and dynamic analyses, molecular docking, and molecular dynamics simulation. Ultimately, we identified NfsB K181hib and ThiF K83hib as potential functional sites. This work has established a novel analytical approach for the identification of functional protein PTM sites, thereby contributing to the understanding of Khib functions.
Collapse
Affiliation(s)
- Xiaoxia Tan
- The
Province and Ministry Co-Sponsored Collaborative Innovation Center
for Medical Epigenetics, Key Laboratory of Immune Microenvironment
and Disease (Ministry of Education), Tianjin Key Laboratory of Medical
Epigenetics, Department of Biochemistry and Molecular Biology, School
of Basic Medical Sciences, Tianjin Medical
University, Tianjin 300070, China
| | - Yue Han
- The
Province and Ministry Co-Sponsored Collaborative Innovation Center
for Medical Epigenetics, Key Laboratory of Immune Microenvironment
and Disease (Ministry of Education), Tianjin Key Laboratory of Medical
Epigenetics, Department of Biochemistry and Molecular Biology, School
of Basic Medical Sciences, Tianjin Medical
University, Tianjin 300070, China
| | - Shengrui Zhai
- The
Province and Ministry Co-Sponsored Collaborative Innovation Center
for Medical Epigenetics, Key Laboratory of Immune Microenvironment
and Disease (Ministry of Education), Tianjin Key Laboratory of Medical
Epigenetics, Department of Biochemistry and Molecular Biology, School
of Basic Medical Sciences, Tianjin Medical
University, Tianjin 300070, China
| | - Hanyang Dong
- The
Province and Ministry Co-Sponsored Collaborative Innovation Center
for Medical Epigenetics, Key Laboratory of Immune Microenvironment
and Disease (Ministry of Education), Tianjin Key Laboratory of Medical
Epigenetics, Department of Biochemistry and Molecular Biology, School
of Basic Medical Sciences, Tianjin Medical
University, Tianjin 300070, China
| | - Tao Zhang
- School
of Biomedical Engineering, Tianjin Medical
University, Tianjin 300070, China
| | - Kai Zhang
- The
Province and Ministry Co-Sponsored Collaborative Innovation Center
for Medical Epigenetics, Key Laboratory of Immune Microenvironment
and Disease (Ministry of Education), Tianjin Key Laboratory of Medical
Epigenetics, Department of Biochemistry and Molecular Biology, School
of Basic Medical Sciences, Tianjin Medical
University, Tianjin 300070, China
| |
Collapse
|
36
|
Zhao H, Kumar P, Sobreira TJP, Smith M, Novick S, Johansson A, Luchniak A, Zhang A, Woollard KJ, Larsson N, Kawatkar A. Integrated Proteomics Characterization of NLRP3 Inflammasome Inhibitor MCC950 in Monocytic Cell Line Confirms Direct MCC950 Engagement with Endogenous NLRP3. ACS Chem Biol 2024; 19:962-972. [PMID: 38509779 DOI: 10.1021/acschembio.3c00777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
Inhibition of the NLRP3 inflammasome is a promising strategy for the development of new treatments for inflammatory diseases. MCC950 is a potent and selective small-molecule inhibitor of the NLRP3 pathway and has been validated in numerous species and disease models. Although the capacity of MCC950 to block NLRP3 signaling is well-established, it is still critical to identify the mechanism of action and molecular targets of MCC950 to inform and derisk drug development. Quantitative proteomics performed in disease-relevant systems provides a powerful method to study both direct and indirect pharmacological responses to small molecules to elucidate the mechanism of action and confirm target engagement. A comprehensive target deconvolution campaign requires the use of complementary chemical biology techniques. Here we applied two orthogonal chemical biology techniques: compressed Cellular Thermal Shift Assay (CETSA) and photoaffinity labeling chemoproteomics, performed under biologically relevant conditions with LPS-primed THP-1 cells, thereby deconvoluting, for the first time, the molecular targets of MCC950 using chemical biology techniques. In-cell chemoproteomics with inlysate CETSA confirmed the suspected mechanism as the disruption of inflammasome formation via NLRP3. Further cCETSA (c indicates compressed) in live cells mapped the stabilization of NLRP3 inflammasome pathway proteins, highlighting modulation of the targeted pathway. This is the first evidence of direct MCC950 engagement with endogenous NLRP3 in a human macrophage cellular system using discovery proteomics chemical biology techniques, providing critical information for inflammasome studies.
Collapse
Affiliation(s)
- Heng Zhao
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, 02451 Waltham, Massachusetts, United States
| | - Praveen Kumar
- Quantitative Biology, Discovery Sciences, R&D, AstraZeneca, 02451 Waltham, Massachusetts, United States
| | | | - Mackenzie Smith
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, 02451 Waltham, Massachusetts, United States
| | - Steven Novick
- Quantitative Biology, Discovery Sciences, R&D, AstraZeneca, 02451 Waltham, Massachusetts, United States
| | - Anders Johansson
- Medicinal Chemistry, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, 43183 Mölndal, Sweden
| | - Anna Luchniak
- Mechanistic and Structural Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, 43183 Mölndal, Sweden
| | - Andrew Zhang
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, 02451 Waltham, Massachusetts, United States
| | - Kevin J Woollard
- Bioscience Renal, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, CB2 OAA Cambridge, U.K
| | - Niklas Larsson
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, 43183 Mölndal, Sweden
| | - Aarti Kawatkar
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, 02451 Waltham, Massachusetts, United States
| |
Collapse
|
37
|
Mansfield CR, Quan B, Chirgwin ME, Eduful B, Hughes PF, Neveu G, Sylvester K, Ryan DH, Kafsack BFC, Haystead TAJ, Leahy JW, Fitzgerald MC, Derbyshire ER. Selective targeting of Plasmodium falciparum Hsp90 disrupts the 26S proteasome. Cell Chem Biol 2024; 31:729-742.e13. [PMID: 38492573 PMCID: PMC11031320 DOI: 10.1016/j.chembiol.2024.02.008] [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: 03/01/2023] [Revised: 11/09/2023] [Accepted: 02/22/2024] [Indexed: 03/18/2024]
Abstract
The molecular chaperone heat shock protein 90 (Hsp90) has an essential but largely undefined role in maintaining proteostasis in Plasmodium falciparum, the most lethal malaria parasite. Herein, we identify BX-2819 and XL888 as potent P. falciparum (Pf)Hsp90 inhibitors. Derivatization of XL888's scaffold led to the development of Tropane 1, as a PfHsp90-selective binder with nanomolar affinity. Hsp90 inhibitors exhibit anti-Plasmodium activity against the liver, asexual blood, and early gametocyte life stages. Thermal proteome profiling was implemented to assess PfHsp90-dependent proteome stability, and the proteasome-the main site of cellular protein recycling-was enriched among proteins with perturbed stability upon PfHsp90 inhibition. Subsequent biochemical and cellular studies suggest that PfHsp90 directly promotes proteasome hydrolysis by chaperoning the active 26S complex. These findings expand our knowledge of the PfHsp90-dependent proteome and protein quality control mechanisms in these pathogenic parasites, as well as further characterize this chaperone as a potential antimalarial drug target.
Collapse
Affiliation(s)
- Christopher R Mansfield
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA
| | - Baiyi Quan
- Department of Chemistry, Duke University, Durham, NC, USA
| | | | - Benjamin Eduful
- Department of Chemistry, University of South Florida, Tampa, FL, USA
| | - Philip F Hughes
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - Gaëlle Neveu
- Department of Microbiology & Immunology, Weill Cornell Medicine, New York, NY, USA
| | - Kayla Sylvester
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA
| | - Daniel H Ryan
- Department of Chemistry, Duke University, Durham, NC, USA
| | - Björn F C Kafsack
- Department of Microbiology & Immunology, Weill Cornell Medicine, New York, NY, USA
| | - Timothy A J Haystead
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - James W Leahy
- Department of Chemistry, University of South Florida, Tampa, FL, USA; Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA; Center for Drug Discovery and Innovation, University of South Florida, Tampa, FL, USA
| | | | - Emily R Derbyshire
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA; Department of Chemistry, Duke University, Durham, NC, USA.
| |
Collapse
|
38
|
George AL, Dueñas ME, Marín-Rubio JL, Trost M. Stability-based approaches in chemoproteomics. Expert Rev Mol Med 2024; 26:e6. [PMID: 38604802 PMCID: PMC11062140 DOI: 10.1017/erm.2024.6] [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: 08/29/2023] [Revised: 01/17/2024] [Accepted: 02/22/2024] [Indexed: 04/13/2024]
Abstract
Target deconvolution can help understand how compounds exert therapeutic effects and can accelerate drug discovery by helping optimise safety and efficacy, revealing mechanisms of action, anticipate off-target effects and identifying opportunities for therapeutic expansion. Chemoproteomics, a combination of chemical biology with mass spectrometry has transformed target deconvolution. This review discusses modification-free chemoproteomic approaches that leverage the change in protein thermodynamics induced by small molecule ligand binding. Unlike modification-based methods relying on enriching specific protein targets, these approaches offer proteome-wide evaluations, driven by advancements in mass spectrometry sensitivity, increasing proteome coverage and quantitation methods. Advances in methods based on denaturation/precipitation by thermal or chemical denaturation, or by protease degradation are evaluated, emphasising the evolving landscape of chemoproteomics and its potential impact on future drug-development strategies.
Collapse
Affiliation(s)
- Amy L. George
- Laboratory for Biomedical Mass Spectrometry, Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, NE2 4HH, UK
| | - Maria Emilia Dueñas
- Laboratory for Biomedical Mass Spectrometry, Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, NE2 4HH, UK
| | - José Luis Marín-Rubio
- Laboratory for Biomedical Mass Spectrometry, Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, NE2 4HH, UK
| | - Matthias Trost
- Laboratory for Biomedical Mass Spectrometry, Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, NE2 4HH, UK
| |
Collapse
|
39
|
Shanley HT, Taki AC, Nguyen N, Wang T, Byrne JJ, Ang CS, Leeming MG, Nie S, Williamson N, Zheng Y, Young ND, Korhonen PK, Hofmann A, Chang BCH, Wells TNC, Häberli C, Keiser J, Jabbar A, Sleebs BE, Gasser RB. Structure-activity relationship and target investigation of 2-aryl quinolines with nematocidal activity. Int J Parasitol Drugs Drug Resist 2024; 24:100522. [PMID: 38295619 PMCID: PMC10845918 DOI: 10.1016/j.ijpddr.2024.100522] [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: 11/02/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 02/02/2024]
Abstract
Within the context of our anthelmintic discovery program, we recently identified and evaluated a quinoline derivative, called ABX464 or obefazimod, as a nematocidal candidate; synthesised a series of analogues which were assessed for activity against the free-living nematode Caenorhabditis elegans; and predicted compound-target relationships by thermal proteome profiling (TPP) and in silico docking. Here, we logically extended this work and critically evaluated the anthelmintic activity of ABX464 analogues on Haemonchus contortus (barber's pole worm) - a highly pathogenic nematode of ruminant livestock. First, we tested a series of 44 analogues on H. contortus (larvae and adults) to investigate the nematocidal pharmacophore of ABX464, and identified one compound with greater potency than the parent compound and showed moderate activity against a select number of other parasitic nematodes (including Ancylostoma, Heligmosomoides and Strongyloides species). Using TPP and in silico modelling studies, we predicted protein HCON_00074590 (a predicted aldo-keto reductase) as a target candidate for ABX464 in H. contortus. Future work aims to optimise this compound as a nematocidal candidate and investigate its pharmacokinetic properties. Overall, this study presents a first step toward the development of a new nematocide.
Collapse
Affiliation(s)
- Harrison T Shanley
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria, 3010, Australia; Chemical Biology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia
| | - Aya C Taki
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Nghi Nguyen
- Chemical Biology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia
| | - Tao Wang
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Joseph J Byrne
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Ching-Seng Ang
- Melbourne Mass Spectrometry and Proteomics Facility, The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Michael G Leeming
- Melbourne Mass Spectrometry and Proteomics Facility, The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Shuai Nie
- Melbourne Mass Spectrometry and Proteomics Facility, The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Nicholas Williamson
- Melbourne Mass Spectrometry and Proteomics Facility, The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Yuanting Zheng
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Neil D Young
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Pasi K Korhonen
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Andreas Hofmann
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria, 3010, Australia; National Reference Centre for Authentic Food, Max Rubner-Institut, 95326, Kulmbach, Germany
| | - Bill C H Chang
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Tim N C Wells
- Medicines for Malaria Venture (MMV), 1215, Geneva, Switzerland
| | - Cécile Häberli
- Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, 4123, Allschwil, Switzerland; University of Basel, 4001, Basel, Switzerland
| | - Jennifer Keiser
- Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, 4123, Allschwil, Switzerland; University of Basel, 4001, Basel, Switzerland
| | - Abdul Jabbar
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Brad E Sleebs
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria, 3010, Australia; Chemical Biology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia.
| | - Robin B Gasser
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria, 3010, Australia.
| |
Collapse
|
40
|
Burtscher ML, Gade S, Garrido-Rodriguez M, Rutkowska A, Werner T, Eberl HC, Petretich M, Knopf N, Zirngibl K, Grandi P, Bergamini G, Bantscheff M, Fälth-Savitski M, Saez-Rodriguez J. Network integration of thermal proteome profiling with multi-omics data decodes PARP inhibition. Mol Syst Biol 2024; 20:458-474. [PMID: 38454145 PMCID: PMC10987601 DOI: 10.1038/s44320-024-00025-w] [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: 01/17/2024] [Revised: 02/13/2024] [Accepted: 02/20/2024] [Indexed: 03/09/2024] Open
Abstract
Complex disease phenotypes often span multiple molecular processes. Functional characterization of these processes can shed light on disease mechanisms and drug effects. Thermal Proteome Profiling (TPP) is a mass-spectrometry (MS) based technique assessing changes in thermal protein stability that can serve as proxies of functional protein changes. These unique insights of TPP can complement those obtained by other omics technologies. Here, we show how TPP can be integrated with phosphoproteomics and transcriptomics in a network-based approach using COSMOS, a multi-omics integration framework, to provide an integrated view of transcription factors, kinases and proteins with altered thermal stability. This allowed us to recover consequences of Poly (ADP-ribose) polymerase (PARP) inhibition in ovarian cancer cells on cell cycle and DNA damage response as well as interferon and hippo signaling. We found that TPP offers a complementary perspective to other omics data modalities, and that its integration allowed us to obtain a more complete molecular overview of PARP inhibition. We anticipate that this strategy can be used to integrate functional proteomics with other omics to study molecular processes.
Collapse
Affiliation(s)
- Mira L Burtscher
- Heidelberg University, Faculty of Medicine, Heidelberg University Hospital, Institute for Computational Biomedicine, Heidelberg, Germany
- Cellzome, a GSK company, Heidelberg, Germany
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | | | - Martin Garrido-Rodriguez
- Heidelberg University, Faculty of Medicine, Heidelberg University Hospital, Institute for Computational Biomedicine, Heidelberg, Germany
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | | | | | | | | | | | - Katharina Zirngibl
- Heidelberg University, Faculty of Medicine, Heidelberg University Hospital, Institute for Computational Biomedicine, Heidelberg, Germany
- Cellzome, a GSK company, Heidelberg, Germany
| | | | | | | | | | - Julio Saez-Rodriguez
- Heidelberg University, Faculty of Medicine, Heidelberg University Hospital, Institute for Computational Biomedicine, Heidelberg, Germany.
| |
Collapse
|
41
|
Reed TJ, Tyl MD, Tadych A, Troyanskaya OG, Cristea IM. Tapioca: a platform for predicting de novo protein-protein interactions in dynamic contexts. Nat Methods 2024; 21:488-500. [PMID: 38361019 PMCID: PMC11249048 DOI: 10.1038/s41592-024-02179-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 01/12/2024] [Indexed: 02/17/2024]
Abstract
Protein-protein interactions (PPIs) drive cellular processes and responses to environmental cues, reflecting the cellular state. Here we develop Tapioca, an ensemble machine learning framework for studying global PPIs in dynamic contexts. Tapioca predicts de novo interactions by integrating mass spectrometry interactome data from thermal/ion denaturation or cofractionation workflows with protein properties and tissue-specific functional networks. Focusing on the thermal proximity coaggregation method, we improved the experimental workflow. Finely tuned thermal denaturation afforded increased throughput, while cell lysis optimization enhanced protein detection from different subcellular compartments. The Tapioca workflow was next leveraged to investigate viral infection dynamics. Temporal PPIs were characterized during the reactivation from latency of the oncogenic Kaposi's sarcoma-associated herpesvirus. Together with functional assays, NUCKS was identified as a proviral hub protein, and a broader role was uncovered by integrating PPI networks from alpha- and betaherpesvirus infections. Altogether, Tapioca provides a web-accessible platform for predicting PPIs in dynamic contexts.
Collapse
Affiliation(s)
- Tavis J Reed
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Carl Icahn Laboratory, Princeton, NJ, USA
- Department of Computer Science, Princeton University, Princeton, NJ, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Matthew D Tyl
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Alicja Tadych
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Carl Icahn Laboratory, Princeton, NJ, USA
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | - Olga G Troyanskaya
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Carl Icahn Laboratory, Princeton, NJ, USA.
- Department of Computer Science, Princeton University, Princeton, NJ, USA.
- Flatiron Institute, Simons Foundation, New York City, NY, USA.
| | - Ileana M Cristea
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
42
|
Patel S, Karlsson M, Klahn JT, Gambino F, Costa H, McGuire KA, Baumgartner CK, Williams J, Sandoz S, Kath JE. Quantitative target engagement of RIPK1 in human whole blood via the cellular thermal shift assay for potential pre-clinical and clinical applications. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2024; 29:100135. [PMID: 38101572 DOI: 10.1016/j.slasd.2023.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/29/2023] [Accepted: 12/12/2023] [Indexed: 12/17/2023]
Abstract
The cellular thermal shift assay (CETSA®) is a target engagement method widely used for preclinical characterization of small molecule compounds. CETSA® has been used for semi-quantitative readouts in whole blood with PBMC isolation, and quantitative, plate-based readouts using cell lines. However, there has been no quantitative evaluation of CETSA® in unprocessed human whole blood, which is preferred for clinical applications. Here we report two separate assay formats - Alpha CETSA® and MSD CETSA® - that require less than 100 μL of whole blood per sample without PBMC isolation. We chose RIPK1 as a proof-of-concept target and, by measuring engagement of seven different inhibitors, demonstrate high assay sensitivity and robustness. These quantitative CETSA® platforms enable possible applications in preclinical pharmacokinetic-pharmacodynamic studies, and direct target engagement with small molecules in clinical trials.
Collapse
|
43
|
Figueroa-Navedo AM, Ivanov AR. Experimental and data analysis advances in thermal proteome profiling. CELL REPORTS METHODS 2024; 4:100717. [PMID: 38412830 PMCID: PMC10921035 DOI: 10.1016/j.crmeth.2024.100717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/17/2023] [Accepted: 02/05/2024] [Indexed: 02/29/2024]
Abstract
Method development for mass spectrometry (MS)-based thermal shift proteomic assays have advanced to probe small molecules with known and unknown protein-ligand interaction mechanisms and specificity, which is predominantly used in characterization of drug-protein interactions. In the discovery of target and off-target protein-ligand interactions, a thorough investigation of method development and their impact on the sensitivity and accuracy of protein-small molecule and protein-protein interactions is warranted. In this review, we discuss areas of improvement at each stage of thermal proteome profiling data analysis that includes processing of MS-based data, method development, and their effect on the overall quality of thermal proteome profiles. We also overview the optimization of experimental strategies and prioritization of an increased number of independent biological replicates over the number of evaluated temperatures.
Collapse
Affiliation(s)
- Amanda M Figueroa-Navedo
- Barnett Institute of Chemical and Biological Analysis, Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA 02115, USA
| | - Alexander R Ivanov
- Barnett Institute of Chemical and Biological Analysis, Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA 02115, USA.
| |
Collapse
|
44
|
Bailey MA, Martyr JG, Hargrove AE, Fitzgerald MC. Stability-Based Proteomics for Investigation of Structured RNA-Protein Interactions. Anal Chem 2024:10.1021/acs.analchem.3c04978. [PMID: 38341805 PMCID: PMC11316846 DOI: 10.1021/acs.analchem.3c04978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2024]
Abstract
RNA-protein interactions are essential to RNA function throughout biology. Identifying the protein interactions associated with a specific RNA, however, is currently hindered by the need for RNA labeling or costly tiling-based approaches. Conventional strategies, which commonly rely on affinity pull-down approaches, are also skewed to the detection of high affinity interactions and frequently miss weaker interactions that may be biologically important. Reported here is the first adaptation of stability-based mass spectrometry methods for the global analysis of RNA-protein interactions. The stability of proteins from rates of oxidation (SPROX) and thermal protein profiling (TPP) methods are used to identify the protein targets of three RNA ligands, the MALAT1 triple helix (TH), a viral stem loop (SL), and an unstructured RNA (PolyU), in LNCaP nuclear lysate. The 315 protein hits with RNA-induced conformational and stability changes detected by TPP and/or SPROX were enriched in previously annotated RNA-binding proteins and included new proteins for hypothesis generation. Also demonstrated are the orthogonality of the SPROX and TPP approaches and the utility of the domain-specific information available with SPROX. This work establishes a novel platform for the global discovery and interrogation of RNA-protein interactions that is generalizable to numerous biological contexts and RNA targets.
Collapse
Affiliation(s)
- Morgan A Bailey
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Justin G Martyr
- Department of Biochemistry, Duke University School of Medicine, Durham, North Carolina 27710, United States
| | - Amanda E Hargrove
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
- Department of Biochemistry, Duke University School of Medicine, Durham, North Carolina 27710, United States
| | - Michael C Fitzgerald
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
- Department of Biochemistry, Duke University School of Medicine, Durham, North Carolina 27710, United States
| |
Collapse
|
45
|
Yin K, Wu R. Systematic Investigation of Dose-Dependent Protein Thermal Stability Changes to Uncover the Mechanisms of the Pleiotropic Effects of Metformin. ACS Pharmacol Transl Sci 2024; 7:467-477. [PMID: 38357277 PMCID: PMC10863438 DOI: 10.1021/acsptsci.3c00298] [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: 10/24/2023] [Revised: 12/16/2023] [Accepted: 12/22/2023] [Indexed: 02/16/2024]
Abstract
Metformin is a widely used drug to treat type II diabetes. Beyond lowering blood sugar, it has been reported to have pleiotropic effects such as suppressing cancer growth and attenuating cell oxidative stress and inflammation. However, the underlying mechanisms of these effects remain to be explored. Here, we systematically study the thermal stability changes of proteins in liver cells (HepG2) induced by a wide dosage range of metformin by using the proteome integral solubility alteration (PISA) assay. The current results demonstrate that, besides the most accepted target of metformin (complex I), low concentrations of metformin (such as 0.2 μM) stabilize the complex IV subunits, suggesting its important role in the sugar-lowering effect. Low-dose metformin also results in stability alterations of ribosomal proteins, correlating with its inhibitive effect on cell proliferation. We further find that low-concentration metformin impacts mitochondrial cargo and vesicle transport, while high-concentration metformin affects cell redox responses and cell membrane protein sorting. This study provides mechanistic insights into the molecular mechanisms of lowering blood sugar and the pleiotropic effects of metformin.
Collapse
Affiliation(s)
- Kejun Yin
- School of Chemistry and Biochemistry
and the Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Ronghu Wu
- School of Chemistry and Biochemistry
and the Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| |
Collapse
|
46
|
Gooran N, Kopra K. Fluorescence-Based Protein Stability Monitoring-A Review. Int J Mol Sci 2024; 25:1764. [PMID: 38339045 PMCID: PMC10855643 DOI: 10.3390/ijms25031764] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
Proteins are large biomolecules with a specific structure that is composed of one or more long amino acid chains. Correct protein structures are directly linked to their correct function, and many environmental factors can have either positive or negative effects on this structure. Thus, there is a clear need for methods enabling the study of proteins, their correct folding, and components affecting protein stability. There is a significant number of label-free methods to study protein stability. In this review, we provide a general overview of these methods, but the main focus is on fluorescence-based low-instrument and -expertise-demand techniques. Different aspects related to thermal shift assays (TSAs), also called differential scanning fluorimetry (DSF) or ThermoFluor, are introduced and compared to isothermal chemical denaturation (ICD). Finally, we discuss the challenges and comparative aspects related to these methods, as well as future opportunities and assay development directions.
Collapse
Affiliation(s)
| | - Kari Kopra
- Department of Chemistry, University of Turku, Henrikinkatu 2, 20500 Turku, Finland;
| |
Collapse
|
47
|
Locke TM, Fields R, Gizinski H, Otto GM, Shechner DM, Berg MD, Villen J, Sancak Y, Schweppe D. High-Throughput Identification of Calcium Regulated Proteins Across Diverse Proteomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.18.575273. [PMID: 38293219 PMCID: PMC10827220 DOI: 10.1101/2024.01.18.575273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Calcium ions play important roles in nearly every biological process, yet whole-proteome analysis of calcium effectors has been hindered by lack of high-throughput, unbiased, and quantitative methods to identify proteins-calcium engagement. To address this, we adapted protein thermostability assays in the budding yeast, human cells, and mouse mitochondria. Based on calcium-dependent thermostability, we identified 2884 putative calcium-regulated proteins across human, mouse, and yeast proteomes. These data revealed calcium engagement of novel signaling hubs and cellular processes, including metabolic enzymes and the spliceosome. Cross-species comparison of calcium-protein engagement and mutagenesis experiments identified residue-specific cation engagement, even within well-known EF-hand domains. Additionally, we found that the dienoyl-CoA reductase DECR1 binds calcium at physiologically-relevant concentrations with substrate-specific affinity, suggesting direct calcium regulation of mitochondrial fatty acid oxidation. These unbiased, proteomic analyses of calcium effectors establish a key resource to dissect cation engagement and its mechanistic effects across multiple species and diverse biological processes.
Collapse
Affiliation(s)
- Timothy M Locke
- Department of Pharmacology, University of Washington, Seattle, Washington 98195, United States
| | - Rose Fields
- Department of Genome Sciences, University of Washington, Seattle, Washington 98195, United States
| | - Hayden Gizinski
- Department of Pharmacology, University of Washington, Seattle, Washington 98195, United States
| | - George M Otto
- Department of Pharmacology, University of Washington, Seattle, Washington 98195, United States
| | - David M Shechner
- Department of Pharmacology, University of Washington, Seattle, Washington 98195, United States
| | - Matthew D Berg
- Department of Genome Sciences, University of Washington, Seattle, Washington 98195, United States
| | - Judit Villen
- Department of Genome Sciences, University of Washington, Seattle, Washington 98195, United States
| | - Yasemin Sancak
- Department of Pharmacology, University of Washington, Seattle, Washington 98195, United States
| | - Devin Schweppe
- Department of Genome Sciences, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
48
|
Shanley HT, Taki AC, Nguyen N, Wang T, Byrne JJ, Ang CS, Leeming MG, Nie S, Williamson N, Zheng Y, Young ND, Korhonen PK, Hofmann A, Wells TNC, Jabbar A, Sleebs BE, Gasser RB. Structure activity relationship and target prediction for ABX464 analogues in Caenorhabditis elegans. Bioorg Med Chem 2024; 98:117540. [PMID: 38134663 DOI: 10.1016/j.bmc.2023.117540] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 11/20/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023]
Abstract
Global challenges with treatment failures and/or widespread resistance in parasitic worms against commercially available anthelmintics lend impetus to the development of new anthelmintics with novel mechanism(s) of action. The free-living nematode Caenorhabditis elegans is an important model organism used for drug discovery, including the screening and structure-activity investigation of new compounds, and target deconvolution. Previously, we conducted a whole-organism phenotypic screen of the 'Pandemic Response Box' (from Medicines for Malaria Venture, MMV) and identified a hit compound, called ABX464, with activity against C. elegans and a related, parasitic nematode, Haemonchus contortus. Here, we tested a series of 44 synthesized analogues to explore the pharmacophore of activity on C. elegans and revealed five compounds whose potency was similar or greater than that of ABX464, but which were not toxic to human hepatoma (HepG2) cells. Subsequently, we employed thermal proteome profiling (TPP), protein structure prediction and an in silico-docking algorithm to predict ABX464-target candidates. Taken together, the findings from this study contribute significantly to the early-stage drug discovery of a new nematocide based on ABX464. Future work is aimed at validating the ABX464-protein interactions identified here, and at assessing ABX464 and associated analogues against a panel of parasitic nematodes, towards developing a new anthelmintic with a mechanism of action that is distinct from any of the compounds currently-available commercially.
Collapse
Affiliation(s)
- Harrison T Shanley
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia; Chemical Biology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Aya C Taki
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Nghi Nguyen
- Chemical Biology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Tao Wang
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Joseph J Byrne
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Ching-Seng Ang
- Melbourne Mass Spectrometry and Proteomics Facility, The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Michael G Leeming
- Melbourne Mass Spectrometry and Proteomics Facility, The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Shuai Nie
- Melbourne Mass Spectrometry and Proteomics Facility, The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Nicholas Williamson
- Melbourne Mass Spectrometry and Proteomics Facility, The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yuanting Zheng
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Neil D Young
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Pasi K Korhonen
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Andreas Hofmann
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia; National Reference Centre for Authentic Food, Max Rubner-Institut, 95326 Kulmbach, Germany
| | - Tim N C Wells
- Medicines for Malaria Venture (MMV), 1215 Geneva, Switzerland
| | - Abdul Jabbar
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Brad E Sleebs
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia; Chemical Biology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.
| | - Robin B Gasser
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia.
| |
Collapse
|
49
|
Fatemi N, Karimpour M, Bahrami H, Zali MR, Chaleshi V, Riccio A, Nazemalhosseini-Mojarad E, Totonchi M. Current trends and future prospects of drug repositioning in gastrointestinal oncology. Front Pharmacol 2024; 14:1329244. [PMID: 38239190 PMCID: PMC10794567 DOI: 10.3389/fphar.2023.1329244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 12/11/2023] [Indexed: 01/22/2024] Open
Abstract
Gastrointestinal (GI) cancers comprise a significant number of cancer cases worldwide and contribute to a high percentage of cancer-related deaths. To improve survival rates of GI cancer patients, it is important to find and implement more effective therapeutic strategies with better prognoses and fewer side effects. The development of new drugs can be a lengthy and expensive process, often involving clinical trials that may fail in the early stages. One strategy to address these challenges is drug repurposing (DR). Drug repurposing is a developmental strategy that involves using existing drugs approved for other diseases and leveraging their safety and pharmacological data to explore their potential use in treating different diseases. In this paper, we outline the existing therapeutic strategies and challenges associated with GI cancers and explore DR as a promising alternative approach. We have presented an extensive review of different DR methodologies, research efforts and examples of repurposed drugs within various GI cancer types, such as colorectal, pancreatic and liver cancers. Our aim is to provide a comprehensive overview of employing the DR approach in GI cancers to inform future research endeavors and clinical trials in this field.
Collapse
Affiliation(s)
- Nayeralsadat Fatemi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mina Karimpour
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hoda Bahrami
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Vahid Chaleshi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Andrea Riccio
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies (DiSTABiF), Università degli Studi della Campania “Luigi Vanvitelli”, Caserta, Italy
- Institute of Genetics and Biophysics (IGB) “Adriano Buzzati-Traverso”, Consiglio Nazionale delle Ricerche (CNR), Naples, Italy
| | - Ehsan Nazemalhosseini-Mojarad
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Totonchi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies (DiSTABiF), Università degli Studi della Campania “Luigi Vanvitelli”, Caserta, Italy
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| |
Collapse
|
50
|
Luan H, Li X, Zhang W, Luan T. Thermal proteome profiling unveils protein targets of deoxycholic acid in living neuronal cells. ADVANCED BIOTECHNOLOGY 2023; 1:7. [PMID: 39883374 PMCID: PMC11727579 DOI: 10.1007/s44307-023-00007-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 11/26/2023] [Accepted: 11/28/2023] [Indexed: 01/31/2025]
Abstract
Bile acids, synthesized in the liver and modified by the gut microbiota, play vital roles in various physiological processes. The dysregulation of bile acids has been extensively documented in patients with neurodegenerative diseases. However, limited attention has been given to the protein targets associated with microbiota-derived bile acids in neurological diseases. To address this knowledge gap, we conducted comprehensive thermal proteomic analyses to elucidate and comprehend the protein targets affected by microbiota-derived bile acids. Our investigation identified sixty-five unique proteins in SH-SY5Y neuronal cells as potential targets of deoxycholic acid (DCA), a primary component of the bile acid pool originating from the gut microbiota. Notably, Nicastrin and Casein kinase 1 epsilon stood out among these proteins. We found that DCA, through its interaction with the Nicastrin subunit of γ-secretase, significantly contributed to the formation of amyloid beta, a key hallmark in the pathology of neurodegenerative diseases. In summary, our findings provide crucial insights into the intricate interplay between microbiota-derived bile acids and the pathogenesis of neurodegenerative diseases, thereby shedding light on potential therapeutic targets for neurodegenerative diseases.
Collapse
Affiliation(s)
- Hemi Luan
- Department of Biomedical Engineering, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, China.
| | - Xuan Li
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Wenyong Zhang
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Tiangang Luan
- Institute of Environmental and Ecological Engineering, Guangdong University of Technology, Guangzhou, 510006, China.
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, 529020, China.
- Sate Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| |
Collapse
|