1
|
Horbach N, Kalinka M, Ćwilichowska-Puślecka N, Al Mamun A, Mikołajczyk-Martinez A, Turk B, Snipas SJ, Kasperkiewicz P, Groborz KM, Poręba M. Visualization of calpain-1 activation during cell death and its role in GSDMD cleavage using chemical probes. Cell Chem Biol 2025; 32:603-619.e7. [PMID: 40157358 DOI: 10.1016/j.chembiol.2025.03.002] [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: 09/07/2024] [Revised: 01/30/2025] [Accepted: 03/10/2025] [Indexed: 04/01/2025]
Abstract
Calpain-1, a calcium-dependent cysteine protease, plays a vital role in cellular processes such as cell death, cytoskeletal remodeling, signal transduction, and cell cycle progression. While its role in apoptosis, including substrate cleavage for orderly disassembly, is well established, its involvement in pyroptosis remains less understood. This study focused on developing chemical tools to detect calpain-1 activity. Using the hybrid combinatorial substrate library (HyCoSuL) approach with unnatural amino acids, we designed fluorescent substrates, inhibitors, and fluorescent activity-based probe (ABP) specific to calpain-1, enabling its visualization in living cells. We further investigated calpain-1's expression alongside cell death proteins in immune cells using mass cytometry and observed strong colocalization with gasdermin D (GSDMD). Additionally, we demonstrated that calpain-1 can hydrolyze GSDMD in vitro. Through fluorescence-based substrate assays and mass spectrometry, we identified putative cleavage sites within the GSDMD sequence that may promote pyroptosis. These findings underscore calpain-1's multifaceted role in cell death pathways, extending beyond apoptosis.
Collapse
Affiliation(s)
- Natalia Horbach
- Faculty of Chemistry, Wroclaw University of Science and Technology, 50-370 Wroclaw, Poland
| | - Małgorzata Kalinka
- Faculty of Chemistry, Wroclaw University of Science and Technology, 50-370 Wroclaw, Poland
| | | | - Abdulla Al Mamun
- Faculty of Chemistry, Wroclaw University of Science and Technology, 50-370 Wroclaw, Poland
| | - Agata Mikołajczyk-Martinez
- Faculty of Chemistry, Wroclaw University of Science and Technology, 50-370 Wroclaw, Poland; Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, 50-375 Wroclaw, Poland
| | - Boris Turk
- Jozef Stefan Institute, Sl-1000 Ljubljana, Slovenia
| | - Scott J Snipas
- SBP Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Paulina Kasperkiewicz
- Faculty of Chemistry, Wroclaw University of Science and Technology, 50-370 Wroclaw, Poland
| | - Katarzyna M Groborz
- Faculty of Chemistry, Wroclaw University of Science and Technology, 50-370 Wroclaw, Poland; SBP Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Marcin Poręba
- Faculty of Chemistry, Wroclaw University of Science and Technology, 50-370 Wroclaw, Poland; Faculty of Medicine, Wroclaw University of Science and Technology, 50-370 Wroclaw, Poland.
| |
Collapse
|
2
|
Kasperkiewicz P, Kołt S, Janiszewski T, Skowron PM, Krefft D, Brodzik R, Koller KP, Drąg M. Substrate specificity profiling of heat-sensitive serine protease from the fungus Onygena corvina. Biochimie 2024; 226:91-98. [PMID: 38971457 DOI: 10.1016/j.biochi.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 06/28/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
Proteases catalyze hydrolysis of amide bonds within peptides and proteins, therefore they play crucial functions for organism functioning, but also in industry to facilitate numerous processes. Feather-degrading fungus Onygena corvina (O. corvina) is loaded with numerous proteases that can be utilized for variety of applications. The most active species of these enzymes is heat-sensitive serine protease (NHSSP), from O. corvina fungi and due to its potential applications in industry is an alternative to proteinase K. The uniqueness of NHSSP relies on the ability of this enzyme to hydrolyze peptides at neutral to acidic pH values between 5.0 and 8.5, with an optimum of 6.8 and a temperature activity ranging from 15 to 50 °C making NHSSP exceptionally universal enzyme. Thus, we have performed the in-depth characterization of NHSSP substrate specificity by using a positional scanning substrate combinatorial library (PS-SCL). Afterward, we obtained a set of fluorescent substrates hydrolyzed by NHSSP that served as a leading sequence for the first tailored covalent inhibitor of this enzyme, containing a diphenylphosphonate as a warhead and MeOSuc amine protecting group. Our first inhibitor for NHSSP binds potently with target protease and is a tool for future study of this enzyme functions.
Collapse
Affiliation(s)
- Paulina Kasperkiewicz
- Department of Chemical Biology and Bioimaging, Faculty of Chemistry, Wroclaw University of Science and Technology, Wyb. Wyspianskiego 27, 50-370, Wroclaw, Poland
| | - Sonia Kołt
- Department of Chemical Biology and Bioimaging, Faculty of Chemistry, Wroclaw University of Science and Technology, Wyb. Wyspianskiego 27, 50-370, Wroclaw, Poland
| | - Tomasz Janiszewski
- Department of Chemical Biology and Bioimaging, Faculty of Chemistry, Wroclaw University of Science and Technology, Wyb. Wyspianskiego 27, 50-370, Wroclaw, Poland
| | - Piotr M Skowron
- Department of Molecular Biotechnology, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-309, Gdansk, Poland
| | - Daria Krefft
- Department of Molecular Biotechnology, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-309, Gdansk, Poland
| | - Robert Brodzik
- QIAGEN Gdańsk Sp. Z o.o., Trzy Lipy 3/2.58 Street, Gdańsk, Poland
| | - Klaus-Peter Koller
- Institute for Molecular BioScience, University Frankfurt, Max-von-Laue-Str. 9, 60438, Frankfurt, Germany
| | - Marcin Drąg
- Department of Chemical Biology and Bioimaging, Faculty of Chemistry, Wroclaw University of Science and Technology, Wyb. Wyspianskiego 27, 50-370, Wroclaw, Poland.
| |
Collapse
|
3
|
Kahler JP, Ji S, Speelman-Rooms F, Vanhoutte R, Verhelst SHL. Phosphinate Esters as Novel Warheads for Quenched Activity-Based Probes Targeting Serine Proteases. ACS Chem Biol 2024; 19:1409-1415. [PMID: 38913607 DOI: 10.1021/acschembio.3c00203] [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: 06/26/2024]
Abstract
Quenched activity-based probes (qABP) are invaluable tools to visualize aberrant protease activity. Unfortunately, most studies so far have only focused on cysteine proteases, and only a few studies describe the synthesis and use of serine protease qABPs. We recently used phosphinate ester electrophiles as a novel type of reactive group to construct ABPs for serine proteases. Here, we report on the construction of qABPs based on the phosphinate warhead, exemplified by probes for the neutrophil serine proteases. The most successful probes show sub-stoichiometric reaction with human neutrophil elastase, efficient fluorescence quenching, and rapid unquenching of fluorescence upon reaction with target proteases.
Collapse
Affiliation(s)
- Jan Pascal Kahler
- Laboratory of Chemical Biology, Department of Cellular and Molecular Medicine, KU Leuven - University of Leuven, Herestraat 49 box 901b, 3000 Leuven, Belgium
| | - Shanping Ji
- Laboratory of Chemical Biology, Department of Cellular and Molecular Medicine, KU Leuven - University of Leuven, Herestraat 49 box 901b, 3000 Leuven, Belgium
| | - Femke Speelman-Rooms
- Laboratory of Chemical Biology, Department of Cellular and Molecular Medicine, KU Leuven - University of Leuven, Herestraat 49 box 901b, 3000 Leuven, Belgium
- Laboratory of Molecular & Cellular Signaling, Department of Cellular and Molecular Medicine, Herestraat 49 box 802, 3000 Leuven, Belgium
| | - Roeland Vanhoutte
- Laboratory of Chemical Biology, Department of Cellular and Molecular Medicine, KU Leuven - University of Leuven, Herestraat 49 box 901b, 3000 Leuven, Belgium
| | - Steven H L Verhelst
- Laboratory of Chemical Biology, Department of Cellular and Molecular Medicine, KU Leuven - University of Leuven, Herestraat 49 box 901b, 3000 Leuven, Belgium
- AG Chemical Proteomics, Leibniz Institute for Analytical Sciences - ISAS, Otto-Hahn-Str. 6b, 44227 Dortmund, Germany
| |
Collapse
|
4
|
Birk D, Siepmann E, Simon S, Sommerhoff CP. Human Neutrophil Elastase: Characterization of Intra- vs. Extracellular Inhibition. Int J Mol Sci 2024; 25:7917. [PMID: 39063160 PMCID: PMC11276905 DOI: 10.3390/ijms25147917] [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: 06/19/2024] [Revised: 07/12/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Neutrophil elastase (HNE), like other members of the so-called GASPIDs (Granule-Associated Serine Peptidases of Immune Defense), is activated during protein biosynthesis in myeloid precursors and stored enzymatically active in cytoplasmic granules of resting neutrophils until secreted at sites of host defense and inflammation. Inhibitors thus could bind to the fully formed active site of the protease intracellularly in immature progenitors, in circulating neutrophils, or to HNE secreted into the extracellular space. Here, we have compared the ability of a panel of diverse inhibitors to inhibit HNE in the U937 progenitor cell line, in human blood-derived neutrophils, and in solution. Most synthetic inhibitors and, surprisingly, even a small naturally occurring proteinaceous inhibitor inhibit HNE intracellularly, but the extent and dynamics differ markedly from classical enzyme kinetics describing extracellular inhibition. Intracellular inhibition of HNE potentially affects neutrophil functions and has side effects, but it avoids competition of inhibitors with extracellular substrates that limit its efficacy. As both intra- and extracellular inhibition have advantages and disadvantages, the quantification of intracellular inhibition, in addition to classical enzyme kinetics, will aid the design of novel, clinically applicable HNE inhibitors with targeted sites of action.
Collapse
Affiliation(s)
- Denise Birk
- Institute of Laboratory Medicine, University Hospital, LMU Munich, 80336 Munich, Germany
- Department of Neurology, LMU University Hospital, LMU Munich, 81377 Munich, Germany
| | - Erika Siepmann
- Institute of Laboratory Medicine, University Hospital, LMU Munich, 80336 Munich, Germany
| | - Stefan Simon
- Institute of Laboratory Medicine, University Hospital, LMU Munich, 80336 Munich, Germany
| | - Christian P. Sommerhoff
- Institute of Laboratory Medicine, University Hospital, LMU Munich, 80336 Munich, Germany
- Institute of Medical Education, University Hospital, LMU Munich, 80336 Munich, Germany
| |
Collapse
|
5
|
Radisky ES. Extracellular proteolysis in cancer: Proteases, substrates, and mechanisms in tumor progression and metastasis. J Biol Chem 2024; 300:107347. [PMID: 38718867 PMCID: PMC11170211 DOI: 10.1016/j.jbc.2024.107347] [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: 02/09/2024] [Revised: 04/08/2024] [Accepted: 04/25/2024] [Indexed: 06/02/2024] Open
Abstract
A vast ensemble of extracellular proteins influences the development and progression of cancer, shaped and reshaped by a complex network of extracellular proteases. These proteases, belonging to the distinct classes of metalloproteases, serine proteases, cysteine proteases, and aspartic proteases, play a critical role in cancer. They often become dysregulated in cancer, with increases in pathological protease activity frequently driven by the loss of normal latency controls, diminished regulation by endogenous protease inhibitors, and changes in localization. Dysregulated proteases accelerate tumor progression and metastasis by degrading protein barriers within the extracellular matrix (ECM), stimulating tumor growth, reactivating dormant tumor cells, facilitating tumor cell escape from immune surveillance, and shifting stromal cells toward cancer-promoting behaviors through the precise proteolysis of specific substrates to alter their functions. These crucial substrates include ECM proteins and proteoglycans, soluble proteins secreted by tumor and stromal cells, and extracellular domains of cell surface proteins, including membrane receptors and adhesion proteins. The complexity of the extracellular protease web presents a significant challenge to untangle. Nevertheless, technological strides in proteomics, chemical biology, and the development of new probes and reagents are enabling progress and advancing our understanding of the pivotal importance of extracellular proteolysis in cancer.
Collapse
Affiliation(s)
- Evette S Radisky
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida, USA.
| |
Collapse
|
6
|
de Moliner F, Nadal-Bufi F, Vendrell M. Recent advances in minimal fluorescent probes for optical imaging. Curr Opin Chem Biol 2024; 80:102458. [PMID: 38670028 DOI: 10.1016/j.cbpa.2024.102458] [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: 03/06/2024] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024]
Abstract
Fluorescent probes have revolutionized biological imaging by enabling the real-time visualization of cellular processes under physiological conditions. However, their size and potential perturbative nature can pose challenges in retaining the integrity of biological functions. This manuscript highlights recent advancements in the development of small fluorescent probes for optical imaging studies. Single benzene-based fluorophores offer versatility with minimal disruption, exhibiting diverse properties like aggregation-induced emission and pH responsiveness. Fluorescent nucleobases enable precise labeling of nucleic acids without compromising function, offering high sensitivity and compatibility with biochemistry studies. Bright yet small fluorescent amino acids provide an interesting alternative to bulky fusion proteins, facilitating non-invasive imaging of cellular events with high precision. These miniaturized fluorophores promise enhanced capabilities for studying biological systems in a non-invasive manner, fostering further innovations in molecular imaging.
Collapse
Affiliation(s)
- Fabio de Moliner
- Centre for Inflammation Research, The University of Edinburgh, EH16 4UU Edinburgh, UK; IRR Chemistry Hub, Institute for Regeneration and Repair, The University of Edinburgh, EH16 4UU Edinburgh, UK
| | - Ferran Nadal-Bufi
- Centre for Inflammation Research, The University of Edinburgh, EH16 4UU Edinburgh, UK; IRR Chemistry Hub, Institute for Regeneration and Repair, The University of Edinburgh, EH16 4UU Edinburgh, UK
| | - Marc Vendrell
- Centre for Inflammation Research, The University of Edinburgh, EH16 4UU Edinburgh, UK; IRR Chemistry Hub, Institute for Regeneration and Repair, The University of Edinburgh, EH16 4UU Edinburgh, UK.
| |
Collapse
|
7
|
Skorenski M, Ji S, Verhelst SHL. Covalent activity-based probes for imaging of serine proteases. Biochem Soc Trans 2024; 52:923-935. [PMID: 38629725 DOI: 10.1042/bst20231450] [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: 02/14/2024] [Revised: 04/03/2024] [Accepted: 04/04/2024] [Indexed: 04/25/2024]
Abstract
Serine proteases are one of the largest mechanistic classes of proteases. They regulate a plethora of biochemical pathways inside and outside the cell. Aberrant serine protease activity leads to a wide variety of human diseases. Reagents to visualize these activities can be used to gain insight into the biological roles of serine proteases. Moreover, they may find future use for the detection of serine proteases as biomarkers. In this review, we discuss small molecule tools to image serine protease activity. Specifically, we outline different covalent activity-based probes and their selectivity against various serine protease targets. We also describe their application in several imaging methods.
Collapse
Affiliation(s)
- Marcin Skorenski
- Department of Cellular and Molecular Medicine, Laboratory of Chemical Biology, KU Leuven - University of Leuven, Herestraat 49 Box 901b, 3000 Leuven, Belgium
| | - Shanping Ji
- Department of Cellular and Molecular Medicine, Laboratory of Chemical Biology, KU Leuven - University of Leuven, Herestraat 49 Box 901b, 3000 Leuven, Belgium
| | - Steven H L Verhelst
- Department of Cellular and Molecular Medicine, Laboratory of Chemical Biology, KU Leuven - University of Leuven, Herestraat 49 Box 901b, 3000 Leuven, Belgium
| |
Collapse
|
8
|
Gartan P, Khorsand F, Mizar P, Vahokovski JI, Cervantes LF, Haug BE, Brenk R, Brooks CL, Reuter N. Investigating Polypharmacology through Targeting Known Human Neutrophil Elastase Inhibitors to Proteinase 3. J Chem Inf Model 2024; 64:621-626. [PMID: 38276895 PMCID: PMC10865350 DOI: 10.1021/acs.jcim.3c01949] [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: 12/07/2023] [Revised: 01/15/2024] [Accepted: 01/19/2024] [Indexed: 01/27/2024]
Abstract
Using a combination of multisite λ-dynamics (MSλD) together with in vitro IC50 assays, we evaluated the polypharmacological potential of a scaffold currently in clinical trials for inhibition of human neutrophil elastase (HNE), targeting cardiopulmonary disease, for efficacious inhibition of Proteinase 3 (PR3), a related neutrophil serine proteinase. The affinities we observe suggest that the dihydropyrimidinone scaffold can serve as a suitable starting point for the establishment of polypharmacologically targeting both enzymes and enhancing the potential for treatments addressing diseases like chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Parveen Gartan
- Department
of Chemistry, University of Bergen, Bergen 5020, Norway
- Computational
Biology Unit, University of Bergen, Bergen 5020, Norway
| | - Fahimeh Khorsand
- Department
of Biomedicine, University of Bergen, Bergen 5020, Norway
| | - Pushpak Mizar
- Department
of Chemistry, University of Bergen, Bergen 5020, Norway
| | - Juha Ilmari Vahokovski
- Core
Facility for Biophysics, Structural Biology, and Screening, Department
of Biomedicine, University of Bergen, Bergen 5020, Norway
| | - Luis F. Cervantes
- Department
of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Bengt Erik Haug
- Department
of Chemistry, University of Bergen, Bergen 5020, Norway
- Centre for
Pharmacy, University of Bergen, Bergen 5020, Norway
| | - Ruth Brenk
- Department
of Biomedicine, University of Bergen, Bergen 5020, Norway
| | - Charles L. Brooks
- Department
of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
- Biophysics
Program, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Nathalie Reuter
- Department
of Chemistry, University of Bergen, Bergen 5020, Norway
- Computational
Biology Unit, University of Bergen, Bergen 5020, Norway
| |
Collapse
|
9
|
Sadowski B, Kaliszewska M, Clermont G, Poronik YM, Blanchard-Desce M, Piątkowski P, Gryko DT. Realization of nitroaromatic chromophores with intense two-photon brightness. Chem Commun (Camb) 2023; 59:11708-11711. [PMID: 37700732 DOI: 10.1039/d3cc03347c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2023]
Abstract
Strong fluorescence is a general feature of dipyrrolonaphthyridinediones bearing two nitrophenyl substituents. Methyl groups simultaneously being weakly electron-donating and inducing steric hindrance appear to be a key structural parameter that allows for significant emission enhancement, whereas Et2N groups cause fluorescence quenching. The magnitude of two-photon absorption increases if 4-nitrophenyl substituents are present while the contribution of Et2N groups is detrimental.
Collapse
Affiliation(s)
- Bartłomiej Sadowski
- Centre of New Technologies, University of Warsaw, S. Banacha 2c, Warsaw 02-097, Poland.
| | - Marzena Kaliszewska
- Department of Chemistry, University of Warsaw, Zwirki i Wigury 101, Warsaw 02-089, Poland.
| | - Guillaume Clermont
- Univ. Bordeaux, CNRS, Bordeaux INP, ISM, UMR 5255, Talence F-33400, France.
| | - Yevgen M Poronik
- Institute of Organic Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, Warsaw 01-224, Poland.
| | | | - Piotr Piątkowski
- Department of Chemistry, University of Warsaw, Zwirki i Wigury 101, Warsaw 02-089, Poland.
| | - Daniel T Gryko
- Institute of Organic Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, Warsaw 01-224, Poland.
| |
Collapse
|
10
|
Goettig P, Koch NG, Budisa N. Non-Canonical Amino Acids in Analyses of Protease Structure and Function. Int J Mol Sci 2023; 24:14035. [PMID: 37762340 PMCID: PMC10531186 DOI: 10.3390/ijms241814035] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/18/2023] [Accepted: 08/20/2023] [Indexed: 09/29/2023] Open
Abstract
All known organisms encode 20 canonical amino acids by base triplets in the genetic code. The cellular translational machinery produces proteins consisting mainly of these amino acids. Several hundred natural amino acids serve important functions in metabolism, as scaffold molecules, and in signal transduction. New side chains are generated mainly by post-translational modifications, while others have altered backbones, such as the β- or γ-amino acids, or they undergo stereochemical inversion, e.g., in the case of D-amino acids. In addition, the number of non-canonical amino acids has further increased by chemical syntheses. Since many of these non-canonical amino acids confer resistance to proteolytic degradation, they are potential protease inhibitors and tools for specificity profiling studies in substrate optimization and enzyme inhibition. Other applications include in vitro and in vivo studies of enzyme kinetics, molecular interactions and bioimaging, to name a few. Amino acids with bio-orthogonal labels are particularly attractive, enabling various cross-link and click reactions for structure-functional studies. Here, we cover the latest developments in protease research with non-canonical amino acids, which opens up a great potential, e.g., for novel prodrugs activated by proteases or for other pharmaceutical compounds, some of which have already reached the clinical trial stage.
Collapse
Affiliation(s)
- Peter Goettig
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Paracelsus Medical University, Strubergasse 21, 5020 Salzburg, Austria
| | - Nikolaj G. Koch
- Biocatalysis Group, Technische Universität Berlin, 10623 Berlin, Germany;
- Bioanalytics Group, Institute of Biotechnology, Technische Universität Berlin, 10623 Berlin, Germany;
| | - Nediljko Budisa
- Bioanalytics Group, Institute of Biotechnology, Technische Universität Berlin, 10623 Berlin, Germany;
- Department of Chemistry, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| |
Collapse
|
11
|
Lee I, Tantisirivat P, Edgington-Mitchell LE. Chemical Tools to Image the Activity of PAR-Cleaving Proteases. ACS BIO & MED CHEM AU 2023; 3:295-304. [PMID: 37599791 PMCID: PMC10436261 DOI: 10.1021/acsbiomedchemau.3c00019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/18/2023] [Accepted: 05/19/2023] [Indexed: 08/22/2023]
Abstract
Protease-activated receptors (PARs) comprise a family of four G protein-coupled receptors (GPCRs) that have broad functions in health and disease. Unlike most GPCRs, PARs are uniquely activated by proteolytic cleavage of their extracellular N termini. To fully understand PAR activation and function in vivo, it is critical to also study the proteases that activate them. As proteases are heavily regulated at the post-translational level, measures of total protease abundance have limited utility. Measures of protease activity are instead required to inform their function. This review will introduce several classes of chemical probes that have been developed to measure the activation of PAR-cleaving proteases. Their strengths, weaknesses, and applications will be discussed, especially as applied to image protease activity at the whole organism, tissue, and cellular level.
Collapse
Affiliation(s)
- Irene
Y. Lee
- Department
of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology
Institute, The University of Melbourne, Parkville, Victoria 3052 Australia
| | - Piyapa Tantisirivat
- Department
of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology
Institute, The University of Melbourne, Parkville, Victoria 3052 Australia
| | - Laura E. Edgington-Mitchell
- Department
of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology
Institute, The University of Melbourne, Parkville, Victoria 3052 Australia
| |
Collapse
|
12
|
Yang J, Korovesis D, Ji S, Kahler JP, Vanhoutte R, Verhelst SHL. Efficient Synthesis of an Alkyne Fluorophosphonate Activity‐Based Probe and Applications in Dual Colour Serine Hydrolase Labelling. Isr J Chem 2023. [DOI: 10.1002/ijch.202200094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Affiliation(s)
- Jian Yang
- Department of Cellular and Molecular Medicine KU Leuven – University of Leuven Herestraat 49, box 901b 3000 Leuven Belgium
| | - Dimitris Korovesis
- Department of Cellular and Molecular Medicine KU Leuven – University of Leuven Herestraat 49, box 901b 3000 Leuven Belgium
| | - Shanping Ji
- Department of Cellular and Molecular Medicine KU Leuven – University of Leuven Herestraat 49, box 901b 3000 Leuven Belgium
| | - Jan Pascal Kahler
- Department of Cellular and Molecular Medicine KU Leuven – University of Leuven Herestraat 49, box 901b 3000 Leuven Belgium
| | - Roeland Vanhoutte
- Department of Cellular and Molecular Medicine KU Leuven – University of Leuven Herestraat 49, box 901b 3000 Leuven Belgium
| | - Steven H. L. Verhelst
- Department of Cellular and Molecular Medicine KU Leuven – University of Leuven Herestraat 49, box 901b 3000 Leuven Belgium
- Leibniz Institut für Analytische Wissenschaften – ISAS – e.V. Otto-Hahn Strasse 6b 44227 Dortmund Germany
| |
Collapse
|
13
|
Janiszewski T, Kołt S, Ciastoń I, Vizovisek M, Poręba M, Turk B, Drąg M, Kozieł J, Kasperkiewicz P. Investigation of osteoclast cathepsin K activity in osteoclastogenesis and bone loss using a set of chemical reagents. Cell Chem Biol 2023; 30:159-174.e8. [PMID: 36696904 DOI: 10.1016/j.chembiol.2023.01.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 08/28/2022] [Accepted: 01/04/2023] [Indexed: 01/26/2023]
Abstract
Cathepsin K (CatK) is a lysosomal cysteine protease whose highest expression is found in osteoclasts, which are the cells responsible for bone resorption. Investigations of the functions and physiological relevance of CatK have often relied on antibody-related techniques, which makes studying its activity patterns a challenging task. Hence, we developed a set of chemical tools for the investigation of CatK activity. We show that our probe is a valuable tool for monitoring the proteolytic activation of CatK during osteoclast formation. Moreover, we demonstrate that our inhibitor of CatK impedes osteoclastogenesis and bone resorption and that CatK is stored in its active form in osteoclasts within their lysosomal compartment and mainly in the ruffled borders of osteoclasts. Given that our probe recognizes active CatK within living cells without exhibiting any observed cytotoxicity in the several models tested, we expect that it would be well suited to theranostic applications in CatK-related diseases.
Collapse
Affiliation(s)
- Tomasz Janiszewski
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology, Wyb. Wyspianskiego 27, 50-370 Wroclaw, Poland
| | - Sonia Kołt
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology, Wyb. Wyspianskiego 27, 50-370 Wroclaw, Poland
| | - Izabela Ciastoń
- Department of Microbiology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian University, ul. Gronostajowa 7, 30-387 Kraków, Poland
| | - Matej Vizovisek
- Department of Biochemistry and Molecular and Structural Biology, Jozef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia
| | - Marcin Poręba
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology, Wyb. Wyspianskiego 27, 50-370 Wroclaw, Poland
| | - Boris Turk
- Department of Biochemistry and Molecular and Structural Biology, Jozef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia; Faculty of Chemistry and Chemical Technology, University of Ljubljana, Vecna pot 113, 1000 Ljubljana, Slovenia
| | - Marcin Drąg
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology, Wyb. Wyspianskiego 27, 50-370 Wroclaw, Poland
| | - Joanna Kozieł
- Department of Microbiology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian University, ul. Gronostajowa 7, 30-387 Kraków, Poland
| | - Paulina Kasperkiewicz
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology, Wyb. Wyspianskiego 27, 50-370 Wroclaw, Poland.
| |
Collapse
|
14
|
Racioppo B, Qiu N, Adibekian A. Serine Hydrolase Activity‐Based Probes for use in Chemical Proteomics. Isr J Chem 2023. [DOI: 10.1002/ijch.202300016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Affiliation(s)
- Brittney Racioppo
- Department of Chemistry University of Illinois Chicago Chicago Illinois 60607 United States
- Skaggs Doctoral Program in the Chemical and Biological Sciences, Scripps Research La Jolla California 92037 United States
| | - Nan Qiu
- Department of Chemistry University of Illinois Chicago Chicago Illinois 60607 United States
- Skaggs Doctoral Program in the Chemical and Biological Sciences, Scripps Research La Jolla California 92037 United States
| | - Alexander Adibekian
- Department of Chemistry University of Illinois Chicago Chicago Illinois 60607 United States
| |
Collapse
|
15
|
Zhang L, Lovell S, De Vita E, Jagtap PKA, Lucy D, Goya Grocin A, Kjær S, Borg A, Hennig J, Miller AK, Tate EW. A KLK6 Activity-Based Probe Reveals a Role for KLK6 Activity in Pancreatic Cancer Cell Invasion. J Am Chem Soc 2022; 144:22493-22504. [PMID: 36413626 DOI: 10.1021/jacs.2c07378] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Pancreatic cancer has the lowest survival rate of all common cancers due to late diagnosis and limited treatment options. Serine hydrolases are known to mediate cancer progression and metastasis through initiation of signaling cascades and cleavage of extracellular matrix proteins, and the kallikrein-related peptidase (KLK) family of secreted serine proteases have emerging roles in pancreatic ductal adenocarcinoma (PDAC). However, the lack of reliable activity-based probes (ABPs) to profile KLK activity has hindered progress in validation of these enzymes as potential targets or biomarkers. Here, we developed potent and selective ABPs for KLK6 by using a positional scanning combinatorial substrate library and characterized their binding mode and interactions by X-ray crystallography. The optimized KLK6 probe IMP-2352 (kobs/I = 11,000 M-1 s-1) enabled selective detection of KLK6 activity in a variety of PDAC cell lines, and we observed that KLK6 inhibition reduced the invasiveness of PDAC cells that secrete active KLK6. KLK6 inhibitors were combined with N-terminomics to identify potential secreted protein substrates of KLK6 in PDAC cells, providing insights into KLK6-mediated invasion pathways. These novel KLK6 ABPs offer a toolset to validate KLK6 and associated signaling partners as targets or biomarkers across a range of diseases.
Collapse
Affiliation(s)
- Leran Zhang
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| | - Scott Lovell
- Department of Life Sciences, University of Bath, Bath BA2 7AX, U.K
| | - Elena De Vita
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| | - Pravin Kumar Ankush Jagtap
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany.,Chair of Biochemistry IV, Biophysical Chemistry, University of Bayreuth, Bayreuth 95447, Germany
| | - Daniel Lucy
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| | - Andrea Goya Grocin
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| | - Svend Kjær
- Structural Biology Science Technology Platform, The Francis Crick Institute, London NW1 1AT, U.K
| | - Annabel Borg
- Structural Biology Science Technology Platform, The Francis Crick Institute, London NW1 1AT, U.K
| | - Janosch Hennig
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany.,Chair of Biochemistry IV, Biophysical Chemistry, University of Bayreuth, Bayreuth 95447, Germany
| | - Aubry K Miller
- Cancer Drug Development Group, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Edward W Tate
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| |
Collapse
|
16
|
Ćwilichowska N, Świderska KW, Dobrzyń A, Drąg M, Poręba M. Diagnostic and therapeutic potential of protease inhibition. Mol Aspects Med 2022; 88:101144. [PMID: 36174281 DOI: 10.1016/j.mam.2022.101144] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 07/20/2022] [Accepted: 09/09/2022] [Indexed: 12/14/2022]
Abstract
Proteases are enzymes that hydrolyze peptide bonds in proteins and peptides; thus, they control virtually all biological processes. Our understanding of protease function has advanced considerably from nonselective digestive enzymes to highly specialized molecular scissors that orchestrate complex signaling networks through a limited proteolysis. The catalytic activity of proteases is tightly regulated at several levels, ranging from gene expression through trafficking and maturation to posttranslational modifications. However, when this delicate balance is disturbed, many diseases develop, including cancer, inflammatory disorders, diabetes, and neurodegenerative diseases. This new understanding of the role of proteases in pathologic physiology indicates that these enzymes represent excellent molecular targets for the development of therapeutic inhibitors, as well as for the design of chemical probes to visualize their redundant activity. Recently, numerous platform technologies have been developed to identify and optimize protease substrates and inhibitors, which were further used as lead structures for the development of chemical probes and therapeutic drugs. Due to this considerable success, the clinical potential of proteases in therapeutics and diagnostics is rapidly growing and is still not completely explored. Therefore, small molecules that can selectively target aberrant protease activity are emerging in diseases cells. In this review, we describe modern trends in the design of protease drugs as well as small molecule activity-based probes to visualize selected proteases in clinical settings.
Collapse
Affiliation(s)
- Natalia Ćwilichowska
- Department of Chemical Biology and Bioimaging, Faculty of Chemistry, Wroclaw University of Science and Technology, Wyb, Wyspianskiego 27, 50-370, Wroclaw, Poland
| | - Karolina W Świderska
- Department of Chemical Biology and Bioimaging, Faculty of Chemistry, Wroclaw University of Science and Technology, Wyb, Wyspianskiego 27, 50-370, Wroclaw, Poland
| | - Agnieszka Dobrzyń
- Nencki Institute of Experimental Biology, Ludwika Pasteura 3, 02-093, Warsaw, Poland
| | - Marcin Drąg
- Department of Chemical Biology and Bioimaging, Faculty of Chemistry, Wroclaw University of Science and Technology, Wyb, Wyspianskiego 27, 50-370, Wroclaw, Poland.
| | - Marcin Poręba
- Department of Chemical Biology and Bioimaging, Faculty of Chemistry, Wroclaw University of Science and Technology, Wyb, Wyspianskiego 27, 50-370, Wroclaw, Poland.
| |
Collapse
|
17
|
Sun Q, Li X, Guo Y, Qiu Y, Luo X, Liu G, Han Y. Coumarin-based turn-on fluorescence probe with a large Stokes shift for detection of endogenous neutrophil elastase in live cells and zebrafish. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2022; 281:121563. [PMID: 35810672 DOI: 10.1016/j.saa.2022.121563] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/21/2022] [Accepted: 06/23/2022] [Indexed: 06/15/2023]
Abstract
Neutrophil elastase (NE), a serine proteinase, is a significant biomarker which is closely related to the progress of diseases. However, only few probes have been reported for detection of NE activity and cell imaging. And these probes have exhibited small Stokes shift, which leads to high fluorescence interferences. Furthermore, only one probe among them is able to image NE in vivo successfully. To overcome the above problems, we designed a novel coumarin-based fluorescent probe HNCOU-NE with large Stokes shift to visualize NE activity in living cells and zebrafish. The new probe HNCOU-NE for NE contains fluorophore HNCOU as the reporter and pentafluoroethyl as the enzyme-active trigger moiety. As expected, HNCOU-NE displays perfect detecting performance for sensing of NE, including good water solubility, large Stokes shift, high affinity and wide linear response concentration. In addition, HNCOU-NE has been successfully utilized for NE real-time detection and imaging in different living cells, exhibiting low cytotoxicity and excellent biocompatibility. Most importantly, endogenous NE fluorescence imaging experiments reveals that HNCOU-NE can distinguish liver cancer cells (HepG2) and other cells (293T, HeLa and SKOV3), illustrating its specific ability to diagnose liver cancer cells. Besides, probe HNCOU-NE also has the ability to specifically detect endogenous NE activity in living zebrafish. All the results indicate that HNCOU-NE is a valuable probe for qualitative and quantitative sensing of NE activity in vitro and in vivo.
Collapse
Affiliation(s)
- Qi Sun
- Key Laboratory for Green Chemical Process of Ministry of Education, Hubei Key Laboratory of Novel Reactor and Green Chemistry Technology, Key Laboratory of Novel Biomass-based Environmental and Energy Materials in Petroleum and Chemical Industry and School of Chemistry and Environmental Engineering, Wuhan Institute of Technology, Wuhan, China
| | - Xiang Li
- Key Laboratory for Green Chemical Process of Ministry of Education, Hubei Key Laboratory of Novel Reactor and Green Chemistry Technology, Key Laboratory of Novel Biomass-based Environmental and Energy Materials in Petroleum and Chemical Industry and School of Chemistry and Environmental Engineering, Wuhan Institute of Technology, Wuhan, China
| | - Yun Guo
- Key Laboratory for Green Chemical Process of Ministry of Education, Hubei Key Laboratory of Novel Reactor and Green Chemistry Technology, Key Laboratory of Novel Biomass-based Environmental and Energy Materials in Petroleum and Chemical Industry and School of Chemistry and Environmental Engineering, Wuhan Institute of Technology, Wuhan, China
| | - Yuan Qiu
- Key Laboratory for Green Chemical Process of Ministry of Education, Hubei Key Laboratory of Novel Reactor and Green Chemistry Technology, Key Laboratory of Novel Biomass-based Environmental and Energy Materials in Petroleum and Chemical Industry and School of Chemistry and Environmental Engineering, Wuhan Institute of Technology, Wuhan, China
| | - Xiaogang Luo
- Key Laboratory for Green Chemical Process of Ministry of Education, Hubei Key Laboratory of Novel Reactor and Green Chemistry Technology, Key Laboratory of Novel Biomass-based Environmental and Energy Materials in Petroleum and Chemical Industry and School of Chemistry and Environmental Engineering, Wuhan Institute of Technology, Wuhan, China; School of Materials Science and Engineering, Zhengzhou University, Zhengzhou, China
| | - Genyan Liu
- Key Laboratory for Green Chemical Process of Ministry of Education, Hubei Key Laboratory of Novel Reactor and Green Chemistry Technology, Key Laboratory of Novel Biomass-based Environmental and Energy Materials in Petroleum and Chemical Industry and School of Chemistry and Environmental Engineering, Wuhan Institute of Technology, Wuhan, China.
| | - Yunfeng Han
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
18
|
Ferguson TEG, Reihill JA, Martin SL, Walker B. Novel inhibitors and activity-based probes targeting serine proteases. Front Chem 2022; 10:1006618. [PMID: 36247662 PMCID: PMC9555310 DOI: 10.3389/fchem.2022.1006618] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 08/18/2022] [Indexed: 11/13/2022] Open
Abstract
Serine proteases play varied and manifold roles in important biological, physiological, and pathological processes. These include viral, bacterial, and parasitic infection, allergic sensitization, tumor invasion, and metastasis. The use of activity-based profiling has been foundational in pinpointing the precise roles of serine proteases across this myriad of processes. A broad range of serine protease-targeted activity-based probe (ABP) chemotypes have been developed and we have recently introduced biotinylated and "clickable" peptides containing P1 N-alkyl glycine arginine N-hydroxy succinimidyl (NHS) carbamates as ABPs for detection/profiling of trypsin-like serine proteases. This present study provides synthetic details for the preparation of additional examples of this ABP chemotype, which function as potent irreversible inhibitors of their respective target serine protease. We describe their use for the activity-based profiling of a broad range of serine proteases including trypsin, the trypsin-like protease plasmin, chymotrypsin, cathepsin G, and neutrophil elastase (NE), including the profiling of the latter protease in clinical samples obtained from patients with cystic fibrosis.
Collapse
Affiliation(s)
| | | | | | - Brian Walker
- Biomolecular Sciences Research Group, School of Pharmacy, Queen’s University Belfast, Belfast, United Kingdom
| |
Collapse
|
19
|
Yu H, Wang S, Huang J, Fu Y, Wagner M, Weil T, Zhong F, Zhao W, Wu Y. Light-Controlled Traceless Protein Labeling via Decaging Thio- o-naphthoquinone Methide Chemistry. Org Lett 2022; 24:6816-6821. [PMID: 36099167 DOI: 10.1021/acs.orglett.2c02742] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We report the molecular design of a novel multifunctional reagent and its application for light-controlled selective protein labeling. This molecule integrates functions of protein-ligand recognition, bioconjugation, ligand cleavage, and photoactivation by merging the photochemistries of 2-nitrophenylpropyloxycarbonyl and 3-hydroxymethyl-2-naphthol with an affinity ligand and fluorescein. Highly electrophilic o-naphthoquinone methide was photochemically released and underwent proximity-driven selective labeling with the protein of interest (e.g., carbonic anhydrases), which retains its native function after labeling.
Collapse
Affiliation(s)
- Huaibin Yu
- Key Laboratory of Material Chemistry for Energy Conversion and Storage, Ministry of Education, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), 1037 Luoyu Road, Wuhan 430074, China
| | - Shuangshuang Wang
- Key Laboratory of Material Chemistry for Energy Conversion and Storage, Ministry of Education, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), 1037 Luoyu Road, Wuhan 430074, China
| | - Jianjian Huang
- Key Laboratory of Material Chemistry for Energy Conversion and Storage, Ministry of Education, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), 1037 Luoyu Road, Wuhan 430074, China
| | - Yu Fu
- Key Laboratory of Material Chemistry for Energy Conversion and Storage, Ministry of Education, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), 1037 Luoyu Road, Wuhan 430074, China
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China
| | - Manfred Wagner
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Tanja Weil
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Fangrui Zhong
- Key Laboratory of Material Chemistry for Energy Conversion and Storage, Ministry of Education, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), 1037 Luoyu Road, Wuhan 430074, China
| | - Weining Zhao
- College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China
| | - Yuzhou Wu
- Key Laboratory of Material Chemistry for Energy Conversion and Storage, Ministry of Education, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), 1037 Luoyu Road, Wuhan 430074, China
| |
Collapse
|
20
|
Benson S, de Moliner F, Tipping W, Vendrell M. Miniaturized Chemical Tags for Optical Imaging. Angew Chem Int Ed Engl 2022; 61:e202204788. [PMID: 35704518 PMCID: PMC9542129 DOI: 10.1002/anie.202204788] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Indexed: 11/06/2022]
Abstract
Recent advances in optical bioimaging have prompted the need for minimal chemical reporters that can retain the molecular recognition properties and activity profiles of biomolecules. As a result, several methodologies to reduce the size of fluorescent and Raman labels to a few atoms (e.g., single aryl fluorophores, Raman-active triple bonds and isotopes) and embed them into building blocks (e.g., amino acids, nucleobases, sugars) to construct native-like supramolecular structures have been described. The integration of small optical reporters into biomolecules has also led to smart molecular entities that were previously inaccessible in an expedite manner. In this article, we review recent chemical approaches to synthesize miniaturized optical tags as well as some of their multiple applications in biological imaging.
Collapse
Affiliation(s)
- Sam Benson
- Centre for Inflammation ResearchThe University of EdinburghEdinburghEH16 4TJUK
| | - Fabio de Moliner
- Centre for Inflammation ResearchThe University of EdinburghEdinburghEH16 4TJUK
| | - William Tipping
- Centre for Molecular NanometrologyThe University of StrathclydeGlasgowG1 1RDUK
| | - Marc Vendrell
- Centre for Inflammation ResearchThe University of EdinburghEdinburghEH16 4TJUK
| |
Collapse
|
21
|
Huang J, Cheng P, Xu C, Liew SS, He S, Zhang Y, Pu K. Chemiluminescent Probes with Long‐Lasting High Brightness for In Vivo Imaging of Neutrophils. Angew Chem Int Ed Engl 2022; 61:e202203235. [DOI: 10.1002/anie.202203235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Indexed: 12/18/2022]
Affiliation(s)
- Jingsheng Huang
- School of Chemical and Biomedical Engineering 70 Nanyang Drive Singapore 637457 Singapore
| | - Penghui Cheng
- School of Chemical and Biomedical Engineering 70 Nanyang Drive Singapore 637457 Singapore
| | - Cheng Xu
- School of Chemical and Biomedical Engineering 70 Nanyang Drive Singapore 637457 Singapore
| | - Si Si Liew
- School of Chemical and Biomedical Engineering 70 Nanyang Drive Singapore 637457 Singapore
| | - Shasha He
- School of Chemical and Biomedical Engineering 70 Nanyang Drive Singapore 637457 Singapore
| | - Yan Zhang
- National Engineering Research Centre for Nanomedicine College of Life Science and Technology Huazhong University of Science and Technology Wuhan 430074 P. R. China
| | - Kanyi Pu
- School of Chemical and Biomedical Engineering 70 Nanyang Drive Singapore 637457 Singapore
- School of Physical and Mathematical Sciences Nanyang Technological University Singapore 637371 Singapore
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore 636921 Singapore
| |
Collapse
|
22
|
Krzeszewski M, Modrzycka S, Bousquet MHE, Jacquemin D, Drąg M, Gryko DT. Green-Emitting 4,5-Diaminonaphthalimides in Activity-Based Probes for the Detection of Thrombin. Org Lett 2022; 24:5602-5607. [PMID: 35863755 PMCID: PMC9361357 DOI: 10.1021/acs.orglett.2c02320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The natures of electron-donating groups as well as the bridge between them determine the fate of substituted 1,8-naphthalimide molecules in the excited state. An activity-based probe constructed from a selective peptide sequence, a reactive warhead, and the brightest green-emitting fluorophore displays impressive performance for thrombin protease detection in a newly constructed series of 1,8-naphthalimides.
Collapse
Affiliation(s)
- Maciej Krzeszewski
- Institute of Organic Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, Warsaw 01-224, Poland
| | - Sylwia Modrzycka
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology, Wybrezeże Wyspiańskiego 27, Wrocław 50-370, Poland
| | | | - Denis Jacquemin
- CEISAM UMR CNRS 6230, Nantes University, Nantes 44000, France
| | - Marcin Drąg
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology, Wybrezeże Wyspiańskiego 27, Wrocław 50-370, Poland
| | - Daniel T Gryko
- Institute of Organic Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, Warsaw 01-224, Poland
| |
Collapse
|
23
|
Benson S, de Moliner F, Tipping W, Vendrell M. Miniaturized Chemical Tags for Optical Imaging. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202204788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Sam Benson
- The University of Edinburgh Centre for Inflammation Research UNITED KINGDOM
| | - Fabio de Moliner
- The University of Edinburgh Centre for Inflammation Research UNITED KINGDOM
| | - William Tipping
- University of Strathclyde Centre for Molecular Nanometrology UNITED KINGDOM
| | - Marc Vendrell
- University of Edinburgh Centre for Inflammation Research 47 Little France Crescent EH16 4TJ Edinburgh UNITED KINGDOM
| |
Collapse
|
24
|
Modrzycka S, Kołt S, Polderdijk SGI, Adams TE, Potoczek S, Huntington JA, Kasperkiewicz P, Drąg M. Parallel imaging of coagulation pathway proteases activated protein C, thrombin, and factor Xa in human plasma. Chem Sci 2022; 13:6813-6829. [PMID: 35774156 PMCID: PMC9200056 DOI: 10.1039/d2sc01108e] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/22/2022] [Indexed: 12/12/2022] Open
Abstract
Activated protein C (APC), thrombin, and factor (f) Xa are vitamin K-dependent serine proteases that are key factors in blood coagulation. Moreover, they play important roles in inflammation, apoptosis, fibrosis, angiogenesis, and viral infections. Abnormal activity of these coagulation factors has been related to multiple conditions, such as bleeding and thrombosis, Alzheimer's disease, sepsis, multiple sclerosis, and COVID-19. The individual activities of APC, thrombin, and fXa in coagulation and in various diseases are difficult to establish since these proteases are related and have similar substrate preferences. Therefore, the development of selective chemical tools that enable imaging and discrimination between coagulation factors in biological samples may provide better insight into their roles in various conditions and potentially aid in the establishment of novel diagnostic tests. In our study, we used a large collection of unnatural amino acids, and this enabled us to extensively explore the binding pockets of the enzymes' active sites. Based on the specificity profiles obtained, we designed highly selective substrates, inhibitors, and fluorescent activity-based probes (ABPs) that were used for fast, direct, and simultaneous detection of APC, thrombin, and fXa in human plasma. Using a collection of natural and unnatural amino acids, we synthesized a set of fluorescent activity-based probes for the fast, direct, and simultaneous detection of coagulation factors in human plasma.![]()
Collapse
Affiliation(s)
- Sylwia Modrzycka
- Department of Chemical Biology and Bioimaging, Faculty of Chemistry, Wrocław University of Science and Technology Wybrzeże Wyspiańskiego 27 50-370 Wrocław Poland
| | - Sonia Kołt
- Department of Chemical Biology and Bioimaging, Faculty of Chemistry, Wrocław University of Science and Technology Wybrzeże Wyspiańskiego 27 50-370 Wrocław Poland
| | - Stéphanie G I Polderdijk
- Department of Haematology, Cambridge Institute for Medical Research, University of Cambridge The Keith Peters Building, Hills Road Cambridge CB2 0XY UK
| | - Ty E Adams
- Department of Haematology, Cambridge Institute for Medical Research, University of Cambridge The Keith Peters Building, Hills Road Cambridge CB2 0XY UK
| | - Stanisław Potoczek
- Department of Haematology, Blood Neoplasms, and Bone Marrow Transplantation, Wrocław Medical University Pasteura 1 50-367 Wrocław Poland
| | - James A Huntington
- Department of Haematology, Cambridge Institute for Medical Research, University of Cambridge The Keith Peters Building, Hills Road Cambridge CB2 0XY UK
| | - Paulina Kasperkiewicz
- Department of Chemical Biology and Bioimaging, Faculty of Chemistry, Wrocław University of Science and Technology Wybrzeże Wyspiańskiego 27 50-370 Wrocław Poland
| | - Marcin Drąg
- Department of Chemical Biology and Bioimaging, Faculty of Chemistry, Wrocław University of Science and Technology Wybrzeże Wyspiańskiego 27 50-370 Wrocław Poland
| |
Collapse
|
25
|
Huang J, Cheng P, Xu C, Liew SS, He S, Zhang Y, Pu K. Chemiluminescent Probes with Long‐Lasting High Brightness for In Vivo Imaging of Neutrophils. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202203235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Jingsheng Huang
- Nanyang Technological University Chemical and Biomedical Engineering SINGAPORE
| | - Penghui Cheng
- Nanyang Technological University Chemical and Biomedical Engineering SINGAPORE
| | - Cheng Xu
- Nanyang Technological University Chemical and Biomedical Engineering SINGAPORE
| | - Si Si Liew
- Nanyang Technological University Chemical and Biomedical Engineering SINGAPORE
| | - Shasha He
- Nanyang Technological University Chemical and Biomedical Engineering SINGAPORE
| | - Yan Zhang
- Huazhong University of Science and Technology College of Life Science and Technology CHINA
| | - Kanyi Pu
- Nanyang Technological University School of Chemical and Biomedical Engieering 70 Nanyang Drive 637457 Singapore SINGAPORE
| |
Collapse
|
26
|
Sotiropoulou G, Zingkou E, Bisyris E, Pampalakis G. Activity-Based Probes for Proteases Pave the Way to Theranostic Applications. Pharmaceutics 2022; 14:pharmaceutics14050977. [PMID: 35631563 PMCID: PMC9145445 DOI: 10.3390/pharmaceutics14050977] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/23/2022] [Accepted: 04/28/2022] [Indexed: 11/16/2022] Open
Abstract
Proteases are important enzymes in health and disease. Their activities are regulated at multiple levels. In fact, proteases are synthesized as inactive proenzymes (zymogens) that are activated by proteolytic removal of their pro-peptide sequence and can remain active or their activity can be attenuated by complex formation with specific endogenous inhibitors or by limited proteolysis or degradation. Consequently, quite often, only a fraction of the protease molecules is in the active/functional form, thus, the abundance of a protease is not always linearly proportional to the (patho)physiological function(s). Therefore, assays to determine the active forms of proteases are needed, not only in research but also in molecular diagnosis and therapy. Activity-based probes (ABPs) are chemical entities that bind covalently to the active enzyme/protease. ABPs carry a detection tag to enable localization and quantification of specific enzymatic/proteolytic activities with applications in molecular imaging and diagnosis. Moreover, ABPs act as suicide inhibitors of proteases, which can be exploited for delineation of the functional role(s) of a given protease in (patho) biological context and as potential therapeutics. In this sense, ABPs represent new theranostic agents. We outline recent developments pertaining to ABPs for proteases with potential therapeutic applications, with the aim to highlight their importance in theranostics.
Collapse
Affiliation(s)
- Georgia Sotiropoulou
- Department of Pharmacy, School of Health Sciences, University of Patras, 26500 Rion-Patras, Greece; (E.Z.); (E.B.)
- Correspondence: (G.S.); (G.P.)
| | - Eleni Zingkou
- Department of Pharmacy, School of Health Sciences, University of Patras, 26500 Rion-Patras, Greece; (E.Z.); (E.B.)
| | - Evangelos Bisyris
- Department of Pharmacy, School of Health Sciences, University of Patras, 26500 Rion-Patras, Greece; (E.Z.); (E.B.)
| | - Georgios Pampalakis
- Department of Pharmacognosy-Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Correspondence: (G.S.); (G.P.)
| |
Collapse
|
27
|
Abstract
![]()
Optical
imaging has become an essential tool to study biomolecular
processes in live systems with unprecedented spatial resolution. New
fluorescent technologies and advances in optical microscopy have revolutionized
the ways in which we can study immune cells in real time. For example,
activatable fluorophores that emit signals after target recognition
have enabled direct imaging of immune cell function with enhanced
readouts and minimal background. In this Account, we summarize recent
advances in the chemical synthesis and implementation of activatable
fluorescent probes to monitor the activity and the role of immune
cells in different pathological processes, from infection to inflammatory
diseases or cancer. In addition to the contributions that our group
has made to this field, we review the most relevant literature disclosed
over the past decade, providing examples of different activatable
architectures and their application in diagnostics and drug discovery.
This Account covers the imaging of the three major cell types in the
immune system, that is, neutrophils, macrophages, and lymphocytes.
Attracted by the tunability and target specificity of peptides, many
groups have designed strategies based on fluorogenic peptides whose
fluorescence emission is regulated by the reaction with enzymes (e.g.,
MMPs, cathepsins, granzymes), or through Förster resonance
energy transfer (FRET) mechanisms. Selective imaging of immune cells
has been also achieved by targeting different intracellular metabolic
routes, such as lipid biogenesis. Other approaches involve the implementation
of diversity-oriented fluorescence libraries or the use of environmentally
sensitive fluorescent scaffolds (e.g., molecular rotors). Our group
has made important progress by constructing probes to image metastasis-associated
macrophages in tumors, apoptotic neutrophils, or cytotoxic natural
killer (NK) cells against cancer cells, among other examples. The
chemical probes covered in this Account have been successfully validated
in vitro in cell culture systems, and in vivo in relevant models of
inflammation and cancer. Overall, the range of chemical structures
and activation mechanisms reported to sense immune cell function is
remarkable. However, the emergence of new strategies based on new
molecular targets or activatable mechanisms that are yet to be discovered
will open the door to track unexplored roles of immune cells in different
biological systems. We anticipate that upcoming generations of activatable
probes will find applications in the clinic to help assessing immunotherapies
and advance precision medicine. We hope that this Account will evoke
new ideas and innovative work in the design of fluorescent probes
for imaging cell function.
Collapse
Affiliation(s)
- Lorena Mendive-Tapia
- Centre for Inflammation Research, The University of Edinburgh, EH16 4TJ Edinburgh, U.K
| | - Marc Vendrell
- Centre for Inflammation Research, The University of Edinburgh, EH16 4TJ Edinburgh, U.K
| |
Collapse
|
28
|
Tagirasa R, Yoo E. Role of Serine Proteases at the Tumor-Stroma Interface. Front Immunol 2022; 13:832418. [PMID: 35222418 PMCID: PMC8873516 DOI: 10.3389/fimmu.2022.832418] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/24/2022] [Indexed: 01/19/2023] Open
Abstract
During tumor development, invasion and metastasis, the intimate interaction between tumor and stroma shapes the tumor microenvironment and dictates the fate of tumor cells. Stromal cells can also influence anti-tumor immunity and response to immunotherapy. Understanding the molecular mechanisms that govern this complex and dynamic interplay, thus is important for cancer diagnosis and therapy. Proteolytic enzymes that are expressed and secreted by both cancer and stromal cells play important roles in modulating tumor-stromal interaction. Among, several serine proteases such as fibroblast activation protein, urokinase-type plasminogen activator, kallikrein-related peptidases, and granzymes have attracted great attention owing to their elevated expression and dysregulated activity in the tumor microenvironment. This review highlights the role of serine proteases that are mainly derived from stromal cells in tumor progression and associated theranostic applications.
Collapse
|
29
|
The roles of cellular protease interactions in viral infections and programmed cell death: a lesson learned from the SARS-CoV-2 outbreak and COVID-19 pandemic. Pharmacol Rep 2022; 74:1149-1165. [PMID: 35997950 PMCID: PMC9395814 DOI: 10.1007/s43440-022-00394-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/03/2022] [Accepted: 07/18/2022] [Indexed: 12/13/2022]
Abstract
The unprecedented pandemic of SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2), which leads to COVID-19, is threatening global health. Over the last 2 years, we have witnessed rapid progress in research focusing on developing new antiviral vaccines and drugs, as well as in academic and clinical efforts to understand the biology and pathology of COVID-19. The roles of proteases among master regulators of SARS-CoV-2 invasion and replication and their pivotal roles in host defence against this pathogen, including programmed cell death, have not been well established. Our understanding of protease function in health and disease has increased considerably over the last two decades, with caspases, matrix metalloproteases, and transmembrane serine proteases representing the most prominent examples. Therefore, during the COVID-19 pandemic, these enzymes have been investigated as potential molecular targets for therapeutic interventions. Proteases that are responsible for SARS-CoV-2 cell entry and replication, such as TMPRSS2, ACE2 or cathepsins, are screened with inhibitor libraries to discover lead structures for further drug design that would prevent virus multiplication. On the other hand, proteases that orchestrate programmed cell death can also be harnessed to enhance the desired demise of infected cells through apoptosis or to attenuate highly inflammatory lytic cell death that leads to undesired cytokine storms, a major hallmark of severe COVID-19. Given the prominent role of proteases in SARS-CoV-2-induced cell death, we discuss the individual roles of these enzymes and their catalytic interactions in the pathology of COVID-19 in this article. We provide a rationale for targeting proteases participating in cell death as potential COVID-19 treatments and identify knowledge gaps that might be investigated to better understand the mechanism underlying SARS-CoV-2-induced cell death.
Collapse
|
30
|
Scott J, Deng Q, Vendrell M. Near-Infrared Fluorescent Probes for the Detection of Cancer-Associated Proteases. ACS Chem Biol 2021; 16:1304-1317. [PMID: 34315210 PMCID: PMC8383269 DOI: 10.1021/acschembio.1c00223] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 06/14/2021] [Indexed: 12/11/2022]
Abstract
Proteases are enzymes capable of catalyzing protein breakdown, which is critical across many biological processes. There are several families of proteases, each of which perform key functions through the degradation of specific proteins. As our understanding of cancer improves, it has been demonstrated that several proteases can be overactivated during the progression of cancer and contribute to malignancy. Optical imaging systems that employ near-infrared (NIR) fluorescent probes to detect protease activity offer clinical promise, both for early detection of cancer as well as for the assessment of personalized therapy. In this Review, we review the design of NIR probes and their successful application for the detection of different cancer-associated proteases.
Collapse
Affiliation(s)
- Jamie
I. Scott
- Centre
for Inflammation Research, The University
of Edinburgh, EH16 4TJ Edinburgh, United Kingdom
| | - Qinyi Deng
- Centre
for Inflammation Research, The University
of Edinburgh, EH16 4TJ Edinburgh, United Kingdom
| | - Marc Vendrell
- Centre
for Inflammation Research, The University
of Edinburgh, EH16 4TJ Edinburgh, United Kingdom
| |
Collapse
|
31
|
Kahler JP, Verhelst SHL. Phosphinate esters as novel warheads for activity-based probes targeting serine proteases. RSC Chem Biol 2021; 2:1285-1290. [PMID: 34458842 PMCID: PMC8341442 DOI: 10.1039/d1cb00117e] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/07/2021] [Indexed: 11/21/2022] Open
Abstract
Activity-based protein profiling enables the specific detection of the active fraction of an enzyme and is of particular use for the profiling of proteases. The technique relies on a mechanism-based reaction between small molecule activity-based probes (ABPs) with the active enzyme. Here we report a set of new ABPs for serine proteases, specifically neutrophil serine proteases. The probes contain a phenylphosphinate warhead that mimics the P1 amino acid recognized by the primary recognition pocket of S1 family serine proteases. The warhead is easily synthesized from commercial starting materials and leads to potent probes which can be used for fluorescent in-gel protease detection and fluorescent microscopy imaging experiments.
Collapse
Affiliation(s)
- Jan Pascal Kahler
- Laboratory of Chemical Biology, Department of Cellular and Molecular Medicine, KU Leuven Herestraat 49 Box 802 3000 Leuven Belgium
| | - Steven H L Verhelst
- Laboratory of Chemical Biology, Department of Cellular and Molecular Medicine, KU Leuven Herestraat 49 Box 802 3000 Leuven Belgium
- Leibniz Institute for Analytical Sciences ISAS Otto-Hahn-Strasse 6b 44227 Dortmund Germany
| |
Collapse
|
32
|
Saidi A, Wartenberg M, Madinier JB, Ilango G, Seren S, Korkmaz B, Lecaille F, Aucagne V, Lalmanach G. Monitoring Human Neutrophil Activation by a Proteinase 3 Near-Infrared Fluorescence Substrate-Based Probe. Bioconjug Chem 2021; 32:1782-1790. [PMID: 34269060 DOI: 10.1021/acs.bioconjchem.1c00267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A near-infrared fluorescent (NIRF) substrate-based probe (SBP) was conceived to monitor secreted human proteinase 3 (hPR3) activity. This probe, called pro3-SBP, is shaped by a fused peptide hairpin loop structure, which associates a hPR3 recognition domain (Val-Ala-Asp-Nva-Ala-Asp-Tyr-Gln, where Nva is norvaline) and an electrostatic zipper (consisting of complementary polyanionic (d-Glu)5 and polycationic (d-Arg)5 sequences) in close vicinity of the N- and C-terminal FRET couple (fluorescent donor, sulfoCy5.5; dark quencher, QSY21). Besides its subsequent stability, no intermolecular fluorescence quenching was detected following its complete hydrolysis by hPR3, advocating that pro3-SBP could further afford unbiased imaging. Pro3-SBP was specifically hydrolyzed by hPR3 (kcat/Km= 440 000 ± 5500 M-1·s-1) and displayed a sensitive detection threshold for hPR3 (subnanomolar concentration range), while neutrophil elastase showed a weaker potency. Conversely, pro3-SBP was not cleaved by cathepsin G. Pro3-SBP was successfully hydrolyzed by conditioned media of activated human neutrophils but not by quiescent neutrophils. Moreover, unlike unstimulated neutrophils, a strong NIRF signal was specifically detected by confocal microscopy following neutrophil ionomycin-induced degranulation. Fluorescence release was abolished in the presence of a selective hPR3 inhibitor, indicating that pro3-SBP is selectively cleaved by extracellular hPR3. Taken together, the present data support that pro3-SBP could be a convenient tool, allowing straightforward monitoring of human neutrophil activation.
Collapse
Affiliation(s)
- Ahlame Saidi
- Université de Tours, Tours 37032, France.,UMR 1100, Research Center for Respiratory Diseases (CEPR), Team: "Proteolytic Mechanisms in Inflammation", INSERM, Tours 37032, France
| | - Mylène Wartenberg
- Université de Tours, Tours 37032, France.,UMR 1100, Research Center for Respiratory Diseases (CEPR), Team: "Proteolytic Mechanisms in Inflammation", INSERM, Tours 37032, France
| | - Jean-Baptiste Madinier
- Center for Molecular Biophysics (CBM), Team: "Molecular, Structural and Chemical Biology″, CNRS UPR 4301, Orléans 45071, France
| | - Guy Ilango
- IBiSA Electron Microscopy Platform, Université de Tours, Tours 37032, France
| | - Seda Seren
- Université de Tours, Tours 37032, France.,UMR 1100, Research Center for Respiratory Diseases (CEPR), Team: "Proteolytic Mechanisms in Inflammation", INSERM, Tours 37032, France
| | - Brice Korkmaz
- Université de Tours, Tours 37032, France.,UMR 1100, Research Center for Respiratory Diseases (CEPR), Team: "Proteolytic Mechanisms in Inflammation", INSERM, Tours 37032, France
| | - Fabien Lecaille
- Université de Tours, Tours 37032, France.,UMR 1100, Research Center for Respiratory Diseases (CEPR), Team: "Proteolytic Mechanisms in Inflammation", INSERM, Tours 37032, France
| | - Vincent Aucagne
- Center for Molecular Biophysics (CBM), Team: "Molecular, Structural and Chemical Biology″, CNRS UPR 4301, Orléans 45071, France
| | - Gilles Lalmanach
- Université de Tours, Tours 37032, France.,UMR 1100, Research Center for Respiratory Diseases (CEPR), Team: "Proteolytic Mechanisms in Inflammation", INSERM, Tours 37032, France
| |
Collapse
|
33
|
Lovell S, Zhang L, Kryza T, Neodo A, Bock N, De Vita E, Williams ED, Engelsberger E, Xu C, Bakker AT, Maneiro M, Tanaka RJ, Bevan CL, Clements JA, Tate EW. A Suite of Activity-Based Probes To Dissect the KLK Activome in Drug-Resistant Prostate Cancer. J Am Chem Soc 2021; 143:8911-8924. [PMID: 34085829 PMCID: PMC9282638 DOI: 10.1021/jacs.1c03950] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
![]()
Kallikrein-related
peptidases (KLKs) are a family of secreted serine
proteases, which form a network (the KLK activome) with an important
role in proteolysis and signaling. In prostate cancer (PCa), increased
KLK activity promotes tumor growth and metastasis through multiple
biochemical pathways, and specific quantification and tracking of
changes in the KLK activome could contribute to validation of KLKs
as potential drug targets. Herein we report a technology platform
based on novel activity-based probes (ABPs) and inhibitors enabling
simultaneous orthogonal analysis of KLK2, KLK3, and KLK14 activity
in hormone-responsive PCa cell lines and tumor homogenates. Importantly,
we identifed a significant decoupling of KLK activity and abundance
and suggest that KLK proteolysis should be considered as an additional
parameter, along with the PSA blood test, for accurate PCa diagnosis
and monitoring. Using selective inhibitors and multiplexed fluorescent
activity-based protein profiling (ABPP), we dissect the KLK activome
in PCa cells and show that increased KLK14 activity leads to a migratory
phenotype. Furthermore, using biotinylated ABPs, we show that active
KLK molecules are secreted into the bone microenvironment by PCa cells
following stimulation by osteoblasts suggesting KLK-mediated signaling
mechanisms could contribute to PCa metastasis to bone. Together our
findings show that ABPP is a powerful approach to dissect dysregulation
of the KLK activome as a promising and previously underappreciated
therapeutic target in advanced PCa.
Collapse
Affiliation(s)
- Scott Lovell
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| | - Leran Zhang
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| | - Thomas Kryza
- Mater Research Institute-The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia.,Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Institute of Health & Biomedical Innovation and School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Anna Neodo
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| | - Nathalie Bock
- Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Institute of Health & Biomedical Innovation and School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Elena De Vita
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| | - Elizabeth D Williams
- Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Institute of Health & Biomedical Innovation and School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Elisabeth Engelsberger
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| | - Congyi Xu
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| | - Alexander T Bakker
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| | - Maria Maneiro
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K
| | - Reiko J Tanaka
- Department of Bioengineering, Imperial College London, London SW7 2AZ, U.K
| | - Charlotte L Bevan
- Department of Surgery and Cancer, Imperial Centre for Translational and Experimental Medicine, Imperial College London, Hammersmith Hospital, Du Cane Road, London W12 0NN, U.K
| | - Judith A Clements
- Australian Prostate Cancer Research Centre-Queensland (APCRC-Q), Institute of Health & Biomedical Innovation and School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Edward W Tate
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London W12 0BZ, U.K.,The Francis Crick Institute, London NW1 1AT, U.K
| |
Collapse
|
34
|
Kasperkiewicz P. Peptidyl Activity-Based Probes for Imaging Serine Proteases. Front Chem 2021; 9:639410. [PMID: 33996745 PMCID: PMC8117214 DOI: 10.3389/fchem.2021.639410] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 04/15/2021] [Indexed: 01/12/2023] Open
Abstract
Proteases catalyze the hydrolysis of peptide bonds. Products of this breakdown mediate signaling in an enormous number of biological processes. Serine proteases constitute the most numerous group of proteases, accounting for 40%, and they are prevalent in many physiological functions, both normal and disease-related functions, making them one of the most important enzymes in humans. The activity of proteases is controlled at the expression level by posttranslational modifications and/or endogenous inhibitors. The study of serine proteases requires specific reagents not only for detecting their activity but also for their imaging. Such tools include inhibitors or substrate-related chemical molecules that allow the detection of proteolysis and visual observation of active enzymes, thus facilitating the characterization of the activity of proteases in the complex proteome. Peptidyl activity-based probes (ABPs) have been extensively studied recently, and this review describes the basic principles in the design of peptide-based imaging agents for serine proteases, provides examples of activity-based probe applications and critically discusses their strengths, weaknesses, challenges and limitations.
Collapse
Affiliation(s)
- Paulina Kasperkiewicz
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology, Wroclaw, Poland
| |
Collapse
|
35
|
Kim HR, Tagirasa R, Yoo E. Covalent Small Molecule Immunomodulators Targeting the Protease Active Site. J Med Chem 2021; 64:5291-5322. [PMID: 33904753 DOI: 10.1021/acs.jmedchem.1c00172] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Cells of the immune system utilize multiple proteases to regulate cell functions and orchestrate innate and adaptive immune responses. Dysregulated protease activities are implicated in many immune-related disorders; thus, protease inhibitors have been actively investigated for pharmaceutical development. Although historically considered challenging with concerns about toxicity, compounds that covalently modify the protease active site represent an important class of agents, emerging not only as chemical probes but also as approved drugs. Here, we provide an overview of technologies useful for the study of proteases with the focus on recent advances in chemoproteomic methods and screening platforms. By highlighting covalent inhibitors that have been designed to target immunomodulatory proteases, we identify opportunities for the development of small molecule immunomodulators.
Collapse
Affiliation(s)
- Hong-Rae Kim
- Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Ravichandra Tagirasa
- Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Euna Yoo
- Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| |
Collapse
|
36
|
Lentz CS. What you see is what you get: activity-based probes in single-cell analysis of enzymatic activities. Biol Chem 2021; 401:233-248. [PMID: 31939273 DOI: 10.1515/hsz-2019-0262] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 06/25/2019] [Indexed: 11/15/2022]
Abstract
Molecular imaging methods can provide spatio-temporal information about the distribution of biomolecules or biological processes, such as certain enzymatic activities, in single cells. Within a cell, it is possible to define the subcellular location of a target, its trafficking through the cell, colocalization with other biomolecules of interest and involvement in certain cell biological processes. On the other hand, single-cell imaging promises to distinguish cells that are phenotypically different from each other. The corresponding cellular diversity comprises the presence of functionally distinct cells in a population ('phenotypic heterogeneity'), as well as dynamic cellular responses to external stimuli ('phenotypic plasticity'), which is highly relevant, e.g. during cell differentiation, activation (of immune cells), or cell death. This review focuses on applications of a certain class of chemical probes, the so-called activity-based probes (ABPs), for visualization of enzymatic activities in the single-cell context. It discusses the structure of ABPs and other chemical probes, exemplary applications of ABPs in single-cell studies in human, mouse and bacterial systems and considerations to be made with regard to data interpretation.
Collapse
Affiliation(s)
- Christian S Lentz
- Department of Chemical Biology (CBIO), Helmholtz-Centre for Infection Research, Inhoffenstr. 7, D-38102 Braunschweig, Germany
| |
Collapse
|
37
|
Kasperkiewicz P, Hempel A, Janiszewski T, Kołt S, Snipas SJ, Drag M, Salvesen GS. NETosis occurs independently of neutrophil serine proteases. J Biol Chem 2021; 295:17624-17631. [PMID: 33454002 DOI: 10.1074/jbc.ra120.015682] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/08/2020] [Indexed: 12/12/2022] Open
Abstract
Neutrophils are primary host innate immune cells defending against pathogens. One proposed mechanism by which neutrophils prevent the spread of pathogens is NETosis, the extrusion of cellular DNA resulting in neutrophil extracellular traps (NETs). The protease neutrophil elastase (NE) has been implicated in the formation of NETs through proteolysis of nuclear proteins leading to chromatin decondensation. In addition to NE, neutrophils contain three other serine proteases that could compensate if the activity of NE was neutralized. However, whether they do play such a role is unknown. Thus, we deployed recently described specific inhibitors against all four of the neutrophil serine proteases (NSPs). Using specific antibodies to the NSPs along with our labeled inhibitors, we show that catalytic activity of these enzymes is not required for the formation of NETs. Moreover, the NSPs that decorate NETs are in an inactive conformation and thus cannot participate in further catalytic events. These results indicate that NSPs play no role in either NETosis or arming NETs with proteolytic activity.
Collapse
Affiliation(s)
- Paulina Kasperkiewicz
- Sanford Burnham Prebys Medical Discovery Institute, NCI-Designated Cancer Center, La Jolla, California, USA; Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology, Wroclaw, Poland.
| | - Anne Hempel
- Sanford Burnham Prebys Medical Discovery Institute, NCI-Designated Cancer Center, La Jolla, California, USA
| | - Tomasz Janiszewski
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Sonia Kołt
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Scott J Snipas
- Sanford Burnham Prebys Medical Discovery Institute, NCI-Designated Cancer Center, La Jolla, California, USA
| | - Marcin Drag
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Guy S Salvesen
- Sanford Burnham Prebys Medical Discovery Institute, NCI-Designated Cancer Center, La Jolla, California, USA.
| |
Collapse
|
38
|
Vizovisek M, Ristanovic D, Menghini S, Christiansen MG, Schuerle S. The Tumor Proteolytic Landscape: A Challenging Frontier in Cancer Diagnosis and Therapy. Int J Mol Sci 2021; 22:ijms22052514. [PMID: 33802262 PMCID: PMC7958950 DOI: 10.3390/ijms22052514] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/24/2021] [Accepted: 02/25/2021] [Indexed: 02/06/2023] Open
Abstract
In recent decades, dysregulation of proteases and atypical proteolysis have become increasingly recognized as important hallmarks of cancer, driving community-wide efforts to explore the proteolytic landscape of oncologic disease. With more than 100 proteases currently associated with different aspects of cancer development and progression, there is a clear impetus to harness their potential in the context of oncology. Advances in the protease field have yielded technologies enabling sensitive protease detection in various settings, paving the way towards diagnostic profiling of disease-related protease activity patterns. Methods including activity-based probes and substrates, antibodies, and various nanosystems that generate reporter signals, i.e., for PET or MRI, after interaction with the target protease have shown potential for clinical translation. Nevertheless, these technologies are costly, not easily multiplexed, and require advanced imaging technologies. While the current clinical applications of protease-responsive technologies in oncologic settings are still limited, emerging technologies and protease sensors are poised to enable comprehensive exploration of the tumor proteolytic landscape as a diagnostic and therapeutic frontier. This review aims to give an overview of the most relevant classes of proteases as indicators for tumor diagnosis, current approaches to detect and monitor their activity in vivo, and associated therapeutic applications.
Collapse
|
39
|
Burster T, Gärtner F, Knippschild U, Zhanapiya A. Activity-Based Probes to Utilize the Proteolytic Activity of Cathepsin G in Biological Samples. Front Chem 2021; 9:628295. [PMID: 33732686 PMCID: PMC7959752 DOI: 10.3389/fchem.2021.628295] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 01/07/2021] [Indexed: 12/30/2022] Open
Abstract
Neutrophils, migrating to the site of infection, are able to release serine proteases after being activated. These serine proteases comprise cathepsin G (CatG), neutrophil elastase protease 3 (PR3), and neutrophil serine protease 4 (NSP4). A disadvantage of the uncontrolled proteolytic activity of proteases is the outcome of various human diseases, including cardiovascular diseases, thrombosis, and autoimmune diseases. Activity-based probes (ABPs) are used to determine the proteolytic activity of proteases, containing a set of three essential elements: Warhead, recognition sequence, and the reporter tag for detection of the covalent enzyme activity–based probe complex. Here, we summarize the latest findings of ABP-mediated detection of proteases in both locations intracellularly and on the cell surface of cells, thereby focusing on CatG. Particularly, application of ABPs in regular flow cytometry, imaging flow cytometry, and mass cytometry by time-of-flight (CyTOF) approaches is advantageous when distinguishing between immune cell subsets. ABPs can be included in a vast panel of markers to detect proteolytic activity and determine whether proteases are properly regulated during medication. The use of ABPs as a detection tool opens the possibility to interfere with uncontrolled proteolytic activity of proteases by employing protease inhibitors.
Collapse
Affiliation(s)
- Timo Burster
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Fabian Gärtner
- Department of General and Visceral Surgery, Surgery Center, Ulm University Hospital, Ulm, Germany
| | - Uwe Knippschild
- Department of General and Visceral Surgery, Surgery Center, Ulm University Hospital, Ulm, Germany
| | - Anuar Zhanapiya
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Nur-Sultan, Kazakhstan
| |
Collapse
|
40
|
AhYoung AP, Eckard SC, Gogineni A, Xi H, Lin SJ, Gerhardy S, Cox C, Phung QT, Hackney JA, Katakam AK, Reichelt M, Caplazi P, Manzanillo P, Zhang J, Roose-Girma M, Tam LW, Newman RJ, Murthy A, Weimer RM, Lill JR, Lee WP, Grimbaldeston M, Kirchhofer D, van Lookeren Campagne M. Neutrophil serine protease 4 is required for mast cell-dependent vascular leakage. Commun Biol 2020; 3:687. [PMID: 33214666 PMCID: PMC7677402 DOI: 10.1038/s42003-020-01407-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 10/17/2020] [Indexed: 02/06/2023] Open
Abstract
Vascular leakage, or edema, is a serious complication of acute allergic reactions. Vascular leakage is triggered by the release of histamine and serotonin from granules within tissue-resident mast cells. Here, we show that expression of Neutrophil Serine Protease 4 (NSP4) during the early stages of mast cell development regulates mast cell-mediated vascular leakage. In myeloid precursors, the granulocyte-macrophage progenitors (GMPs), loss of NSP4 results in the decrease of cellular levels of histamine, serotonin and heparin/heparan sulfate. Mast cells that are derived from NSP4-deficient GMPs have abnormal secretory granule morphology and a sustained reduction in histamine and serotonin levels. Consequently, in passive cutaneous anaphylaxis and acute arthritis models, mast cell-mediated vascular leakage in the skin and joints is substantially reduced in NSP4-deficient mice. Our findings reveal that NSP4 is required for the proper storage of vasoactive amines in mast cell granules, which impacts mast cell-dependent vascular leakage in mouse models of immune complex-mediated diseases.
Collapse
Affiliation(s)
- Andrew P AhYoung
- Department of Early Discovery Biochemistry, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Sterling C Eckard
- Department of Immunology, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Alvin Gogineni
- Department of Biomedical Imaging, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Hongkang Xi
- Department of Immunology, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - S Jack Lin
- Department of Early Discovery Biochemistry, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Stefan Gerhardy
- Department of Early Discovery Biochemistry, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Christian Cox
- Department of Immunology, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Qui T Phung
- Department of Microchemistry, Proteomics, Lipidomics, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Jason A Hackney
- Department of Bioinformatics, 1 DNA Way, South San Francisco, CA, 94080, USA
| | | | - Mike Reichelt
- Department of Pathology, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Patrick Caplazi
- Department of Pathology, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Paolo Manzanillo
- Department of Immunology, 1 DNA Way, South San Francisco, CA, 94080, USA
- Department of Inflammation and Oncology, Amgen Research, Amgen, 1120 Veterans Boulevard, South San Francisco, CA, 94080, USA
| | - Juan Zhang
- Department of Translational Immunology, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Merone Roose-Girma
- Department of Molecular Biology, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Lucinda W Tam
- Department of Molecular Biology, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Robert J Newman
- Department of Molecular Biology, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Aditya Murthy
- Department of Cancer Immunology, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Robby M Weimer
- Department of Biomedical Imaging, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Jennie R Lill
- Department of Microchemistry, Proteomics, Lipidomics, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Wyne P Lee
- Department of Translational Immunology, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Michele Grimbaldeston
- OMNI-Biomarker Development, Genentech Inc, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Daniel Kirchhofer
- Department of Early Discovery Biochemistry, 1 DNA Way, South San Francisco, CA, 94080, USA.
| | - Menno van Lookeren Campagne
- Department of Immunology, 1 DNA Way, South San Francisco, CA, 94080, USA.
- Department of Inflammation and Oncology, Amgen Research, Amgen, 1120 Veterans Boulevard, South San Francisco, CA, 94080, USA.
| |
Collapse
|
41
|
Hira J, Uddin MJ, Haugland MM, Lentz CS. From Differential Stains to Next Generation Physiology: Chemical Probes to Visualize Bacterial Cell Structure and Physiology. Molecules 2020; 25:E4949. [PMID: 33114655 PMCID: PMC7663024 DOI: 10.3390/molecules25214949] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 12/16/2022] Open
Abstract
Chemical probes have been instrumental in microbiology since its birth as a discipline in the 19th century when chemical dyes were used to visualize structural features of bacterial cells for the first time. In this review article we will illustrate the evolving design of chemical probes in modern chemical biology and their diverse applications in bacterial imaging and phenotypic analysis. We will introduce and discuss a variety of different probe types including fluorogenic substrates and activity-based probes that visualize metabolic and specific enzyme activities, metabolic labeling strategies to visualize structural features of bacterial cells, antibiotic-based probes as well as fluorescent conjugates to probe biomolecular uptake pathways.
Collapse
Affiliation(s)
- Jonathan Hira
- Research Group for Host-Microbe Interactions, Department of Medical Biology and Centre for New Antibacterial Strategies (CANS), UiT—The Arctic University of Norway, 9019 Tromsø, Norway; (J.H.); (M.J.U.)
| | - Md. Jalal Uddin
- Research Group for Host-Microbe Interactions, Department of Medical Biology and Centre for New Antibacterial Strategies (CANS), UiT—The Arctic University of Norway, 9019 Tromsø, Norway; (J.H.); (M.J.U.)
| | - Marius M. Haugland
- Department of Chemistry and Centre for New Antibacterial Strategies (CANS), UiT—The Arctic University of Norway, 9019 Tromsø, Norway;
| | - Christian S. Lentz
- Research Group for Host-Microbe Interactions, Department of Medical Biology and Centre for New Antibacterial Strategies (CANS), UiT—The Arctic University of Norway, 9019 Tromsø, Norway; (J.H.); (M.J.U.)
| |
Collapse
|
42
|
Rut W, Groborz K, Zhang L, Sun X, Zmudzinski M, Pawlik B, Wang X, Jochmans D, Neyts J, Młynarski W, Hilgenfeld R, Drag M. SARS-CoV-2 M pro inhibitors and activity-based probes for patient-sample imaging. Nat Chem Biol 2020; 17:222-228. [PMID: 33093684 DOI: 10.1038/s41589-020-00689-z] [Citation(s) in RCA: 208] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 09/23/2020] [Accepted: 10/05/2020] [Indexed: 12/12/2022]
Abstract
In December 2019, the first cases of infection with a novel coronavirus, SARS-CoV-2, were diagnosed. Currently, there is no effective antiviral treatment for COVID-19. To address this emerging problem, we focused on the SARS-CoV-2 main protease that constitutes one of the most attractive antiviral drug targets. We have synthesized a combinatorial library of fluorogenic substrates with glutamine in the P1 position. We used it to determine the substrate preferences of the SARS-CoV and SARS-CoV-2 main proteases. On the basis of these findings, we designed and synthesized a potent SARS-CoV-2 inhibitor (Ac-Abu-DTyr-Leu-Gln-VS, half-maximal effective concentration of 3.7 µM) and two activity-based probes, for one of which we determined the crystal structure of its complex with the SARS-CoV-2 Mpro. We visualized active SARS-CoV-2 Mpro in nasopharyngeal epithelial cells of patients suffering from COVID-19 infection. The results of our work provide a structural framework for the design of inhibitors as antiviral agents and/or diagnostic tests.
Collapse
Affiliation(s)
- Wioletta Rut
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology, Wroclaw, Poland.
| | - Katarzyna Groborz
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Linlin Zhang
- Institute of Molecular Medicine, University of Lübeck, Lübeck, Germany.,Institute of Biochemistry, Center for Structural and Cell Biology in Medicine, University of Lübeck, Lübeck, Germany.,German Center for Infection Research (DZIF), Hamburg-Lübeck-Borstel-Riems Site, University of Lübeck, Lübeck, Germany
| | - Xinyuanyuan Sun
- Institute of Molecular Medicine, University of Lübeck, Lübeck, Germany.,Institute of Biochemistry, Center for Structural and Cell Biology in Medicine, University of Lübeck, Lübeck, Germany.,German Center for Infection Research (DZIF), Hamburg-Lübeck-Borstel-Riems Site, University of Lübeck, Lübeck, Germany
| | - Mikolaj Zmudzinski
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Bartlomiej Pawlik
- Department of Pediatrics, Oncology & Hematology, Medical University of Lodz, Lodz, Poland.,Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Xinyu Wang
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Dirk Jochmans
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Johan Neyts
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Wojciech Młynarski
- Department of Pediatrics, Oncology & Hematology, Medical University of Lodz, Lodz, Poland
| | - Rolf Hilgenfeld
- Institute of Molecular Medicine, University of Lübeck, Lübeck, Germany.,German Center for Infection Research (DZIF), Hamburg-Lübeck-Borstel-Riems Site, University of Lübeck, Lübeck, Germany.,Center for Brain, Behavior, and Metabolism, University of Lübeck, Lübeck, Germany
| | - Marcin Drag
- Department of Chemical Biology and Bioimaging, Wroclaw University of Science and Technology, Wroclaw, Poland.
| |
Collapse
|
43
|
Ahmad J, Ikram S, Hafeez AB, Durdagi S. Physics-driven identification of clinically approved and investigation drugs against human neutrophil serine protease 4 (NSP4): A virtual drug repurposing study. J Mol Graph Model 2020; 101:107744. [PMID: 33032202 DOI: 10.1016/j.jmgm.2020.107744] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 08/29/2020] [Accepted: 08/31/2020] [Indexed: 01/19/2023]
Abstract
Neutrophils synthesize four immune associated serine proteases: Cathepsin G (CTSG), Elastase (ELANE), Proteinase 3 (PRTN3) and Neutrophil Serine Protease 4 (NSP4). While previously considered to be immune modulators, overexpression of neutrophil serine proteases correlates with various disease conditions. Therefore, identifying novel small molecules that can potentially control or inhibit the proteolytic activity of these proteases is crucial to revert or temper the aggravated disease phenotype. To the best of our knowledge, although there is limited data for inhibitors of other neutrophil protease members, there is no previous clinical study of a synthetic small molecule inhibitor targeting NSP4. In this study, an integrated molecular modeling algorithm was performed within a virtual drug repurposing study to identify novel inhibitors for NSP4, using clinically approved and investigation drugs library (∼8000 compounds). Based on our rigorous filtration, we found that following molecules Becatecarin, Iogulamide, Delprostenate and Iralukast are predicted to block the activity of NSP4 by interacting with core catalytic residues. The selected ligands were energetically more favorable compared to the reference molecule. The result of this study identifies promising molecules as potential lead candidates.
Collapse
Affiliation(s)
- Jamshaid Ahmad
- Center of Biotechnology & Microbiology, University of Peshawar, Pakistan.
| | - Saima Ikram
- Center of Biotechnology & Microbiology, University of Peshawar, Pakistan; Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahcesehir University, Istanbul, Turkey
| | - Ahmer Bin Hafeez
- Center of Biotechnology & Microbiology, University of Peshawar, Pakistan
| | - Serdar Durdagi
- Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahcesehir University, Istanbul, Turkey.
| |
Collapse
|
44
|
Poreba M, Groborz KM, Rut W, Pore M, Snipas SJ, Vizovisek M, Turk B, Kuhn P, Drag M, Salvesen GS. Multiplexed Probing of Proteolytic Enzymes Using Mass Cytometry-Compatible Activity-Based Probes. J Am Chem Soc 2020; 142:16704-16715. [PMID: 32870676 PMCID: PMC7595764 DOI: 10.1021/jacs.0c06762] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The subset of the proteome that contains enzymes in their catalytically active form can be interrogated by using probes targeted toward individual specific enzymes. A subset of such enzymes are proteases that are frequently studied with activity-based probes, small inhibitors equipped with a detectable tag, commonly a fluorophore. Due to the spectral overlap of these commonly used fluorophores, multiplex analysis becomes limited. To overcome this, we developed a series of protease-selective lanthanide-labeled probes compatible with mass cytometry giving us the ability to monitor the activity of multiple proteases in parallel. Using these probes, we were able to identify the distribution of four proteases with different active site geometries in three cell lines and peripheral blood mononuclear cells. This provides a framework for the use of mass cytometry for multiplexed enzyme activity detection.
Collapse
Affiliation(s)
- Marcin Poreba
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Katarzyna M. Groborz
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Wioletta Rut
- Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Milind Pore
- University of Southern California, USC Michelson Center for Convergent Biosciences, Los Angeles, CA, USA
| | - Scott J. Snipas
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | | | - Boris Turk
- Jozef Stefan Institute, Ljubljana, Slovenia
| | - Peter Kuhn
- University of Southern California, USC Michelson Center for Convergent Biosciences, Los Angeles, CA, USA
| | - Marcin Drag
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Guy S. Salvesen
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| |
Collapse
|
45
|
Kahler JP, Lenders S, van de Plassche MAT, Verhelst SHL. Facile Synthesis of Aminomethyl Phosphinate Esters as Serine Protease Inhibitors with Primed Site Interaction. ACS Med Chem Lett 2020; 11:1739-1744. [PMID: 32944141 DOI: 10.1021/acsmedchemlett.0c00284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 08/10/2020] [Indexed: 12/22/2022] Open
Abstract
Serine proteases comprise about one-third of all proteases, and defective regulation of serine proteases is involved in numerous diseases. Therefore, serine protease inhibitors are promising drug candidates. Aminomethyl diphenyl phosphonates have been regularly used as scaffolds for covalent serine protease inhibition and the design of activity-based probes. However, they cannot make use of a protease's primed site. Therefore, we developed a facile two-step synthesis toward a set of phenyl phosphinates, which is a related scaffold but can interact with the primed site. We tested their inhibitory activity on five different serine proteases and found that a phenyl group directly attached to the phosphorus atom leads to superior activity compared with phosphonates.
Collapse
Affiliation(s)
- Jan Pascal Kahler
- Laboratory of Chemical Biology, Department of Cellular and Molecular Medicine, KU Leuven − University of Leuven, Herestraat 49 Box
802, 3000 Leuven, Belgium
| | - Stijn Lenders
- Laboratory of Chemical Biology, Department of Cellular and Molecular Medicine, KU Leuven − University of Leuven, Herestraat 49 Box
802, 3000 Leuven, Belgium
| | - Merel A. T. van de Plassche
- Laboratory of Chemical Biology, Department of Cellular and Molecular Medicine, KU Leuven − University of Leuven, Herestraat 49 Box
802, 3000 Leuven, Belgium
| | - Steven H. L. Verhelst
- Laboratory of Chemical Biology, Department of Cellular and Molecular Medicine, KU Leuven − University of Leuven, Herestraat 49 Box
802, 3000 Leuven, Belgium
- AG Chemical Proteomics, Leibniz Institute for Analytical Sciences − ISAS, Otto-Hahn-Str. 6b, 44227 Dortmund, Germany
| |
Collapse
|
46
|
Faucher F, Bennett JM, Bogyo M, Lovell S. Strategies for Tuning the Selectivity of Chemical Probes that Target Serine Hydrolases. Cell Chem Biol 2020; 27:937-952. [PMID: 32726586 PMCID: PMC7484133 DOI: 10.1016/j.chembiol.2020.07.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 07/06/2020] [Accepted: 07/08/2020] [Indexed: 02/08/2023]
Abstract
Serine hydrolases comprise a large family of enzymes that have diverse roles in key cellular processes, such as lipid metabolism, cell signaling, and regulation of post-translation modifications of proteins. They are also therapeutic targets for multiple human pathologies, including viral infection, diabetes, hypertension, and Alzheimer disease; however, few have well-defined substrates and biological functions. Activity-based probes (ABPs) have been used as effective tools to both profile activity and screen for selective inhibitors of serine hydrolases. One broad-spectrum ABP containing a fluorophosphonate electrophile has been used extensively to advance our understanding of diverse serine hydrolases. Due to the success of this single reagent, several robust chemistries have been developed to further diversify and tune the selectivity of ABPs used to target serine hydrolases. In this review, we highlight approaches to identify selective serine hydrolase ABPs and suggest new synthetic methodologies that could be applied to further advance probe development.
Collapse
Affiliation(s)
- Franco Faucher
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - John M Bennett
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Matthew Bogyo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Scott Lovell
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
47
|
Kahler JP, Vanhoutte R, Verhelst SHL. Activity-Based Protein Profiling of Serine Proteases in Immune Cells. Arch Immunol Ther Exp (Warsz) 2020; 68:23. [DOI: 10.1007/s00005-020-00586-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 06/11/2020] [Indexed: 12/14/2022]
|
48
|
Rut W, Zmudzinski M, Snipas SJ, Bekes M, Huang TT, Drag M. Engineered unnatural ubiquitin for optimal detection of deubiquitinating enzymes. Chem Sci 2020; 11:6058-6069. [PMID: 32953009 PMCID: PMC7477763 DOI: 10.1039/d0sc01347a] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 05/27/2020] [Indexed: 12/15/2022] Open
Abstract
Herein we present a workflow for design and synthesis of novel selective Ub-based tools for DUBs. Selectivity is achieved by incorporation of unnatural amino acids into the Ub C-terminal epitope.
Deubiquitinating enzymes (DUBs) are responsible for removing ubiquitin (Ub) from its protein conjugates. DUBs have been implicated as attractive therapeutic targets in the treatment of viral diseases, neurodegenerative disorders and cancer. The lack of selective chemical tools for the exploration of these enzymes significantly impairs the determination of their roles in both normal and pathological states. Commercially available fluorogenic substrates are based on the C-terminal Ub motif or contain Ub coupled to a fluorophore (Z-LRGG-AMC, Ub-AMC); therefore, these substrates suffer from lack of selectivity. By using a hybrid combinatorial substrate library (HyCoSuL) and a defined P2 library containing a wide variety of nonproteinogenic amino acids, we established a full substrate specificity profile for two DUBs—MERS PLpro and human UCH-L3. Based on these results, we designed and synthesized Ub-based substrates and activity-based probes (ABPs) containing selected unnatural amino acids located in the C-terminal Ub motif. Biochemical analysis and cell lysate experiments confirmed the activity and selectivity of engineered Ub-based substrates and probes. Using this approach, we propose that for any protease that recognizes Ub and Ub-like substrates, a highly active and selective unnatural substrate or probe can be engineered.
Collapse
Affiliation(s)
- Wioletta Rut
- Department of Chemical Biology and Bioimaging , Wroclaw University of Science and Technology , Wyb. Wyspianskiego 27 , 50-370 Wroclaw , Poland . ;
| | - Mikolaj Zmudzinski
- Department of Chemical Biology and Bioimaging , Wroclaw University of Science and Technology , Wyb. Wyspianskiego 27 , 50-370 Wroclaw , Poland . ;
| | - Scott J Snipas
- Sanford Burnham Prebys Medical Discovery Institute , 10901 North Torrey Pines Road , La Jolla , CA 92037 , USA
| | - Miklos Bekes
- Department of Biochemistry & Molecular Pharmacology , New York University School of Medicine , New York , NY 10016 , USA
| | - Tony T Huang
- Department of Biochemistry & Molecular Pharmacology , New York University School of Medicine , New York , NY 10016 , USA
| | - Marcin Drag
- Department of Chemical Biology and Bioimaging , Wroclaw University of Science and Technology , Wyb. Wyspianskiego 27 , 50-370 Wroclaw , Poland . ; .,Sanford Burnham Prebys Medical Discovery Institute , 10901 North Torrey Pines Road , La Jolla , CA 92037 , USA
| |
Collapse
|
49
|
Janiszewski T, Kołt S, Kaiserman D, Snipas SJ, Li S, Kulbacka J, Saczko J, Bovenschen N, Salvesen G, Drąg M, Bird PI, Kasperkiewicz P. Noninvasive optical detection of granzyme B from natural killer cells with enzyme-activated fluorogenic probes. J Biol Chem 2020; 295:9567-9582. [PMID: 32439802 DOI: 10.1074/jbc.ra120.013204] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 05/11/2020] [Indexed: 12/31/2022] Open
Abstract
Natural killer (NK) cells are key innate immunity effectors that combat viral infections and control several cancer types. For their immune function, human NK cells rely largely on five different cytotoxic proteases, called granzymes (A/B/H/K/M). Granzyme B (GrB) initiates at least three distinct cell death pathways, but key aspects of its function remain unexplored because selective probes that detect its activity are currently lacking. In this study, we used a set of unnatural amino acids to fully map the substrate preferences of GrB, demonstrating previously unknown GrB substrate preferences. We then used these preferences to design substrate-based inhibitors and a GrB-activatable activity-based fluorogenic probe. We show that our GrB probes do not significantly react with caspases, making them ideal for in-depth analyses of GrB localization and function in cells. Using our quenched fluorescence substrate, we observed GrB within the cytotoxic granules of human YT cells. When used as cytotoxic effectors, YT cells loaded with GrB attacked MDA-MB-231 target cells, and active GrB influenced its target cell-killing efficiency. In summary, we have developed a set of molecular tools for investigating GrB function in NK cells and demonstrate noninvasive visual detection of GrB with an enzyme-activated fluorescent substrate.
Collapse
Affiliation(s)
- Tomasz Janiszewski
- Wroclaw University of Science and Technology, Department of Chemical Biology and Bioimaging, Wroclaw, Poland
| | - Sonia Kołt
- Wroclaw University of Science and Technology, Department of Chemical Biology and Bioimaging, Wroclaw, Poland
| | - Dion Kaiserman
- Monash University, Monash Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Clayton, VIC, Australia
| | - Scott J Snipas
- Sanford-Burnham Prebys Medical Discovery Institute, NCI-designated Cancer Center, La Jolla, California, USA
| | - Shuang Li
- University Medical Center Utrecht, Department of Pathology, Utrecht, The Netherlands
| | - Julita Kulbacka
- Wroclaw Medical University, Department of Molecular and Cellular Biology, Wroclaw, Poland
| | - Jolanta Saczko
- Wroclaw Medical University, Department of Molecular and Cellular Biology, Wroclaw, Poland
| | - Niels Bovenschen
- University Medical Center Utrecht, Department of Pathology, Utrecht, The Netherlands
| | - Guy Salvesen
- Sanford-Burnham Prebys Medical Discovery Institute, NCI-designated Cancer Center, La Jolla, California, USA
| | - Marcin Drąg
- Wroclaw University of Science and Technology, Department of Chemical Biology and Bioimaging, Wroclaw, Poland.,Sanford-Burnham Prebys Medical Discovery Institute, NCI-designated Cancer Center, La Jolla, California, USA
| | - Phillip I Bird
- Monash University, Monash Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Clayton, VIC, Australia
| | - Paulina Kasperkiewicz
- Wroclaw University of Science and Technology, Department of Chemical Biology and Bioimaging, Wroclaw, Poland
| |
Collapse
|
50
|
Breidenbach J, Bartz U, Gütschow M. Coumarin as a structural component of substrates and probes for serine and cysteine proteases. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2020; 1868:140445. [PMID: 32405284 PMCID: PMC7219385 DOI: 10.1016/j.bbapap.2020.140445] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/23/2020] [Accepted: 05/07/2020] [Indexed: 02/08/2023]
Abstract
Coumarins represent well-established structures to introduce fluorescence into tool compounds for biochemical investigations. They are valued for their small size, chemical stability and accessibility as well as their tunable photochemical properties. As components of fluorophore/quencher pairs or FRET donor/acceptor pairs, coumarins have frequently been applied in substrate mapping approaches for serine and cysteine proteases. This review also focuses on the incorporation of coumarins into the side chain of amino acids and the exploitation of the resulting fluorescent amino acids for the positional profiling of protease substrates. The protease-inhibiting properties of certain coumarin derivatives and the utilization of coumarin moieties to assemble activity-based probes for serine and cysteine proteases are discussed as well. Coumarins represent well-established structures to introduce fluorescence into tool compounds for biochemical investigations. They are valued for their small size, chemical stability and accessibility as well as their tunable photochemical properties. Coumarins are components of fluorophore/quencher pairs or FRET donor/acceptor pairs in substrate mapping of proteases. Coumarins have been incorporated into amino acids side chains to be used for the positional profiling of protease substrates. Coumarins have protease-inhibiting properties and are used for activity-based probes for serine and cysteine proteases.
Collapse
Affiliation(s)
- Julian Breidenbach
- Pharmaceutical Institute, Department of Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Ulrike Bartz
- Department of Natural Sciences, University of Applied Sciences Bonn-Rhein-Sieg, von-Liebig-Str. 20, 53359 Rheinbach, Germany
| | - Michael Gütschow
- Pharmaceutical Institute, Department of Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany.
| |
Collapse
|