1
|
McKenna SM, Florea BI, Zisterer DM, van Kasteren SI, McGouran JF. Probing the metalloproteome: an 8-mercaptoquinoline motif enriches minichromosome maintenance complex components as significant metalloprotein targets in live cells. RSC Chem Biol 2024; 5:776-786. [PMID: 39092446 PMCID: PMC11289876 DOI: 10.1039/d4cb00053f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 06/18/2024] [Indexed: 08/04/2024] Open
Abstract
Affinity-based probes are valuable tools for detecting binding interactions between small molecules and proteins in complex biological environments. Metalloproteins are a class of therapeutically significant biomolecules which bind metal ions as part of key structural or catalytic domains and are compelling targets for study. However, there is currently a limited range of chemical tools suitable for profiling the metalloproteome. Here, we describe the preparation and application of a novel, photoactivatable affinity-based probe for detection of a subset of previously challenging to engage metalloproteins. The probe, bearing an 8-mercaptoquinoline metal chelator, was anticipated to engage several zinc metalloproteins, including the 26S-proteasome subunit Rpn11. Upon translation of the labelling experiment to mammalian cell lysate and live cell experiments, proteomic analysis revealed that several metalloproteins were competitively enriched. The diazirine probe SMK-24 was found to effectively enrich multiple components of the minichromosome maintenance complex, a zinc metalloprotein assembly with helicase activity essential to DNA replication. Cell cycle analysis experiments revealed that HEK293 cells treated with SMK-24 experienced stalling in G0/G1 phase, consistent with inactivation of the DNA helicase complex. This work represents an important contribution to the library of cell-permeable chemical tools for studying a collection of metalloproteins for which no previous probe existed.
Collapse
Affiliation(s)
- Sean M McKenna
- School of Chemistry, Trinity Biomedical Sciences Institute, Trinity College Dublin 152-160 Pearse St Dublin 2 Ireland
- Synthesis and Solid State Pharmaceutical Centre (SSPC) Ireland
| | - Bogdan I Florea
- Department of Bioorganic Synthesis, Leiden Institute of Chemistry, Leiden University Einsteinweg 55 2333 CC Leiden The Netherlands
| | - Daniela M Zisterer
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin 152-160 Pearse St Dublin 2 Ireland
| | - Sander I van Kasteren
- Department of Bioorganic Synthesis, Leiden Institute of Chemistry, Leiden University Einsteinweg 55 2333 CC Leiden The Netherlands
| | - Joanna F McGouran
- School of Chemistry, Trinity Biomedical Sciences Institute, Trinity College Dublin 152-160 Pearse St Dublin 2 Ireland
- Synthesis and Solid State Pharmaceutical Centre (SSPC) Ireland
| |
Collapse
|
2
|
Walrant A, Sachon E. Photoaffinity labeling coupled to MS to identify peptide biological partners: Secondary reactions, for better or for worse? MASS SPECTROMETRY REVIEWS 2024. [PMID: 38576378 DOI: 10.1002/mas.21880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 02/22/2024] [Accepted: 03/13/2024] [Indexed: 04/06/2024]
Abstract
Affinity photolabeling is a smart method to study noncovalent and transient interactions and provide a submolecular picture of the contacts between interacting partners. In this review, we will focus on the identification of peptide partners using photoaffinity labeling coupled to mass spectrometry in different contexts such as in vitro with a purified potential partner, in model systems such as model membranes, and with live cells using both targeted and nontargeted proteomics studies. Different biological partners will be described, among which glycoconjugates, oligonucleotides, peptides, proteins, and lipids, with the photoreactive label inserted either on the peptide of interest or on the potential partner. Particular attention will be paid to the observation and characterization of specific rearrangements following the photolabeling reaction, which can help characterize photoadducts and provide a better understanding of the interacting systems and environment.
Collapse
Affiliation(s)
- Astrid Walrant
- Laboratoire des Biomolécules, LBM, Sorbonne Université, École normale supérieure, PSL University, CNRS, Paris, France
| | - Emmanuelle Sachon
- Laboratoire des Biomolécules, LBM, Sorbonne Université, École normale supérieure, PSL University, CNRS, Paris, France
- Sorbonne Université, Mass Spectrometry Sciences Sorbonne Université, MS3U platform, Fédération de Chimie moléculaire de Paris centre, Paris, France
| |
Collapse
|
3
|
Georgiadis D, Skoulikas N, Papakyriakou A, Stratikos E. Phosphinic Peptides as Tool Compounds for the Study of Pharmacologically Relevant Zn-Metalloproteases. ACS Pharmacol Transl Sci 2022; 5:1228-1253. [PMID: 36524013 PMCID: PMC9745897 DOI: 10.1021/acsptsci.2c00183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Indexed: 11/29/2022]
Abstract
Phosphinic peptides constitute an important class of bioactive compounds that have found a wide range of applications in the field of biology and pharmacology of Zn-metalloproteases, the largest family of proteases in humans. They are designed to mimic the structure of natural substrates during their proteolysis, thus acting as mechanism-based, transition state analogue inhibitors. A combination of electrostatic interactions between the phosphinic acid group and the Zn cation as well as optimal noncovalent enzyme-ligand interactions can result in both high binding affinity for the desired target and selectivity against other proteases. Due to these unique properties, phosphinic peptides have been mainly employed as tool compounds for (a) the purposes of rational drug design by serving as ligands in X-ray crystal structures of target enzymes and allowing the identification of crucial interactions that govern optimal molecular recognition, and (b) the delineation of biological pathways where Zn-metalloproteases are key regulators. For the latter objective, inhibitors of the phosphinopeptidic type have been used either unmodified or after being transformed to probes of various types, thus expanding the arsenal of functional tools available to researchers. The aim of this review is to summarize all recent research achievements in which phosphinic peptides have played a central role as tool compounds in the understanding of the mechanism and biological functions of Zn-metalloproteases in both health and disease.
Collapse
Affiliation(s)
- Dimitris Georgiadis
- Department
of Chemistry, National and Kapodistrian
University of Athens, GR-15784 Athens, Greece
| | - Nikolaos Skoulikas
- Department
of Chemistry, National and Kapodistrian
University of Athens, GR-15784 Athens, Greece
| | - Athanasios Papakyriakou
- National
Centre for Scientific Research “Demokritos”, Agia Paraskevi GR-15341 Athens, Greece
| | - Efstratios Stratikos
- Department
of Chemistry, National and Kapodistrian
University of Athens, GR-15784 Athens, Greece
- National
Centre for Scientific Research “Demokritos”, Agia Paraskevi GR-15341 Athens, Greece
| |
Collapse
|
4
|
de Almeida LGN, Thode H, Eslambolchi Y, Chopra S, Young D, Gill S, Devel L, Dufour A. Matrix Metalloproteinases: From Molecular Mechanisms to Physiology, Pathophysiology, and Pharmacology. Pharmacol Rev 2022; 74:712-768. [PMID: 35738680 DOI: 10.1124/pharmrev.121.000349] [Citation(s) in RCA: 126] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The first matrix metalloproteinase (MMP) was discovered in 1962 from the tail of a tadpole by its ability to degrade collagen. As their name suggests, matrix metalloproteinases are proteases capable of remodeling the extracellular matrix. More recently, MMPs have been demonstrated to play numerous additional biologic roles in cell signaling, immune regulation, and transcriptional control, all of which are unrelated to the degradation of the extracellular matrix. In this review, we will present milestones and major discoveries of MMP research, including various clinical trials for the use of MMP inhibitors. We will discuss the reasons behind the failures of most MMP inhibitors for the treatment of cancer and inflammatory diseases. There are still misconceptions about the pathophysiological roles of MMPs and the best strategies to inhibit their detrimental functions. This review aims to discuss MMPs in preclinical models and human pathologies. We will discuss new biochemical tools to track their proteolytic activity in vivo and ex vivo, in addition to future pharmacological alternatives to inhibit their detrimental functions in diseases. SIGNIFICANCE STATEMENT: Matrix metalloproteinases (MMPs) have been implicated in most inflammatory, autoimmune, cancers, and pathogen-mediated diseases. Initially overlooked, MMP contributions can be both beneficial and detrimental in disease progression and resolution. Thousands of MMP substrates have been suggested, and a few hundred have been validated. After more than 60 years of MMP research, there remain intriguing enigmas to solve regarding their biological functions in diseases.
Collapse
Affiliation(s)
- Luiz G N de Almeida
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Hayley Thode
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Yekta Eslambolchi
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Sameeksha Chopra
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Daniel Young
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Sean Gill
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Laurent Devel
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Antoine Dufour
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| |
Collapse
|
5
|
Kaminska M, Bruyat P, Malgorn C, Doladilhe M, Cassar‐Lajeunesse E, Fruchart Gaillard C, De Souza M, Beau F, Thai R, Correia I, Galat A, Georgiadis D, Lequin O, Dive V, Bregant S, Devel L. Ligand‐Directed Modification of Active Matrix Metalloproteases: Activity‐based Probes with no Photolabile Group. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202106117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Monika Kaminska
- Université Paris-Saclay CEA, INRAE, Medicaments et Technologies pour la Sante (MTS), SIMoS 91191 Gif-sur-Yvette France
| | - Pierrick Bruyat
- Université Paris-Saclay CEA, INRAE, Medicaments et Technologies pour la Sante (MTS), SIMoS 91191 Gif-sur-Yvette France
| | - Carole Malgorn
- Université Paris-Saclay CEA, INRAE, Medicaments et Technologies pour la Sante (MTS), SIMoS 91191 Gif-sur-Yvette France
| | - Marion Doladilhe
- Université Paris-Saclay CEA, INRAE, Medicaments et Technologies pour la Sante (MTS), SIMoS 91191 Gif-sur-Yvette France
| | - Evelyne Cassar‐Lajeunesse
- Université Paris-Saclay CEA, INRAE, Medicaments et Technologies pour la Sante (MTS), SIMoS 91191 Gif-sur-Yvette France
| | - Carole Fruchart Gaillard
- Université Paris-Saclay CEA, INRAE, Medicaments et Technologies pour la Sante (MTS), SIMoS 91191 Gif-sur-Yvette France
| | - Mélissa De Souza
- Université Paris-Saclay CEA, INRAE, Medicaments et Technologies pour la Sante (MTS), SIMoS 91191 Gif-sur-Yvette France
| | - Fabrice Beau
- Université Paris-Saclay CEA, INRAE, Medicaments et Technologies pour la Sante (MTS), SIMoS 91191 Gif-sur-Yvette France
| | - Robert Thai
- Université Paris-Saclay CEA, INRAE, Medicaments et Technologies pour la Sante (MTS), SIMoS 91191 Gif-sur-Yvette France
| | - Isabelle Correia
- CNRS, Laboratoire des Biomolécules, LBM Sorbonne Université Ecole Normale Supérieure PSL University 75005 Paris France
| | - Andrzej Galat
- Université Paris-Saclay CEA, INRAE, Medicaments et Technologies pour la Sante (MTS), SIMoS 91191 Gif-sur-Yvette France
| | - Dimitris Georgiadis
- Department of Chemistry Laboratory of Organic Chemistry University of Athens Panepistimiopolis Zografou 15771 Athens Greece
| | - Olivier Lequin
- CNRS, Laboratoire des Biomolécules, LBM Sorbonne Université Ecole Normale Supérieure PSL University 75005 Paris France
| | - Vincent Dive
- Université Paris-Saclay CEA, INRAE, Medicaments et Technologies pour la Sante (MTS), SIMoS 91191 Gif-sur-Yvette France
| | - Sarah Bregant
- Université Paris-Saclay CEA, INRAE, Medicaments et Technologies pour la Sante (MTS), SIMoS 91191 Gif-sur-Yvette France
| | - Laurent Devel
- Université Paris-Saclay CEA, INRAE, Medicaments et Technologies pour la Sante (MTS), SIMoS 91191 Gif-sur-Yvette France
| |
Collapse
|
6
|
Kaminska M, Bruyat P, Malgorn C, Doladilhe M, Cassar-Lajeunesse E, Fruchart Gaillard C, De Souza M, Beau F, Thai R, Correia I, Galat A, Georgiadis D, Lequin O, Dive V, Bregant S, Devel L. Ligand-Directed Modification of Active Matrix Metalloproteases: Activity-based Probes with no Photolabile Group. Angew Chem Int Ed Engl 2021; 60:18272-18279. [PMID: 34096148 DOI: 10.1002/anie.202106117] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Indexed: 12/12/2022]
Abstract
Activity-based probes enable discrimination between the active enzyme and its inactive or inactivated counterparts. Since metalloproteases catalysis is non-covalent, activity-based probes targeting them have been systematically developed by decorating reversible inhibitors with photo-crosslinkers. By exploiting two types of ligand-guided chemistry, we identified novel activity-based probes capable of covalently modifying the active site of matrix metalloproteases (MMPs) without any external trigger. The ability of these probes to label recombinant MMPs was validated in vitro and the identity of the main labelling sites within their S3 ' region unambiguously assigned. We also demonstrated that our affinity probes can react with rhMMP12 at nanogram scale (that is, at 0.07 % (w/w)) in complex proteomes. Finally, this ligand-directed chemistry was successfully applied to label active MMP-12 secreted by eukaryote cells. We believe that this approach could be transferred more widely to many other metalloproteases, thus contributing to tackle their unresolved proteomic profiling in vivo.
Collapse
Affiliation(s)
- Monika Kaminska
- Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Sante (MTS), SIMoS, 91191, Gif-sur-Yvette, France
| | - Pierrick Bruyat
- Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Sante (MTS), SIMoS, 91191, Gif-sur-Yvette, France
| | - Carole Malgorn
- Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Sante (MTS), SIMoS, 91191, Gif-sur-Yvette, France
| | - Marion Doladilhe
- Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Sante (MTS), SIMoS, 91191, Gif-sur-Yvette, France
| | - Evelyne Cassar-Lajeunesse
- Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Sante (MTS), SIMoS, 91191, Gif-sur-Yvette, France
| | - Carole Fruchart Gaillard
- Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Sante (MTS), SIMoS, 91191, Gif-sur-Yvette, France
| | - Mélissa De Souza
- Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Sante (MTS), SIMoS, 91191, Gif-sur-Yvette, France
| | - Fabrice Beau
- Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Sante (MTS), SIMoS, 91191, Gif-sur-Yvette, France
| | - Robert Thai
- Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Sante (MTS), SIMoS, 91191, Gif-sur-Yvette, France
| | - Isabelle Correia
- CNRS, Laboratoire des Biomolécules, LBM, Sorbonne Université, Ecole Normale Supérieure, PSL University, 75005, Paris, France
| | - Andrzej Galat
- Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Sante (MTS), SIMoS, 91191, Gif-sur-Yvette, France
| | - Dimitris Georgiadis
- Department of Chemistry, Laboratory of Organic Chemistry, University of Athens, Panepistimiopolis, Zografou, 15771 Athens, Greece
| | - Olivier Lequin
- CNRS, Laboratoire des Biomolécules, LBM, Sorbonne Université, Ecole Normale Supérieure, PSL University, 75005, Paris, France
| | - Vincent Dive
- Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Sante (MTS), SIMoS, 91191, Gif-sur-Yvette, France
| | - Sarah Bregant
- Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Sante (MTS), SIMoS, 91191, Gif-sur-Yvette, France
| | - Laurent Devel
- Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Sante (MTS), SIMoS, 91191, Gif-sur-Yvette, France
| |
Collapse
|
7
|
Talma M, Maślanka M, Mucha A. Recent developments in the synthesis and applications of phosphinic peptide analogs. Bioorg Med Chem Lett 2019; 29:1031-1042. [PMID: 30846252 DOI: 10.1016/j.bmcl.2019.02.034] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 02/25/2019] [Accepted: 02/27/2019] [Indexed: 01/20/2023]
Abstract
Synthetic pseudopeptides that fit well with the active site architecture allow the most effective binding to enzymes, similar to native substrates in high-energy transition states. Phosphinic acid peptide analogs that comprise the tetrahedral phosphorus moiety introduced to replace an internal amide bond exert such an isosteric or isoelectronic resemblance, combined with providing other advantageous features, for example, metal complexing properties. Accordingly, they are capable of inhibiting metal-dependent enzymes involved in biological functions in eukaryotic and prokaryotic cells. These enzymes are associated with notorious human diseases, such as cancer, e.g., matrix metalloproteinases, or are etiological factors of protozoal and bacterial infections, e.g., metalloaminopeptidases. The affinity and selectivity of these compounds can be conveniently adjusted, either by structural modification of dedicated side chains or by backbone elongation to enhance specific interactions with the corresponding binding pockets. Recent approaches to the synthesis of these compounds are illustrated by examples of the preparation of rationally designed structures of inhibitors of particular enzymes. Activity against appealing enzymatic targets is presented, along with the molecular mechanisms of action and therapeutic implications. Innovative aspects of phosphinic peptide application, e.g., as activity-based probes, and ligands of complexes of radioisotopes for nuclear medicine are also outlined.
Collapse
Affiliation(s)
- Michał Talma
- Wrocław University of Science and Technology, Department of Bioorganic Chemistry, Wybrzeże Wyspiańskiego 27, 50-370 Wrocław, Poland
| | - Marta Maślanka
- Wrocław University of Science and Technology, Department of Bioorganic Chemistry, Wybrzeże Wyspiańskiego 27, 50-370 Wrocław, Poland
| | - Artur Mucha
- Wrocław University of Science and Technology, Department of Bioorganic Chemistry, Wybrzeże Wyspiańskiego 27, 50-370 Wrocław, Poland.
| |
Collapse
|
8
|
Zhu Y, Huang RZ, Wang CG, Ouyang XL, Jing XT, Liang D, Wang HS. New inhibitors of matrix metalloproteinases 9 (MMP-9): Lignans from Selaginella moellendorffii. Fitoterapia 2018; 130:281-289. [DOI: 10.1016/j.fitote.2018.09.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 09/16/2018] [Accepted: 09/17/2018] [Indexed: 02/01/2023]
|
9
|
Wang CG, Yao WN, Zhang B, Hua J, Liang D, Wang HS. Lung cancer and matrix metalloproteinases inhibitors of polyphenols from Selaginella tamariscina with suppression activity of migration. Bioorg Med Chem Lett 2018. [DOI: 10.1016/j.bmcl.2018.06.024 pmid: 29921475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
10
|
Lung cancer and matrix metalloproteinases inhibitors of polyphenols from Selaginella tamariscina with suppression activity of migration. Bioorg Med Chem Lett 2018; 28:2413-2417. [DOI: 10.1016/j.bmcl.2018.06.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 06/09/2018] [Accepted: 06/12/2018] [Indexed: 12/15/2022]
|
11
|
Huang RZ, Liang GB, Huang XC, Zhang B, Zhou MM, Liao ZX, Wang HS. Discovery of dehydroabietic acid sulfonamide based derivatives as selective matrix metalloproteinases inactivators that inhibit cell migration and proliferation. Eur J Med Chem 2017; 138:979-992. [PMID: 28756264 DOI: 10.1016/j.ejmech.2017.07.020] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 07/11/2017] [Accepted: 07/12/2017] [Indexed: 01/28/2023]
Abstract
A series of dehydroabietic acid (DHAA) dipeptide derivatives containing the sulfonamide moiety were designed, synthesized and evaluated for inhibition of MMPs as well as the effects of in vitro cell migration. These compounds exhibited relatively good inhibition activity against MMPs with IC50 values in low micromolar range. A docking study of the most active compound 8k revealed key interactions between 8k and MMP-3 in which the sulfonamide moiety and the dipeptide group were important for improving activity. It is noteworthy that further antitumor activity screening revealed that some compounds exhibited better inhibitory activity than the commercial anticancer drug 5-FU. In particular, compound 8k appeared to be the most potent compound against the HepG2 cell line, at least partly, by inhibition of the activity of MMP-3 and apoptosis induction. The treatment of HepG2 cells with compound 8k resulted in inhibition of in vitro cell migration through wound healing assay and G1 phase of cell cycle arrested. In addition, 8k-induced apoptosis was significantly facilitated in HepG2 cells. Thus, we conclude that DHAA dipeptide derivatives containing the sulfonamide moiety may be the potential MMPs inhibitors with the ability to suppress cells migration.
Collapse
Affiliation(s)
- Ri-Zhen Huang
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Gui-Bin Liang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University, Guilin 541004, China
| | - Xiao-Chao Huang
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Bin Zhang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University, Guilin 541004, China
| | - Mei-Mei Zhou
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University, Guilin 541004, China
| | - Zhi-Xin Liao
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China.
| | - Heng-Shan Wang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University, Guilin 541004, China.
| |
Collapse
|
12
|
Huang X, Huang R, Gou S, Wang Z, Wang H. Anticancer Platinum(IV) Prodrugs Containing Monoaminophosphonate Ester as a Targeting Group Inhibit Matrix Metalloproteinases and Reverse Multidrug Resistance. Bioconjug Chem 2017; 28:1305-1323. [PMID: 28276682 DOI: 10.1021/acs.bioconjchem.7b00117] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A novel class of platinum(IV) complexes comprising a monoaminophosphonate ester moiety, which can not only act as a bone-targeting group but also inhibit matrix metalloproteinases (MMPs), were designed and synthesized. Biological assay of these compounds showed that they had potent antitumor activities against the tested cancer cell lines compared with cisplatin and oxaliplatin and indicated low cytotoxicity to human normal liver cells. Particularly, the platinum(IV) complexes were very sensitive to cisplatin resistant cancer cell lines. The corresponding structure-activity relationships were studied and discussed. Related mechanism study revealed that the typical complex 11 caused cell cycle arrest at S phase and induced apoptosis in Bel-7404 cells via a mitochondrial-dependent apoptosis pathway. Moreover, complex 11 had potent ability to inhibit the tumor growth in the NCI-H460 xenograft model comparable to cisplatin.
Collapse
Affiliation(s)
| | | | | | | | - Hengshan Wang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University , Guilin 541004, China
| |
Collapse
|
13
|
Prely L, Klein T, Geurink PP, Paal K, Overkleeft HS, Bischoff R. Activity-Dependent Photoaffinity Labeling of Metalloproteases. Methods Mol Biol 2017; 1491:103-111. [PMID: 27778284 DOI: 10.1007/978-1-4939-6439-0_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Metalloproteases, notably members of the matrix metalloprotease (MMP) and A Disintegrin And Metalloprotease (ADAM) families play crucial roles in tissue remodeling, the liberation of growth factors and cytokines from cell membranes (shedding) and cell-cell or cell-matrix interactions. Activity of MMPs or ADAMs must therefore be tightly controlled in time and space by activation of pro-enzymes upon appropriate stimuli and inhibition by endogenous tissue inhibitors of metalloproteases (TIMPs) or α2-macroglobulin to prevent irreversible tissue damage due to excessive degradation or uncontrolled release of potent inflammatory mediators, such as tumor necrosis factor-α (TNF-α).Although there is a wide range of methods to measure the amount of metalloproteases based on immunological approaches, relatively little is known about the activation status of a given enzyme at any given time and location. This information is, however, critical in order to understand the function and possible implication of these enzymes in disease. Since metalloproteases use an active-site bound water molecule to cleave the peptide bond, it is not possible to apply known active-site-directed labeling approaches with electrophilic "warheads." We therefore developed novel metalloprotease inhibitors that contain a photoactivatable trifluoromethylphenyldiazirine group and show that such inhibitors are suitable for activity-dependent photoaffinity labeling of MMPs and ADAMs.
Collapse
Affiliation(s)
- Laurette Prely
- Department of Pharmacy, Analytical Biochemistry, University of Groningen, Antonius Deusinglaan 1, 9713, AV, Groningen, The Netherlands
| | - Theo Klein
- Department of Pharmacy, Analytical Biochemistry, University of Groningen, Antonius Deusinglaan 1, 9713, AV, Groningen, The Netherlands
| | - Paul P Geurink
- Leiden Institute of Chemistry and the Netherlands Proteomics Center, Leiden University, Einsteinweg 55, 2300, RA, Leiden, The Netherlands
| | - Krisztina Paal
- Department of Pharmacy, Analytical Biochemistry, University of Groningen, Antonius Deusinglaan 1, 9713, AV, Groningen, The Netherlands
| | - Herman S Overkleeft
- Bio-Organic Synthesis Groups, Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2300, RA, Leiden, The Netherlands
| | - Rainer Bischoff
- Department of Pharmacy, Analytical Biochemistry, University of Groningen, Antonius Deusinglaan 1, 9713, AV, Groningen, The Netherlands.
| |
Collapse
|
14
|
Rouanet-Mehouas C, Czarny B, Beau F, Cassar-Lajeunesse E, Stura EA, Dive V, Devel L. Zinc–Metalloproteinase Inhibitors: Evaluation of the Complex Role Played by the Zinc-Binding Group on Potency and Selectivity. J Med Chem 2016; 60:403-414. [DOI: 10.1021/acs.jmedchem.6b01420] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Cecile Rouanet-Mehouas
- Service d’Ingénierie
Moléculaire des Protéines (SIMOPRO), IBITECS, CEA, Université Paris-Saclay, Gif/Yvette F-91191, France
| | - Bertrand Czarny
- Service d’Ingénierie
Moléculaire des Protéines (SIMOPRO), IBITECS, CEA, Université Paris-Saclay, Gif/Yvette F-91191, France
| | - Fabrice Beau
- Service d’Ingénierie
Moléculaire des Protéines (SIMOPRO), IBITECS, CEA, Université Paris-Saclay, Gif/Yvette F-91191, France
| | - Evelyne Cassar-Lajeunesse
- Service d’Ingénierie
Moléculaire des Protéines (SIMOPRO), IBITECS, CEA, Université Paris-Saclay, Gif/Yvette F-91191, France
| | - Enrico A. Stura
- Service d’Ingénierie
Moléculaire des Protéines (SIMOPRO), IBITECS, CEA, Université Paris-Saclay, Gif/Yvette F-91191, France
| | - Vincent Dive
- Service d’Ingénierie
Moléculaire des Protéines (SIMOPRO), IBITECS, CEA, Université Paris-Saclay, Gif/Yvette F-91191, France
| | - Laurent Devel
- Service d’Ingénierie
Moléculaire des Protéines (SIMOPRO), IBITECS, CEA, Université Paris-Saclay, Gif/Yvette F-91191, France
| |
Collapse
|
15
|
Wen H, Qin Y, Zhong W, Li C, Liu X, Shen Y. Trivalent metal ions based on inorganic compounds with in vitro inhibitory activity of matrix metalloproteinase 13. Enzyme Microb Technol 2016; 92:9-17. [PMID: 27542739 DOI: 10.1016/j.enzmictec.2016.06.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 06/07/2016] [Accepted: 06/10/2016] [Indexed: 02/02/2023]
Abstract
Collagenase-3 (MMP-13) inhibitors have attracted considerable attention in recent years and have been developed as a therapeutic target for a variety of diseases, including cancer. Matrix metalloproteinases (MMPs) can be inhibited by a multitude of compounds, including hydroxamic acids. Studies have shown that materials and compounds containing trivalent metal ions, particularly potassium hexacyanoferrate (III) (K3[Fe(CN)6]), exhibit cdMMP-13 inhibitory potential with a half maximal inhibitory concentration (IC50) of 1.3μM. The target protein was obtained by refolding the recombinant histidine-tagged cdMMP-13 using size exclusion chromatography (SEC). The secondary structures of the refolded cdMMP-13 with or without metal ions were further analyzed via circular dichroism and the results indicate that upon binding with metal ions, an altered structure with increased domain stability was obtained. Furthermore, isothermal titration calorimetry (ITC) experiments demonstrated that K3[Fe(CN)6]is able to bind to MMP-13 and endothelial cell tube formation tests provide further evidence for this interaction to exhibit anti-angiogenesis potential. To the best of our knowledge, no previous report of an inorganic compound featuring a MMP-13 inhibitory activity has ever been reported in the literature. Our results demonstrate that K3[Fe(CN)6] is useful as a new effective and specific inhibitor for cdMMP-13 which may be of great potential for future drug screening applications.
Collapse
Affiliation(s)
- Hanyu Wen
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry and Materials Science, Northwest University, Shaanxi Alcohol Ether and Biomass Energy Engineering Research Center, Key laboratory of Yulin Desert Plants Resources, 229 Taibai North Road, Xi'an 710069, PR China
| | - Yuan Qin
- College of Pharmacy, Nankai University, PR China
| | | | - Cong Li
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry and Materials Science, Northwest University, Shaanxi Alcohol Ether and Biomass Energy Engineering Research Center, Key laboratory of Yulin Desert Plants Resources, 229 Taibai North Road, Xi'an 710069, PR China
| | - Xiang Liu
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry and Materials Science, Northwest University, Shaanxi Alcohol Ether and Biomass Energy Engineering Research Center, Key laboratory of Yulin Desert Plants Resources, 229 Taibai North Road, Xi'an 710069, PR China; College of Pharmacy, Nankai University, PR China.
| | - Yehua Shen
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry and Materials Science, Northwest University, Shaanxi Alcohol Ether and Biomass Energy Engineering Research Center, Key laboratory of Yulin Desert Plants Resources, 229 Taibai North Road, Xi'an 710069, PR China; College of Pharmacy, Nankai University, PR China.
| |
Collapse
|
16
|
Zhou B, Yu X, Zhuang C, Villalta P, Lin Y, Lu J, Xing C. Unambiguous Identification of β-Tubulin as the Direct Cellular Target Responsible for the Cytotoxicity of Chalcone by Photoaffinity Labeling. ChemMedChem 2016; 11:1436-45. [PMID: 27203512 DOI: 10.1002/cmdc.201600150] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 04/12/2016] [Indexed: 12/25/2022]
Abstract
Chalcone is a simple and potentially privileged structure in medicinal chemistry with a diverse repertoire of biological activities, among which cytotoxicity is of particular interest. The sharp structure-activity relationship (SAR) for chalcone's cytotoxicity suggests structure-specific target interactions. Despite the numerous putative targets proposed, evidence for direct target interactions in cells is unavailable. In this study, guided by the sharp cytotoxic SAR, we developed a cytotoxic chalcone-based photoaffinity labeling (PAL) probe, (E)-3-(3-azidophenyl)-1-[3,5-dimethoxy-4-(prop-2-yn-1-yloxy)phenyl]-2-methylprop-2-en-1-one (C95; IC50 : 0.38±0.01 μm), along with two structurally similar non-cytotoxic probes. These probes were used to search for the direct cellular target responsible for chalcone's cytotoxicity through intact cell-based PAL experiments, in which β-tubulin was identified to specifically interact with the cytotoxic probe (i.e., C95) but not the non-cytotoxic probes. A set of phenotypical and biochemical assays further reinforced β-tubulin as the cytotoxic target of chalcones. Peptide mass quantitation by mass spectrometric analysis revealed one peptide potentially labeled by C95, providing information on chalcone's binding site on β-tubulin.
Collapse
Affiliation(s)
- Bo Zhou
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, 2231 6th Street SE, Minneapolis, MN, 55455, USA
| | - Xingxin Yu
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, 2231 6th Street SE, Minneapolis, MN, 55455, USA.,Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Chunlin Zhuang
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, 2231 6th Street SE, Minneapolis, MN, 55455, USA. .,Research Center for Marine Drugs, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, China.
| | - Peter Villalta
- Masonic Cancer Center, University of Minnesota, 2231 6th Street SE, Minneapolis, MN, 55455, USA
| | - Yong Lin
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, NM, 87108, USA
| | - Junxuan Lu
- Department of Pharmacology and Cancer Institute, Penn State University College of Medicine, Hershey, PA, 17033, USA
| | - Chengguo Xing
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, 2231 6th Street SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
17
|
Czarny B, Stura EA, Devel L, Vera L, Cassar-Lajeunesse E, Beau F, Calderone V, Fragai M, Luchinat C, Dive V. Molecular Determinants of a Selective Matrix Metalloprotease-12 Inhibitor: Insights from Crystallography and Thermodynamic Studies. J Med Chem 2013; 56:1149-59. [DOI: 10.1021/jm301574d] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Bertrand Czarny
- CEA, Service
d’Ingénierie
Moléculaire des Protéines (SIMOPRO), Gif/Yvette 91191
Cedex, France
| | - Enrico A. Stura
- CEA, Service
d’Ingénierie
Moléculaire des Protéines (SIMOPRO), Gif/Yvette 91191
Cedex, France
| | - Laurent Devel
- CEA, Service
d’Ingénierie
Moléculaire des Protéines (SIMOPRO), Gif/Yvette 91191
Cedex, France
| | - Laura Vera
- CEA, Service
d’Ingénierie
Moléculaire des Protéines (SIMOPRO), Gif/Yvette 91191
Cedex, France
| | | | - Fabrice Beau
- CEA, Service
d’Ingénierie
Moléculaire des Protéines (SIMOPRO), Gif/Yvette 91191
Cedex, France
| | - Vito Calderone
- Magnetic
Resonance Center (CERM),
University of Florence, Via L. Sacconi 6, 50019 Sesto Fiorentino,
Italy
| | - Marco Fragai
- Magnetic
Resonance Center (CERM),
University of Florence, Via L. Sacconi 6, 50019 Sesto Fiorentino,
Italy
| | - Claudio Luchinat
- Magnetic
Resonance Center (CERM),
University of Florence, Via L. Sacconi 6, 50019 Sesto Fiorentino,
Italy
| | - Vincent Dive
- CEA, Service
d’Ingénierie
Moléculaire des Protéines (SIMOPRO), Gif/Yvette 91191
Cedex, France
| |
Collapse
|
18
|
Nury C, Bregant S, Czarny B, Berthon F, Cassar-Lajeunesse E, Dive V. Detection of endogenous matrix metalloprotease-12 active form with a novel broad spectrum activity-based probe. J Biol Chem 2012; 288:5636-44. [PMID: 23271741 DOI: 10.1074/jbc.m112.419499] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Matrix metalloproteases (MMPs) have attracted considerable attention as critical mediators of pathological tissue remodeling processes. However it remains an unresolved challenge to detect their active forms in biological samples. To prove the efficacy of a recently developed MMP activity-based probe, we examined the content in MMP active forms of bronchoalveolar lavage fluids (BALf) from male C57BL/6 mice exposed to ultrafine carbon black nanoparticles, a model of chronic obstructive pulmonary disease. This probe was shown to label proteins, mostly expressed in BALf of mice exposed to nanoparticles. Using competition assays with a selective MMP-12 inhibitor as well as MMP-12 knock-out mice, one of these proteins was identified as the active form of the catalytic domain of MMP-12. This new probe can detect the active form of MMP-12 down to a threshold of 1 fmol. Radioactive counting showed the concentration of the active form of MMP-12 to be around 1 fmol/μl in BALf from nanoparticle-treated mice. A less sensitive probe would therefore not have detected MMP-12. As the probe can detect other MMPs in the femtomolar range, it is a potentially powerful tool for monitoring the levels of MMP active forms in various diseases.
Collapse
Affiliation(s)
- Catherine Nury
- CEA (Commissariat à l'Energie Atomique), iBiTec-S, Service d'Ingénierie Moléculaire de Protéines (SIMOPRO), CE-Saclay, 91191 Gif /Yvette, Cedex, France
| | | | | | | | | | | |
Collapse
|
19
|
Nury C, Czarny B, Cassar-Lajeunesse E, Georgiadis D, Bregant S, Dive V. A Pan Photoaffinity Probe for Detecting Active Forms of Matrix Metalloproteinases. Chembiochem 2012. [DOI: 10.1002/cbic.201200583] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
20
|
Torkar A, Bregant S, Devel L, Novinec M, Lenarčič B, Lah T, Dive V. A novel photoaffinity-based probe for selective detection of cathepsin L active form. Chembiochem 2012; 13:2616-21. [PMID: 23125066 DOI: 10.1002/cbic.201200389] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Indexed: 01/04/2023]
Abstract
Detecting the active forms of proteases by using activity-based probes in complex proteomes has become an intensively investigated field of research over the past years because many pathogenic conditions involve alterations in protease activities. The detection of lysosomal cysteine proteases, the cathepsins, has mostly relied on the use of probes that incorporate reactive electrophilic moieties to modify a cysteine in the active site covalently. Here we report the first example of an activity-based probe that targets the cathepsins and incorporates a photoactivatable benzophenone group for covalent labelling. When tested on a set of five cathepsins (B, K, L, S and V), this probe selectively labelled the active site of cathepsin L. Furthermore, when tested on crude cell extracts, the probe specifically detected cathepsin L quantities as low as a few picomoles. This study suggests that photoaffinity labelling is a promising approach for developing highly selective and useful cathepsin L probes. In particular, this probe might allow the detection of small amounts of the secreted active cathepsin L form in the cellular microenvironment in vitro and ex vivo.
Collapse
Affiliation(s)
- Ana Torkar
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna pot 111, 1000 Ljubljana, Slovenia
| | | | | | | | | | | | | |
Collapse
|
21
|
Knapinska A, Fields GB. Chemical biology for understanding matrix metalloproteinase function. Chembiochem 2012; 13:2002-20. [PMID: 22933318 PMCID: PMC3951272 DOI: 10.1002/cbic.201200298] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Indexed: 12/20/2022]
Abstract
The matrix metalloproteinase (MMP) family has long been associated with normal physiological processes such as embryonic implantation, tissue remodeling, organ development, and wound healing, as well as multiple aspects of cancer initiation and progression, osteoarthritis, inflammatory and vascular diseases, and neurodegenerative diseases. The development of chemically designed MMP probes has advanced our understanding of the roles of MMPs in disease in addition to shedding considerable light on the mechanisms of MMP action. The first generation of protease-activated agents has demonstrated proof of principle as well as providing impetus for in vivo applications. One common problem has been a lack of agent stability at nontargeted tissues and organs due to activation by multiple proteases. The present review considers how chemical biology has impacted the progress made in understanding the roles of MMPs in disease and the basic mechanisms of MMP action.
Collapse
Affiliation(s)
| | - Gregg B. Fields
- Departments of Chemistry and Biology Torrey Pines Institute for Molecular Studies 11350 SW Village Parkway, Port St. Lucie, FL 34987 (USA)
| |
Collapse
|
22
|
Abstract
Traditionally, proteomics is the high-throughput characterization of the global complement of proteins in a biological system using cutting-edge technologies (robotics and mass spectrometry) and bioinformatics tools (Internet-based search engines and databases). As the field of proteomics has matured, a diverse range of strategies have evolved to answer specific problems. Chemical proteomics is one such direction that provides the means to enrich and detect less abundant proteins (the 'hidden' proteome) from complex mixtures of wide dynamic range (the 'deep' proteome). In pharmacology, chemical proteomics has been utilized to determine the specificity of drugs and their analogues, for anticipated known targets, only to discover other proteins that bind and could account for side effects observed in preclinical and clinical trials. As a consequence, chemical proteomics provides a valuable accessory in refinement of second- and third-generation drug design for treatment of many diseases. However, determining definitive affinity capture of proteins by a drug immobilized on soft gel chromatography matrices has highlighted some of the challenges that remain to be addressed. Examples of the different strategies that have emerged using well-established drugs against pharmaceutically important enzymes, such as protein kinases, metalloproteases, PDEs, cytochrome P450s, etc., indicate the potential opportunity to employ chemical proteomics as an early-stage screening approach in the identification of new targets.
Collapse
Affiliation(s)
- Chris W Sutton
- Institute of Cancer Therapeutics, University of Bradford, Tumbling Hill Street, Bradford, West Yorkshire, UK.
| |
Collapse
|
23
|
Bijakowski C, Vadon-Le Goff S, Delolme F, Bourhis JM, Lécorché P, Ruggiero F, Becker-Pauly C, Yiallouros I, Stöcker W, Dive V, Hulmes DJS, Moali C. Sizzled is unique among secreted frizzled-related proteins for its ability to specifically inhibit bone morphogenetic protein-1 (BMP-1)/tolloid-like proteinases. J Biol Chem 2012; 287:33581-93. [PMID: 22825851 DOI: 10.1074/jbc.m112.380816] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
BMP-1/tolloid-like proteinases (BTPs) are major enzymes involved in extracellular matrix assembly and activation of bioactive molecules, both growth factors and anti-angiogenic molecules. Although the control of BTP activity by several enhancing molecules is well established, the possibility that regulation also occurs through endogenous inhibitors is still debated. Secreted frizzled-related proteins (sFRPs) have been studied as possible candidates, with highly contradictory results, after the demonstration that sizzled, a sFRP found in Xenopus and zebrafish, was a potent inhibitor of Xenopus and zebrafish tolloid-like proteases. In this study, we demonstrate that mammalian sFRP-1, -2, and -4 do not modify human BMP-1 activity on several of its known substrates including procollagen I, procollagen III, pN-collagen V, and prolysyl oxidase. In contrast, Xenopus sizzled appears as a tight binding inhibitor of human BMP-1, with a K(i) of 1.5 ± 0.5 nM, and is shown to strongly inhibit other human tolloid isoforms mTLD and mTLL-1. Because sizzled is the most potent inhibitor of human tolloid-like proteinases known to date, we have studied its mechanism of action in detail and shown that the frizzled domain of sizzled is both necessary and sufficient for inhibitory activity and that it acts directly on the catalytic domain of BMP-1. Residues in sizzled required for inhibition include Asp-92, which is shared by sFRP-1 and -2, and also Phe-94, Ser-43, and Glu-44, which are specific to sizzled, thereby providing a rational basis for the absence of inhibitory activity of human sFRPs.
Collapse
Affiliation(s)
- Cécile Bijakowski
- Institut de Biologie et Chimie des Protéines, CNRS/Université de Lyon FRE3310/FR3302, 69367 Lyon cedex 7, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Devel L, Beau F, Amoura M, Vera L, Cassar-Lajeunesse E, Garcia S, Czarny B, Stura EA, Dive V. Simple pseudo-dipeptides with a P2' glutamate: a novel inhibitor family of matrix metalloproteases and other metzincins. J Biol Chem 2012; 287:26647-56. [PMID: 22689580 DOI: 10.1074/jbc.m112.380782] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
A series of pseudo-peptides with general formula X-l-Glu-NH(2) (with X corresponding to an acyl moiety with a long aryl-alkyl side chain) have been synthesized, evaluated as inhibitors of matrix metalloproteases (MMPs), and found to display remarkable nanomolar affinity. The loss in potency associated with a substitution of the P(2)' l-glutamate by a l-glutamine corroborates the importance of a carboxylate at this position. The binding mode of some of these inhibitors was characterized in solution and by x-ray crystallography in complex with various MMPs. The x-ray crystal structures reveal an unusual binding mode with the glutamate side chain chelating the active site zinc ion. Competition experiments between these inhibitors and acetohydroxamic acid, a small zinc-binding molecule, are in accord with the crystallographic results. One of these pseudo-dipeptides displays potency and selectivity toward MMP-12 similar to the best MMP-12 inhibitors reported to date. This novel family of pseudo peptides opens new opportunities to develop potent and selective inhibitors for several metzincins.
Collapse
Affiliation(s)
- Laurent Devel
- CEA (Commissariat à l'Energie Atomique), iBiTec-S, Service d'Ingénierie Moléculaire de Protéines (SIMOPRO), CE Saclay, 91191 Gif/Yvette, Cedex, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Geurink PP, Prely LM, van der Marel GA, Bischoff R, Overkleeft HS. Photoaffinity labeling in activity-based protein profiling. Top Curr Chem (Cham) 2011; 324:85-113. [PMID: 22028098 DOI: 10.1007/128_2011_286] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Activity-based protein profiling has come to the fore in recent years as a powerful strategy for studying enzyme activities in their natural surroundings. Substrate analogs that bind covalently and irreversibly to an enzyme active site and that are equipped with an identification or affinity tag can be used to unearth new enzyme activities, to establish whether and at what subcellular location the enzymes are active, and to study the inhibitory effects of small compounds. A specific class of activity-based protein probes includes those that employ a photo-activatable group to create the covalent bond. Such probes are targeted to those enzymes that do not employ a catalytic nucleophile that is part of the polypeptide backbone. An overview of the various photo-activatable groups that are available to chemical biology researchers is presented, with a focus on their (photo)chemistry and their application in various research fields. A number of comparative studies are described in which the efficiency of various photo-activatable groups are compared.
Collapse
Affiliation(s)
- Paul P Geurink
- Leiden Institute of Chemistry and the Netherlands Proteomics Centre, Leiden University, The Netherlands
| | | | | | | | | |
Collapse
|
26
|
Devel L, Garcia S, Czarny B, Beau F, Lajeunesse E, Vera L, Georgiadis D, Stura E, Dive V. Insights from selective non-phosphinic inhibitors of MMP-12 tailored to fit with an S1' loop canonical conformation. J Biol Chem 2010; 285:35900-9. [PMID: 20817735 PMCID: PMC2975213 DOI: 10.1074/jbc.m110.139634] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2010] [Revised: 07/21/2010] [Indexed: 01/28/2023] Open
Abstract
After the disappointment of clinical trials with early broad spectrum synthetic inhibitors of matrix metalloproteinases (MMPs), the field is now resurging with a new focus on the development of selective inhibitors that fully discriminate between different members of the MMP family with several therapeutic applications in perspective. Here, we report a novel class of highly selective MMP-12 inhibitors, without a phosphinic zinc-binding group, designed to plunge deeper into the S(1)' cavity of the enzyme. The best inhibitor from this series, identified through a systematic chemical exploration, displays nanomolar potency toward MMP-12 and selectivity factors that range between 2 and 4 orders of magnitude toward a large set of MMPs. Comparison of the high resolution x-ray structures of MMP-12 in free state or bound to this new MMP-12 selective inhibitor reveals that this compound fits deeply within the S(1)' specificity cavity, maximizing surface/volume ratios, without perturbing the S(1)' loop conformation. This is in contrast with highly selective MMP-13 inhibitors that were shown to select a particular S(1)' loop conformation. The search for such compounds that fit precisely to preponderant S(1)' loop conformation of a particular MMP may prove to be an alternative effective strategy for developing selective inhibitors of MMPs.
Collapse
Affiliation(s)
- Laurent Devel
- From the Commissariat à l'Energie Atomique, Service d'Ingénierie Moléculaire de Protéines, CE-Saclay, 91191 Gif/Yvette Cedex, France and
| | - Sandra Garcia
- From the Commissariat à l'Energie Atomique, Service d'Ingénierie Moléculaire de Protéines, CE-Saclay, 91191 Gif/Yvette Cedex, France and
| | - Bertrand Czarny
- From the Commissariat à l'Energie Atomique, Service d'Ingénierie Moléculaire de Protéines, CE-Saclay, 91191 Gif/Yvette Cedex, France and
| | - Fabrice Beau
- From the Commissariat à l'Energie Atomique, Service d'Ingénierie Moléculaire de Protéines, CE-Saclay, 91191 Gif/Yvette Cedex, France and
| | - Evelyne Lajeunesse
- From the Commissariat à l'Energie Atomique, Service d'Ingénierie Moléculaire de Protéines, CE-Saclay, 91191 Gif/Yvette Cedex, France and
| | - Laura Vera
- From the Commissariat à l'Energie Atomique, Service d'Ingénierie Moléculaire de Protéines, CE-Saclay, 91191 Gif/Yvette Cedex, France and
| | - Dimitris Georgiadis
- the Laboratory of Organic Chemistry, Department of Organic Chemistry, University of Athens, Panepistimiopolis, Zografou, 15771 Athens, Greece
| | - Enrico Stura
- From the Commissariat à l'Energie Atomique, Service d'Ingénierie Moléculaire de Protéines, CE-Saclay, 91191 Gif/Yvette Cedex, France and
| | - Vincent Dive
- From the Commissariat à l'Energie Atomique, Service d'Ingénierie Moléculaire de Protéines, CE-Saclay, 91191 Gif/Yvette Cedex, France and
| |
Collapse
|
27
|
Bregant S, Huillet C, Devel L, Dabert-Gay AS, Beau F, Thai R, Czarny B, Yiotakis A, Dive V. Detection of matrix metalloproteinase active forms in complex proteomes: evaluation of affinity versus photoaffinity capture. J Proteome Res 2009; 8:2484-94. [PMID: 19271733 DOI: 10.1021/pr801069c] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Various attempts to detect matrix metalloproteinase (MMP) active forms from complex proteomes, based on the use of specific photoactivatable affinity probes, have up to now failed. To overcome this failure, an affinity approach has been evaluated as an alternative to the photoaffinity one. For this purpose, two probes were synthesized to interact specifically with the active site of MMPs and allow isolation of MMP/probe complexes on magnetic beads through a biotin linker. Using phosphinic peptide chemistry, we prepared an affinity probe displaying picomolar potency toward several MMPs, and a related photoaffinity probe incorporating a photoactivatable azido group exhibiting subnanomolar affinity toward these targets. By a combination of silver-staining detection and MALDI peptide mass fingerprints, a systematic comparison was made of both strategies in terms of hMMP-12 and hMMP-8 recovery and identification when present in mixtures of different complexity. The results obtained show that the affinity protocol is superior to the photoaffinity strategy in terms of quantity of captured MMPs and number of MMP tryptic fragments detected in MALDI-MS. The specificity and efficiency of the affinity capture protocol developed in this study allowed easy, fast, and unambiguous detection by MALDI-MS of three hMMPs (2, 8, and 12), from a single affinity capture experiment, when added (10-36 ng of MMPs) to a tumor extract (10 microg). Thus, the tools and approaches reported should enable us to progress in the detection of endogenous active forms of MMPs in complex proteomes, an important objective with many diagnostic applications.
Collapse
Affiliation(s)
- Sarah Bregant
- CEA, iBiTecS, Service d'Ingénierie Moléculaire des Protéines (SIMOPRO), Bat 152, CE-Saclay Gif/Yvette, F-91191, France
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Dabert-Gay AS, Czarny B, Lajeunesse E, Thai R, Nagase H, Dive V. Covalent modification of matrix metalloproteinases by a photoaffinity probe: influence of nucleophilicity and flexibility of the residue in position 241. Bioconjug Chem 2009; 20:367-75. [PMID: 19138112 DOI: 10.1021/bc800478b] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
A photoaffinity probe, developed for the specific labeling of matrix metalloproteinase (MMP) active sites, was recently shown to covalently modify a single residue in human MMP-12, namely, Lys(241), by reacting selectively with the side chain epsilon-amino group of that residue. The residue in position 241 of MMPs is not conserved; thus, variability in this position may be responsible for the dispersion in cross-linking yield observed between MMPs when labeled by this photoaffinity probe. By studying the pH dependence of the labeling properties of this probe toward different MMPs (MMP-12, MMP-3, MMP-9, and various mutants of human MMP-12) and identifying the site of covalent modification of MMP-3 by this probe, our new data demonstrated that the nucleophilicity of the residue in position 241 plays a key role in determining the cross-linking yield of MMP modification by the probe. However, these studies also reveal that subtle additional structural parameters, including local conformation and flexibility, of the residue in position 241 should also be taken into consideration, a property adding a further degree of complexity in our understanding of the photolabeling probe reactivity and in designing optimal photoaffinity probes for performing functional proteomic studies of zinc proteinases like MMPs.
Collapse
Affiliation(s)
- Anne-Sophie Dabert-Gay
- CEA, iBiTecS, Service d'Ingenierie Moleculaire des Proteines (SIMOPRO), CE-Saclay Gif/Yvette, F-91191, France
| | | | | | | | | | | |
Collapse
|