1
|
Mazzocato Y, Perin S, Morales-Sanfrutos J, Romanyuk Z, Pluda S, Acquasaliente L, Borsato G, De Filippis V, Scarso A, Angelini A. A novel genetically-encoded bicyclic peptide inhibitor of human urokinase-type plasminogen activator with better cross-reactivity toward the murine orthologue. Bioorg Med Chem 2023; 95:117499. [PMID: 37879145 DOI: 10.1016/j.bmc.2023.117499] [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: 03/25/2023] [Revised: 08/30/2023] [Accepted: 10/10/2023] [Indexed: 10/27/2023]
Abstract
The inhibition of human urokinase-type plasminogen activator (huPA), a serine protease that plays an important role in pericellular proteolysis, is a promising strategy to decrease the invasive and metastatic activity of tumour cells. However, the generation of selective small molecule huPA inhibitors has proven to be challenging due to the high structural similarity of huPA to other paralogue serine proteases. Efforts to generate more specific therapies have led to the development of cyclic peptide-based inhibitors with much higher selectivity against huPA. While this latter property is desired, the sparing of the orthologue murine poses difficulties for the testing of the inhibitor in preclinical mouse model. In this work, we have applied a Darwinian evolution-based approach to identify phage-encoded bicyclic peptide inhibitors of huPA with better cross-reactivity towards murine uPA (muPA). The best selected bicyclic peptide (UK132) inhibited huPA and muPA with Ki values of 0.33 and 12.58 µM, respectively. The inhibition appears to be specific for uPA, as UK132 only weakly inhibits a panel of structurally similar serine proteases. Removal or substitution of the second loop with one not evolved in vitro led to monocyclic and bicyclic peptide analogues with lower potency than UK132. Moreover, swapping of 1,3,5-tris-(bromomethyl)-benzene with different small molecules not used in the phage selection, resulted in an 80-fold reduction of potency, revealing the important structural role of the branched cyclization linker. Further substitution of an arginine in UK132 to a lysine resulted in a bicyclic peptide UK140 with enhanced inhibitory potency against both huPA (Ki = 0.20 µM) and murine orthologue (Ki = 2.79 µM). By combining good specificity, nanomolar affinity and a low molecular mass, the bicyclic peptide inhibitor developed in this work may provide a novel human and murine cross-reactive lead for the development of a potent and selective anti-metastatic therapy.
Collapse
Affiliation(s)
- Ylenia Mazzocato
- Department of Molecular Sciences and Nanosystems, Ca' Foscari University of Venice, Via Torino 155, 30172 Venice, Italy
| | - Stefano Perin
- Department of Molecular Sciences and Nanosystems, Ca' Foscari University of Venice, Via Torino 155, 30172 Venice, Italy
| | - Julia Morales-Sanfrutos
- Proteomics Unit, Spanish National Cancer Research Centre (CNIO), C. de Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Zhanna Romanyuk
- Department of Molecular Sciences and Nanosystems, Ca' Foscari University of Venice, Via Torino 155, 30172 Venice, Italy
| | - Stefano Pluda
- Department of Molecular Sciences and Nanosystems, Ca' Foscari University of Venice, Via Torino 155, 30172 Venice, Italy; Fidia Farmaceutici S.p.A., Via Ponte della Fabbrica 3/A, Abano Terme 35031, Italy
| | - Laura Acquasaliente
- Department of Pharmaceutical and Pharmacological Sciences, School of Medicine, University of Padova, Via Marzolo 5, 35131 Padova, Italy
| | - Giuseppe Borsato
- Department of Molecular Sciences and Nanosystems, Ca' Foscari University of Venice, Via Torino 155, 30172 Venice, Italy
| | - Vincenzo De Filippis
- Department of Pharmaceutical and Pharmacological Sciences, School of Medicine, University of Padova, Via Marzolo 5, 35131 Padova, Italy
| | - Alessandro Scarso
- Department of Molecular Sciences and Nanosystems, Ca' Foscari University of Venice, Via Torino 155, 30172 Venice, Italy
| | - Alessandro Angelini
- Department of Molecular Sciences and Nanosystems, Ca' Foscari University of Venice, Via Torino 155, 30172 Venice, Italy; European Centre for Living Technology (ECLT), Ca' Bottacin, Dorsoduro 3911, Calle Crosera, 30123 Venice, Italy.
| |
Collapse
|
2
|
Xu Y, Chen D, Liu P, Hu Y, Peng S, Chen S, Li Y, Lin W, Jiang L, Yuan C, Huang M. A triple fusion tissue-type plasminogen activator (TriF-ΔtPA) enhanced thrombolysis in carotid embolism-induced stroke model. Int J Pharm 2023; 637:122878. [PMID: 36958614 DOI: 10.1016/j.ijpharm.2023.122878] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 02/06/2023] [Accepted: 03/18/2023] [Indexed: 03/25/2023]
Abstract
Recombinant tissue-type plasminogen activator (rtPA) is the first approved thrombolytic agent in acute ischemic stroke, but suffers from a short half-life and poor resistance to plasminogen activator inhibitor (PAI-1), limiting its clinical use. Thus, the development of novel thrombolytic agents with improved benefit/risk balance has always been of great significance. In this study, We identified a serine protease domain of tPA mutant (named ΔtPAA146V) capable of escaping the inhibition by endogenous PAI-1 with 66-fold increased resistance compared to the wild type. Based on this mutant, we generated a triple fusion ΔtPA (TriF-ΔtPA), including albumin and fibrin binding peptide(FBP). The fusion with albumin effectively prolonged the plasma half-life of ΔtPA in mice to 144 minutes, which is much longer than ΔtPA and did not affect its thrombolytic activity. Furthermore, FBP rendered fibrin specificity of the fusion protein, giving a KD of ∼25 ± 0.9 μM. In a novel murine carotid embolism-induced stroke (CES) model, i.v. administration of TriF-ΔtPA promoted vascular recanalization, reduced infarct volume, and mitigated neurobehavioral deficits more significantly compared to ΔtPA-HSA or Alteplase, showing little bleeding risk. Together, this long-acting PAI-1-resistant thrombolytic agent holds great potential for clinical applications.
Collapse
Affiliation(s)
- Yanyan Xu
- College of Chemical Engineering, Fuzhou University, Fujian 350108, China
| | - Dan Chen
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Peiwen Liu
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Yinping Hu
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Shuangzhou Peng
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, Fujian 361102, China
| | - Shanli Chen
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Yongkun Li
- Department of Neurology, Fujian Provincial Hospital, Shengli Clinical College of Fujian Medical University, No. 134 Dong Street, Fuzhou, Fujian 350001, P. R. China
| | - Wei Lin
- Fujian Institute of integrated traditional Chinese and Western Medicine, Fujian University of Traditional Chinese Medicine, Minhou District, Fuzhou, Fujian, 350122 P. R. China
| | - Longguang Jiang
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Cai Yuan
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian, 350108, China; Fujian Key Laboratory of Marine Enzyme Engineering, Fuzhou University, Fuzhou, Fujian, 350108, China.
| | - Mingdong Huang
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China.
| |
Collapse
|
3
|
Zhou Y, Wu J, Xue G, Li J, Jiang L, Huang M. Structural study of the uPA-nafamostat complex reveals a covalent inhibitory mechanism of nafamostat. Biophys J 2022; 121:3940-3949. [PMID: 36039386 PMCID: PMC9674978 DOI: 10.1016/j.bpj.2022.08.034] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 08/02/2022] [Accepted: 08/24/2022] [Indexed: 11/23/2022] Open
Abstract
Nafamostat mesylate (NM) is a synthetic compound that inhibits various serine proteases produced during the coagulation cascade and inflammation. Previous studies showed that NM was a highly safe drug for the treatment of different cancers, but the precise functions and mechanisms of NM are not clear. In this study, we determined a series of crystal structures of NM and its hydrolysates in complex with a serine protease (urokinase-type plasminogen activator [uPA]). These structures reveal that NM was cleaved by uPA and that a hydrolyzed product (4-guanidinobenzoic acid [GBA]) remained covalently linked to Ser195 of uPA, and the other hydrolyzed product (6-amidino-2-naphthol [6A2N]) released from uPA. Strikingly, in the inactive uPA (uPA-S195A):NM structure, the 6A2N side of intact NM binds to the specific pocket of uPA. Molecular dynamics simulations and end-point binding free-energy calculations show that the conf1 of NM (6A2N as P1 group) in the uPA-S195A:NM complex may be more stable than conf2 of NM (GBA as P1 group). Moreover, in the structure of uPA:NM complex, the imidazole group of His57 flips further away from Ser195 and disrupts the stable canonical catalytic triad conformation. These results not only reveal the inhibitory mechanism of NM as an efficient serine protease inhibitor but also might provide the structural basis for the further development of serine protease inhibitors.
Collapse
Affiliation(s)
- Yang Zhou
- College of Chemistry, Fuzhou University, Fuzhou, Fujian, P.R. China
| | - Juhong Wu
- College of Chemistry, Fuzhou University, Fuzhou, Fujian, P.R. China
| | - Guangpu Xue
- College of Chemistry, Fuzhou University, Fuzhou, Fujian, P.R. China
| | - Jinyu Li
- College of Chemistry, Fuzhou University, Fuzhou, Fujian, P.R. China
| | - Longguang Jiang
- College of Chemistry, Fuzhou University, Fuzhou, Fujian, P.R. China; Fujian Key Laboratory of Marine Enzyme Engineering, Fuzhou University, Fuzhou, Fujian, P.R. China.
| | - Mingdong Huang
- College of Chemistry, Fuzhou University, Fuzhou, Fujian, P.R. China.
| |
Collapse
|
4
|
El Salamouni NS, Buckley BJ, Ranson M, Kelso MJ, Yu H. Urokinase plasminogen activator as an anti-metastasis target: inhibitor design principles, recent amiloride derivatives, and issues with human/mouse species selectivity. Biophys Rev 2022; 14:277-301. [PMID: 35340592 PMCID: PMC8921380 DOI: 10.1007/s12551-021-00921-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 11/18/2021] [Indexed: 01/09/2023] Open
Abstract
The urokinase plasminogen activator (uPA) is a widely studied anticancer drug target with multiple classes of inhibitors reported to date. Many of these inhibitors contain amidine or guanidine groups, while others lacking these groups show improved oral bioavailability. Most of the X-ray co-crystal structures of small molecule uPA inhibitors show a key salt bridge with the side chain carboxylate of Asp189 in the S1 pocket of uPA. This review summarises the different classes of uPA inhibitors, their binding interactions and experimentally measured inhibitory potencies and highlights species selectivity issues with attention to recently described 6-substituted amiloride and 5‑N,N-(hexamethylene)amiloride (HMA) derivatives.
Collapse
Affiliation(s)
- Nehad S El Salamouni
- School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522 Australia
- Molecular Horizons, University of Wollongong, Wollongong, NSW 2522 Australia
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522 Australia
| | - Benjamin J. Buckley
- School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522 Australia
- Molecular Horizons, University of Wollongong, Wollongong, NSW 2522 Australia
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522 Australia
| | - Marie Ranson
- School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522 Australia
- Molecular Horizons, University of Wollongong, Wollongong, NSW 2522 Australia
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522 Australia
| | - Michael J. Kelso
- School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522 Australia
- Molecular Horizons, University of Wollongong, Wollongong, NSW 2522 Australia
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522 Australia
| | - Haibo Yu
- School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522 Australia
- Molecular Horizons, University of Wollongong, Wollongong, NSW 2522 Australia
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522 Australia
| |
Collapse
|
5
|
S El Salamouni N, Buckley BJ, Jiang L, Huang M, Ranson M, Kelso MJ, Yu H. Disruption of Water Networks is the Cause of Human/Mouse Species Selectivity in Urokinase Plasminogen Activator (uPA) Inhibitors Derived from Hexamethylene Amiloride (HMA). J Med Chem 2021; 65:1933-1945. [PMID: 34898192 DOI: 10.1021/acs.jmedchem.1c01423] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The urokinase plasminogen activator (uPA) plays a critical role in tumor cell invasion and migration and is a promising antimetastasis target. 6-Substituted analogues of 5-N,N-(hexamethylene)amiloride (HMA) are potent and selective uPA inhibitors that lack the diuretic and antikaliuretic properties of the parent drug amiloride. However, the compounds display pronounced selectivity for human over mouse uPA, thus confounding interpretation of data from human xenograft mouse models of cancer. Here, computational and experimental findings reveal that residue 99 is a key contributor to the observed species selectivity, whereby enthalpically unfavorable expulsion of a water molecule by the 5-N,N-hexamethylene ring occurs when residue 99 is Tyr (as in mouse uPA). Analogue 7 lacking the 5-N,N-hexamethylene ring maintained similar water networks when bound to human and mouse uPA and displayed reduced selectivity, thus supporting this conclusion. The study will guide further optimization of dual-potent human/mouse uPA inhibitors from the amiloride class as antimetastasis drugs.
Collapse
Affiliation(s)
- Nehad S El Salamouni
- School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia.,Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia.,Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
| | - Benjamin J Buckley
- School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia.,Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia.,Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia.,CONCERT-Translational Cancer Research Centre, Sydney, NSW 2750, Australia
| | - Longguang Jiang
- National Joint Biomedical Engineering Research Centre on Photodynamic Technologies, Fuzhou University, Fujian 350116, China
| | - Mingdong Huang
- National Joint Biomedical Engineering Research Centre on Photodynamic Technologies, Fuzhou University, Fujian 350116, China
| | - Marie Ranson
- School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia.,Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia.,Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia.,CONCERT-Translational Cancer Research Centre, Sydney, NSW 2750, Australia
| | - Michael J Kelso
- School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia.,Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia.,Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
| | - Haibo Yu
- School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW 2522, Australia.,Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia.,Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia
| |
Collapse
|
6
|
Structural basis of covalent inhibitory mechanism of TMPRSS2-related serine proteases by camostat. J Virol 2021; 95:e0086121. [PMID: 34160253 PMCID: PMC8428381 DOI: 10.1128/jvi.00861-21] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the viral pathogen causing the coronavirus disease 2019 (COVID-19) global pandemic. No effective treatment for COVID-19 has been established yet. The serine protease transmembrane protease serine 2 (TMPRSS2) is essential for viral spread and pathogenicity by facilitating the entry of SARS-CoV-2 into host cells. The protease inhibitor camostat, an anticoagulant used in the clinic, has potential anti-inflammatory and antiviral activities against COVID-19. However, the potential mechanisms of viral resistance and antiviral activity of camostat are unclear. Herein, we demonstrate high inhibitory potencies of camostat for a panel of serine proteases, indicating that camostat is a broad-spectrum inhibitor of serine proteases. In addition, we determined the crystal structure of camostat in complex with a serine protease (uPA [urokinase-type plasminogen activator]), which reveals that camostat is inserted in the S1 pocket of uPA but is hydrolyzed by uPA, and the cleaved camostat covalently binds to Ser195. We also generated a homology model of the structure of the TMPRSS2 serine protease domain. The model shows that camostat uses the same inhibitory mechanism to inhibit the activity of TMPRSS2, subsequently preventing SARS-CoV-2 spread. IMPORTANCE Serine proteases are a large family of enzymes critical for multiple physiological processes and proven diagnostic and therapeutic targets in several clinical indications. The serine protease transmembrane protease serine 2 (TMPRSS2) was recently found to mediate SARS-CoV-2 entry into the host. Camostat mesylate (FOY 305), a serine protease inhibitor active against TMPRSS2 and used for the treatment of oral squamous cell carcinoma and chronic pancreatitis, inhibits SARS-CoV-2 infection of human lung cells. However, the direct inhibition mechanism of camostat mesylate for TMPRSS2 is unclear. Herein, we demonstrate that camostat uses the same inhibitory mechanism to inhibit the activity of TMPRSS2 as uPA, subsequently preventing SARS-CoV-2 spread.
Collapse
|
7
|
Vu QN, Young R, Sudhakar HK, Gao T, Huang T, Tan YS, Lau YH. Cyclisation strategies for stabilising peptides with irregular conformations. RSC Med Chem 2021; 12:887-901. [PMID: 34263169 PMCID: PMC8230030 DOI: 10.1039/d1md00098e] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 04/12/2021] [Indexed: 11/21/2022] Open
Abstract
Cyclisation is a common synthetic strategy for enhancing the therapeutic potential of peptide-based molecules. While there are extensive studies on peptide cyclisation for reinforcing regular secondary structures such as α-helices and β-sheets, there are remarkably few reports of cyclising peptides which adopt irregular conformations in their bioactive target-bound state. In this review, we highlight examples where cyclisation techniques have been successful in stabilising irregular conformations, then discuss how the design of cyclic constraints for irregularly structured peptides can be informed by existing β-strand stabilisation approaches, new computational design techniques, and structural principles extracted from cyclic peptide library screening hits. Through this analysis, we demonstrate how existing peptide cyclisation techniques can be adapted to address the synthetic design challenge of stabilising irregularly structured binding motifs.
Collapse
Affiliation(s)
- Quynh Ngoc Vu
- School of Chemistry, Eastern Ave, The University of Sydney NSW 2006 Australia
| | - Reginald Young
- School of Chemistry, Eastern Ave, The University of Sydney NSW 2006 Australia
| | | | - Tianyi Gao
- School of Chemistry, Eastern Ave, The University of Sydney NSW 2006 Australia
| | - Tiancheng Huang
- School of Chemistry, Eastern Ave, The University of Sydney NSW 2006 Australia
| | - Yaw Sing Tan
- Bioinformatics Institute, Agency for Science, Technology and Research (ASTAR) 30 Biopolis Street, #07-01, Matrix Singapore 138671 Singapore
| | - Yu Heng Lau
- School of Chemistry, Eastern Ave, The University of Sydney NSW 2006 Australia
| |
Collapse
|
8
|
McAllister TE, Coleman OD, Roper G, Kawamura A. Structural diversity in
de novo
cyclic peptide ligands from genetically encoded library technologies. Pept Sci (Hoboken) 2020. [DOI: 10.1002/pep2.24204] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Tom E. McAllister
- Chemistry – School of Natural and Environmental Sciences Newcastle University Newcastle upon Tyne UK
| | - Oliver D. Coleman
- Chemistry – School of Natural and Environmental Sciences Newcastle University Newcastle upon Tyne UK
| | - Grace Roper
- Chemistry – School of Natural and Environmental Sciences Newcastle University Newcastle upon Tyne UK
- Chemistry Research Laboratory, Department of Chemistry University of Oxford Oxford UK
| | - Akane Kawamura
- Chemistry – School of Natural and Environmental Sciences Newcastle University Newcastle upon Tyne UK
- Chemistry Research Laboratory, Department of Chemistry University of Oxford Oxford UK
| |
Collapse
|
9
|
Zuo K, Qi Y, Yuan C, Jiang L, Xu P, Hu J, Huang M, Li J. Specifically targeting cancer proliferation and metastasis processes: the development of matriptase inhibitors. Cancer Metastasis Rev 2020; 38:507-524. [PMID: 31471691 DOI: 10.1007/s10555-019-09802-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Matriptase is a type II transmembrane serine protease, which has been suggested to play critical roles in numerous pathways of biological developments. Matriptase is the activator of several oncogenic proteins, including urokinase-type plasminogen activator (uPA), hepatocyte growth factor (HGF) and protease-activated receptor 2 (PAR-2). The activations of these matriptase substrates subsequently lead to the generation of plasmin, matrix metalloproteases (MMPs), and the triggers for many other signaling pathways related to cancer proliferation and metastasis. Accordingly, matriptase is considered an emerging target for the treatments of cancer. Thus far, inhibitors of matriptase have been developed as potential anti-cancer agents, which include small-molecule inhibitors, peptide-based inhibitors, and monoclonal antibodies. This review covers established literature to summarize the chemical and biochemical aspects, especially the inhibitory mechanisms and structure-activity relationships (SARs) of matriptase inhibitors with the goal of proposing the strategies for their future developments in anti-cancer therapy.
Collapse
Affiliation(s)
- Ke Zuo
- College of Chemistry, Fuzhou University, Fuzhou, 350116, Fujian, People's Republic of China
| | - Yingying Qi
- College of Chemistry, Fuzhou University, Fuzhou, 350116, Fujian, People's Republic of China
| | - Cai Yuan
- College of Chemistry, Fuzhou University, Fuzhou, 350116, Fujian, People's Republic of China
| | - Longguang Jiang
- College of Chemistry, Fuzhou University, Fuzhou, 350116, Fujian, People's Republic of China
| | - Peng Xu
- Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), 61 Biopolis Dr, 138673, Singapore, Singapore.
| | - Jianping Hu
- College of Pharmacy and Biological Engineering, Chengdu University, Chengdu, 610106, Sichuan, People's Republic of China.
| | - Mingdong Huang
- College of Chemistry, Fuzhou University, Fuzhou, 350116, Fujian, People's Republic of China.
| | - Jinyu Li
- College of Chemistry, Fuzhou University, Fuzhou, 350116, Fujian, People's Republic of China.
| |
Collapse
|
10
|
Xue G, Xie X, Zhou Y, Yuan C, Huang M, Jiang L. Insight to the residue in P2 position prevents the peptide inhibitor from being hydrolyzed by serine proteases. Biosci Biotechnol Biochem 2020; 84:1153-1159. [PMID: 32019421 DOI: 10.1080/09168451.2020.1723405] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Peptidic inhibitors of proteases are attracting increasing interest not only as drug candidates but also for studying the function and regulation mechanisms of these enzymes. Previously, we screened out a cyclic peptide inhibitor of human uPA [Formula: see text] and found that Ala substitution of P2 residue turns upain-1 to a substrate. To further investigate the effect of P2 residue on the peptide behavior transformation, we constructed upain-1-W3F, which has Phe replacement in the P2 position. We determined KD and Ki of upain-1-W3F and found that upain-1-W3F might still exist as an inhibitor. Furthermore, the high-resolution crystal structure of upain-1-W3F·uPA reveals that upain-1-W3F indeed stays as an intact inhibitor bind to uPA. We thus propose that the P2 residue plays a nonnegligible role in the conversion of upain-1 to a substrate. These results also proposed a strategy to optimize the pharmacological properties of peptide-based drug candidates by hydrophobicity and steric hindrance.Abbreviations : uPA: urokinase-type plasminogen activator; SPD: serine protease domain; S1 pocket: specific substrate-binding pocket.
Collapse
Affiliation(s)
- Guangpu Xue
- College of Chemistry, Fuzhou University, Fuzhou, China.,National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou, China
| | - Xie Xie
- College of Chemistry, Fuzhou University, Fuzhou, China.,National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou, China
| | - Yang Zhou
- College of Chemistry, Fuzhou University, Fuzhou, China.,National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou, China
| | - Cai Yuan
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou, China.,College of Biological Science and Engineering, Fuzhou University, Fuzhou, China
| | - Mingdong Huang
- College of Chemistry, Fuzhou University, Fuzhou, China.,National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou, China.,College of Biological Science and Engineering, Fuzhou University, Fuzhou, China
| | - Longguang Jiang
- College of Chemistry, Fuzhou University, Fuzhou, China.,National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou, China
| |
Collapse
|
11
|
Shang L, Xue G, Gong L, Zhang Y, Peng S, Yuan C, Huang M. A novel ELISA for the detection of active form of plasminogen activator inhibitor-1 based on a highly specific trapping agent. Anal Chim Acta 2019; 1053:98-104. [DOI: 10.1016/j.aca.2018.12.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 11/26/2018] [Accepted: 12/08/2018] [Indexed: 10/27/2022]
|
12
|
Xu P, Huang M. Small Peptides as Modulators of Serine Proteases. Curr Med Chem 2018; 27:3686-3705. [PMID: 30332941 DOI: 10.2174/0929867325666181016163630] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 09/26/2018] [Accepted: 10/09/2018] [Indexed: 02/08/2023]
Abstract
Serine proteases play critical roles in many physiological and pathological processes, and are proven diagnostic and therapeutic targets in a number of clinical indications. Suppression of the aberrant proteolytic activities of these proteases has been clinically used for the treatments of relevant diseases. Polypeptides with 10-20 residues are of great interests as medicinal modulators of serine proteases, because these peptides demonstrate the characteristics of both small molecule drugs and macromolecular drugs. In this review, we summarized the recent development of peptide-based inhibitors against serine proteases with potent inhibitory and high specificity comparable to monoclonal antibodies. In addition, we also discussed the strategies of enhancing plasma half-life and bioavailability of peptides in vivo, which is the main hurdle that limits the clinical translation of peptide-based drugs. This review advocates new avenue for the development of effective serine protease inhibitors and highlights the prospect of the medicinal use of these inhibitors.
Collapse
Affiliation(s)
- Peng Xu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350116, China
| | - Mingdong Huang
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China
| |
Collapse
|
13
|
Buckley BJ, Aboelela A, Minaei E, Jiang LX, Xu Z, Ali U, Fildes K, Cheung CY, Cook SM, Johnson DC, Bachovchin DA, Cook GM, Apte M, Huang M, Ranson M, Kelso MJ. 6-Substituted Hexamethylene Amiloride (HMA) Derivatives as Potent and Selective Inhibitors of the Human Urokinase Plasminogen Activator for Use in Cancer. J Med Chem 2018; 61:8299-8320. [PMID: 30130401 DOI: 10.1021/acs.jmedchem.8b00838] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Metastasis is the cause of death in the majority (∼90%) of malignant cancers. The oral potassium-sparing diuretic amiloride and its 5-substituted derivative 5 -N, N-(hexamethylene)amiloride (HMA) reportedly show robust antitumor/metastasis effects in multiple in vitro and animal models. These effects are likely due, at least in part, to inhibition of the urokinase plasminogen activator (uPA), a key protease determinant of cell invasiveness and metastasis. This study reports the discovery of 6-substituted HMA analogs that show nanomolar potency against uPA, high selectivity over related trypsin-like serine proteases, and minimal inhibitory effects against epithelial sodium channels (ENaC), the diuretic and antikaliuretic target of amiloride. Reductions in lung metastases were demonstrated for two analogs in a late-stage experimental mouse metastasis model, and one analog completely inhibited formation of liver metastases in an orthotopic xenograft mouse model of pancreatic cancer. The results support further evaluation of 6-substituted HMA derivatives as uPA-targeting anticancer drugs.
Collapse
Affiliation(s)
- Benjamin J Buckley
- Molecular Horizons and School of Chemistry & Molecular Bioscience , University of Wollongong , Wollongong , NSW 2522 , Australia.,Illawarra Health & Medical Research Institute , Wollongong , NSW 2522 , Australia
| | - Ashraf Aboelela
- Molecular Horizons and School of Chemistry & Molecular Bioscience , University of Wollongong , Wollongong , NSW 2522 , Australia.,Illawarra Health & Medical Research Institute , Wollongong , NSW 2522 , Australia
| | - Elahe Minaei
- Molecular Horizons and School of Chemistry & Molecular Bioscience , University of Wollongong , Wollongong , NSW 2522 , Australia.,Illawarra Health & Medical Research Institute , Wollongong , NSW 2522 , Australia
| | - Longguang X Jiang
- National Joint Biomedical Engineering Research Centre on Photodynamic Technologies , Fuzhou University , Fujian 350116 , China
| | - Zhihong Xu
- Pancreatic Research Group, South Western Sydney Clinical School , University of New South Wales, and Ingham Institute for Applied Medical Research , Liverpool , NSW 2170 , Australia
| | - Umar Ali
- Molecular Horizons and School of Chemistry & Molecular Bioscience , University of Wollongong , Wollongong , NSW 2522 , Australia.,Illawarra Health & Medical Research Institute , Wollongong , NSW 2522 , Australia
| | - Karen Fildes
- Illawarra Health & Medical Research Institute , Wollongong , NSW 2522 , Australia.,Graduate School of Medicine , University of Wollongong , Wollongong , NSW 2522 , Australia
| | - Chen-Yi Cheung
- Department of Microbiology and Immunology , University of Otago , Otago 9016 , New Zealand
| | - Simon M Cook
- Illawarra Health & Medical Research Institute , Wollongong , NSW 2522 , Australia
| | - Darren C Johnson
- Tri-Institutional PhD Program in Chemical Biology , Memorial Sloan Kettering Cancer Center , New York , New York 10065 , United States
| | - Daniel A Bachovchin
- Tri-Institutional PhD Program in Chemical Biology , Memorial Sloan Kettering Cancer Center , New York , New York 10065 , United States.,Chemical Biology Program , Memorial Sloan Kettering Cancer Center , New York , New York 10065 , United States
| | - Gregory M Cook
- Department of Microbiology and Immunology , University of Otago , Otago 9016 , New Zealand
| | - Minoti Apte
- Pancreatic Research Group, South Western Sydney Clinical School , University of New South Wales, and Ingham Institute for Applied Medical Research , Liverpool , NSW 2170 , Australia
| | - Mingdong Huang
- National Joint Biomedical Engineering Research Centre on Photodynamic Technologies , Fuzhou University , Fujian 350116 , China
| | - Marie Ranson
- Molecular Horizons and School of Chemistry & Molecular Bioscience , University of Wollongong , Wollongong , NSW 2522 , Australia.,Illawarra Health & Medical Research Institute , Wollongong , NSW 2522 , Australia
| | - Michael J Kelso
- Molecular Horizons and School of Chemistry & Molecular Bioscience , University of Wollongong , Wollongong , NSW 2522 , Australia.,Illawarra Health & Medical Research Institute , Wollongong , NSW 2522 , Australia
| |
Collapse
|
14
|
Jiang L, Oldenburg E, Kromann-Hansen T, Xu P, Jensen JK, Andreasen PA, Huang M. Cleavage of peptidic inhibitors by target protease is caused by peptide conformational transition. Biochim Biophys Acta Gen Subj 2018; 1862:2017-2023. [DOI: 10.1016/j.bbagen.2018.06.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 06/08/2018] [Accepted: 06/25/2018] [Indexed: 11/30/2022]
|
15
|
Jiang L, Zhang X, Zhou Y, Chen Y, Luo Z, Li J, Yuan C, Huang M. Halogen bonding for the design of inhibitors by targeting the S1 pocket of serine proteases. RSC Adv 2018; 8:28189-28197. [PMID: 35542712 PMCID: PMC9083945 DOI: 10.1039/c8ra03145b] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 07/24/2018] [Indexed: 12/14/2022] Open
Abstract
Halogen bonding (or X bonding) has attracted increasing interest due to its significant role in molecular recognition in biological systems. Trypsin-like serine proteases have many physiological and pathophysiological functions. There is therefore extensive interest in generating specific inhibitors for pharmacological intervention in their enzymatic activity. We study here if it is possible to use halogenated compounds as the P1 group to bind to the S1 specificity pocket of trypsin-like serine proteases to avoid the low bioavailability of the amidine or guanidine P1 group that is typically used in many inhibitors. We used 4-chlorobenzylamine (ClBA), 4-bromobenzylamine (BrBA) and 4-iodobenzylamine (IBA) as probes to test their binding modes to a trypsin-like serine protease, urokinase-type plasminogen activator (uPA), which has been recognized as a marker for breast cancer and an important target for inhibitor development. The results showed that these compounds inhibited uPA with stronger efficacies compared with their non-halogenated analogues. We also determined the high-resolution crystal structures of uPA in complex with BrBA and IBA, respectively. The structures revealed that BrBA bound to the S1 pocket of uPA via halogen bonds, but IBA did not make halogen bonds with uPA, demonstrating that the iodine may not be the best choice as a target moiety for serine proteases. These results advocate halogen bonding, especially bromine bonding, as an efficient strategy for the future design of novel inhibitors against trypsin-like serine proteases to provide strong potency and promote bioavailability.
Collapse
Affiliation(s)
| | - Xu Zhang
- Center for Life Science, School of Life Sciences, Yunnan University Kunming 650021 China
| | - Yang Zhou
- College of Chemistry, Fuzhou University Fuzhou 350116 China
| | - Yayu Chen
- College of Chemistry, Fuzhou University Fuzhou 350116 China
| | - Zhipu Luo
- Synchrotron Radiation Research Section, NCI, Argonne National Laboratory Argonne Illinois 60439 USA
| | - Jinyu Li
- College of Chemistry, Fuzhou University Fuzhou 350116 China
| | - Cai Yuan
- College of Biological Science and Engineering, Fuzhou University Fuzhou 350116 China
| | - Mingdong Huang
- College of Chemistry, Fuzhou University Fuzhou 350116 China
| |
Collapse
|
16
|
Xu P, Andreasen PA, Huang M. Structural Principles in the Development of Cyclic Peptidic Enzyme Inhibitors. Int J Biol Sci 2017; 13:1222-1233. [PMID: 29104489 PMCID: PMC5666521 DOI: 10.7150/ijbs.21597] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 08/28/2017] [Indexed: 01/23/2023] Open
Abstract
This review summarizes our studies in the development of small cyclic peptides for specifically modulating enzyme activity. Serine proteases share highly similar active sites but perform diverse physiological and pathological functions. From a phage-display peptide library, we isolated two mono-cyclic peptides, upain-1 (CSWRGLENHRMC) and mupain-1 (CPAYSRYLDC), which inhibit the activity of human and murine urokinase-type plasminogen activators (huPA and muPA) with Ki values in the micromolar or sub-micromolar range, respectively. The following affinity maturations significantly enhanced the potencies of the two peptides, 10-fold and >250-fold for upain-1 and mupain-1, respectively. The most potent muPA inhibitor has a potency (Ki = 2 nM) and specificity comparable to mono-clonal antibodies. Furthermore, we also found an unusual feature of mupain-1 that its inhibitory potency can be enhanced by increasing the flexibility, which challenges the traditional viewpoint that higher rigidity leading to higher affinity. Moreover, by changing a few key residues, we converted mupain-1 from a uPA inhibitor to inhibitors of other serine proteases, including plasma kallikrein (PK) and coagulation factor XIa (fXIa). PK and fXIa inhibitors showed Ki values in the low nanomolar range and high specificity. Our studies demonstrate the versatility of small cyclic peptides to engineer inhibitory potency against serine proteases and to provide a new strategy for generating peptide inhibitors of serine proteases.
Collapse
Affiliation(s)
- Peng Xu
- State Key Laboratory of Structural Chemistry and Danish-Chinese Centre for Proteases and Cancer, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian, 350002, P.R. China
| | - Peter A Andreasen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, 8000, Denmark
| | - Mingdong Huang
- State Key Laboratory of Structural Chemistry and Danish-Chinese Centre for Proteases and Cancer, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian, 350002, P.R. China.,College of Chemistry, Fuzhou University, Fuzhou, Fujian, 350116, P.R. China
| |
Collapse
|
17
|
Xue G, Gong L, Yuan C, Xu M, Wang X, Jiang L, Huang M. A structural mechanism of flavonoids in inhibiting serine proteases. Food Funct 2017. [DOI: 10.1039/c6fo01825d] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The crystal structure of quercerin:uPA reveals that catechol serves as the functional group in inhibiting serine proteases.
Collapse
Affiliation(s)
- Guangpu Xue
- State Key Laboratory of Structural Chemistry
- Fujian Institute of Research on the Structure of Matter
- Chinese Academy of Sciences
- Fuzhou 350002
- China
| | - Lihu Gong
- State Key Laboratory of Structural Chemistry
- Fujian Institute of Research on the Structure of Matter
- Chinese Academy of Sciences
- Fuzhou 350002
- China
| | - Cai Yuan
- College of Biological Science and Engineering
- Fuzhou University
- Fuzhou 350116
- China
| | - Mingming Xu
- State Key Laboratory of Structural Chemistry
- Fujian Institute of Research on the Structure of Matter
- Chinese Academy of Sciences
- Fuzhou 350002
- China
| | - Xu Wang
- State Key Laboratory of Structural Chemistry
- Fujian Institute of Research on the Structure of Matter
- Chinese Academy of Sciences
- Fuzhou 350002
- China
| | | | - Mingdong Huang
- State Key Laboratory of Structural Chemistry
- Fujian Institute of Research on the Structure of Matter
- Chinese Academy of Sciences
- Fuzhou 350002
- China
| |
Collapse
|
18
|
Maini R, Umemoto S, Suga H. Ribosome-mediated synthesis of natural product-like peptides via cell-free translation. Curr Opin Chem Biol 2016; 34:44-52. [DOI: 10.1016/j.cbpa.2016.06.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 06/06/2016] [Indexed: 11/29/2022]
|
19
|
Kromann-Hansen T, Oldenburg E, Yung KWY, Ghassabeh GH, Muyldermans S, Declerck PJ, Huang M, Andreasen PA, Ngo JCK. A Camelid-derived Antibody Fragment Targeting the Active Site of a Serine Protease Balances between Inhibitor and Substrate Behavior. J Biol Chem 2016; 291:15156-68. [PMID: 27226628 DOI: 10.1074/jbc.m116.732503] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Indexed: 01/15/2023] Open
Abstract
A peptide segment that binds the active site of a serine protease in a substrate-like manner may behave like an inhibitor or a substrate. However, there is sparse information on which factors determine the behavior a particular peptide segment will exhibit. Here, we describe the first x-ray crystal structure of a nanobody in complex with a serine protease. The nanobody displays a new type of interaction between an antibody and a serine protease as it inserts its complementary determining region-H3 loop into the active site of the protease in a substrate-like manner. The unique binding mechanism causes the nanobody to behave as a strong inhibitor as well as a poor substrate. Intriguingly, its substrate behavior is incomplete, as 30-40% of the nanobody remained intact and inhibitory after prolonged incubation with the protease. Biochemical analysis reveals that an intra-loop interaction network within the complementary determining region-H3 of the nanobody balances its inhibitor versus substrate behavior. Collectively, our results unveil molecular factors, which may be a general mechanism to determine the substrate versus inhibitor behavior of other protease inhibitors.
Collapse
Affiliation(s)
- Tobias Kromann-Hansen
- From the Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, 8000 Aarhus C, Denmark,
| | - Emil Oldenburg
- From the Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, 8000 Aarhus C, Denmark
| | - Kristen Wing Yu Yung
- the School of Life Sciences, Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region, China
| | - Gholamreza H Ghassabeh
- the Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussels, 1050 Brussels, Belgium, Nanobody Service Facility, Flanders Institute for Biotechnology, 1050 Brussels, Belgium
| | - Serge Muyldermans
- the Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussels, 1050 Brussels, Belgium
| | - Paul J Declerck
- the Laboratory for Therapeutic and Diagnostic Antibodies, Department of Pharmaceutical and Pharmacological Sciences, Katholieke Universiteit, 3000 Leuven, Belgium, and
| | - Mingdong Huang
- the State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Science, Fuzhou, Fujian 350002, China
| | - Peter A Andreasen
- From the Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, 8000 Aarhus C, Denmark
| | - Jacky Chi Ki Ngo
- the School of Life Sciences, Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region, China
| |
Collapse
|
20
|
Gong L, Proulle V, Fang C, Hong Z, Lin Z, Liu M, Xue G, Yuan C, Lin L, Furie B, Flaumenhaft R, Andreasen P, Furie B, Huang M. A specific plasminogen activator inhibitor-1 antagonist derived from inactivated urokinase. J Cell Mol Med 2016; 20:1851-60. [PMID: 27197780 PMCID: PMC4876229 DOI: 10.1111/jcmm.12875] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 03/13/2016] [Indexed: 12/27/2022] Open
Abstract
Fibrinolysis is a process responsible for the dissolution of formed thrombi to re‐establish blood flow after thrombus formation. Plasminogen activator inhibitor‐1 (PAI‐1) inhibits urokinase‐type and tissue‐type plasminogen activator (uPA and tPA) and is the major negative regulator of fibrinolysis. Inhibition of PAI‐1 activity prevents thrombosis and accelerates fibrinolysis. However, a specific antagonist of PAI‐1 is currently unavailable for therapeutic use. We screened a panel of uPA variants with mutations at and near the active site to maximize their binding to PAI‐1 and identified a potent PAI‐1 antagonist, PAItrap. PAItrap is the serine protease domain of urokinase containing active‐site mutation (S195A) and four additional mutations (G37bR–R217L–C122A–N145Q). PAItrap inhibits human recombinant PAI‐1 with high potency (Kd = 0.15 nM) and high specificity. In vitro using human plasma, PAItrap showed significant thrombolytic activity by inhibiting endogenous PAI‐1. In addition, PAItrap inhibits both human and murine PAI‐1, allowing the evaluation in murine models. In vivo, using a laser‐induced thrombosis mouse model in which thrombus formation and fibrinolysis are monitored by intravital microscopy, PAItrap reduced fibrin generation and inhibited platelet accumulation following vascular injury. Therefore, this work demonstrates the feasibility to generate PAI‐1 inhibitors using inactivated urokinase.
Collapse
Affiliation(s)
- Lihu Gong
- State Key Laboratory of Structural Chemistry and Danish-Chinese Centre for Proteases and Cancer, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Valerie Proulle
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Chao Fang
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Zebin Hong
- State Key Laboratory of Structural Chemistry and Danish-Chinese Centre for Proteases and Cancer, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian, China
| | - Zhonghui Lin
- State Key Laboratory of Structural Chemistry and Danish-Chinese Centre for Proteases and Cancer, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian, China
| | - Min Liu
- State Key Laboratory of Structural Chemistry and Danish-Chinese Centre for Proteases and Cancer, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Guangpu Xue
- State Key Laboratory of Structural Chemistry and Danish-Chinese Centre for Proteases and Cancer, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian, China
| | - Cai Yuan
- State Key Laboratory of Structural Chemistry and Danish-Chinese Centre for Proteases and Cancer, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian, China
| | - Lin Lin
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Barbara Furie
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Robert Flaumenhaft
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Peter Andreasen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, Denmark
| | - Bruce Furie
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Mingdong Huang
- State Key Laboratory of Structural Chemistry and Danish-Chinese Centre for Proteases and Cancer, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian, China. .,University of Chinese Academy of Sciences, Beijing, China. .,Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
21
|
Dupont DM, Bjerregaard N, Verpaalen B, Andreasen PA, Jensen JK. Building a Molecular Trap for a Serine Protease from Aptamer and Peptide Modules. Bioconjug Chem 2016; 27:918-26. [PMID: 26926041 DOI: 10.1021/acs.bioconjchem.6b00007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In drug development, molecular intervention strategies are usually based on interference with a single protein function, such as enzyme activity or receptor binding. However, in many cases, protein drug targets are multifunctional, with several molecular functions contributing to their pathophysiological actions. Aptamers and peptides are interesting synthetic building blocks for the design of multivalent molecules capable of modulating multiple functions of a target protein. Here, we report a molecular trap with the ability to interfere with the activation, catalytic activity, receptor binding, etc. of the serine protease urokinase-type plasminogen activator (uPA) by a rational combination of two RNA aptamers and a peptide with different inhibitory properties. The assembly of these artificial inhibitors into one molecule enhanced the inhibitory activity between 10- and 10,000-fold toward several functions of uPA. The study highlights the potential of multivalent designs and illustrates how they can easily be constructed from aptamers and peptides using nucleic acid engineering, chemical synthesis, and bioconjugation chemistry. By aptamer to aptamer and aptamer to peptide conjugation, we created, to the best of our knowledge, the first trivalent molecule which combines three artificial inhibitors binding to three different sites in a protein target. We hypothesize that by simultaneously preventing all of the functional interactions and activities of the target protein, this approach may represent an alternative to siRNA technology for a functional knockout.
Collapse
Affiliation(s)
- Daniel M Dupont
- Department of Molecular Biology and Genetics, Aarhus University , Gustav Wieds Vej 10C, 8000 Aarhus, Denmark
| | - Nils Bjerregaard
- Department of Molecular Biology and Genetics, Aarhus University , Gustav Wieds Vej 10C, 8000 Aarhus, Denmark
| | - Ben Verpaalen
- Department of Molecular Biology and Genetics, Aarhus University , Gustav Wieds Vej 10C, 8000 Aarhus, Denmark
| | - Peter A Andreasen
- Department of Molecular Biology and Genetics, Aarhus University , Gustav Wieds Vej 10C, 8000 Aarhus, Denmark
| | - Jan K Jensen
- Department of Molecular Biology and Genetics, Aarhus University , Gustav Wieds Vej 10C, 8000 Aarhus, Denmark
| |
Collapse
|
22
|
Jiang L, Andersen LM, Andreasen PA, Chen L, Huang M. Insights into the serine protease mechanism based on structural observations of the conversion of a peptidyl serine protease inhibitor to a substrate. Biochim Biophys Acta Gen Subj 2015; 1860:599-606. [PMID: 26691138 DOI: 10.1016/j.bbagen.2015.12.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Revised: 12/02/2015] [Accepted: 12/11/2015] [Indexed: 11/28/2022]
Abstract
BACKGROUND Serine proteases are one of the most studied group of enzymes. Despite the extensive mechanistic studies, some crucial details remain controversial, for example, how the cleaved product is released in the catalysis reaction. A cyclic peptidyl inhibitor (CSWRGLENHRMC, upain-1) of a serine protease, urokinase-type plasminogen activator (uPA), was found to become a slow substrate and cleaved slowly upon the replacement of single residue (W3A). METHODS By taking advantage of the unique property of this peptide, we report the high-resolution structures of uPA in complex with upain-1-W3A peptide at four different pH values by X-ray crystallography. RESULTS In the structures obtained at low pH (pH4.6 and 5.5), the cyclic peptide upain-1-W3A was found to be intact and remained in the active site of uPA. At 7.4, the scissile bond of the peptide was found cleaved, showing that the peptide became a uPA substrate. At pH9.0, the C-terminal part of the substrate was no longer visible, and only the P1 residue occupying the S1 pocket was identified. CONCLUSIONS The analysis of these structures provides explanations why the upain-1-W3A is a slow substrate. In addition, we clearly identified the cleaved fragments of the peptide at both sides of the scissile bond in the active site of the enzyme, showing a slow release of the cleaved peptide. GENERAL SIGNIFICANCE This work indicates that the quick release of the cleaved P' fragment after the first step of hydrolysis may not always be needed for the second hydrolysis.
Collapse
Affiliation(s)
- Longguang Jiang
- State Key Lab of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, 155 Yang Qiao West Road, Fuzhou, Fujian 350002, China; Danish-Chinese Centre for Proteases and Cancer, Denmark. http://www.proteasesandcancer.org
| | - Lisbeth Moreau Andersen
- Danish-Chinese Centre for Proteases and Cancer, Denmark; Department of Molecular Biology and Genetics, Aarhus University, Aarhus 8000-DK, Denmark. http://www.proteasesandcancer.org
| | - Peter A Andreasen
- Danish-Chinese Centre for Proteases and Cancer, Denmark; Department of Molecular Biology and Genetics, Aarhus University, Aarhus 8000-DK, Denmark. http://www.proteasesandcancer.org
| | - Liqing Chen
- University of Alabama in Huntsville, Huntsville, AL 35899, USA
| | - Mingdong Huang
- State Key Lab of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, 155 Yang Qiao West Road, Fuzhou, Fujian 350002, China; Danish-Chinese Centre for Proteases and Cancer, Denmark.
| |
Collapse
|
23
|
Selection of High-Affinity Peptidic Serine Protease Inhibitors with Increased Binding Entropy from a Back-Flip Library of Peptide-Protease Fusions. J Mol Biol 2015; 427:3110-22. [PMID: 26281711 DOI: 10.1016/j.jmb.2015.08.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Revised: 07/19/2015] [Accepted: 08/07/2015] [Indexed: 11/21/2022]
Abstract
We have developed a new concept for designing peptidic protein modulators, by recombinantly fusing the peptidic modulator, with randomized residues, directly to the target protein via a linker and screening for internal modulation of the activity of the protein. We tested the feasibility of the concept by fusing a 10-residue-long, disulfide-bond-constrained inhibitory peptide, randomized in selected positions, to the catalytic domain of the serine protease murine urokinase-type plasminogen activator. High-affinity inhibitory peptide variants were identified as those that conferred to the fusion protease the lowest activity for substrate hydrolysis. The usefulness of the strategy was demonstrated by the selection of peptidic inhibitors of murine urokinase-type plasminogen activator with a low nanomolar affinity. The high affinity could not have been predicted by rational considerations, as the high affinity was associated with a loss of polar interactions and an increased binding entropy.
Collapse
|
24
|
Distinctive binding modes and inhibitory mechanisms of two peptidic inhibitors of urokinase-type plasminogen activator with isomeric P1 residues. Int J Biochem Cell Biol 2015; 62:88-92. [DOI: 10.1016/j.biocel.2015.02.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 12/02/2014] [Accepted: 02/23/2015] [Indexed: 01/29/2023]
|
25
|
Jongkees SAK, Hipolito CJ, Rogers JM, Suga H. Model foldamers: applications and structures of stable macrocyclic peptides identified using in vitro selection. NEW J CHEM 2015. [DOI: 10.1039/c4nj01633e] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
A survey of crystal- and solution-structure information for macrocyclic peptides, illustrating common folding patterns and target binding effects.
Collapse
Affiliation(s)
- Seino A. K. Jongkees
- Department of Chemistry
- Graduate School of Science
- University of Tokyo
- Tokyo 113-0033
- Japan
| | | | - Joseph M. Rogers
- Department of Chemistry
- Graduate School of Science
- University of Tokyo
- Tokyo 113-0033
- Japan
| | - Hiroaki Suga
- Department of Chemistry
- Graduate School of Science
- University of Tokyo
- Tokyo 113-0033
- Japan
| |
Collapse
|
26
|
Wang F, Li S, Wang Y, Zhu H, Zhang X, Zhao M, Wu J, Peng S. Enantiomeric diketopiperazines: getting insight into the impact of the configuration on the conformation, nanoimage, u-PA inhibition and anti-metastatic activity. MEDCHEMCOMM 2015. [DOI: 10.1039/c5md00007f] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The impact of the configuration of enantiomeric diketopiperazines on the conformation, nanoimage, u-PA inhibition and anti-metastatic activity was explored.
Collapse
Affiliation(s)
- Feng Wang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China
- Beijing Laboratory of Biomedical Materials
- College of Pharmaceutical Sciences, of Capital Medical University
- Beijing
| | - Shan Li
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China
- Beijing Laboratory of Biomedical Materials
- College of Pharmaceutical Sciences, of Capital Medical University
- Beijing
| | - Yuji Wang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China
- Beijing Laboratory of Biomedical Materials
- College of Pharmaceutical Sciences, of Capital Medical University
- Beijing
| | - Haimei Zhu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China
- Beijing Laboratory of Biomedical Materials
- College of Pharmaceutical Sciences, of Capital Medical University
- Beijing
| | - Xiaoyi Zhang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China
- Beijing Laboratory of Biomedical Materials
- College of Pharmaceutical Sciences, of Capital Medical University
- Beijing
| | - Ming Zhao
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China
- Beijing Laboratory of Biomedical Materials
- College of Pharmaceutical Sciences, of Capital Medical University
- Beijing
| | - Jianhui Wu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China
- Beijing Laboratory of Biomedical Materials
- College of Pharmaceutical Sciences, of Capital Medical University
- Beijing
| | - Shiqi Peng
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China
- Beijing Laboratory of Biomedical Materials
- College of Pharmaceutical Sciences, of Capital Medical University
- Beijing
| |
Collapse
|
27
|
Zhao B, Xu P, Jiang L, Paaske B, Kromann-Hansen T, Jensen JK, Sørensen HP, Liu Z, Nielsen JT, Christensen A, Hosseini M, Sørensen KK, Nielsen NC, Jensen KJ, Huang M, Andreasen PA. A cyclic peptidic serine protease inhibitor: increasing affinity by increasing peptide flexibility. PLoS One 2014; 9:e115872. [PMID: 25545505 PMCID: PMC4278837 DOI: 10.1371/journal.pone.0115872] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 11/19/2014] [Indexed: 11/19/2022] Open
Abstract
Peptides are attracting increasing interest as protease inhibitors. Here, we demonstrate a new inhibitory mechanism and a new type of exosite interactions for a phage-displayed peptide library-derived competitive inhibitor, mupain-1 (CPAYSRYLDC), of the serine protease murine urokinase-type plasminogen activator (uPA). We used X-ray crystal structure analysis, site-directed mutagenesis, liquid state NMR, surface plasmon resonance analysis, and isothermal titration calorimetry and wild type and engineered variants of murine and human uPA. We demonstrate that Arg6 inserts into the S1 specificity pocket, its carbonyl group aligning improperly relative to Ser195 and the oxyanion hole, explaining why the peptide is an inhibitor rather than a substrate. Substitution of the P1 Arg with novel unnatural Arg analogues with aliphatic or aromatic ring structures led to an increased affinity, depending on changes in both P1 - S1 and exosite interactions. Site-directed mutagenesis showed that exosite interactions, while still supporting high affinity binding, differed substantially between different uPA variants. Surprisingly, high affinity binding was facilitated by Ala-substitution of Asp9 of the peptide, in spite of a less favorable binding entropy and loss of a polar interaction. We conclude that increased flexibility of the peptide allows more favorable exosite interactions, which, in combination with the use of novel Arg analogues as P1 residues, can be used to manipulate the affinity and specificity of this peptidic inhibitor, a concept different from conventional attempts at improving inhibitor affinity by reducing the entropic burden.
Collapse
Affiliation(s)
- Baoyu Zhao
- Danish-Chinese Centre for Proteases and Cancer, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, China
| | - Peng Xu
- Danish-Chinese Centre for Proteases and Cancer, Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Longguang Jiang
- Danish-Chinese Centre for Proteases and Cancer, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, China
| | - Berit Paaske
- Nanoscience Center and Department of Chemistry, University of Aarhus, Aarhus, Denmark
| | - Tobias Kromann-Hansen
- Danish-Chinese Centre for Proteases and Cancer, Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Jan K. Jensen
- Danish-Chinese Centre for Proteases and Cancer, Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Hans Peter Sørensen
- Danish-Chinese Centre for Proteases and Cancer, Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Zhuo Liu
- Danish-Chinese Centre for Proteases and Cancer, Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Jakob T. Nielsen
- Nanoscience Center and Department of Chemistry, University of Aarhus, Aarhus, Denmark
| | - Anni Christensen
- Danish-Chinese Centre for Proteases and Cancer, Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Masood Hosseini
- Department of Chemistry, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Kasper K. Sørensen
- Department of Chemistry, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | | | - Knud J. Jensen
- Department of Chemistry, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Mingdong Huang
- Danish-Chinese Centre for Proteases and Cancer, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, China
| | - Peter A. Andreasen
- Danish-Chinese Centre for Proteases and Cancer, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, China
- Danish-Chinese Centre for Proteases and Cancer, Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
- * E-mail:
| |
Collapse
|
28
|
Wyganowska-Świątkowska M, Surdacka A, Skrzypczak-Jankun E, Jankun J. The plasminogen activation system in periodontal tissue (Review). Int J Mol Med 2014; 33:763-8. [PMID: 24535478 DOI: 10.3892/ijmm.2014.1653] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 01/28/2014] [Indexed: 11/05/2022] Open
Abstract
The plasminogen activation system (PAS) plays an essential role in tissue proteolysis in physiological and pathological processes. Periodontitis is a chronic infection associated with increased proteolysis driven by plasminogen activation. In this comprehensive review, we summarise the effects of PAS in wound healing, tissue remodelling, inflammation, bacterial infection, and in the initiation and progression of periodontal disease. Specifically, we discuss the role of plasminogen activators (PAs), including urokinase PA (uPA), tissue-type PA (tPA), PA inhibitor type 1 (PAI-1) and 2 (PAI-2) and activated plasminogen in periodontal tissue, where their concentrations can reach much higher values than those found in other parts of the body. We also discuss whether PA deficiencies can have effects on periodontal tissue. We conclude that in periodontal disease, PAS is unbalanced and equalizing its function can improve the clinical periodontal tissue condition.
Collapse
Affiliation(s)
| | - Anna Surdacka
- Department of Conservative Dentistry and Periodontology, Poznań University of Medical Sciences, Poznań 60-820, Poland
| | - Ewa Skrzypczak-Jankun
- Urology Research Center, Department of Urology, College of Medicine, University of Toledo, Toledo, OH 43614, USA
| | - Jerzy Jankun
- Protein Research Chair, Department of Biochemistry, College of Sciences, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| |
Collapse
|
29
|
Roodbeen R, Paaske B, Jiang L, Jensen JK, Christensen A, Nielsen JT, Huang M, Mulder FAA, Nielsen NC, Andreasen PA, Jensen KJ. Bicyclic Peptide Inhibitor of Urokinase-Type Plasminogen Activator: Mode of Action. Chembiochem 2013; 14:2179-88. [DOI: 10.1002/cbic.201300335] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Indexed: 01/24/2023]
|
30
|
Kromann-Hansen T, Lund IK, Liu Z, Andreasen PA, Høyer-Hansen G, Sørensen HP. Allosteric inactivation of a trypsin-like serine protease by an antibody binding to the 37- and 70-loops. Biochemistry 2013; 52:7114-26. [PMID: 24079451 DOI: 10.1021/bi400491k] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Serine protease catalytic activity is in many cases regulated by conformational changes initiated by binding of physiological modulators to exosites located distantly from the active site. Inhibitory monoclonal antibodies binding to such exosites are potential therapeutics and offer opportunities for elucidating fundamental allosteric mechanisms. The monoclonal antibody mU1 has previously been shown to be able to inhibit the function of murine urokinase-type plasminogen activator in vivo. We have now mapped the epitope of mU1 to the catalytic domain's 37- and 70-loops, situated about 20 Å from the S1 specificity pocket of the active site. Our data suggest that binding of mU1 destabilizes the catalytic domain and results in conformational transition into a state, in which the N-terminal amino group of Ile16 is less efficiently stabilizing the oxyanion hole and in which the active site has a reduced affinity for substrates and inhibitors. Furthermore, we found evidence for functional interactions between residues in uPA's C-terminal catalytic domain and its N-terminal A-chain, as deletion of the A-chain facilitates the mU1-induced conformational distortion. The inactive, distorted state is by several criteria similar to the E* conformation described for other serine proteases. Hence, agents targeting serine protease conformation through binding to exosites in the 37- and 70-loops represent a new class of potential therapeutics.
Collapse
Affiliation(s)
- Tobias Kromann-Hansen
- Danish-Chinese Centre for Proteases and Cancer and ‡Department of Molecular Biology and Genetics, Aarhus University , DK-8000 Aarhus C, Denmark
| | | | | | | | | | | |
Collapse
|
31
|
Frost JR, Smith JM, Fasan R. Design, synthesis, and diversification of ribosomally derived peptide macrocycles. Curr Opin Struct Biol 2013; 23:571-80. [PMID: 23856642 DOI: 10.1016/j.sbi.2013.06.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Revised: 06/19/2013] [Accepted: 06/20/2013] [Indexed: 10/26/2022]
Abstract
Ring topologies are widespread structural motifs among biologically active peptides found in nature. The recurrence of this motif is linked to the inherent advantages resulting from backbone cyclization, which include increased resistance against proteolytic degradation, improved cell permeability, and tighter and more specific interaction with the respective biomolecular target. Inspired by these natural product topologies, a number of groups have recently focused on developing methodologies that hinge upon the chemical elaboration of ribosomally derived polypeptides toward the synthesis and diversification of macrocyclic peptide structures. In this review, we highlight recent advances in this emerging new area and discuss the opportunities created by these methods toward the discovery of new functional entities.
Collapse
Affiliation(s)
- John R Frost
- Department of Chemistry, University of Rochester, Rochester, NY 14627, USA
| | | | | |
Collapse
|
32
|
Chen S, Rentero Rebollo I, Buth SA, Morales-Sanfrutos J, Touati J, Leiman PG, Heinis C. Bicyclic peptide ligands pulled out of cysteine-rich peptide libraries. J Am Chem Soc 2013; 135:6562-9. [PMID: 23560397 DOI: 10.1021/ja400461h] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Bicyclic peptide ligands were found to have good binding affinity and target specificity. However, the method applied to generate bicyclic ligands based on phage-peptide alkylation is technically complex and limits its application to specialized laboratories. Herein, we report a method that involves a simpler and more robust procedure that additionally allows screening of structurally more diverse bicyclic peptide libraries. In brief, phage-encoded combinatorial peptide libraries of the format X(m)CX(n)CX(o)CX(p) are oxidized to connect two pairs of cysteines (C). This allows the generation of 3 × (m + n + o + p) different peptide topologies because the fourth cysteine can appear in any of the (m + n + o + p) randomized amino acid positions (X). Panning of such libraries enriched strongly peptides with four cysteines and yielded tight binders to protein targets. X-ray structure analysis revealed an important structural role of the disulfide bridges. In summary, the presented approach offers facile access to bicyclic peptide ligands with good binding affinities.
Collapse
Affiliation(s)
- Shiyu Chen
- Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
An important regulatory mechanism of serine proteases is the proteolytic conversion of the inactive pro-enzyme, or zymogen, into the active enzyme. This activation process is generally considered an irreversible process. In the present study, we demonstrate that an active enzyme can be converted back into its zymogen form. We determined the crystal structure of uPA (urokinase-type plasminogen activator) in complex with an inhibitory antibody, revealing that the antibody 'rezymogenizes' already activated uPA. The present study demonstrates a new regulatory mechanism of protease activity, which is also an extreme case of protein allostery. Mechanistically, the antibody binds a single surface-exposed loop, named the autolysis loop, thereby preventing the stabilization of uPA in its active conformation. We argue that this autolysis loop is a key structural element for rezymogenation of other proteases, and will be a new target site for pharmacological intervention with serine protease activity.
Collapse
|
34
|
Angelini A, Cendron L, Chen S, Touati J, Winter G, Zanotti G, Heinis C. Bicyclic peptide inhibitor reveals large contact interface with a protease target. ACS Chem Biol 2012; 7:817-21. [PMID: 22304751 DOI: 10.1021/cb200478t] [Citation(s) in RCA: 138] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
From a large combinatorial library of chemically constrained bicyclic peptides we isolated a selective and potent (K(i) = 53 nM) inhibitor of human urokinase-type plasminogen activator (uPA) and crystallized the complex. This revealed an extended structure of the peptide with both peptide loops engaging the target to form a large interaction surface of 701 Å(2) with multiple hydrogen bonds and complementary charge interactions, explaining the high affinity and specificity of the inhibitor. The interface resembles that between two proteins and suggests that these constrained peptides have the potential to act as small protein mimics.
Collapse
Affiliation(s)
- Alessandro Angelini
- Institute
of Chemical Sciences
and Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Laura Cendron
- Department of Biological Chemistry, University of Padua, Viale G. Colombo 3, 35131 Padua,
Italy
- Venetian Institute of Molecular Medicine (VIMM), Via Giuseppe Orus 2, 35129
Padua, Italy
| | - Shiyu Chen
- Institute
of Chemical Sciences
and Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Jeremy Touati
- Institute
of Chemical Sciences
and Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Greg Winter
- Laboratory of
Molecular Biology, Medical Research Council, Hills Road, Cambridge CB2
0QH, U.K
| | - Giuseppe Zanotti
- Department of Biological Chemistry, University of Padua, Viale G. Colombo 3, 35131 Padua,
Italy
- Venetian Institute of Molecular Medicine (VIMM), Via Giuseppe Orus 2, 35129
Padua, Italy
| | - Christian Heinis
- Institute
of Chemical Sciences
and Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| |
Collapse
|
35
|
Baeriswyl V, Rapley H, Pollaro L, Stace C, Teufel D, Walker E, Chen S, Winter G, Tite J, Heinis C. Bicyclic peptides with optimized ring size inhibit human plasma kallikrein and its orthologues while sparing paralogous proteases. ChemMedChem 2012; 7:1173-6. [PMID: 22492508 DOI: 10.1002/cmdc.201200071] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Indexed: 11/10/2022]
Affiliation(s)
- Vanessa Baeriswyl
- Institute of Chemical Sciences and Engineering (ISIC), Ecole Polytechnique Fédérale de Lausanne (EPFL), CH B3 391, Station 6, 1015 Lausanne, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Targeting the autolysis loop of urokinase-type plasminogen activator with conformation-specific monoclonal antibodies. Biochem J 2011; 438:39-51. [PMID: 21635223 DOI: 10.1042/bj20110129] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Tight regulation of serine proteases is essential for their physiological function, and unbalanced states of protease activity have been implicated in a variety of human diseases. One key example is the presence of uPA (urokinase-type plasminogen activator) in different human cancer types, with high levels correlating with a poor prognosis. This observation has stimulated efforts into finding new principles for intervening with uPA's activity. In the present study we characterize the so-called autolysis loop in the catalytic domain of uPA as a potential inhibitory target. This loop was found to harbour the epitopes for three conformation-specific monoclonal antibodies, two with a preference for the zymogen form pro-uPA, and one with a preference for active uPA. All three antibodies were shown to have overlapping epitopes, with three common residues being crucial for all three antibodies, demonstrating a direct link between conformational changes of the autolysis loop and the creation of a catalytically mature active site. All three antibodies are potent inhibitors of uPA activity, the two pro-uPA-specific ones by inhibiting conversion of pro-uPA to active uPA and the active uPA-specific antibody by shielding the access of plasminogen to the active site. Furthermore, using immunofluorescence, the conformation-specific antibodies mAb-112 and mAb-12E6B10 enabled us to selectively stain pro-uPA or active uPA on the surface of cultured cells. Moreover, in various independent model systems, the antibodies inhibited tumour cell invasion and dissemination, providing evidence for the feasibility of pharmaceutical intervention with serine protease activity by targeting surface loops that undergo conformational changes during zymogen activation.
Collapse
|
37
|
The Binding Mechanism of a Peptidic Cyclic Serine Protease Inhibitor. J Mol Biol 2011; 412:235-50. [DOI: 10.1016/j.jmb.2011.07.028] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Revised: 07/08/2011] [Accepted: 07/14/2011] [Indexed: 11/19/2022]
|
38
|
Hosseini M, Jiang L, Sørensen HP, Jensen JK, Christensen A, Fogh S, Yuan C, Andersen LM, Huang M, Andreasen PA, Jensen KJ. Elucidation of the Contribution of Active Site and Exosite Interactions to Affinity and Specificity of Peptidylic Serine Protease Inhibitors Using Non-Natural Arginine Analogs. Mol Pharmacol 2011; 80:585-97. [DOI: 10.1124/mol.111.072280] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
39
|
Yuan C, Chen L, Meehan EJ, Daly N, Craik DJ, Huang M, Ngo JC. Structure of catalytic domain of Matriptase in complex with Sunflower trypsin inhibitor-1. BMC STRUCTURAL BIOLOGY 2011; 11:30. [PMID: 21693064 PMCID: PMC3141381 DOI: 10.1186/1472-6807-11-30] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Accepted: 06/22/2011] [Indexed: 12/17/2022]
Abstract
Background Matriptase is a type II transmembrane serine protease that is found on the surfaces of epithelial cells and certain cancer cells. Matriptase has been implicated in the degradation of certain extracellular matrix components as well as the activation of various cellular proteins and proteases, including hepatocyte growth factor and urokinase. Sunflower trypsin inhibitor-1 (SFTI-1), a cyclic peptide inhibitor originally isolated from sunflower seeds, exhibits potent inhibitory activity toward matriptase. Results We have engineered and produced recombinant proteins of the matriptase protease domain, and have determined the crystal structures of the protease:SFTI-1 complex at 2.0 Å as well as the protease:benzamidine complex at 1.2 Å. These structures elaborate the structural basis of substrate selectivity of matriptase, and show that the matriptase S1 substrate specificity pocket is larger enough to allow movement of benzamidine inside the S1 pocket. Our study also reveals that SFTI-1 binds to matriptase in a way similar to its binding to trypsin despite the significantly different isoelectric points of the two proteins (5.6 vs. 8.2). Conclusions This work helps to define the structural basis of substrate specificity of matriptase and the interactions between the inhibitor and protease. The complex structure also provides a structural template for designing new SFTI-1 derivatives with better potency and selectivity against matriptase and other proteases.
Collapse
Affiliation(s)
- Cai Yuan
- State Key Lab of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian 350002, China
| | | | | | | | | | | | | |
Collapse
|
40
|
Sukhwal A, Bhattacharyya M, Vishveshwara S. Network approach for capturing ligand-induced subtle global changes in protein structures. ACTA CRYSTALLOGRAPHICA SECTION D: BIOLOGICAL CRYSTALLOGRAPHY 2011; 67:429-39. [PMID: 21543845 DOI: 10.1107/s0907444911007062] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Accepted: 02/24/2011] [Indexed: 01/12/2023]
Abstract
Ligand-induced conformational changes in proteins are of immense functional relevance. It is a major challenge to elucidate the network of amino acids that are responsible for the percolation of ligand-induced conformational changes to distal regions in the protein from a global perspective. Functionally important subtle conformational changes (at the level of side-chain noncovalent interactions) upon ligand binding or as a result of environmental variations are also elusive in conventional studies such as those using root-mean-square deviations (r.m.s.d.s). In this article, the network representation of protein structures and their analyses provides an efficient tool to capture these variations (both drastic and subtle) in atomistic detail in a global milieu. A generalized graph theoretical metric, using network parameters such as cliques and/or communities, is used to determine similarities or differences between structures in a rigorous manner. The ligand-induced global rewiring in the protein structures is also quantified in terms of network parameters. Thus, a judicious use of graph theory in the context of protein structures can provide meaningful insights into global structural reorganizations upon perturbation and can also be helpful for rigorous structural comparison. Data sets for the present study include high-resolution crystal structures of serine proteases from the S1A family and are probed to quantify the ligand-induced subtle structural variations.
Collapse
Affiliation(s)
- Anshul Sukhwal
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore 560012, India
| | | | | |
Collapse
|
41
|
Lin Z, Jiang L, Yuan C, Jensen JK, Zhang X, Luo Z, Furie BC, Furie B, Andreasen PA, Huang M. Structural basis for recognition of urokinase-type plasminogen activator by plasminogen activator inhibitor-1. J Biol Chem 2011; 286:7027-32. [PMID: 21199867 PMCID: PMC3044959 DOI: 10.1074/jbc.m110.204537] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2010] [Revised: 12/15/2010] [Indexed: 11/06/2022] Open
Abstract
Plasminogen activator inhibitor-1 (PAI-1), together with its physiological target urokinase-type plasminogen activator (uPA), plays a pivotal role in fibrinolysis, cell migration, and tissue remodeling and is currently recognized as being among the most extensively validated biological prognostic factors in several cancer types. PAI-1 specifically and rapidly inhibits uPA and tissue-type PA (tPA). Despite extensive structural/functional studies on these two reactions, the underlying structural mechanism has remained unknown due to the technical difficulties of obtaining the relevant structures. Here, we report a strategy to generate a PAI-1·uPA(S195A) Michaelis complex and present its crystal structure at 2.3-Å resolution. In this structure, the PAI-1 reactive center loop serves as a bait to attract uPA onto the top of the PAI-1 molecule. The P4-P3' residues of the reactive center loop interact extensively with the uPA catalytic site, accounting for about two-thirds of the total contact area. Besides the active site, almost all uPA exosite loops, including the 37-, 60-, 97-, 147-, and 217-loops, are involved in the interaction with PAI-1. The uPA 37-loop makes an extensive interaction with PAI-1 β-sheet B, and the 147-loop directly contacts PAI-1 β-sheet C. Both loops are important for initial Michaelis complex formation. This study lays down a foundation for understanding the specificity of PAI-1 for uPA and tPA and provides a structural basis for further functional studies.
Collapse
Affiliation(s)
- Zhonghui Lin
- From the State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences and
- the Danish-Chinese Centre for Proteases and Cancer, Fuzhou 350002, China
| | - Longguang Jiang
- From the State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences and
- the Danish-Chinese Centre for Proteases and Cancer, Fuzhou 350002, China
| | - Cai Yuan
- From the State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences and
- the Danish-Chinese Centre for Proteases and Cancer, Fuzhou 350002, China
- the Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215
| | - Jan K. Jensen
- the Danish-Chinese Centre for Proteases and Cancer, Fuzhou 350002, China
- the Department of Molecular Biology, Aarhus University, Gustav Wieds Vej 10C, 8000 Aarhus C, Denmark, and
| | - Xu Zhang
- From the State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences and
- the Danish-Chinese Centre for Proteases and Cancer, Fuzhou 350002, China
| | - Zhipu Luo
- From the State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences and
- the Danish-Chinese Centre for Proteases and Cancer, Fuzhou 350002, China
| | - Barbara C. Furie
- the Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215
| | - Bruce Furie
- the Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215
| | - Peter A. Andreasen
- the Danish-Chinese Centre for Proteases and Cancer, Fuzhou 350002, China
- the Department of Molecular Biology, Aarhus University, Gustav Wieds Vej 10C, 8000 Aarhus C, Denmark, and
| | - Mingdong Huang
- From the State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences and
- the Danish-Chinese Centre for Proteases and Cancer, Fuzhou 350002, China
- the Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215
| |
Collapse
|
42
|
Bøtkjaer KA, Byszuk AA, Andersen LM, Christensen A, Andreasen PA, Blouse GE. Nonproteolytic induction of catalytic activity into the single-chain form of urokinase-type plasminogen activator by dipeptides. Biochemistry 2009; 48:9606-17. [PMID: 19705874 DOI: 10.1021/bi900510f] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Serine proteases are initially synthesized as single-chain proenzymes with activities that are many orders of magnitude lower than those of the mature enzyme. Proteolytic cleavage of an exposed loop liberates a new amino terminus that inserts into a hydrophobic pocket and forms a stabilizing salt bridge with a ubiquitously conserved aspartate residue, resulting in a conformational change organizing the mature oxyanion hole. In a decisive 1976 work, Huber and Bode [Bode, W., and Huber, R. (1976) FEBS Lett. 68, 231-236] demonstrated that peptides sequentially similar to the new amino terminus in combination with a catalytic site inhibitor could specifically induce a trypsin-like conformation in trypsinogen. We now demonstrate that an Ile-Ile or Ile-Val dipeptide can induce limited enzyme activity in the single-chain zymogen form of urokinase-type plasminogen activator (uPA) or its K158A variant, which cannot be activated proteolytically. Furthermore, the slow formation of a covalent serpin-protease complex between single-chain uPA and PAI-1 is significantly accelerated in the presence of specific dipeptide sequences. The technique of using a dipeptide mimic as a surrogate for the liberated amino terminus further provides a novel means by which to covalently label the immature active site of single-chain uPA with a fluorescent probe, permitting fluorescence approaches for direct observations of conformational changes within the protease domain during zymogen activation. These data demonstrate the structural plasticity of the protease domain, reinforce the notion of "molecular sexuality", and provide a novel way of studying conformational changes of zymogens during proteolytic activation.
Collapse
|
43
|
A cyclic peptidylic inhibitor of murine urokinase-type plasminogen activator: changing species specificity by substitution of a single residue. Biochem J 2008; 412:447-57. [PMID: 18318660 DOI: 10.1042/bj20071646] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
uPA (urokinase-type plasminogen activator) is a potential therapeutic target in a variety of pathological conditions, including cancer. In order to find new principles for inhibiting uPA in murine cancer models, we screened a phage-displayed peptide library with murine uPA as bait. We thereby isolated several murine uPA-binding peptide sequences, the predominant of which was the disulfide-bridged constrained sequence CPAYSRYLDC, which we will refer to as mupain-1. A chemically synthesized peptide corresponding to this sequence was found to be a competitive inhibitor of murine uPA, inhibiting its activity towards a low-molecular-mass chromogenic substrate as well as towards its natural substrate plasminogen. The K(i) value for inhibition as well as the K(D) value for binding were approx. 400 nM. Among a variety of other murine and human serine proteases, including trypsin, mupain-1 was found to be highly selective for murine uPA and did not even measurably inhibit human uPA. The cyclic structure of mupain-1 was indispensable for binding. Alanine scanning mutagenesis identified Arg(6) of mupain-1 as the P1 residue and indicated an extended binding interaction including the P5, P3, P2, P1 and P1' residues of mupain-1 and the specificity pocket, the catalytic triad and amino acids 41, 99 and 192 located in and around the active site of murine uPA. Exchanging His(99) of human uPA by a tyrosine residue, the corresponding residue in murine uPA, conferred mupain-1 susceptibility on to the latter. Peptide-derived inhibitors, such as mupain-1, may provide novel mechanistic information about enzyme-inhibitor interactions, provide alternative methodologies for designing effective protease inhibitors, and be used for target validation in murine model systems.
Collapse
|
44
|
Farady CJ, Egea PF, Schneider EL, Darragh MR, Craik CS. Structure of an Fab-protease complex reveals a highly specific non-canonical mechanism of inhibition. J Mol Biol 2008; 380:351-60. [PMID: 18514224 DOI: 10.1016/j.jmb.2008.05.009] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2008] [Revised: 05/01/2008] [Accepted: 05/02/2008] [Indexed: 11/17/2022]
Abstract
The vast majority of protein protease inhibitors bind their targets in a substrate-like manner. This is a robust and efficient mechanism of inhibition but, due to the highly conserved architecture of protease active sites, these inhibitors often exhibit promiscuity. Inhibitors that show strict specificity for one protease usually achieve this selectivity by combining substrate-like binding in the active site with exosite binding on the protease surface. The development of new, specific inhibitors can be aided greatly by binding to non-conserved regions of proteases if potency can be maintained. Due to their ability to bind specifically to nearly any antigen, antibodies provide an excellent scaffold for creating inhibitors targeted to a single member of a family of highly homologous enzymes. The 2.2 A resolution crystal structure of an Fab antibody inhibitor in complex with the serine protease membrane-type serine protease 1 (MT-SP1/matriptase) reveals the molecular basis of its picomolar potency and specificity. The inhibitor has a distinct mechanism of inhibition; it gains potency and specificity through interactions with the protease surface loops, and inhibits by binding in the active site in a catalytically non-competent manner. In contrast to most naturally occurring protease inhibitors, which have diverse structures but converge to a similar inhibitory archetype, antibody inhibitors provide an opportunity to develop divergent mechanisms of inhibition from a single scaffold.
Collapse
Affiliation(s)
- Christopher J Farady
- Graduate Group in Biophysics, University of California, San Francisco, 600 16th St. Genentech Hall, San Francisco, CA 94143-2240, USA
| | | | | | | | | |
Collapse
|