45 results on '"Cysteine Proteases chemistry"'
Search Results
2. Substitution-Induced Mechanistic Switching in S N Ar-Warheads for Cysteine Proteases.
- Author
-
Zimmer C, Brauer J, Ferenc D, Meyr J, Müller P, Räder HJ, Engels B, Opatz T, and Schirmeister T
- Subjects
- Structure-Activity Relationship, Cysteine Endopeptidases metabolism, Cysteine Endopeptidases chemistry, Molecular Structure, Cysteine Proteinase Inhibitors chemistry, Cysteine Proteinase Inhibitors pharmacology, Cysteine Proteinase Inhibitors metabolism, Molecular Docking Simulation, Cysteine Proteases metabolism, Cysteine Proteases chemistry
- Abstract
The aim of this study was to investigate the transition from non-covalent reversible over covalent reversible to covalent irreversible inhibition of cysteine proteases by making delicate structural changes to the warhead scaffold. To this end, dipeptidic rhodesain inhibitors with different N -terminal electrophilic arenes as warheads relying on the S
N Ar mechanism were synthesized and investigated. Strong structure-activity relationships of the inhibition potency, the degree of covalency, and the reversibility of binding on the arene substitution pattern were found. The studies were complemented and substantiated by molecular docking and quantum-mechanical calculations of model systems. Furthermore, the improvement in the membrane permeability of peptide esters in comparison to their corresponding carboxylic acids was exemplified.- Published
- 2024
- Full Text
- View/download PDF
3. The Power of Molecular Dynamics Simulations and Their Applications to Discover Cysteine Protease Inhibitors.
- Author
-
Dos Santos Nascimento IJ, Gomes JNS, de Oliveira Viana J, de Medeiros E Silva YMS, Barbosa EG, and de Moura RO
- Subjects
- Humans, Cysteine Proteases metabolism, Cysteine Proteases chemistry, Drug Discovery, SARS-CoV-2 enzymology, SARS-CoV-2 drug effects, Molecular Dynamics Simulation, Cysteine Proteinase Inhibitors chemistry, Cysteine Proteinase Inhibitors pharmacology, Cysteine Proteinase Inhibitors metabolism
- Abstract
A large family of enzymes with the function of hydrolyzing peptide bonds, called peptidases or cysteine proteases (CPs), are divided into three categories according to the peptide chain involved. CPs catalyze the hydrolysis of amide, ester, thiol ester, and thioester peptide bonds. They can be divided into several groups, such as papain-like (CA), viral chymotrypsin-like CPs (CB), papainlike endopeptidases of RNA viruses (CC), legumain-type caspases (CD), and showing active residues of His, Glu/Asp, Gln, Cys (CE). The catalytic mechanism of CPs is the essential cysteine residue present in the active site. These mechanisms are often studied through computational methods that provide new information about the catalytic mechanism and identify inhibitors. The role of computational methods during drug design and development stages is increasing. Methods in Computer-Aided Drug Design (CADD) accelerate the discovery process, increase the chances of selecting more promising molecules for experimental studies, and can identify critical mechanisms involved in the pathophysiology and molecular pathways of action. Molecular dynamics (MD) simulations are essential in any drug discovery program due to their high capacity for simulating a physiological environment capable of unveiling significant inhibition mechanisms of new compounds against target proteins, especially CPs. Here, a brief approach will be shown on MD simulations and how the studies were applied to identify inhibitors or critical information against cysteine protease from several microorganisms, such as Trypanosoma cruzi (cruzain), Trypanosoma brucei (rhodesain), Plasmodium spp . (falcipain), and SARS-CoV-2 (M
pro ). We hope the readers will gain new insights and use our study as a guide for potential compound identifications using MD simulations., (Copyright© Bentham Science Publishers; For any queries, please email at epub@benthamscience.net.)- Published
- 2024
- Full Text
- View/download PDF
4. Mechanisms Applied by Protein Inhibitors to Inhibit Cysteine Proteases.
- Author
-
Tušar L, Usenik A, Turk B, and Turk D
- Subjects
- Animals, Cystatins chemistry, Cystatins metabolism, Cystatins pharmacology, Cysteine Proteases metabolism, Cysteine Proteinase Inhibitors metabolism, Cysteine Proteinase Inhibitors pharmacology, Humans, Protein Binding, Securin chemistry, Securin metabolism, Securin pharmacology, X-Linked Inhibitor of Apoptosis Protein chemistry, X-Linked Inhibitor of Apoptosis Protein metabolism, X-Linked Inhibitor of Apoptosis Protein pharmacology, Cysteine Proteases chemistry, Cysteine Proteinase Inhibitors chemistry
- Abstract
Protein inhibitors of proteases are an important tool of nature to regulate and control proteolysis in living organisms under physiological and pathological conditions. In this review, we analyzed the mechanisms of inhibition of cysteine proteases on the basis of structural information and compiled kinetic data. The gathered structural data indicate that the protein fold is not a major obstacle for the evolution of a protease inhibitor. It appears that nature can convert almost any starting fold into an inhibitor of a protease. In addition, there appears to be no general rule governing the inhibitory mechanism. The structural data make it clear that the "lock and key" mechanism is a historical concept with limited validity. However, the analysis suggests that the shape of the active site cleft of proteases imposes some restraints. When the S1 binding site is shaped as a pocket buried in the structure of protease, inhibitors can apply substrate-like binding mechanisms. In contrast, when the S1 binding site is in part exposed to solvent, the substrate-like inhibition cannot be employed. It appears that all proteases, with the exception of papain-like proteases, belong to the first group of proteases. Finally, we show a number of examples and provide hints on how to engineer protein inhibitors., Competing Interests: The authors declare no conflict of interest.
- Published
- 2021
- Full Text
- View/download PDF
5. Mycotoxins from Fusarium proliferatum: new inhibitors of papain-like cysteine proteases.
- Author
-
Silva TL, Toffano L, Fernandes JB, das Graças Fernandes da Silva MF, de Sousa LRF, and Vieira PC
- Subjects
- Ananas chemistry, Ananas microbiology, Cysteine Proteinase Inhibitors metabolism, Fusarium metabolism, Kinetics, Mass Spectrometry, Mycotoxins metabolism, Secondary Metabolism, Ananas enzymology, Cysteine Proteases chemistry, Cysteine Proteinase Inhibitors chemistry, Fusarium chemistry, Mycotoxins chemistry, Plant Proteins chemistry
- Abstract
Papain-like cysteine proteases (PLCPs) in plants are essential to prevent phytopathogen invasion. In order to search for cysteine protease inhibitors and to investigate compounds that could be associated to pineapple Fusarium disease, a chemistry investigation was performed on Fusarium proliferatum isolated from Ananas comosus (pineapple) and cultivated in Czapek medium. From F. proliferatum extracts, nine secondary metabolites were isolated and characterized by nuclear magnetic resonance spectroscopy and mass spectrometry experiments: beauvericin (1), fusaric acid (2), N-ethyl-3-phenylacetamide (3), N-acetyltryptamine (4), cyclo(L-Val-L-Pro) cyclodipeptide (5), cyclo(L-Leu-L-Pro) cyclodipeptide (6), cyclo(L-Leu-L-Pro) diketopiperazine (7), 2,4-dihydroxypyrimidine (8), and 1H-indole-3-carbaldehyde (9). Compounds 1, 3, and 6 showed significant inhibition of papain, with IC
50 values of 25.3 ± 1.9, 39.4 ± 2.5, and 7.4 ± 0.5 μM, respectively. Compound 1 also showed significant inhibition against human cathepsins V and B with IC50 of 46.0 ± 3.0 and 6.8 ± 0.7 μM, respectively. The inhibition of papain by mycotoxins (fusaric acid and beauvericin) may indicate a mechanism of Fusarium in the roles of infection process.- Published
- 2020
- Full Text
- View/download PDF
6. Naphthoquinones as Covalent Reversible Inhibitors of Cysteine Proteases-Studies on Inhibition Mechanism and Kinetics.
- Author
-
Klein P, Barthels F, Johe P, Wagner A, Tenzer S, Distler U, Le TA, Schmid P, Engel V, Engels B, Hellmich UA, Opatz T, and Schirmeister T
- Subjects
- Cathepsin L chemistry, Cysteine Endopeptidases chemistry, Cysteine Proteinase Inhibitors chemistry, Dipeptides, Electrons, Esters, Hydrolysis, Inhibitory Concentration 50, Kinetics, Mass Spectrometry, Prodrugs chemistry, Quantum Theory, Structure-Activity Relationship, Trypanosoma brucei brucei drug effects, Cysteine Proteases chemistry, Cysteine Proteinase Inhibitors chemical synthesis, Naphthoquinones chemistry, Trypanocidal Agents pharmacology
- Abstract
The facile synthesis and detailed investigation of a class of highly potent protease inhibitors based on 1,4-naphthoquinones with a dipeptidic recognition motif (HN-l-Phe-l-Leu-OR) in the 2-position and an electron-withdrawing group (EWG) in the 3-position is presented. One of the compound representatives, namely the acid with EWG = CN and with R = H proved to be a highly potent rhodesain inhibitor with nanomolar affinity. The respective benzyl ester (R = Bn) was found to be hydrolyzed by the target enzyme itself yielding the free acid. Detailed kinetic and mass spectrometry studies revealed a reversible covalent binding mode. Theoretical calculations with different density functionals (DFT) as well as wavefunction-based approaches were performed to elucidate the mode of action., Competing Interests: The authors declare no conflict of interest.
- Published
- 2020
- Full Text
- View/download PDF
7. Evaluating QM/MM Free Energy Surfaces for Ranking Cysteine Protease Covalent Inhibitors.
- Author
-
da Costa CHS, Bonatto V, Dos Santos AM, Lameira J, Leitão A, and Montanari CA
- Subjects
- Cysteine Proteases chemistry, Ligands, Models, Molecular, Protein Conformation, Thermodynamics, Cysteine Proteases metabolism, Cysteine Proteinase Inhibitors metabolism, Quantum Theory
- Abstract
One tactic for cysteine protease inhibition is to form a covalent bond between an electrophilic atom of the inhibitor and the thiol of the catalytic cysteine. In this study, we evaluate the reaction free energy obtained from a hybrid quantum mechanical/molecular mechanical (QM/MM) free energy profile as a predictor of affinity for reversible, covalent inhibitors of rhodesain. We demonstrate that the reaction free energy calculated with the PM6/MM potential is in agreement with the experimental data and suggest that the free energy profile for covalent bond formation in a protein environment may be a useful tool for the inhibitor design.
- Published
- 2020
- Full Text
- View/download PDF
8. Fragment-Based Discovery of Irreversible Covalent Inhibitors of Cysteine Proteases Using Chlorofluoroacetamide Library.
- Author
-
Miura C, Shindo N, Okamoto K, Kuwata K, and Ojida A
- Subjects
- Acetamides metabolism, Chromatography, High Pressure Liquid, Cysteine Proteases metabolism, Cysteine Proteinase Inhibitors metabolism, Drug Discovery, Fluorescent Dyes chemistry, Kinetics, Ligands, Papain antagonists & inhibitors, Papain metabolism, Spectrometry, Mass, Electrospray Ionization, Acetamides chemistry, Cysteine Proteases chemistry, Cysteine Proteinase Inhibitors chemistry
- Abstract
Fragment-based approach combined with electrophilic reactive compounds is a powerful strategy to discover novel covalent ligands for protein target. However, the promiscuous reactivity often interferes with identification of the fragments possessing specific binding affinity to the targeted protein. In our study, we report the fragment-based covalent drug discovery using the chemically tuned weak reactivity of chlorofluoroacetamide (CFA). We constructed a small fragment library composed of 30 CFA-appended compounds and applied it to the covalent ligand screening for cysteine protease papain as a model protein target. Using the fluorescence enzymatic assay, we identified CFA-benzothiazole 30 as a papain inhibitor, which was found to irreversibly inactivate papain upon enzyme kinetic analysis. The formation of the covalent papain-30 adduct was confirmed using electrospray ionization mass spectrometry analysis. The activity-based protein profiling (ABPP) experiment using an alkynylated analog of 30 (i.e., 30-yne) revealed that 30-yne covalently labeled papain with high selectivity. These data demonstrate potential utility of the CFA-fragment library for de novo discovery of target selective covalent inhibitors.
- Published
- 2020
- Full Text
- View/download PDF
9. Putative structural rearrangements associated with the interaction of macrocyclic inhibitors with norovirus 3CL protease.
- Author
-
Galasiti Kankanamalage AC, Weerawarna PM, Rathnayake AD, Kim Y, Mehzabeen N, Battaile KP, Lovell S, Chang KO, and Groutas WC
- Subjects
- Animals, Caliciviridae Infections drug therapy, Caliciviridae Infections virology, Catalytic Domain drug effects, Cell Line, Crystallography, X-Ray, Cysteine Proteases chemistry, Cysteine Proteinase Inhibitors chemistry, Gastroenteritis drug therapy, Gastroenteritis virology, Humans, Macrocyclic Compounds chemistry, Mice, Models, Molecular, Norovirus chemistry, Norovirus drug effects, Protein Conformation drug effects, RAW 264.7 Cells, Cysteine Proteases metabolism, Cysteine Proteinase Inhibitors pharmacology, Macrocyclic Compounds pharmacology, Norovirus enzymology
- Abstract
Human noroviruses are the primary cause of outbreaks of acute gastroenteritis worldwide. The problem is further compounded by the current lack of norovirus-specific antivirals or vaccines. Noroviruses have a single-stranded, positive sense 7 to 8 kb RNA genome which encodes a polyprotein precursor that is processed by a virus-encoded 3C-like cysteine protease (NV 3CLpro) to generate at least six mature nonstructural proteins. Processing of the polyprotein is essential for virus replication, consequently, NV 3CLpro has emerged as an attractive target for the discovery of norovirus therapeutics and prophylactics. We have recently described the structure-based design of macrocyclic transition state inhibitors of NV 3CLpro. In order to gain insight and understanding into the interaction of macrocyclic inhibitors with the enzyme, as well as probe the effect of ring size on pharmacological activity and cellular permeability, additional macrocyclic inhibitors were synthesized and high resolution cocrystal structures determined. The results of our studies tentatively suggest that the macrocyclic scaffold may hamper optimal binding to the active site by impeding concerted cross-talk between the S
2 and S4 subsites., (© 2019 Wiley Periodicals, Inc.)- Published
- 2019
- Full Text
- View/download PDF
10. X-ray Structures of Two Bacteroides thetaiotaomicron C11 Proteases in Complex with Peptide-Based Inhibitors.
- Author
-
Roncase EJ, González-Páez GE, and Wolan DW
- Subjects
- Bacteroides Infections microbiology, Bacteroides thetaiotaomicron chemistry, Bacteroides thetaiotaomicron drug effects, Bacteroides thetaiotaomicron metabolism, Catalytic Domain drug effects, Crystallography, X-Ray, Cysteine Proteases metabolism, Humans, Molecular Docking Simulation, Protein Conformation drug effects, Bacteroides thetaiotaomicron enzymology, Cysteine Proteases chemistry, Cysteine Proteinase Inhibitors pharmacology, Peptides pharmacology
- Abstract
Commensal bacteria secrete proteins and metabolites to influence host intestinal homeostasis, and proteases represent a significant constituent of the components at the host:microbiome interface. Here, we determined the structures of the two secreted C11 cysteine proteases encoded by the established gut commensal Bacteroides thetaiotaomicron. We employed mutational analysis to demonstrate the two proteases, termed "thetapain" and "iotapain", undergo in trans autoactivation after lysine and/or arginine residues, as observed for other C11 proteases. We determined the structures of the active forms of thetapain and iotapain in complex with irreversible peptide inhibitors, Ac-VLTK-AOMK and biotin-VLTK-AOMK, respectively. Structural comparisons revealed key active-site interactions important for peptide recognition are more extensive for thetapain; however, both proteases employ a glutamate residue to preferentially bind small polar residues at the P2 position. Our results will aid in the design of protease-specific probes to ultimately understand the biological role of C11 proteases in bacterial fitness, elucidate their host and/or microbial substrates, and interrogate their involvement in microbiome-related diseases.
- Published
- 2019
- Full Text
- View/download PDF
11. Experimental study and computational modelling of cruzain cysteine protease inhibition by dipeptidyl nitriles.
- Author
-
Dos Santos AM, Cianni L, De Vita D, Rosini F, Leitão A, Laughton CA, Lameira J, and Montanari CA
- Subjects
- Drug Design, Molecular Dynamics Simulation, Protein Binding, Quantum Theory, Thermodynamics, Cysteine Endopeptidases chemistry, Cysteine Proteases chemistry, Cysteine Proteinase Inhibitors chemistry, Nitriles chemical synthesis, Protozoan Proteins chemistry, Trypanocidal Agents chemistry, Trypanosoma cruzi enzymology
- Abstract
Chagas disease affects millions of people in Latin America. This disease is caused by the protozoan parasite Trypanossoma cruzi. The cysteine protease cruzain is a key enzyme for the survival and propagation of this parasite lifecycle. Nitrile-based inhibitors are efficient inhibitors of cruzain that bind by forming a covalent bond with this enzyme. Here, three nitrile-based inhibitors dubbed Neq0409, Neq0410 and Neq0570 were synthesized, and the thermodynamic profile of the bimolecular interaction with cruzain was determined using isothermal titration calorimetry (ITC). The result suggests the inhibition process is enthalpy driven, with a detrimental contribution of entropy. In addition, we have used hybrid Quantum Mechanical/Molecular Mechanical (QM/MM) and Molecular Dynamics (MD) simulations to investigate the reaction mechanism of reversible covalent modification of cruzain by Neq0409, Neq0410 and Neq0570. The computed free energy profile shows that the nucleophilic attack of Cys25 on the carbon C1 of inhibitiors and the proton transfer from His162 to N1 of the dipeptidyl nitrile inhibitor take place in a single step. The calculated free energy of the inhibiton reaction is in agreement with covalent experimental binding. Altogether, the results reported here suggests that nitrile-based inhibitors are good candidates for the development of reversible covalent inhibitors of cruzain and other cysteine proteases.
- Published
- 2018
- Full Text
- View/download PDF
12. Discovery of benzimidazole-based Leishmania mexicana cysteine protease CPB2.8ΔCTE inhibitors as potential therapeutics for leishmaniasis.
- Author
-
De Luca L, Ferro S, Buemi MR, Monforte AM, Gitto R, Schirmeister T, Maes L, Rescifina A, and Micale N
- Subjects
- Antiprotozoal Agents chemical synthesis, Antiprotozoal Agents metabolism, Antiprotozoal Agents therapeutic use, Antiprotozoal Agents toxicity, Benzimidazoles metabolism, Benzimidazoles therapeutic use, Benzimidazoles toxicity, Binding Sites, Cell Line, Cell Survival drug effects, Cysteine Proteases chemistry, Cysteine Proteinase Inhibitors metabolism, Cysteine Proteinase Inhibitors therapeutic use, Cysteine Proteinase Inhibitors toxicity, Drug Evaluation, Preclinical, Enzyme Assays, Humans, Hydrogen Bonding, Inhibitory Concentration 50, Leishmaniasis drug therapy, Molecular Docking Simulation, Protein Structure, Tertiary, Protozoan Proteins metabolism, Benzimidazoles chemistry, Cysteine Proteases metabolism, Cysteine Proteinase Inhibitors chemistry, Leishmania mexicana enzymology, Protozoan Proteins antagonists & inhibitors
- Abstract
Chemotherapy is currently the only effective approach to treat all forms of leishmaniasis. However, its effectiveness is severely limited due to high toxicity, long treatment length, drug resistance, or inadequate mode of administration. As a consequence, there is a need to identify new molecular scaffolds and targets as potential therapeutics for the treatment of this disease. We report a small series of 1,2-substituted-1H-benzo[d]imidazole derivatives (9a-d) showing affinity in the submicromolar range (K
i = 0.15-0.69 μM) toward Leishmania mexicanaCPB2.8ΔCTE, one of the more promising targets for antileishmanial drug design. The compounds confirmed activity in vitro against intracellular amastigotes of Leishmania infantum with the best result being obtained with derivative 9d (IC50 = 6.8 μM), although with some degree of cytotoxicity (CC50 = 8.0 μM on PMM and CC50 = 32.0 μM on MCR-5). In silico molecular docking studies and ADME-Tox properties prediction were performed to validate the hypothesis of the interaction with the intended target and to assess the drug-likeness of these derivatives., (© 2018 John Wiley & Sons A/S.)- Published
- 2018
- Full Text
- View/download PDF
13. Ensemble-based ADME-Tox profiling and virtual screening for the discovery of new inhibitors of the Leishmania mexicana cysteine protease CPB2.8ΔCTE.
- Author
-
Scala A, Rescifina A, Micale N, Piperno A, Schirmeister T, Maes L, and Grassi G
- Subjects
- Binding Sites, Catalytic Domain, Cell Line, Cell Survival drug effects, Cysteine Proteases chemistry, Cysteine Proteinase Inhibitors metabolism, Cysteine Proteinase Inhibitors pharmacology, Humans, Inhibitory Concentration 50, Leishmania mexicana drug effects, Molecular Docking Simulation, Protozoan Proteins antagonists & inhibitors, Structure-Activity Relationship, Thiophenes metabolism, Thiophenes pharmacology, Cysteine Proteases metabolism, Cysteine Proteinase Inhibitors chemistry, Leishmania mexicana enzymology, Protozoan Proteins metabolism, Thiophenes chemistry
- Abstract
In an effort to identify novel molecular warheads able to inhibit Leishmania mexicana cysteine protease CPB2.8ΔCTE, fused benzo[b]thiophenes and β,β'-triketones emerged as covalent inhibitors binding the active site cysteine residue. Enzymatic screening showed a moderate-to-excellent activity (12%-90% inhibition of the target enzyme at 20 μm). The most promising compounds were selected for further profiling including in vitro cell-based assays and docking studies. Computational data suggest that benzo[b]thiophenes act immediately as non-covalent inhibitors and then as irreversible covalent inhibitors, whereas a reversible covalent mechanism emerged for the 1,3,3'-triketones with a Y-topology. Based on the predicted physicochemical and ADME-Tox properties, compound 2b has been identified as a new drug-like, non-mutagen, non-carcinogen, and non-neurotoxic lead candidate., (© 2017 John Wiley & Sons A/S.)
- Published
- 2018
- Full Text
- View/download PDF
14. Quantum mechanics/molecular mechanics studies of the mechanism of cysteine protease inhibition by peptidyl-2,3-epoxyketones.
- Author
-
Arafet K, Ferrer S, González FV, and Moliner V
- Subjects
- Cysteine Endopeptidases chemistry, Cysteine Endopeptidases metabolism, Cysteine Proteases chemistry, Cysteine Proteinase Inhibitors chemistry, Epoxy Compounds chemistry, Ketones chemistry, Protozoan Proteins chemistry, Protozoan Proteins metabolism, Thermodynamics, Trypanosoma cruzi metabolism, Cysteine Proteases metabolism, Cysteine Proteinase Inhibitors metabolism, Ketones metabolism, Molecular Dynamics Simulation, Quantum Theory
- Abstract
Cysteine proteases are the most abundant proteases in parasitic protozoa and they are essential enzymes to the life cycle of several of them, thus becoming attractive therapeutic targets for the development of new inhibitors. In this paper, a computational study of the inhibition mechanism of cysteine protease by dipeptidyl-2,3-epoxyketone Cbz-Phe-Hph-(S), a recently proposed inhibitor, has been carried out by means of molecular dynamics (MD) simulations with hybrid QM/MM potentials. The computed free energy surfaces of the inhibition mechanism of cysteine proteases by peptidyl epoxyketones showing how the activation of the epoxide ring and the attack of Cys25 on either C2 or C3 atoms take place in a concerted manner. According to our results, the acid species responsible for the protonation of the oxygen atom of the ring would be able to conserve His159, in contrast to previous studies that proposed a water molecule as the activating species. The low activation free energies for the reaction where Cys25 attacks the C2 atom of the epoxide ring (12.1 kcal mol
-1 ) or to the C3 atom (15.4 kcal mol-1 ), together with the high negative reaction energies suggest that the derivatives of peptidyl-2,3-epoxyketones can be used to develop new potent inhibitors for the treatment of Chagas disease.- Published
- 2017
- Full Text
- View/download PDF
15. Cysteine protease inhibitor of Schistosoma japonicum - A parasite-derived negative immunoregulatory factor.
- Author
-
Chen L, He B, Hou W, and He L
- Subjects
- Animals, Cysteine Proteases chemistry, Cysteine Proteases immunology, Cysteine Proteinase Inhibitors immunology, Cysteine Proteinase Inhibitors metabolism, Dendritic Cells enzymology, Dendritic Cells immunology, Female, Flow Cytometry, Humans, Immune Tolerance, Immunosuppressive Agents immunology, Interleukin-4 genetics, Interleukin-4 immunology, Lysosomes chemistry, Lysosomes enzymology, Mice, Mice, Inbred BALB C, Schistosoma japonicum chemistry, Schistosoma japonicum genetics, Schistosomiasis japonica enzymology, Schistosomiasis japonica genetics, Schistosomiasis japonica immunology, T-Lymphocytes, Regulatory immunology, Transforming Growth Factor beta genetics, Transforming Growth Factor beta immunology, Cysteine Proteinase Inhibitors chemistry, Immunosuppressive Agents chemistry, Schistosoma japonicum immunology, Schistosomiasis japonica parasitology
- Abstract
Studies have shown that cysteine protease inhibitors from some parasites have immunosuppressive effects on the host. We previously have cloned a novel cysteine protease inhibitor from Schistosoma japonicum and purified its recombinant version (protein named rSj-C). Its possible inhibitory effect on the host immune response has not been described.This study shows that rSj-C inhibits lysosomal cysteine protease of murine dendritic cells (DCs). After DCs were incubated with rSj-C and then with soluble adult worm antigen (AWA) of S. japonicum, the mean fluorescence intensity of MHC class II antigens on the surface of DCs decreased significantly by flow cytometry. These results indirectly proved that rSj-C can suppress exogenous-antigen presentation by DCs. The flow cytometric assay revealed that in comparison with control groups, the proportion of CD4
+ CD25+ Foxp3+ T cells among CD4+ CD25+ T cells of Schistosom-infected mice increased significantly 8 weeks after the infected mice were injected with rSj-C (p ˂ 0.05). Additionally, the expression levels of cytokines IL-4 and TGF-β produced by T cells increased significantly as compared with these levels in the normal group (p ˂ 0.05). These results clearly show that the cysteine protease inhibitor from S. japonicum is a new parasite-derived immunosuppressive factor.- Published
- 2017
- Full Text
- View/download PDF
16. Analysis of non-peptidic compounds as potential malarial inhibitors against Plasmodial cysteine proteases via integrated virtual screening workflow.
- Author
-
Musyoka TM, Kanzi AM, Lobb KA, and Tastan Bishop Ö
- Subjects
- Amino Acid Sequence, Antimalarials pharmacology, Binding Sites, Catalytic Domain, Conserved Sequence, Cysteine Proteases metabolism, Cysteine Proteinase Inhibitors pharmacology, Drug Discovery, Hydrophobic and Hydrophilic Interactions, Ligands, Molecular Conformation, Molecular Docking Simulation, Molecular Dynamics Simulation, Protein Binding, Workflow, Antimalarials chemistry, Cysteine Proteases chemistry, Cysteine Proteinase Inhibitors chemistry, Models, Molecular, Plasmodium enzymology
- Abstract
Falcipain-2 (FP-2) and falcipain-3 (FP-3), haemoglobin-degrading enzymes in Plasmodium falciparum, are validated drug targets for the development of effective inhibitors against malaria. However, no commercial drug-targeting falcipains has been developed despite their central role in the life cycle of the parasites. In this work, in silico approaches are used to identify key structural elements that control the binding and selectivity of a diverse set of non-peptidic compounds onto FP-2, FP-3 and homologues from other Plasmodium species as well as human cathepsins. Hotspot residues and the underlying non-covalent interactions, important for the binding of ligands, are identified by interaction fingerprint analysis between the proteases and 2-cyanopyridine derivatives (best hits). It is observed that the size and chemical type of substituent groups within 2-cyanopyridine derivatives determine the strength of protein-ligand interactions. This research presents novel results that can further be exploited in the structure-based molecular-guided design of more potent antimalarial drugs.
- Published
- 2016
- Full Text
- View/download PDF
17. Differential expression of a cysteine proteinase and cystatin pair as sidebyside fusion forms in Escherichia coli.
- Author
-
Gholizadeh A
- Subjects
- Arabidopsis enzymology, Arabidopsis Proteins genetics, Arabidopsis Proteins metabolism, Binding Sites, Cloning, Molecular, Cysteine Proteases genetics, Cysteine Proteases metabolism, Cysteine Proteinase Inhibitors metabolism, Escherichia coli genetics, Escherichia coli metabolism, Gene Expression, Genetic Vectors chemistry, Genetic Vectors metabolism, Kinetics, Maltose-Binding Proteins chemistry, Maltose-Binding Proteins genetics, Maltose-Binding Proteins metabolism, Models, Molecular, Protein Binding, Protein Conformation, alpha-Helical, Protein Conformation, beta-Strand, Protein Interaction Domains and Motifs, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Substrate Specificity, Thermodynamics, Arabidopsis chemistry, Arabidopsis Proteins chemistry, Cysteine Proteases chemistry, Cysteine Proteinase Inhibitors chemistry, Recombinant Fusion Proteins chemistry
- Abstract
As a basic study, the fusion expressions of two functionally related proteins were described. The side by side fusion construction, expression, purification and functional characterization of Arabidopsis papain-like cysteine proteinase (CP) and cysteine proteinase inhibitor (CPI) were successfully carried out by using an Escherichia coli expression system without affecting the recombinant bacterial growth. The purification products of two different fused constructs designated as «R1: H2N-maltose binding protein-CPI-CP-COOH and R2: H2N-maltose binding protein-CP-CPI-COOH» showed inverse enzymatic/inhibitory activities, in vitro. Analysis of the constructs by using computational tools revealed that the arrangement of CP/CPI pair in fusion forms might be the important criteria for proper tertiary organization, structural folding and functional property. The overall results showed that the C-terminally located molecule could be the active folded structure in each fusion construct. The achievements of the present work may be utilized in a specific protein engineering application such as manufacturing the novel switchable expression systems in the future.
- Published
- 2016
18. Modeling of babesipain-1 and identification of natural and synthetic leads for bovine babesiosis drug development.
- Author
-
Meetei PA, Rathore RS, Prabhu NP, and Vindal V
- Subjects
- Amino Acid Sequence, Babesia drug effects, Catalytic Domain, Conserved Sequence, Cysteine Endopeptidases chemistry, Drug Discovery, High-Throughput Screening Assays, Molecular Dynamics Simulation, Protein Structure, Quaternary, Protein Structure, Secondary, Protozoan Proteins chemistry, Sequence Alignment, Structural Homology, Protein, User-Computer Interface, Antiprotozoal Agents chemistry, Cysteine Proteases chemistry, Cysteine Proteinase Inhibitors chemistry, Molecular Docking Simulation, Protozoan Proteins antagonists & inhibitors, Small Molecule Libraries chemistry
- Abstract
Babesiosis is a tick-borne, zoonotic disease caused by species of the intraerythrocytic protozoan Babesia. It is distributed all around the world and affects various domestic and wild animals, mainly cattle. Recently, the cysteine protease enzyme, babesipain-1 from Babesia bigemina has been identified as a potential target for designing new anti-babesiosis drugs. In the present study, a three-dimensional structural model of babesipain-1 was developed. An active site with three pockets (S1, S2, and S3), which is congruent with its homolog, falcipain-3, was also identified. Moreover, the conservation of active site residues was consistent with the cysteine protease family. In order to identify potential inhibitors, a virtual screening workflow was employed with a chemical library containing natural and synthetic compounds. Potential inhibitors interacting with all the three subsites were identified. Further, molecular dynamic simulations were carried out to assess the interactions and stability of the inhibitors. The informatics approach, and the findings presented in this study will assist researchers in further development of potential anti-babesiosis molecules.
- Published
- 2016
- Full Text
- View/download PDF
19. Synthetic Studies on the Preparation of Alanyl Epoxysulfones as Cathepsin Cysteine Protease Electrophilic Traps.
- Author
-
Latorre A, Rodríguez S, González FV, Florea BI, and Overkleeft HS
- Subjects
- Alanine chemistry, Cysteine Proteases chemistry, Cysteine Proteinase Inhibitors pharmacology, Epoxy Compounds chemistry, Molecular Structure, Oxidation-Reduction, Alanine analogs & derivatives, Chlorohydrins chemistry, Cysteine Proteases chemical synthesis, Cysteine Proteinase Inhibitors chemistry, Epoxy Compounds chemical synthesis
- Abstract
A Darzens reaction between tert-butoxycarbonyl alaninal and chloromethyl phenyl sulfone afforded chlorohydrins, which were converted into epoxysulfones by reaction with sodium tert-butoxide. Epoxysulfone 10 and chloroketone 14 derived from chlorohydrins by oxidation proved to be inhibitors of cathepsins H, S, and C as determined by competitive activity-based protein profiling.
- Published
- 2015
- Full Text
- View/download PDF
20. Thiopurine analogs and mycophenolic acid synergistically inhibit the papain-like protease of Middle East respiratory syndrome coronavirus.
- Author
-
Cheng KW, Cheng SC, Chen WY, Lin MH, Chuang SJ, Cheng IH, Sun CY, and Chou CY
- Subjects
- Coronavirus 3C Proteases, Cysteine Endopeptidases, Cysteine Proteases chemistry, Drug Synergism, Middle East Respiratory Syndrome Coronavirus enzymology, Models, Molecular, Severe acute respiratory syndrome-related coronavirus drug effects, Severe acute respiratory syndrome-related coronavirus enzymology, Viral Proteins antagonists & inhibitors, Antiviral Agents pharmacology, Cysteine Proteases metabolism, Cysteine Proteinase Inhibitors pharmacology, Mercaptopurine pharmacology, Middle East Respiratory Syndrome Coronavirus drug effects, Mycophenolic Acid pharmacology, Thioguanine pharmacology
- Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV) is a new highly pathogenic human coronaviruses that emerged in Jeddah and Saudi Arabia and has quickly spread to other countries in Middle East, Europe and North Africa since 2012. Up to 17 December 2014, it has infected at least 938 people with a fatality rate of about 36% globally. This has resulted in an urgent need to identify antiviral drugs that are active against MERS-CoV. The papain-like protease (PL(pro)) of MERS-CoV represents an important antiviral target as it is not only essential for viral maturation, but also antagonizes interferon stimulation of the host via its deubiquitination activity. Here, we report the discovery that two SARS-CoV PL(pro) inhibitors, 6-mercaptopurine (6MP) and 6-thioguanine (6TG), as well as the immunosuppressive drug mycophenolic acid, are able to inhibit MERS-CoV PL(pro). Their inhibition mechanisms and mutually binding synergistic effect were also investigated. Our results identify for the first time three inhibitors targeting MERS-CoV PL(pro) and these can now be used as lead compounds for further antiviral drug development., (Copyright © 2014 Elsevier B.V. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
21. A novel sulfate-reducing bacteria detection method based on inhibition of cysteine protease activity.
- Author
-
Qi P, Zhang D, and Wan Y
- Subjects
- Buffers, Catalysis, Catalytic Domain, Culture Media chemistry, Desulfovibrio isolation & purification, Desulfovibrio metabolism, Escherichia coli isolation & purification, Escherichia coli metabolism, Glutathione chemistry, Hydrogen-Ion Concentration, Hydrolysis, Oxygen chemistry, Papain chemistry, Staphylococcus aureus isolation & purification, Staphylococcus aureus metabolism, Sulfides chemistry, Sulfur-Reducing Bacteria metabolism, Temperature, Vibrio alginolyticus isolation & purification, Vibrio alginolyticus metabolism, Cysteine Proteases chemistry, Cysteine Proteinase Inhibitors chemistry, Sulfates chemistry, Sulfur-Reducing Bacteria isolation & purification
- Abstract
Sulfate-reducing bacteria (SRB) have been extensively studied in corrosion and environmental science. However, fast enumeration of SRB population is still a difficult task. This work presents a novel specific SRB detection method based on inhibition of cysteine protease activity. The hydrolytic activity of cysteine protease was inhibited by taking advantage of sulfide, the characteristic metabolic product of SRB, to attack active cysteine thiol group in cysteine protease catalytic sites. The active thiol S-sulfhydration process could be used for SRB detection, since the amount of sulfide accumulated in culture medium was highly related with initial bacterial concentration. The working conditions of cysteine protease have been optimized to obtain better detection capability, and the SRB detection performances have been evaluated in this work. The proposed SRB detection method based on inhibition of cysteine protease activity avoided the use of biological recognition elements. In addition, compared with the widely used most probable number (MPN) method which would take up to at least 15days to accomplish whole detection process, the method based on inhibition of papain activity could detect SRB in 2 days, with a detection limit of 5.21×10(2) cfu mL(-1). The detection time for SRB population quantitative analysis was greatly shortened., (Copyright © 2014 Elsevier B.V. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
22. A knowledge-based approach for identification of drugs against vivapain-2 protein of Plasmodium vivax through pharmacophore-based virtual screening with comparative modelling.
- Author
-
Yadav MK, Singh A, and Swati D
- Subjects
- Amino Acid Sequence, Antimalarials metabolism, Binding Sites, Catalytic Domain, Cysteine Proteases genetics, Cysteine Proteases metabolism, Databases, Factual, Drug Evaluation, Preclinical instrumentation, Humans, Malaria, Vivax parasitology, Models, Molecular, Molecular Sequence Data, Plasmodium vivax chemistry, Plasmodium vivax drug effects, Plasmodium vivax genetics, Protozoan Proteins antagonists & inhibitors, Protozoan Proteins genetics, Protozoan Proteins metabolism, Sequence Alignment, Antimalarials chemistry, Antimalarials pharmacology, Cysteine Proteases chemistry, Cysteine Proteinase Inhibitors chemistry, Cysteine Proteinase Inhibitors pharmacology, Drug Evaluation, Preclinical methods, Plasmodium vivax enzymology, Protozoan Proteins chemistry
- Abstract
Malaria is one of the most infectious diseases in the world. Plasmodium vivax, the pathogen causing endemic malaria in humans worldwide, is responsible for extensive disease morbidity. Due to the emergence of resistance to common anti-malarial drugs, there is a continuous need to develop a new class of drugs for this pathogen. P. vivax cysteine protease, also known as vivapain-2, plays an important role in haemoglobin hydrolysis and is considered essential for the survival of the parasite. The three-dimensional (3D) structure of vivapain-2 is not predicted experimentally, so its structure is modelled by using comparative modelling approach and further validated by Qualitative Model Energy Analysis (QMEAN) and RAMPAGE tools. The potential binding site of selected vivapain-2 structure has been detected by grid-based function prediction method. Drug targets and their respective drugs similar to vivapain-2 have been identified using three publicly available databases: STITCH 3.1, DrugBank and Therapeutic Target Database (TTD). The second approach of this work focuses on docking study of selected drug E-64 against vivapain-2 protein. Docking reveals crucial information about key residues (Asn281, Cys283, Val396 and Asp398) that are responsible for holding the ligand in the active site. The similarity-search criterion is used for the preparation of our in-house database of drugs, obtained from filtering the drugs from the DrugBank database. A five-point 3D pharmacophore model is generated for the docked complex of vivapain-2 with E-64. This study of 3D pharmacophore-based virtual screening results in identifying three new drugs, amongst which one is approved and the other two are experimentally proved. The ADMET properties of these drugs are found to be in the desired range. These drugs with novel scaffolds may act as potent drugs for treating malaria caused by P. vivax.
- Published
- 2014
- Full Text
- View/download PDF
23. Cross-talk between malarial cysteine proteases and falstatin: the BC loop as a hot-spot target.
- Author
-
Sundararaj S, Saxena AK, Sharma R, Vashisht K, Sharma S, Anvikar A, Dixit R, Rosenthal PJ, and Pandey KC
- Subjects
- Amino Acid Sequence, Cross Reactions, Cysteine Proteases chemistry, Erythrocytes pathology, Hemoglobins metabolism, Humans, Hydrogen Bonding, Hydrolysis, Malaria, Falciparum pathology, Models, Molecular, Molecular Sequence Data, Mutagenesis, Site-Directed, Mutation genetics, Peptide Fragments chemistry, Peptide Fragments metabolism, Plasmodium falciparum genetics, Protein Conformation, Protein Multimerization, Protozoan Proteins chemistry, Protozoan Proteins genetics, Sequence Homology, Amino Acid, Cysteine Proteases metabolism, Cysteine Proteinase Inhibitors metabolism, Erythrocytes parasitology, Malaria, Falciparum parasitology, Plasmodium falciparum enzymology, Plasmodium falciparum pathogenicity, Protozoan Proteins metabolism
- Abstract
Cysteine proteases play a crucial role in the development of the human malaria parasites Plasmodium falciparum and Plasmodium vivax. Our earlier studies demonstrated that these enzymes are equipped with specific domains for defined functions and further suggested the mechanism of activation of cysteine proteases. The activities of these proteases are regulated by a new class of endogenous inhibitors of cysteine proteases (ICPs). Structural studies of the ICPs of Trypanosoma cruzi (chagasin) and Plasmodium berghei (PbICP) indicated that three loops (termed BC, DE, and FG) are crucial for binding to target proteases. Falstatin, an ICP of P. falciparum, appears to play a crucial role in invasion of erythrocytes and hepatocytes. However, the mechanism of inhibition of cysteine proteases by falstatin has not been established. Our study suggests that falstatin is the first known ICP to function as a multimeric protein. Using site-directed mutagenesis, hemoglobin hydrolysis assays and peptide inhibition studies, we demonstrate that the BC loop, but not the DE or FG loops, inhibits cysteine proteases of P. falciparum and P. vivax via hydrogen bonds. These results suggest that the BC loop of falstatin acts as a hot-spot target for inhibiting malarial cysteine proteases. This finding suggests new strategies for the development of anti-malarial agents based on protease-inhibitor interactions.
- Published
- 2014
- Full Text
- View/download PDF
24. Quantum mechanics-based scoring rationalizes the irreversible inactivation of parasitic Schistosoma mansoni cysteine peptidase by vinyl sulfone inhibitors.
- Author
-
Fanfrlík J, Brahmkshatriya PS, Řezáč J, Jílková A, Horn M, Mareš M, Hobza P, and Lepšík M
- Subjects
- Animals, Crystallography, X-Ray, Cysteine Proteases chemistry, Cysteine Proteinase Inhibitors chemistry, Dose-Response Relationship, Drug, Models, Molecular, Molecular Structure, Structure-Activity Relationship, Sulfones chemistry, Vinyl Compounds chemistry, Cysteine Proteases metabolism, Cysteine Proteinase Inhibitors pharmacology, Quantum Theory, Schistosoma mansoni enzymology, Sulfones pharmacology, Vinyl Compounds pharmacology
- Abstract
The quantum mechanics (QM)-based scoring function that we previously developed for the description of noncovalent binding in protein-ligand complexes has been modified and extended to treat covalent binding of inhibitory ligands. The enhancements are (i) the description of the covalent bond breakage and formation using hybrid QM/semiempirical QM (QM/SQM) restrained optimizations and (ii) the addition of the new ΔG(cov)' term to the noncovalent score, describing the "free" energy difference between the covalent and noncovalent complexes. This enhanced QM-based scoring function is applied to a series of 20 vinyl sulfone-based inhibitory compounds inactivating the cysteine peptidase cathepsin B1 of the Schistosoma mansoni parasite (SmCB1). The available X-ray structure of the SmCB1 in complex with a potent vinyl sulfone inhibitor K11017 is used as a template to build the other covalently bound complexes and to model the derived noncovalent complexes. We present the correlation of the covalent score and its constituents with the experimental binding data. Four outliers are identified. They contain bulky R1' substituents structurally divergent from the template, which might induce larger protein rearrangements than could be accurately modeled. In summary, we propose a new computational approach and an optimal protocol for the rapid evaluation and prospective design of covalent inhibitors with a conserved binding mode.
- Published
- 2013
- Full Text
- View/download PDF
25. A cysteine protease inhibitor rescues mice from a lethal Cryptosporidium parvum infection.
- Author
-
Ndao M, Nath-Chowdhury M, Sajid M, Marcus V, Mashiyama ST, Sakanari J, Chow E, Mackey Z, Land KM, Jacobson MP, Kalyanaraman C, McKerrow JH, Arrowood MJ, and Caffrey CR
- Subjects
- Administration, Oral, Animals, Antiprotozoal Agents chemistry, Cryptosporidiosis mortality, Cryptosporidiosis parasitology, Cryptosporidium parvum drug effects, Cryptosporidium parvum enzymology, Cryptosporidium parvum growth & development, Cysteine Proteases chemistry, Cysteine Proteinase Inhibitors chemistry, Dipeptides chemistry, Drug Administration Schedule, Female, Injections, Intraperitoneal, Isoenzymes antagonists & inhibitors, Isoenzymes chemistry, Isoenzymes metabolism, Male, Mice, Mice, Knockout, Molecular Docking Simulation, Phenylalanine analogs & derivatives, Piperazines, Protozoan Proteins chemistry, Protozoan Proteins metabolism, Receptors, Interferon deficiency, Receptors, Interferon genetics, Survival Analysis, Tosyl Compounds, Vinyl Compounds chemistry, Interferon gamma Receptor, Antiprotozoal Agents pharmacology, Cryptosporidiosis drug therapy, Cysteine Proteases metabolism, Cysteine Proteinase Inhibitors pharmacology, Dipeptides pharmacology, Protozoan Proteins antagonists & inhibitors, Vinyl Compounds pharmacology
- Abstract
Cryptosporidiosis, caused by the protozoan parasite Cryptosporidium parvum, can stunt infant growth and can be lethal in immunocompromised individuals. The most widely used drugs for treating cryptosporidiosis are nitazoxanide and paromomycin, although both exhibit limited efficacy. To investigate an alternative approach to therapy, we demonstrate that the clan CA cysteine protease inhibitor N-methyl piperazine-Phe-homoPhe-vinylsulfone phenyl (K11777) inhibits C. parvum growth in mammalian cell lines in a concentration-dependent manner. Further, using the C57BL/6 gamma interferon receptor knockout (IFN-γR-KO) mouse model, which is highly susceptible to C. parvum, oral or intraperitoneal treatment with K11777 for 10 days rescued mice from otherwise lethal infections. Histologic examination of untreated mice showed intestinal inflammation, villous blunting, and abundant intracellular parasite stages. In contrast, K11777-treated mice (210 mg/kg of body weight/day) showed only minimal inflammation and no epithelial changes. Three putative protease targets (termed cryptopains 1 to 3, or CpaCATL-1, -2, and -3) were identified in the C. parvum genome, but only two are transcribed in infected mammals. A homology model predicted that K11777 would bind to cryptopain 1. Recombinant enzymatically active cryptopain 1 was successfully targeted by K11777 in a competition assay with a labeled active-site-directed probe. K11777 exhibited no toxicity in vitro and in vivo, and surviving animals remained free of parasites 3 weeks after treatment. The discovery that a cysteine protease inhibitor provides potent anticryptosporidial activity in an animal model of infection encourages the investigation and development of this biocide class as a new, and urgently needed, chemotherapy for cryptosporidiosis.
- Published
- 2013
- Full Text
- View/download PDF
26. Proteolysis of noncollagenous proteins in sea cucumber, Stichopus japonicus, body wall: characterisation and the effects of cysteine protease inhibitors.
- Author
-
Wu HT, Li DM, Zhu BW, Sun JJ, Zheng J, Wang FL, Konno K, and Jiang X
- Subjects
- Animals, Hydrogen-Ion Concentration, Molecular Weight, Proteins isolation & purification, Proteolysis, Tandem Mass Spectrometry, Cysteine Proteases chemistry, Cysteine Proteinase Inhibitors chemistry, Proteins chemistry, Seafood analysis, Stichopus chemistry
- Abstract
Proteolysis of noncollagenous proteins in sea cucumber, Stichopus Japonicus, body wall (sjBW) was investigated. The proteins removed from sjBW by SDS and urea extraction were mainly noncollagenous proteins with molecular weights about 200kDa (Band I) and 44kDa (Band II), respectively. Band I and Band II were identified as major yolk protein (MYP) and actin, respectively, from holothurian species by liquid chromatography-mass spectrometry (LC-MS/MS) with significant scores. Based on TCA-soluble oligopeptide assay, the optimum proteolysis condition of noncollagenous proteins was at 46.3°C and pH 6.1, by response surface methodology. The proteolysis of MYP, and actin, was partially inhibited by cysteine protease inhibitors, including Trans-epoxysuccinyl-l-leucyl-amido (4-guanidino) butane (E-64), iodoacetic acid, antipain and whey protein concentrate. These results suggest that cysteine proteases are partially involved in the proteolysis of noncollagenous proteins in body wall of sea cucumber, S. japonicus., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
27. Identification of semicarbazones, thiosemicarbazones and triazine nitriles as inhibitors of Leishmania mexicana cysteine protease CPB.
- Author
-
Schröder J, Noack S, Marhöfer RJ, Mottram JC, Coombs GH, and Selzer PM
- Subjects
- Binding Sites, Catalytic Domain, Cysteine Proteases metabolism, Cysteine Proteinase Inhibitors pharmacology, Drug Discovery, High-Throughput Screening Assays, Inhibitory Concentration 50, Leishmania mexicana enzymology, Models, Molecular, Molecular Docking Simulation, Molecular Structure, Nitriles pharmacology, Protein Binding, Recombinant Proteins chemistry, Recombinant Proteins metabolism, Semicarbazones pharmacology, Thiosemicarbazones pharmacology, Cysteine Proteases chemistry, Cysteine Proteinase Inhibitors chemistry, Leishmania mexicana drug effects, Nitriles chemistry, Semicarbazones chemistry, Thiosemicarbazones chemistry
- Abstract
Cysteine proteases of the papain superfamily are present in nearly all eukaryotes. They play pivotal roles in the biology of parasites and inhibition of cysteine proteases is emerging as an important strategy to combat parasitic diseases such as sleeping sickness, Chagas' disease and leishmaniasis. Homology modeling of the mature Leishmania mexicana cysteine protease CPB2.8 suggested that it differs significantly from bovine cathepsin B and thus could be a good drug target. High throughput screening of a compound library against this enzyme and bovine cathepsin B in a counter assay identified four novel inhibitors, containing the warhead-types semicarbazone, thiosemicarbazone and triazine nitrile, that can be used as leads for antiparasite drug design. Covalent docking experiments confirmed the SARs of these lead compounds in an effort to understand the structural elements required for specific inhibition of CPB2.8. This study has provided starting points for the design of selective and highly potent inhibitors of L. mexicana cysteine protease CPB that may also have useful efficacy against other important cysteine proteases.
- Published
- 2013
- Full Text
- View/download PDF
28. Toward the discovery of inhibitors of babesipain-1, a Babesia bigemina cysteine protease: in vitro evaluation, homology modeling and molecular docking studies.
- Author
-
Pérez B, Antunes S, Gonçalves LM, Domingos A, Gomes JR, Gomes P, and Teixeira C
- Subjects
- Amino Acid Sequence, Animals, Antiprotozoal Agents chemical synthesis, Antiprotozoal Agents pharmacology, Babesia genetics, Cattle, Cloning, Molecular, Cysteine Endopeptidases chemistry, Cysteine Proteases genetics, Cysteine Proteases metabolism, In Vitro Techniques, Models, Molecular, Molecular Sequence Data, Molecular Structure, Protein Conformation, Protozoan Proteins genetics, Protozoan Proteins metabolism, Sequence Homology, Amino Acid, Babesia enzymology, Cysteine Proteases chemistry, Cysteine Proteinase Inhibitors pharmacology, Drug Discovery, Drug Evaluation, Preclinical, Protozoan Proteins antagonists & inhibitors
- Abstract
Babesia bigemina is a protozoan parasite that causes babesiosis, a disease with a world-wide distribution in mammals, principally affecting cattle and man. The unveiling of the genome of B. bigemina is a project in active progress that has already revealed a number of new targets with potential interest for the design of anti-babesiosis drugs. In this context, babesipain-1 has been identified as a proteolytically active enzyme whose three-dimensional structure has not been resolved yet, but which is known to be inhibited by cysteine proteases inhibitors such as E64, ALLN, leupeptin, and vinyl sulfones. In this work, we introduce (1) a homology model of babesipain-1; (2) a comparison between babesipain-1 and falcipain-2, a cysteine protease of the malaria parasite Plasmodium falciparum; (3) in vitro data for babesipain-1 inhibition by HEDICINs and HECINs, previously reported as modest inhibitors of falcipain-2; and (4) the docked binding conformations of HEDICINs and HECINs in the model of babesipain-1. HEDICINs presented similar preferred binding conformations for both babesipain-1 and falcipain-2. However, in vitro bioassay shows that HEDICINs and HECINs are better inhibitors of babesipain-1 than of falcipain-2, which could be explained by observed differences between the active pockets of these proteins in silico. Results presented herein provide a valuable contribution to future computer-aided molecular design of new babesipain-1 inhibitors.
- Published
- 2013
- Full Text
- View/download PDF
29. Cysteine peptidases and their inhibitors in Tetranychus urticae: a comparative genomic approach.
- Author
-
Santamaría ME, Hernández-Crespo P, Ortego F, Grbic V, Grbic M, Diaz I, and Martinez M
- Subjects
- Animals, Arthropods classification, Arthropods genetics, Cathepsin B genetics, Cathepsin B metabolism, Cathepsin L genetics, Cathepsin L metabolism, Cystatins classification, Cystatins genetics, Cysteine Endopeptidases genetics, Cysteine Endopeptidases metabolism, Cysteine Proteases chemistry, Cysteine Proteases metabolism, Cysteine Proteinase Inhibitors classification, Cysteine Proteinase Inhibitors metabolism, Embryo, Nonmammalian metabolism, Evolution, Molecular, Phylogeny, Tetranychidae classification, Cysteine Proteases genetics, Cysteine Proteinase Inhibitors genetics, Genomics, Tetranychidae genetics
- Abstract
Background: Cysteine peptidases in the two-spotted spider mite Tetranychus urticae are involved in essential physiological processes, including proteolytic digestion. Cystatins and thyropins are inhibitors of cysteine peptidases that modulate their activity, although their function in this species has yet to be investigated. Comparative genomic analyses are powerful tools to obtain advanced knowledge into the presence and evolution of both, peptidases and their inhibitors, and could aid to elucidate issues concerning the function of these proteins., Results: We have performed a genomic comparative analysis of cysteine peptidases and their inhibitors in T. urticae and representative species of different arthropod taxonomic groups. The results indicate: i) clade-specific proliferations are common to C1A papain-like peptidases and for the I25B cystatin family of inhibitors, whereas the C1A inhibitors thyropins are evolutionarily more conserved among arthropod clades; ii) an unprecedented extensive expansion for C13 legumain-like peptidases is found in T. urticae; iii) a sequence-structure analysis of the spider mite cystatins suggests that diversification may be related to an expansion of their inhibitory range; and iv) an in silico transcriptomic analysis shows that most cathepsin B and L cysteine peptidases, legumains and several members of the cystatin family are expressed at a higher rate in T. urticae feeding stages than in embryos., Conclusion: Comparative genomics has provided valuable insights on the spider mite cysteine peptidases and their inhibitors. Mite-specific proliferations of C1A and C13 peptidase and I25 cystatin families and their over-expression in feeding stages of mites fit with a putative role in mite's feeding and could have a key role in its broad host feeding range.
- Published
- 2012
- Full Text
- View/download PDF
30. Cryptostatin, a chagasin-family cysteine protease inhibitor of Cryptosporidium parvum.
- Author
-
Kang JM, Ju HL, Yu JR, Sohn WM, and Na BK
- Subjects
- Amino Acid Motifs, Cathepsin B antagonists & inhibitors, Cathepsin B chemistry, Cathepsin L antagonists & inhibitors, Cathepsin L chemistry, Cryptosporidium parvum metabolism, Cystatins genetics, Cystatins metabolism, Cysteine Proteases chemistry, Cysteine Proteases genetics, Cysteine Proteinase Inhibitors genetics, Cysteine Proteinase Inhibitors metabolism, Hot Temperature, Humans, Kinetics, Models, Molecular, Molecular Sequence Data, Papain antagonists & inhibitors, Papain chemistry, Protein Stability, Protein Structure, Secondary, Protozoan Proteins chemistry, Protozoan Proteins genetics, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins metabolism, Sequence Alignment, Cryptosporidium parvum genetics, Cystatins chemistry, Cysteine Proteases metabolism, Cysteine Proteinase Inhibitors chemistry, Protozoan Proteins metabolism
- Abstract
Cysteine proteases of pathogenic protozoan parasites play pivotal roles in the life cycle of parasites, but strict regulation of their activities is also essential for maintenance of parasite physiology and interaction with hosts. In this study, we identified and characterized cryptostatin, a novel inhibitor of cysteine protease (ICP) of Cryptosporidium parvum. Cryptostatin showed low sequence identity to other chagasin-family ICPs, but 3 motifs (NPTTG, GXGG, and RPW/F motifs), which are evolutionarily conserved in chagasin-family ICPs, were found in the sequence. The overall structure of cryptostatin consisted of 8 β-strands that progressed in parallel and closely resembled the immunoglobulin fold. Recombinant cryptostatin inhibited various cysteine proteases, including papain, human cathepsin B, human cathepsin L, and cryptopain-1, with K i's in the picomolar range. Cryptostatin was active over a wide pH range and was highly stable under physiological conditions. The protein was thermostable and retained its inhibitory activity even after incubation at 95°C. Cryptostatin formed tight complexes with cysteine proteases, so the complexes remained intact in the presence of sodium dodecyl sulfate and β-mercaptoethanol, but they were disassembled by boiling. An immunogold electron microscopy analysis demonstrated diffused localization of cryptostatin within oocystes and meronts, but not within trophozoites, which suggests a possible role for cryptostatin in host cell invasion by C. parvum.
- Published
- 2012
- Full Text
- View/download PDF
31. Study of protein complexes via homology modeling, applied to cysteine proteases and their protein inhibitors.
- Author
-
Tastan Bishop O and Kroon M
- Subjects
- Amino Acid Sequence, Animals, Antimalarials metabolism, Antimalarials pharmacology, Crystallography, X-Ray, Cysteine Proteases chemistry, Cysteine Proteinase Inhibitors metabolism, Cysteine Proteinase Inhibitors pharmacology, Databases, Protein, Humans, Malaria, Falciparum drug therapy, Malaria, Falciparum parasitology, Malaria, Vivax drug therapy, Malaria, Vivax parasitology, Molecular Sequence Data, Sequence Alignment, Sequence Homology, Amino Acid, Thermodynamics, Antimalarials chemistry, Cysteine Proteases metabolism, Cysteine Proteinase Inhibitors chemistry, Models, Molecular, Plasmodium falciparum enzymology, Plasmodium vivax enzymology
- Abstract
This paper develops and evaluates large-scale calculation of 3D structures of protein complexes by homology modeling as a promising new approach for protein docking. The complexes investigated were papain-like cysteine proteases and their protein inhibitors, which play numerous roles in human and parasitic metabolisms. The structural modeling was performed in two parts. For the first part (evaluation set), nine crystal structure complexes were selected, 1325 homology models of known complexes were rebuilt by various templates including hybrids, allowing an analysis of the factors influencing the accuracy of the models. The important considerations for modeling the interface were protease coverage and inhibitor sequence identity. In the second part (study set), the findings of the evaluation set were used to select appropriate templates to model novel cysteine protease-inhibitor complexes from human and malaria parasites Plasmodium falciparum and Plasmodium vivax. The energy scores, considering the evaluation set, indicate that the models are of high accuracy.
- Published
- 2011
- Full Text
- View/download PDF
32. Terpyridine platinum(II) complexes inhibit cysteine proteases by binding to active-site cysteine.
- Author
-
Lo YC, Su WC, Ko TP, Wang NC, and Wang AH
- Subjects
- Amino Acid Sequence, Catalysis, Catalytic Domain drug effects, Cysteine Proteases metabolism, Cysteine Proteinase Inhibitors metabolism, Drug Design, Enzyme Activation drug effects, Humans, Models, Molecular, Molecular Sequence Data, Organoplatinum Compounds metabolism, Peptides chemistry, Peptides metabolism, Protein Binding, Protein Conformation, Cysteine chemistry, Cysteine Proteases chemistry, Cysteine Proteinase Inhibitors chemistry, Cysteine Proteinase Inhibitors pharmacology, Organoplatinum Compounds chemistry, Organoplatinum Compounds pharmacology
- Abstract
Platinum(II) complexes have been demonstrated to form covalent bonds with sulfur-donating ligands (in glutathione, metallothionein and other sulfur-containing biomolecules) or coordination bonds with nitrogen-donating ligands (such as histidine and guanine). To investigate how these compounds interact with cysteine proteases, we chose terpyridine platinum(II) (TP-Pt(II)) complexes as a model system. By using X-ray crystallography, we demonstrated that TP-Pt(II) formed a covalent bond with the catalytic cysteine residue in pyroglutamyl peptidase I. Moreover, by using MALDI (matrix-assisted laser desorption/ionization) and TOF-TOF (time of flight) mass spectrometry, we elucidated that the TP-Pt(II) complex formed a covalent bond with the active-site cysteine residue in two other types of cysteine protease. Taken together, the results unequivocally showed that TP-Pt(II) complexes can selectively bind to the active site of most cysteine proteases. Our findings here can be useful in the design of new anti-cancer, anti-parasite or anti-virus platinum(II) compounds.
- Published
- 2011
- Full Text
- View/download PDF
33. Molecular docking and 3D-quantitative structure activity relationship analyses of peptidyl vinyl sulfones: Plasmodium Falciparum cysteine proteases inhibitors.
- Author
-
Teixeira C, Gomes JR, Couesnon T, and Gomes P
- Subjects
- Allosteric Site, Antimalarials chemistry, Antimalarials pharmacology, Binding Sites, Computer Simulation, Cysteine Endopeptidases drug effects, Cysteine Endopeptidases metabolism, Cysteine Proteases drug effects, Cysteine Proteases metabolism, Cysteine Proteinase Inhibitors pharmacology, Ligands, Molecular Conformation, Plasmodium falciparum, Protein Structure, Secondary, Sulfones pharmacology, Cysteine Endopeptidases chemistry, Cysteine Proteases chemistry, Cysteine Proteinase Inhibitors chemistry, Drug Design, Models, Molecular, Quantitative Structure-Activity Relationship, Sulfones chemistry
- Abstract
Comparative molecular field analysis (CoMFA) and comparative molecular similarity indices analysis (CoMSIA) based on three-dimensional quantitative structure-activity relationship (3D-QSAR) studies were conducted on a series (39 molecules) of peptidyl vinyl sulfone derivatives as potential Plasmodium Falciparum cysteine proteases inhibitors. Two different methods of alignment were employed: (i) a receptor-docked alignment derived from the structure-based docking algorithm GOLD and (ii) a ligand-based alignment using the structure of one of the ligands derived from a crystal structure from the PDB databank. The best predictions were obtained for the receptor-docked alignment with a CoMFA standard model (q (2) = 0.696 and r (2) = 0.980) and with CoMSIA combined electrostatic, and hydrophobic fields (q (2) = 0.711 and r (2) = 0.992). Both models were validated by a test set of nine compounds and gave satisfactory predictive r (2) (pred) values of 0.76 and 0.74, respectively. CoMFA and CoMSIA contour maps were used to identify critical regions where any change in the steric, electrostatic, and hydrophobic fields may affect the inhibitory activity, and to highlight the key structural features required for biological activity. Moreover, the results obtained from 3D-QSAR analyses were superimposed on the Plasmodium Falciparum cysteine proteases active site and the main interactions were studied. The present work provides extremely useful guidelines for future structural modifications of this class of compounds towards the development of superior antimalarials.
- Published
- 2011
- Full Text
- View/download PDF
34. Consensus models of activity landscapes with multiple chemical, conformer, and property representations.
- Author
-
Yongye AB, Byler K, Santos R, Martínez-Mayorga K, Maggiora GM, and Medina-Franco JL
- Subjects
- Cysteine Proteases chemistry, Cysteine Proteases metabolism, Humans, Inhibitory Concentration 50, Protein Conformation, Structure-Activity Relationship, Trypanosoma brucei brucei enzymology, Cysteine Proteinase Inhibitors chemistry, Cysteine Proteinase Inhibitors pharmacology, Models, Molecular
- Abstract
We report consensus Structure-Activity Similarity (SAS) maps that address the dependence of activity landscapes on molecular representation. As a case study, we characterized the activity landscape of 54 compounds with activities against human cathepsin B (hCatB), human cathepsin L (hCatL), and Trypanosoma brucei cathepsin B (TbCatB). Starting from an initial set of 28 descriptors we selected ten representations that capture different aspects of the chemical structures. These included four 2D (MACCS keys, GpiDAPH3, pairwise, and radial fingerprints) and six 3D (4p and piDAPH4 fingerprints with each including three conformers) representations. Multiple conformers are used for the first time in consensus activity landscape modeling. The results emphasize the feasibility of identifying consensus data points that are consistently formed in different reference spaces generated with several fingerprint models, including multiple 3D conformers. Consensus data points are not meant to eliminate data, disregarding, for example, "true" activity cliffs that are not identified by some molecular representations. Instead, consensus models are designed to prioritize the SAR analysis of activity cliffs and other consistent regions in the activity landscape that are captured by several molecular representations. Systematic description of the SARs of two targets give rise to the identification of pairs of compounds located in the same region of the activity landscape of hCatL and TbCatB suggesting similar mechanisms of action for the pairs involved. We also explored the relationship between property similarity and activity similarity and found that property similarities are suitable to characterize SARs. We also introduce the concept of structure-property-activity (SPA) similarity in SAR studies.
- Published
- 2011
- Full Text
- View/download PDF
35. A novel transglutaminase substrate from Streptomyces mobaraensis inhibiting papain-like cysteine proteases.
- Author
-
Sarafeddinov A, Arif A, Peters A, and Fuchsbauer HL
- Subjects
- Amino Acid Sequence, Animals, Bacterial Proteins antagonists & inhibitors, Bacterial Proteins chemistry, Bacterial Proteins isolation & purification, Cysteine Proteases chemistry, Cysteine Proteases metabolism, Cysteine Proteinase Inhibitors chemistry, Cysteine Proteinase Inhibitors pharmacology, Molecular Sequence Data, Papain chemistry, Papain isolation & purification, Papain metabolism, Sequence Homology, Streptomyces classification, Streptomyces enzymology, Streptomyces growth & development, Substrate Specificity, Cysteine Proteases drug effects, Cysteine Proteinase Inhibitors isolation & purification, Papain antagonists & inhibitors, Streptomyces metabolism, Transglutaminases metabolism
- Abstract
Transglutaminase from Streptomyces mobaraensis is an enzyme of unknown function that cross-links proteins to high molecular weight aggregates. Previously, we characterized two intrinsic transglutaminase substrates with inactivating activities against subtilisin and dispase. This report now describes a novel substrate that inhibits papain, bromelain, and trypsin. Papain was the most sensitive protease; thus, the protein was designated Streptomyces papain inhibitor (SPI). To avoid transglutaminase-mediated glutamine deamidation during culture, SPI was produced by Streptomyces mobaraensis at various growth temperatures. The best results were achieved by culturing for 30-50 h at 42 degrees C, which yielded high SPI concentrations and negligibly small amounts of mature transglutaminase. Transglutaminasespecific biotinylation displayed largely unmodified glutamine and lysine residues. In contrast, purified SPI from the 28 degrees C culture lost the potential to be cross-linked, but exhibited higher inhibitory activity as indicated by a significantly lower Ki (60 nM vs. 140 nM). Despite similarities in molecular mass (12 kDa) and high thermostability, SPI exhibits clear differences in comparison with all members of the wellknown family of Streptomyces subtilisin inhibitors. The neutral protein (pI of 7.3) shares sequence homology with a putative protein from Streptomyces lavendulae, whose conformation is most likely stabilized by two disulfide bridges. However, cysteine residues are not localized in the typical regions of subtilisin inhibitors. SPI and the formerly characterized dispase-inactivating substrate are unique proteins of distinct Streptomycetes such as Streptomyces mobaraensis. Along with the subtilisin inhibitory protein, they could play a crucial role in the defense of vulnerable protein layers that are solidified by transglutaminase.
- Published
- 2011
36. Potent and selective inhibition of cysteine proteases from Plasmodium falciparum and Trypanosoma brucei.
- Author
-
Ehmke V, Heindl C, Rottmann M, Freymond C, Schweizer WB, Brun R, Stich A, Schirmeister T, and Diederich F
- Subjects
- Animals, Crystallography, X-Ray, Cysteine Proteases chemistry, Models, Molecular, Cysteine Proteases metabolism, Cysteine Proteinase Inhibitors pharmacology, Plasmodium falciparum enzymology, Trypanosoma brucei brucei enzymology
- Published
- 2011
- Full Text
- View/download PDF
37. Crystal structure of the cysteine protease inhibitor 2 from Entamoeba histolytica: functional convergence of a common protein fold.
- Author
-
Casados-Vázquez LE, Lara-González S, and Brieba LG
- Subjects
- Amino Acid Sequence, Base Sequence, Cloning, Molecular, Crystallography, Crystallography, X-Ray methods, Cysteine Proteases chemistry, Cysteine Proteases genetics, Cysteine Proteinase Inhibitors chemistry, Entamoeba histolytica enzymology, Models, Molecular, Molecular Sequence Data, Oligodeoxyribonucleotides chemistry, Peptide Hydrolases metabolism, Polymerase Chain Reaction, Protein Conformation, Protozoan Proteins chemistry, Protozoan Proteins genetics, Protozoan Proteins metabolism, Sequence Alignment, Sequence Homology, Amino Acid, Cysteine Proteinase Inhibitors genetics, Entamoeba histolytica genetics
- Abstract
Cysteine proteases (CP) are key pathogenesis and virulence determinants of protozoan parasites. Entamoeba histolytica contains at least 50 cysteine proteases; however, only three (EhCP1, EhCP2 and EhCP5) are responsible for approximately 90% of the cysteine protease activity in this parasite. CPs are expressed as inactive zymogens. Because the processed proteases are potentially cytotoxic, protozoan parasites have developed mechanisms to regulate their activity. Inhibitors of cysteine proteases (ICP) of the chagasin-like inhibitor family (MEROPS family I42) were recently identified in bacteria and protozoan parasites. E. histolytica contains two ICP-encoding genes of the chagasin-like inhibitor family. EhICP1 localizes to the cytosol, whereas EhICP2 is targeted to phagosomes. Herein, we report two crystal structures of EhICP2. The overall structure of EhICP2 consists of eight β-strands and closely resembles the immunoglobulin fold. A comparison between the two crystal forms of EhICP2 indicates that the conserved BC, DE and FG loops form a flexible wedge that may block the active site of CPs. The positively charged surface of the wedge-forming loops in EhICP2 contrasts with the neutral surface of the wedge-forming loops in chagasin. We postulate that the flexibility and positive charge observed in the DE and FG loops of EhICP2 may be important to facilitate the initial binding of this inhibitor to the battery of CPs present in E. histolytica., (Copyright © 2010 Elsevier B.V. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
38. Identification of potent and reversible cruzipain inhibitors for the treatment of Chagas disease.
- Author
-
Beaulieu C, Isabel E, Fortier A, Massé F, Mellon C, Méthot N, Ndao M, Nicoll-Griffith D, Lee D, Park H, and Black WC
- Subjects
- Biphenyl Compounds chemical synthesis, Biphenyl Compounds therapeutic use, Cathepsins antagonists & inhibitors, Cathepsins metabolism, Cysteine Endopeptidases metabolism, Cysteine Proteases chemistry, Cysteine Proteases metabolism, Cysteine Proteinase Inhibitors chemical synthesis, Cysteine Proteinase Inhibitors therapeutic use, Humans, Protozoan Proteins, Structure-Activity Relationship, Valine chemical synthesis, Valine chemistry, Valine therapeutic use, Biphenyl Compounds chemistry, Chagas Disease drug therapy, Cysteine Endopeptidases chemistry, Cysteine Proteinase Inhibitors chemistry, Valine analogs & derivatives
- Abstract
Identification of potent and reversible cruzipain inhibitors for the treatment of Chagas disease is described. The identified inhibitors bearing an amino nitrile warhead in P1 exhibit low nanomolar in vitro potency against cruzipain. Further SAR in P2 portion led to the identification of compounds, such as 26, that have a unique selectivity profile against other cysteine proteases and offering new opportunities for safer treatment of Chagas disease., (Copyright © 2010 Elsevier Ltd. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
39. Cysteine protease inhibitors: from evolutionary relationships to modern chemotherapeutic design for the treatment of infectious diseases.
- Author
-
Toh EC, Huq NL, Dashper SG, and Reynolds EC
- Subjects
- Amino Acid Sequence, Cysteine Proteases chemistry, Cysteine Proteases metabolism, Cysteine Proteinase Inhibitors metabolism, Humans, Models, Molecular, Molecular Sequence Data, Sequence Alignment, Communicable Diseases drug therapy, Cysteine Proteinase Inhibitors chemistry, Cysteine Proteinase Inhibitors therapeutic use, Drug Design
- Abstract
Cysteine proteases are one of the largest groups of proteases and are involved in many important biological functions in all kingdoms of life. They are virulence factors of a range of eukaryotic, bacterial and viral pathogens and are involved in host invasion, pathogen replication and disruption of the host immune response. Their activity is regulated by a range of protease inhibitors. This review discusses the various families of cysteine protease inhibitors, their different modes of inhibition and their evolutionary relationships. These inhibitors as well as the recent discovery of propeptide and propeptide-like inhibitors provide insights into the structures that are important for particular inhibitory mechanisms, thus forming the foundation for the design of future therapeutics.
- Published
- 2010
- Full Text
- View/download PDF
40. Molecular cloning of a multidomain cysteine protease and protease inhibitor precursor gene from the tobacco hornworm (Manduca sexta) and functional expression of the cathepsin F-like cysteine protease domain.
- Author
-
Miyaji T, Murayama S, Kouzuma Y, Kimura N, Kanost MR, Kramer KJ, and Yonekura M
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Cathepsin F chemistry, Cathepsin F genetics, Cystatins chemistry, Cystatins metabolism, Cysteine Proteases chemistry, Cysteine Proteases metabolism, Cysteine Proteinase Inhibitors chemistry, Cysteine Proteinase Inhibitors metabolism, DNA, Complementary genetics, Genes, Insect, Hemolymph chemistry, Insect Proteins chemistry, Insect Proteins metabolism, Larva enzymology, Larva genetics, Manduca enzymology, Manduca metabolism, Molecular Sequence Data, Protein Precursors genetics, Cloning, Molecular, Cystatins genetics, Cysteine Proteases genetics, Cysteine Proteinase Inhibitors genetics, Insect Proteins genetics, Manduca genetics
- Abstract
A Manduca sexta (tobacco hornworm) cysteine protease inhibitor, MsCPI, purified from larval hemolymph has an apparent molecular mass of 11.5 kDa, whereas the size of the mRNA is very large (∼9 kilobases). MsCPI cDNA consists of a 9,273 nucleotides that encode a polypeptide of 2,676 amino acids, which includes nine tandemly repeated MsCPI domains, four cystatin-like domains and one procathepsin F-like domain. The procathepsin F-like domain protein was expressed in Escherichia coli and processed to its active mature form by incubation with pepsin. The mature enzyme hydrolyzed Z-Leu-Arg-MCA, Z-Phe-Arg-MCA and Boc-Val-Leu-Lys-MCA rapidly, whereas hydrolysis of Suc-Leu-Tyr-MCA and Z-Arg-Arg-MCA was very slow. The protease was strongly inhibited by MsCPI, egg-white cystatin and sunflower cystatin with K(i) values in the nanomolar range. When the MsCPI tandem protein linked to two MsCPI domains was treated with proteases, it was degraded by the cathepsin F-like protease. However, tryptic digestion converted the MsCPI tandem protein to an active inhibitory form. These data support the hypothesis that the mature MsCPI protein is produced from the MsCPI precursor protein by trypsin-like proteases. The resulting mature MsCPI protein probably plays a role in the regulation of the activity of endogenous cysteine proteases., (Copyright © 2010 Elsevier Ltd. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
41. "Click" synthesis of small molecule-peptide conjugates for organelle-specific delivery and inhibition of lysosomal cysteine proteases.
- Author
-
Loh Y, Shi H, Hu M, and Yao SQ
- Subjects
- Aza Compounds chemistry, Click Chemistry, Cysteine Proteases metabolism, Cysteine Proteinase Inhibitors pharmacology, Hep G2 Cells, Humans, Microscopy, Fluorescence, Peptides pharmacology, tat Gene Products, Human Immunodeficiency Virus chemistry, Cysteine Proteases chemistry, Cysteine Proteinase Inhibitors chemistry, Lysosomes enzymology, Peptides chemistry
- Abstract
A click chemistry approach for the synthesis of small molecule inhibitor-peptide conjugates to achieve organelle-specific delivery has been developed. Biological testing showed that the inhibitor-Tat conjugate was successfully delivered to the lysosomes, leading to potent inhibition of lysosomal cysteine proteases in cultured cells.
- Published
- 2010
- Full Text
- View/download PDF
42. Rational design of inhibitors and activity-based probes targeting Clostridium difficile virulence factor TcdB.
- Author
-
Puri AW, Lupardus PJ, Deu E, Albrow VE, Garcia KC, Bogyo M, and Shen A
- Subjects
- Allosteric Regulation, Anti-Bacterial Agents pharmacology, Bacterial Proteins metabolism, Bacterial Toxins metabolism, Binding Sites, Clostridioides difficile enzymology, Computer Simulation, Crystallography, X-Ray, Cysteine Proteases chemistry, Cysteine Proteinase Inhibitors pharmacology, Drug Design, Molecular Probes chemistry, Molecular Probes pharmacology, Phytic Acid chemistry, Protein Binding, Protein Structure, Tertiary, Structure-Activity Relationship, Anti-Bacterial Agents chemistry, Bacterial Proteins antagonists & inhibitors, Bacterial Toxins antagonists & inhibitors, Cysteine Proteinase Inhibitors chemistry
- Abstract
Clostridium difficile is a leading cause of nosocomial infections. The major virulence factors of this pathogen are the multi-domain toxins TcdA and TcdB. These toxins contain a cysteine protease domain (CPD) that autoproteolytically releases a cytotoxic effector domain upon binding intracellular inositol hexakisphosphate. Currently, there are no known inhibitors of this protease. Here, we describe the rational design of covalent small molecule inhibitors of TcdB CPD. We identified compounds that inactivate TcdB holotoxin function in cells and solved the structure of inhibitor-bound protease to 2.0 Å. This structure reveals the molecular basis of CPD substrate recognition and informed the synthesis of activity-based probes for this enzyme. The inhibitors presented will guide the development of therapeutics targeting C. difficile, and the probes will serve as tools for studying the unique activation mechanism of bacterial toxin CPDs., (Copyright © 2010 Elsevier Ltd. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
43. α-ketoheterocycles as inhibitors of Leishmania mexicana cysteine protease CPB.
- Author
-
Steert K, Berg M, Mottram JC, Westrop GD, Coombs GH, Cos P, Maes L, Joossens J, Van der Veken P, Haemers A, and Augustyns K
- Subjects
- Antiprotozoal Agents chemical synthesis, Antiprotozoal Agents pharmacology, Cell Line, Cysteine Proteases metabolism, Cysteine Proteinase Inhibitors chemical synthesis, Cysteine Proteinase Inhibitors pharmacology, Heterocyclic Compounds chemical synthesis, Heterocyclic Compounds pharmacology, Humans, Kinetics, Leishmania infantum drug effects, Plasmodium falciparum drug effects, Trypanosoma brucei brucei drug effects, Trypanosoma cruzi drug effects, Antiprotozoal Agents chemistry, Cysteine Proteases chemistry, Cysteine Proteinase Inhibitors chemistry, Heterocyclic Compounds chemistry, Leishmania mexicana enzymology
- Abstract
Cysteine proteases of the papain superfamily are present in nearly all eukaryotes and also play pivotal roles in the biology of parasites. Inhibition of cysteine proteases is emerging as an important strategy to combat parasitic diseases such as sleeping sickness, Chagas disease, and leishmaniasis. Inspired by the in vivo antiparasitic activity of the vinylsulfone-based cysteine protease inhibitors, a series of α-ketoheterocycles were developed as reversible inhibitors of a recombinant L. mexicana cysteine protease, CPB2.8. Three isoxazoles and especially one oxadiazole compound are potent reversible inhibitors of CPB2.8; however, in vitro whole-organism screening against a panel of protozoan parasites did not fully correlate with the observed inhibition of the cysteine protease.
- Published
- 2010
- Full Text
- View/download PDF
44. Biochemical comparison of two proteolytic enzymes from Carica candamarcensis: structural motifs underlying resistance to cystatin inhibition.
- Author
-
Gomes MT, Ribeiro HA, Lopes MT, Guzman F, and Salas CE
- Subjects
- Animals, Chickens, Cysteine Proteases metabolism, Iodoacetamide, Mitogens metabolism, Peptide Hydrolases metabolism, Plant Proteins metabolism, Protein Isoforms, Structure-Activity Relationship, Caricaceae enzymology, Cystatins, Cysteine Proteases chemistry, Cysteine Proteinase Inhibitors, Mitogens chemistry, Peptide Hydrolases chemistry, Plant Proteins chemistry
- Abstract
The lattices of Carica candamarcensis and Carica papaya, members of the Caricaceae family, contain isoforms of cysteine proteinases that help protect these plants against injury. In a prior study, we fractionated 14 discrete proteinaceous components from C. candamarcensis, two of them displaying mitogenic activity in mammalian cells. In this study, we compared the kinetic parameters of one of the mitogenic proteinases (CMS2MS2) with one of the isoforms displaying the highest enzyme activity of this group (CMS1MS2). Both enzymes display a similar Km value with either BAPNA (Benzoyl-Arg-pNA) or PFLPNA (Pyr-Phe-Leu-pNA), but the kcat of CMS1MS2 is about 14-fold higher for BAPNA and 129-fold higher with PFLPNA. While both enzymes are inhibited by E-64 and iodoacetamide, chicken cystatin fully inhibits CMS1MS2, but scarcely affects activity of CMS2MS2. Based on the structure of these proteins and other enzymes from the Caricaceae family whose structures have been resolved, it is proposed that Arg(180) located in the cleft at the active site in CMS2MS2 is responsible for its resistance to cystatin., (Copyright 2010 Elsevier Ltd. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
45. The medicinal chemistry of the cathepsin cysteine proteases.
- Author
-
Masuya K and Teno N
- Subjects
- Cathepsins chemistry, Cathepsins metabolism, Cysteine Proteases chemistry, Humans, Cathepsins antagonists & inhibitors, Cysteine Proteases metabolism, Cysteine Proteinase Inhibitors pharmacology
- Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.