Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
Add more filters

Publication year range
1.
EMBO J ; 37(7)2018 04 03.
Article in English | MEDLINE | ID: mdl-29519896

ABSTRACT

Toxoplasma gondii aspartyl protease 3 (TgASP3) phylogenetically clusters with Plasmodium falciparum Plasmepsins IX and X (PfPMIX, PfPMX). These proteases are essential for parasite survival, acting as key maturases for secreted proteins implicated in invasion and egress. A potent antimalarial peptidomimetic inhibitor (49c) originally developed against Plasmepsin II selectively targets TgASP3, PfPMIX, and PfPMX To unravel the molecular basis for the selectivity of 49c, we constructed homology models of PfPMIX, PfPMX, and TgASP3 that were first validated by identifying the determinants of microneme and rhoptry substrate recognition. The flap and flap-like structures of several reported Plasmepsins are highly flexible and critically modulate the access to the binding cavity. Molecular docking of 49c to TgASP3, PfPMIX, and PfPMX models predicted that the conserved phenylalanine residues in the flap, F344, F291, and F305, respectively, account for the sensitivity toward 49c. Concordantly, phenylalanine mutations in the flap of the three proteases increase twofold to 15-fold the IC50 values of 49c. Compellingly the selection of mutagenized T. gondii resistant strains to 49c reproducibly converted F344 to a cysteine residue.


Subject(s)
Antimalarials/pharmacology , Aspartic Acid Proteases/antagonists & inhibitors , Aspartic Acid Proteases/metabolism , Drug Resistance/physiology , Protease Inhibitors/pharmacology , Protozoan Proteins/chemistry , Antimalarials/chemistry , Aspartic Acid Endopeptidases/chemistry , Aspartic Acid Endopeptidases/metabolism , Cysteine , Dose-Response Relationship, Drug , Drug Resistance/drug effects , Inhibitory Concentration 50 , Models, Molecular , Molecular Docking Simulation , Mutation , Parasitic Sensitivity Tests , Phenylalanine/drug effects , Phenylalanine/genetics , Plasmodium falciparum/drug effects , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , Recombinant Proteins/genetics , Sequence Alignment , Toxoplasma/drug effects , Toxoplasma/genetics
2.
J Enzyme Inhib Med Chem ; 35(1): 629-638, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32037904

ABSTRACT

Phialophora verrucosa causes several fungal human diseases, mainly chromoblastomycosis, which is extremely difficult to treat. Several studies have shown that human immunodeficiency virus peptidase inhibitors (HIV-PIs) are attractive candidates for antifungal therapies. This work focused on studying the action of HIV-PIs on peptidase activity secreted by P. verrucosa and their effects on fungal proliferation and macrophage interaction. We detected a peptidase activity from P. verrucosa able to cleave albumin, sensitive to pepstatin A and HIV-PIs, especially lopinavir, ritonavir and amprenavir, showing for the first time that this fungus secretes aspartic-type peptidase. Furthermore, lopinavir, ritonavir and nelfinavir reduced the fungal growth, causing remarkable ultrastructural alterations. Lopinavir and ritonavir also affected the conidia-macrophage adhesion and macrophage killing. Interestingly, P. verrucosa had its growth inhibited by ritonavir combined with either itraconazole or ketoconazole. Collectively, our results support the antifungal action of HIV-PIs and their relevance as a possible alternative therapy for fungal infections.


Subject(s)
Antifungal Agents/pharmacology , Aspartic Acid Proteases/antagonists & inhibitors , HIV Protease Inhibitors/pharmacology , Macrophages/drug effects , Phialophora/drug effects , Antifungal Agents/chemical synthesis , Antifungal Agents/chemistry , Aspartic Acid Proteases/metabolism , Carbamates/chemical synthesis , Carbamates/chemistry , Carbamates/pharmacology , Dose-Response Relationship, Drug , Furans , HIV Protease Inhibitors/chemical synthesis , HIV Protease Inhibitors/chemistry , Humans , Lopinavir/chemical synthesis , Lopinavir/chemistry , Lopinavir/pharmacology , Macrophages/metabolism , Microbial Sensitivity Tests , Molecular Structure , Phialophora/enzymology , Phialophora/growth & development , Ritonavir/chemical synthesis , Ritonavir/chemistry , Ritonavir/pharmacology , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/chemistry , Sulfonamides/pharmacology
3.
Apoptosis ; 24(7-8): 662-672, 2019 08.
Article in English | MEDLINE | ID: mdl-31134446

ABSTRACT

Mutations in the DNA damage repair (DDR) pathway are frequently detected in colorectal cancer (CRC). The dysregulation of miRNAs, such as oncogenes or tumor suppressors, participates in CRC tumorigenesis. A previous study showed that low miR-3607 expression correlated with poor survival in prostate cancer patients, but its role in CRC remains unclear. In this study, we analyzed miR-3607 expression Pan-Cancer data from the NCI's Genomic Data Commons (GDC) and found that miR-3607 was downregulated in lymphatic invasion patients and in recurrent cancer and correlated with Pan-Cancer patient survival. Functional studies indicated that the overexpression of miR-3607 decreased CRC cell proliferation, migration and invasion. Additionally, we used gene set enrichment analysis (GSEA), Gene Ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis and a protein-protein interaction network to demonstrate that miR-3607 affects the DDR pathway. Luciferase reporter and apoptosis assays confirmed that DNA damage inducible 1 homolog 2 (DDI2) is the functional target of miR-3607. Therefore, miR-3607 inhibits the tumorigenesis of CRC probably by suppressing the oncogene DDI2, and it might serve as a novel target for CRC prediction and therapy.


Subject(s)
Aspartic Acid Proteases/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA Damage/genetics , MicroRNAs/genetics , Apoptosis , Aspartic Acid Proteases/antagonists & inhibitors , Aspartic Acid Proteases/metabolism , Binding Sites , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Colorectal Neoplasms/metabolism , Gene Expression , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HCT116 Cells , HT29 Cells , Humans , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Mutation , Neoplasm Invasiveness , Protein Interaction Maps
4.
PLoS Biol ; 12(7): e1001897, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24983235

ABSTRACT

The malaria parasite Plasmodium falciparum exports several hundred proteins into the infected erythrocyte that are involved in cellular remodeling and severe virulence. The export mechanism involves the Plasmodium export element (PEXEL), which is a cleavage site for the parasite protease, Plasmepsin V (PMV). The PMV gene is refractory to deletion, suggesting it is essential, but definitive proof is lacking. Here, we generated a PEXEL-mimetic inhibitor that potently blocks the activity of PMV isolated from P. falciparum and Plasmodium vivax. Assessment of PMV activity in P. falciparum revealed PEXEL cleavage occurs cotranslationaly, similar to signal peptidase. Treatment of P. falciparum-infected erythrocytes with the inhibitor caused dose-dependent inhibition of PEXEL processing as well as protein export, including impaired display of the major virulence adhesin, PfEMP1, on the erythrocyte surface, and cytoadherence. The inhibitor killed parasites at the trophozoite stage and knockdown of PMV enhanced sensitivity to the inhibitor, while overexpression of PMV increased resistance. This provides the first direct evidence that PMV activity is essential for protein export in Plasmodium spp. and for parasite survival in human erythrocytes and validates PMV as an antimalarial drug target.


Subject(s)
Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Proteases/antagonists & inhibitors , Oligopeptides/pharmacology , Protozoan Proteins/antagonists & inhibitors , Sulfonamides/pharmacology , Endoplasmic Reticulum/metabolism , Erythrocytes/parasitology , Humans , Protein Transport/drug effects , Protozoan Proteins/metabolism
5.
Biochem J ; 473(6): 769-77, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26759376

ABSTRACT

Pharmacological challenges to oncogenic Ras-expressing cancer cells have shown a novel type of cell death, ferroptosis, which requires intracellular iron. In the present study, we assessed ferroptosis following treatment of human fibrosarcoma HT1080 cells with several inhibitors of lysosomal activity and found that they prevented cell death induced by the ferroptosis-inducing compounds erastin and RSL3. Fluorescent analyses with a reactive oxygen species (ROS) sensor revealed constitutive generation of ROS in lysosomes, and treatment with lysosome inhibitors decreased both lysosomal ROS and a ferroptotic cell-death-associated ROS burst. These inhibitors partially prevented intracellular iron provision by attenuating intracellular transport of transferrin or autophagic degradation of ferritin. Furthermore, analyses with a fluorescent sensor that detects oxidative changes in cell membranes revealed that formation of lipid ROS in perinuclear compartments probably represented an early event in ferroptosis. These results suggest that lysosomal activity is involved in lipid ROS-mediated ferroptotic cell death through regulation of cellular iron equilibria and ROS generation.


Subject(s)
Cell Death/physiology , Iron/metabolism , Lysosomes/physiology , Aspartic Acid Proteases/antagonists & inhibitors , Cell Line, Tumor , Deferoxamine/pharmacology , Humans , Pepstatins/pharmacology , Piperazines/pharmacology , Reactive Oxygen Species
6.
J Enzyme Inhib Med Chem ; 31(1): 126-31, 2016.
Article in English | MEDLINE | ID: mdl-25807296

ABSTRACT

OBJECTIVE: This work describes the anti-enzymatic activity of (7-chloroquinolin-4-yl)arylhydrazones against Candida albicans and examines their cytotoxicity. MATERIAL AND METHODS: Ten C. albicans strains [nine isolates and one azole-resistant standard strain (ATCC 62342)] were used to assess the anti-enzymatic activity. Fifteen compounds at sub-antifungal concentrations ranging from 12.5 to 100 µg/ml were assessed after a 30-min exposure. The strains were seeded onto petri dishes with selective agar media for aspartyl proteases (Saps) and phospholipases (PLs). Enzymatic inhibition was measured by the reduction of the precipitation zone (Pz) against untreated strains (positive control). A colorimetric MTT assay was used with 3T3/NIH mouse fibroblasts to evaluate cytotoxicity. Cells were exposed to 15 compounds in concentrations from 6.25 to 100 µg/ml for 24 and 48 h. RESULTS: Four hydrazones showed enzymatic repression values over 40% to Pl and three over 20% to Saps. The cell viability was over 50% at hydrazone concentrations of 25-100 µg/ml. CONCLUSION: These results revealed that select (7-chloroquinolin-4-yl)arylhydrazones may be potential antifungal agents for the control of C. albicans infections.


Subject(s)
Antifungal Agents/pharmacology , Aspartic Acid Proteases/antagonists & inhibitors , Candida albicans/drug effects , Candida albicans/enzymology , Enzyme Inhibitors/pharmacology , Hydrazones/pharmacology , Phospholipases/antagonists & inhibitors , Quinolines/pharmacology , Animals , Antifungal Agents/chemical synthesis , Antifungal Agents/chemistry , Aspartic Acid Proteases/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Colorimetry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Fibroblasts/drug effects , Fibroblasts/microbiology , Hydrazones/chemical synthesis , Hydrazones/chemistry , Mice , Molecular Structure , NIH 3T3 Cells , Phospholipases/metabolism , Quinolines/chemical synthesis , Quinolines/chemistry , Structure-Activity Relationship
7.
Bioorg Med Chem Lett ; 25(8): 1782-1786, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25782742

ABSTRACT

Recently, we reported on the discovery of (3S,4S)-disubstituted pyrrolidines (e.g., 2) as inhibitors of the human aspartyl protease renin. In our effort to further expand the scope of this novel class of direct renin inhibitors, a new sub-series was designed in which the prime site substituents are linked to the pyrrolidine core by a (3S)-amino functional group. In particular, analogs bearing the corresponding sulfonamide spacer (50, 51 and 54a) demonstrated a pronounced increase in in vitro potency compared to compound 2.


Subject(s)
Protease Inhibitors/chemistry , Pyrrolidines/chemistry , Renin/antagonists & inhibitors , Aspartic Acid Proteases/antagonists & inhibitors , Aspartic Acid Proteases/metabolism , Binding Sites , Crystallography, X-Ray , Drug Design , Half-Life , Humans , Isomerism , Molecular Dynamics Simulation , Protease Inhibitors/chemical synthesis , Protease Inhibitors/metabolism , Protein Binding , Protein Structure, Tertiary , Pyrrolidines/chemical synthesis , Pyrrolidines/metabolism , Renin/metabolism , Structure-Activity Relationship
8.
Bioorg Med Chem Lett ; 25(8): 1787-1791, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25754490

ABSTRACT

Inhibition of the aspartyl protease renin is considered as an efficient approach for treating hypertension. Lately, we described the discovery of a novel class of direct renin inhibitors which comprised a pyrrolidine scaffold (e.g., 2). Based on the X-ray structure of the lead compound 2 bound to renin we predicted that optimization of binding interactions to the prime site could offer an opportunity to further expand the scope of this chemotype. Pyrrolidine-based inhibitors were synthesized in which the prime site moieties are linked to the pyrrolidine core through an oxygen atom, resulting in an ether or a carbamate linker subseries. Especially the carbamate derivatives showed a pronounced increase in in vitro potency compared to 2. Here we report the structure-activity relationship of both subclasses and demonstrate blood pressure lowering effects for an advanced prototype in a hypertensive double-transgenic rat model after oral dosing.


Subject(s)
Aspartic Acid Proteases/antagonists & inhibitors , Protease Inhibitors/chemistry , Pyrrolidines/chemistry , Renin/antagonists & inhibitors , Animals , Aspartic Acid Proteases/metabolism , Binding Sites , Crystallography, X-Ray , Disease Models, Animal , Humans , Hydrogen Bonding , Hypertension/drug therapy , Isomerism , Molecular Dynamics Simulation , Oxygen/chemistry , Protease Inhibitors/metabolism , Protease Inhibitors/therapeutic use , Protein Binding , Protein Structure, Tertiary , Pyrrolidines/chemical synthesis , Pyrrolidines/metabolism , Rats , Rats, Sprague-Dawley , Renin/metabolism , Structure-Activity Relationship
9.
J Nat Prod ; 77(6): 1275-9, 2014 Jun 27.
Article in English | MEDLINE | ID: mdl-24921156

ABSTRACT

BACE-1 is an aspartic protease involved in the conversion of amyloid precursor protein (APP) to amyloid-ß (Aß) in vivo, which is one of the key steps in the development and progression of Alzheimer's disease. In a previous screening procedure for inhibitors of BACE-1 activity, the oil of Lavandula luisieri was identified as the most potent among several essential oils. The inhibitory effect of this essential oil on Aß production was also demonstrated in a cellular assay. The composition of the volatile oil and the isolation of the compound responsible for the inhibitory activity were also reported. The present work focused on the characterization of the inhibition of BACE-1 by this active compound, a monoterpene necrodane ketone, 2,3,4,4-tetramethyl-5-methylenecyclopent-2-enone (1), with assessment of its Ki value and the type of inhibition. The dose-related effects of the compound were also evaluated using two different cell lines, with determinations of the respective EC50 values. The entire oil and the 2,3,4,4-tetramethyl-5-methylenecyclopent-2-enone (1) were tested on a triple transgenic mouse model of Alzheimer's disease. The overall results showed that compound 1 displayed a dose-dependent inhibition of BACE-1 in cellular and mouse models of Alzheimer's disease and is therefore capable of passing through cellular membranes and the blood-brain barrier.


Subject(s)
Alzheimer Disease/metabolism , Aspartic Acid Proteases/antagonists & inhibitors , Lavandula/chemistry , Monoterpenes/isolation & purification , Monoterpenes/pharmacology , Amyloid beta-Peptides/analysis , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Blood-Brain Barrier/metabolism , Brain/drug effects , Brain/metabolism , Cathepsin D/antagonists & inhibitors , Disease Models, Animal , Dose-Response Relationship, Drug , Mice , Molecular Structure , Monoterpenes/chemistry , Monoterpenes/pharmacokinetics , Plant Oils/chemistry
10.
J Biomol Struct Dyn ; 42(5): 2270-2281, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37139547

ABSTRACT

Glioblastoma, the most severe form of brain tumor and a leading cause of death within a year of diagnosis, is characterized by excessive protein synthesis and folding in the lumen of the endoplasmic reticulum (ER), leading to increased ER stress in the cells of GBM tissues. To mitigate this stress the cancer cells have intelligently adopted a plethora of response mechanisms and Unfolded Protein Response (UPR) is one of those. To bear with this exhaustive situation cells upregulate a strong protein degradation system in form of 26S proteasome and blocking of proteasomal gene synthesis may be a potential therapeutic action against GBM. Proteasomal gene synthesis is exclusively dependent on the transcription factor Nuclear respiratory factor 1 (NRF1) and its activating enzyme DNA damage inducible 1 homolog 2 (DDI2). Here in this study, we performed molecular docking against DDI2 with the 20 FDA-approved drugs and identified Alvimopan and Levocabastine as the top two compounds with the best binding score along with the standard drug Nelfinavir. MD simulation (100 ns) of these protein-ligand docked complexes reveals that the stability and compactness of Alvimopan are high in comparison with Nelfinavir. Our in-silico (Molecular docking and Molecular dynamics simulation) studies pointed out that Alvimopan may be repurposed as a DDI2 inhibitor and can be used as a potential anticancer agent for the treatment of brain tumors.Communicated by Ramaswamy H. Sarma.


Subject(s)
Antineoplastic Agents , Aspartic Acid Proteases , Glioblastoma , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Drug Repositioning , Glioblastoma/drug therapy , Molecular Docking Simulation , Molecular Dynamics Simulation , Nelfinavir/pharmacology , Aspartic Acid Proteases/antagonists & inhibitors
11.
Biochem Biophys Res Commun ; 441(2): 291-6, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24070613

ABSTRACT

Alzheimer's disease (AD) is a devastating neurodegenerative disease affecting millions of people. ß-Secretase-1 (BACE-1), an enzyme involved in the processing of the amyloid precursor protein (APP) to form Aß, is a well validated target for AD. Herein, the authors characterize 10 randomly selected hydroxyethylamine (HEA) BACE-1 inhibitors in terms of their association and dissociation rate constants and thermodynamics of binding using surface plasmon resonance (SPR). Rate constants of association (ka) measured at 25 °C ranged from a low of 2.42×10(4) M(-1) s(-1) to the highest value of 8.3×10(5) M(-1) s(-1). Rate constants of dissociation (kd) ranged from 1.09×10(-4) s(-1) (corresponding to a residence time of close to three hours), to the fastest of 0.028 s(-1). Three compounds were selected for further thermodynamic analysis where it was shown that equilibrium binding was enthalpy driven while unfavorable entropy of binding was observed. Structural analysis revealed that upon ligand binding, the BACE-1flap folds down over the bound ligand causing an induced fit. The maximal difference between alpha carbon positions in the open and closed conformations of the flap was over 5 Å. Thus the negative entropy of binding determined using SPR analysis was consistent with an induced fit observed by structural analysis.


Subject(s)
Alzheimer Disease/enzymology , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Ethanolamines , Protease Inhibitors/pharmacology , Amyloid Precursor Protein Secretases/chemistry , Aspartic Acid Endopeptidases/chemistry , Aspartic Acid Proteases/antagonists & inhibitors , Aspartic Acid Proteases/chemistry , Enzymes, Immobilized/antagonists & inhibitors , Enzymes, Immobilized/chemistry , Humans , Kinetics , Protease Inhibitors/chemistry , Protein Conformation , Thermodynamics
12.
J Antimicrob Chemother ; 68(2): 348-53, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23109184

ABSTRACT

OBJECTIVES: There is a general lack of effective and non-toxic chemotherapeutic agents for leishmaniasis and there is as yet no study about the effect of HIV peptidase inhibitors (HIV PIs) on Leishmania/HIV-coinfected patients. In the present work, we performed a comparative analysis of the spectrum of action of HIV PIs on different Leishmania spp., including strains obtained from HIV-positive patients receiving or not receiving antiretroviral treatment. METHODS: The effects of nelfinavir and saquinavir on Leishmania proliferation were assessed by means of a colorimetric assay (MTT). Subsequently, the effect of nelfinavir on aspartic peptidase activity from Leishmania spp. was assessed by following the degradation of the fluorogenic substrate MCA-G-K-P-I-L-F-F-R-L-K-DNP-Arg-NH(2). RESULTS: Nelfinavir was capable of significantly reducing the multiplication of many Leishmania reference strains and isolates obtained from HIV-positive patients receiving or not receiving antiretroviral treatment. Leishmania major growth was inhibited by ≈ 50%, while all other flagellates were strongly inhibited (at least 94%), except for a Leishmania chagasi strain obtained from an HIV-positive patient under treatment with highly active antiretroviral therapy (HAART). Culture of this isolate in the presence of nelfinavir induced a considerable reduction in the aspartic peptidase activity. In addition, nelfinavir was also capable of inhibiting the aspartic peptidase activity of all Leishmania strains tested. CONCLUSIONS: The present data contribute to the study of the effect of HIV PIs on Leishmania infection and add new insights into the possibility of exploiting aspartic peptidases as promising targets in order to generate novel medications to treat leishmaniasis.


Subject(s)
Antiprotozoal Agents/pharmacology , Aspartic Acid Proteases/antagonists & inhibitors , Leishmania/drug effects , Leishmania/enzymology , Leishmaniasis/parasitology , Nelfinavir/pharmacology , Colorimetry , Fluorometry , HIV Infections/complications , Humans , Leishmania/growth & development , Leishmania/isolation & purification , Microbial Viability , Oligopeptides/metabolism , Saquinavir/pharmacology , Tetrazolium Salts/metabolism , Thiazoles/metabolism
13.
J Enzyme Inhib Med Chem ; 28(5): 1054-60, 2013 Oct.
Article in English | MEDLINE | ID: mdl-22957693

ABSTRACT

The aspartic protease inhibitory efficiency of rBm-33, an aspin from a filarial parasite Brugia malayi was investigated. rBm-33 was found to be thermostable up to 90°C and it forms a stable 'enzyme-product' complex with human pepsin. Aspartic protease inhibitory activity was investigated using UV spectroscopy and isothermal titration calorimetry. Our results suggest that rBm-33 inhibits the activity of important human aspartic proteases that were examined with binding constants (Kb) values between 10.23 × 10(3) and 6.52 × 10(3) M(-1). The binding reactions were enthalpy driven with ΔHb values between -50.99 and -46.07 kJ mol(-1). From kinetic studies, pepsin inhibition by rBm-33 was found to be linear competitive with an inhibition constant (Ki) of 2.5 (±0.8) nM. Because of the inhibitory efficacy of Bm-33 against important human aspartic proteases which play a vital role in immune-regulation along with other functions, Bm-33 can be projected as a drug target for the filariasis.


Subject(s)
Antigens, Helminth/metabolism , Aspartic Acid Proteases/antagonists & inhibitors , Brugia malayi/chemistry , Helminth Proteins/metabolism , Protease Inhibitors/pharmacology , Animals , Antigens, Helminth/chemistry , Antigens, Helminth/isolation & purification , Aspartic Acid Proteases/metabolism , Chemistry, Physical , Dose-Response Relationship, Drug , Helminth Proteins/chemistry , Helminth Proteins/isolation & purification , Humans , Kinetics , Molecular Structure , Protease Inhibitors/chemistry , Protease Inhibitors/isolation & purification , Protein Stability , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Structure-Activity Relationship , Temperature
14.
Acta Crystallogr D Biol Crystallogr ; 68(Pt 5): 541-52, 2012 May.
Article in English | MEDLINE | ID: mdl-22525752

ABSTRACT

The analysis reported here describes detailed structural studies of endothiapepsin (the aspartic proteinase from Endothia parasitica), with and without bound inhibitors, and human pepsin 3b. Comparison of multiple crystal structures of members of the aspartic proteinase family has revealed small but significant differences in domain orientation in different crystal forms. In this paper, it is shown that these differences in domain orientation do not necessarily correlate with the presence or absence of bound inhibitors, but appear to stem at least partly from crystal contacts mediated by sulfate ions. However, since the same inherent flexibility of the structure is observed for other enzymes in this family such as human pepsin, the native structure of which is also reported here, the observed domain movements may well have implications for the mechanism of catalysis.


Subject(s)
Aspartic Acid Proteases/chemistry , Ascomycota/enzymology , Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Endopeptidases/chemistry , Aspartic Acid Proteases/antagonists & inhibitors , Crystallography, X-Ray , Humans , Models, Molecular , Pepsin A/antagonists & inhibitors , Pepsin A/chemistry , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Protein Conformation , Protein Structure, Tertiary
15.
Biochem Biophys Res Commun ; 421(3): 413-7, 2012 May 11.
Article in English | MEDLINE | ID: mdl-22469467

ABSTRACT

Two potent inhibitors (compounds 1 and 2) of malarial aspartyl protease, plasmepsin-II, were evaluated against wild type (NL4-3) and multidrug-resistant clinical isolate 769 (MDR) variants of human immunodeficiency virus type-1 (HIV-1) aspartyl protease. Enzyme inhibition assays showed that both 1 and 2 have better potency against NL4-3 than against MDR protease. Crystal structures of MDR protease in complex with 1 and 2 were solved and analyzed. Crystallographic analysis revealed that the MDR protease exhibits a typical wide-open conformation of the flaps (Gly48 to Gly52) causing an overall expansion in the active site cavity, which, in turn caused unstable binding of the inhibitors. Due to the expansion of the active site cavity, both compounds showed loss of direct contacts with the MDR protease compared to the docking models of NL4-3. Multiple water molecules showed a rich network of hydrogen bonds contributing to the stability of the ligand binding in the distorted binding pockets of the MDR protease in both crystal structures. Docking analysis of 1 and 2 showed a decrease in the binding affinity for both compounds against MDR supporting our structure-function studies. Thus, compounds 1 and 2 show promising inhibitory activity against HIV-1 protease variants and hence are good candidates for further development to enhance their potency against NL4-3 as well as MDR HIV-1 protease variants.


Subject(s)
Antimalarials/chemistry , Aspartic Acid Proteases/chemistry , Drug Resistance, Multiple, Viral , HIV Protease Inhibitors/chemistry , HIV Protease/chemistry , Oligopeptides/chemistry , Pyridines/chemistry , Antimalarials/pharmacology , Aspartic Acid Proteases/antagonists & inhibitors , Crystallography, X-Ray , HIV Protease/genetics , HIV Protease/metabolism , HIV Protease Inhibitors/pharmacology , Humans , Oligopeptides/pharmacology , Pepstatins/chemistry , Pepstatins/pharmacology , Protein Conformation/drug effects , Pyridines/pharmacology
16.
Microbiology (Reading) ; 158(Pt 7): 1897-1907, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22493301

ABSTRACT

A low-molecular-mass aspartic protease inhibitor was isolated from a novel Penicillium sp. The inhibitor was purified to homogeneity, as shown by reversed-phase HPLC and SDS-PAGE. The M(r) of the inhibitor was 1585 and the amino acid composition showed the presence of D, D, D, E, A, K, L, Y, H, I and W residues. The steady-state kinetic interactions of Aspergillus saitoi aspartic protease with the inhibitor revealed the reversible, competitive, time-dependent tight-binding nature of the inhibitor, with IC(50) and K(i) values of 1.8 and 0.85 µM, respectively. Fluorescence spectroscopy and circular dichroism analysis showed that inactivation of the enzyme was due to binding of the inhibitor to the active site. The inhibitor was found to inhibit mycelial growth and spore germination of Aspergillus fumigatus and Aspergillus niger in vitro with MIC values of 1.65 and 0.30 µg ml(-1), respectively. This study will potentially open the way towards the development of a tight-binding peptidic inhibitor against fungal aspartic proteases to combat human fungal infections.


Subject(s)
Aspartic Acid Proteases/antagonists & inhibitors , Penicillium/enzymology , Protease Inhibitors/isolation & purification , Antifungal Agents/chemistry , Antifungal Agents/isolation & purification , Antifungal Agents/metabolism , Aspartic Acid Proteases/metabolism , Aspergillus fumigatus/drug effects , Aspergillus fumigatus/growth & development , Aspergillus niger/drug effects , Aspergillus niger/growth & development , Chromatography, High Pressure Liquid , Circular Dichroism , DNA, Fungal/chemistry , DNA, Fungal/genetics , DNA, Ribosomal Spacer/chemistry , DNA, Ribosomal Spacer/genetics , Electrophoresis, Polyacrylamide Gel , Humans , Inhibitory Concentration 50 , Kinetics , Molecular Sequence Data , Penicillium/genetics , Protease Inhibitors/chemistry , Protease Inhibitors/metabolism , Sequence Analysis, DNA , Spectrometry, Fluorescence
18.
Acta Pharmacol Sin ; 33(12): 1459-68, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22842730

ABSTRACT

AIM: To identify a small molecule L655,240 as a novel ß-secretase (BACE1) inhibitor and to investigate its effects on ß-amyloid (Aß) generation in vitro. METHODS: Fluorescence resonance energy transfer (FRET) was used to characterize the inhibitory effect of L655,240 on BACE1. Surface plasmon resonance (SPR) technology-based assay was performed to study the binding affinity of L655,240 for BACE1. The selectivity of L655,240 toward BACE1 over other aspartic proteases was determined with enzymatic assay. The effects of L655,240 on Aß40, Aß42, and sAPPß production were studied in HEK293 cells stably expressing APP695 Swedish mutant(K595N/M596L) (HEK293-APPswe cells). The activities of BACE1, γ-secretase and α-secretase were assayed, and both the mRNA and protein levels of APP and BACE1 were evaluated using real-time PCR (RT-PCR) and Western blot analysis. RESULTS: L655,240 was determined to be a competitive, selective BACE1 inhibitor (IC(50)=4.47±1.37 µmol/L), which bound to BACE1 directly (K(D)=17.9±0.72 µmol/L). L655,240 effectively reduced Aß40, Aß42, and sAPPß production by inhibiting BACE1 without affecting the activities of γ-secretase and α-secretase in HEK293-APPswe cells. L655,240 has no effect on APP and BACE1 mRNA or protein levels in HEK293-APPswe cells. CONCLUSION: The small molecule L655,240 is a novel BACE1 inhibitor that can effectively decreases Aß production in vitro, thereby highlighting its therapeutic potential for the treatment of Alzheimer's disease.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid beta-Peptides/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Indoles/pharmacology , Peptide Fragments/antagonists & inhibitors , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Peptides/biosynthesis , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Proteases/antagonists & inhibitors , Binding, Competitive , Blotting, Western , Cell Culture Techniques , Cell Survival/drug effects , Fluorescence Resonance Energy Transfer , HEK293 Cells , Humans , Indoles/chemistry , Molecular Structure , Peptide Fragments/biosynthesis , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Surface Plasmon Resonance , Transfection
19.
J Antibiot (Tokyo) ; 75(9): 519-522, 2022 09.
Article in English | MEDLINE | ID: mdl-35882959

ABSTRACT

A new pepstatin with a phenylacetyl group, pepstatin Pa (1), and its methyl ester (2) were isolated from Streptomyces varsoviensis DSM 40346. Their structures were determined by high-resolution mass spectrometry and nuclear magnetic resonance techniques. The absolute configuration was determined using the Marfey's method. Both pentapeptide products are inhibitors of pepsin and cathepsin D. Interestingly, the bacterial genome contains no biosynthetic gene cluster for the new pepstatin, suggesting an extrachromosomal origin of the biosynthetic genes.


Subject(s)
Aspartic Acid Proteases , Pepstatins , Streptomyces , Aspartic Acid Proteases/antagonists & inhibitors , Bacterial Proteins , Pepstatins/pharmacology , Protease Inhibitors , Streptomyces/chemistry
20.
Biochim Biophys Acta ; 1800(5): 526-36, 2010 May.
Article in English | MEDLINE | ID: mdl-20138972

ABSTRACT

BACKGROUND: Chitinase inhibitors have chemotherapeutic potential as fungicides, pesticides and antiasthmatics. The majority of chitinase inhibitors reported are natural products like argifin, argifin linear fragments, argadin, allosamidin and disulfide-cyclized peptides. Here, we report a novel peptidic inhibitor API (Aspartic Protease Inhibitor), isolated from Bacillus licheniformis that inhibits chitinase A (ChiA) from Serratia marcescens. METHODS: The binding affinity of API with ChiA and type of inhibition was determined by the inhibition kinetics assays. Fluorescence and CD spectroscopic analysis and chemical modification of API with different affinity reagents elucidated the mechanism of binding of API with ChiA. RESULTS AND CONCLUSIONS: The peptide has an amino acid sequence N-Ile(1)-Cys(2)-Glu(3)-Ala(4)-Glu(5)-His(6)-Lys(7)-Trp(8)-Gly(9)-Asp(10)-Tyr(11)-Leu(12)-Asp(13)-C. The ChiA-API kinetic interactions reveal noncompetitive, irreversible and tight binding nature of API with I(50) = 600 nM and K(i)= 510 nM in the presence of chromogenic substrate p-nitrophenyl-N,N'-diacetyl-beta-chitobioside[p-NP-(GlcNAc)(2)]. The inhibition progress curves show a two-step slow tight binding inhibition mechanism with the rate constant k(5) = 8.7 +/- 1 x 10(-3) s(-1) and k(6) = 7.3 +/- 0.6 x 10(-5) s(-1). CD-spectra and tryptophanyl fluorescence analysis of ChiA incubated with increasing API concentrations confirms conformational changes in enzyme structure which may be due to irreversible denaturation of enzyme upon binding of API. Chemical modifications by WRK abolished the anti-chitinase activity of API and revealed the involvement of carboxyl groups in the enzyme inactivation. Abolished isoindole fluorescence of OPTA-labeled ChiA demonstrates the irreversible denaturation of ChiA upon incubation with API for prolonged time and distortion of active site of the enzyme. GENERAL SIGNIFICANCE: The data provide useful information that could lead to the generation of drug-like, natural product-based chitinase inhibitors.


Subject(s)
Aspartic Acid Proteases , Bacillus/chemistry , Bacterial Proteins , Chitinases , Peptides/chemistry , Protease Inhibitors/chemistry , Serratia marcescens/enzymology , Aspartic Acid Proteases/antagonists & inhibitors , Aspartic Acid Proteases/chemistry , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Chitinases/antagonists & inhibitors , Chitinases/chemistry , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL