Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
J Chem Inf Model ; 61(11): 5626-5643, 2021 11 22.
Article in English | MEDLINE | ID: mdl-34748335

ABSTRACT

PlaF is a cytoplasmic membrane-bound phospholipase A1 from Pseudomonas aeruginosa that alters the membrane glycerophospholipid (GPL) composition and fosters the virulence of this human pathogen. PlaF activity is regulated by a dimer-to-monomer transition followed by tilting of the monomer in the membrane. However, how substrates reach the active site and how the characteristics of the active site tunnels determine the activity, specificity, and regioselectivity of PlaF for natural GPL substrates have remained elusive. Here, we combined unbiased and biased all-atom molecular dynamics (MD) simulations and configurational free-energy computations to identify access pathways of GPL substrates to the catalytic center of PlaF. Our results map out a distinct tunnel through which substrates access the catalytic center. PlaF variants with bulky tryptophan residues in this tunnel revealed decreased catalysis rates due to tunnel blockage. The MD simulations suggest that GPLs preferably enter the active site with the sn-1 acyl chain first, which agrees with the experimentally demonstrated PLA1 activity of PlaF. We propose that the acyl chain-length specificity of PlaF is determined by the structural features of the access tunnel, which results in favorable free energy of binding of medium-chain GPLs. The suggested egress route conveys fatty acid (FA) products to the dimerization interface and, thus, contributes to understanding the product feedback regulation of PlaF by FA-triggered dimerization. These findings open up opportunities for developing potential PlaF inhibitors, which may act as antibiotics against P. aeruginosa.


Subject(s)
Molecular Dynamics Simulation , Phospholipases/chemistry , Pseudomonas aeruginosa , Catalytic Domain , Dimerization , Pseudomonas aeruginosa/enzymology , Substrate Specificity
2.
J Biol Chem ; 293(51): 19874-19885, 2018 12 21.
Article in English | MEDLINE | ID: mdl-30385503

ABSTRACT

Vaccination is devised/formulated to stimulate specific and prolonged immune responses for long-term protection against infection or disease. A vaccine component, namely adjuvant, enhances antigen recognition by the host immune system and thereby stimulates its cellular and adaptive responses. Especially synthetic Toll-like receptor (TLR) agonists having self-assembling properties are considered as good candidates for adjuvant development. Here, a human TLR4-derived 20-residue peptide (TR-433), present in the dimerization interface of the TLR4-myeloid differentiation protein-2 (MD2) complex, displayed self-assembly and adopted a nanostructure. Both in vitro studies and in vivo experiments in mice indicated that TR-433 is nontoxic. TR-433 induced pro-inflammatory responses in THP-1 monocytes and HEK293T cells that were transiently transfected with TLR4/CD14/MD2 and also in BALB/c mice. In light of the self-assembly and pro-inflammatory properties of TR-433, we immunized with a mixture of TR-433 and either ovalbumin or filarial antigen trehalose-6-phosphate phosphatase (TPP). A significant amount of IgG titers was produced, suggesting adjuvanting capability of TR-433 that was comparable with that of Freund's complete adjuvant (FCA) and appreciably higher than that of alum. We found that TR-433 preferentially activates type 1 helper T cell (Th1) response rather than type 2 helper T cell (Th2) response. To our knowledge, this is the first report on the identification of a short TLR4-derived peptide that possesses both self-assembling and pro-inflammatory properties and has significant efficacy as an adjuvant, capable of activating cellular responses in mice. These results indicate that TR-433 possesses significant potential for development as a new adjuvant in therapeutic application.


Subject(s)
Adjuvants, Immunologic/chemistry , Peptide Fragments/chemistry , Peptide Fragments/immunology , Protein Multimerization , Toll-Like Receptor 4/chemistry , Vaccines/chemistry , Vaccines/immunology , Amino Acid Sequence , Animals , Brugia malayi/immunology , Cell Line , Humans , Immunization , Lymphocyte Antigen 96/chemistry , Mice , Molecular Docking Simulation , Molecular Dynamics Simulation , Ovalbumin/immunology , Protein Structure, Quaternary
3.
New Phytol ; 222(2): 1043-1053, 2019 04.
Article in English | MEDLINE | ID: mdl-30565261

ABSTRACT

To obtain insights into the dynamics of nutrient exchange in arbuscular mycorrhizal (AM) symbiosis, we modelled mathematically the two-membrane system at the plant-fungus interface and simulated its dynamics. In computational cell biology experiments, the full range of nutrient transport pathways was tested for their ability to exchange phosphorus (P)/carbon (C)/nitrogen (N) sources. As a result, we obtained a thermodynamically justified, independent and comprehensive model of the dynamics of the nutrient exchange at the plant-fungus contact zone. The predicted optimal transporter network coincides with the transporter set independently confirmed in wet-laboratory experiments previously, indicating that all essential transporter types have been discovered. The thermodynamic analyses suggest that phosphate is released from the fungus via proton-coupled phosphate transporters rather than anion channels. Optimal transport pathways, such as cation channels or proton-coupled symporters, shuttle nutrients together with a positive charge across the membranes. Only in exceptional cases does electroneutral transport via diffusion facilitators appear to be plausible. The thermodynamic models presented here can be generalized and adapted to other forms of mycorrhiza and open the door for future studies combining wet-laboratory experiments with computational simulations to obtain a deeper understanding of the investigated phenomena.


Subject(s)
Mycorrhizae/metabolism , Nitrogen/metabolism , Phosphorus/metabolism , Symbiosis , Biological Transport , Cell Membrane/metabolism , Models, Biological , Thermodynamics
4.
Bioorg Chem ; 87: 276-290, 2019 06.
Article in English | MEDLINE | ID: mdl-30908970

ABSTRACT

Cervical cancer is a leading cause of cancer-related deaths among women in developing countries. Therefore, development of new chemotherapeutic agents is required. Unlike normal cells, cancer cells contain elevated copper levels which play an integral role in angiogenesis. Thus, targeting copper via copper-specific chelators in cancer cells can serve as effective anticancer strategy. In this work, a copper chelator pregnenolone acetate nucleus-based tetrazole derivative (ligand-L) was synthesized and characterized by elemental analysis, ESI-MS, 1H NMR and 13C NMR. DNA binding ability of ligand-L was studied using UV-Vis and fluorescence spectroscopy. Fluorescence spectroscopy studies reveal that quenching constant of ligand-l-DNA and ligand-L-Cu(II) were found to be 7.4 × 103 M-1 and 8.8 × 103 M-1, respectively. In vitro toxicity of ligand-L was studied on human cervical cancer C33A cancer cells. Results showed that ligand-L exhibit significant cytotoxic activity against cervical cancer C33A cells with IC50 value 5.0 ±â€¯1.8 µM. Further, it was found that ligand-L cytotoxicity is due to redox cycling of copper to generate ROS which leads to DNA damage and apoptosis. In conclusion, this is the report where we synthesized pregnenolone acetate-based tetrazole derivative against C33A cells that targets cellular copper to induce pro-oxidant death in cancer cells. These findings will provide significant insights into the development of new chemical molecules with better copper chelating and pro-oxidant properties against cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Chelating Agents/pharmacology , Organometallic Compounds/pharmacology , Reactive Oxygen Species/metabolism , Uterine Cervical Neoplasms/drug therapy , Acetates/chemistry , Acetates/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Chelating Agents/chemical synthesis , Chelating Agents/chemistry , Copper/chemistry , Copper/pharmacology , DNA Cleavage/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Humans , Ligands , Molecular Structure , Organometallic Compounds/chemical synthesis , Organometallic Compounds/chemistry , Pregnenolone/chemistry , Pregnenolone/pharmacology , Structure-Activity Relationship , Tumor Cells, Cultured , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology
5.
Biochem Cell Biol ; 94(6): 584-596, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27845561

ABSTRACT

This study describes the isolation and purification of a phytocystatin from seeds of Brassica juncea (Indian mustard; cultivar RoAgro 5444), which is an important oilseed crop both agriculturally and economically. The protein was purified by gel filtration chromatography with 24.3% yield and 204-fold purification, and visualised by 2D gel electrophoresis. The 18.1 kDa mustard cystatin was highly specific for cysteine proteinases. The plant cystatin inhibited cathepsin B, confirming its role in conferring pest resistance. The inhibitor was highly stable over a pH range of 3-10 and retained significant inhibitory potential up to 70 °C. The stoichiometry of its interaction with papain, determined by isothermal calorimetry, suggests a 1:1 complex. Secondary structural elements calculated by far-UV circular dichroism (CD) spectroscopy show an 18.8% α-helical and 21% ß-sheet structure. The protein was a non-competitive inhibitor of thiol proteinases. The Stokes radius and frictional co-efficient were used to describe the shape and size of the protein. Homology modelling and docking studies proposed a prototype illustrating the Brassica phytocystatin mediated papain inhibition. Molecular dynamics (MD) study revealed the excellent stability of the papain-phytocystatin complex during a simulation for 100 ns. Detailed results identify the mustard cystatin as an important member of the phytocystatin family.


Subject(s)
Cystatins/chemistry , Cystatins/metabolism , Cysteine Proteinase Inhibitors/chemistry , Cysteine Proteinase Inhibitors/metabolism , Mustard Plant/metabolism , Animals , Antibody Formation , Chromatography, Gel , Computer Simulation , Cystatins/immunology , Cystatins/isolation & purification , Cysteine Proteinase Inhibitors/immunology , Cysteine Proteinase Inhibitors/isolation & purification , Immunoglobulin G/immunology , Kinetics , Male , Models, Molecular , Molecular Dynamics Simulation , Mustard Plant/growth & development , Papain/metabolism , Plant Proteins/immunology , Plant Proteins/isolation & purification , Plant Proteins/metabolism , Rabbits
6.
J Mol Graph Model ; 96: 107512, 2020 05.
Article in English | MEDLINE | ID: mdl-31881466

ABSTRACT

Cathepsin S (CatS) is one of the cysteinyl cathepsins widely studied for its clinical significance and found to be a promising therapeutic target for several diseases; to name a few is arthritis, allergic inflammation, cancer, diabetes, obesity, and cystic fibrosis. Elevated CatS level is a contributing factor for related disorders, and therefore among different strategies to regulate the activity of CatS, one is to design a quality inhibitor. Earlier, we have demonstrated a highly selective CatS inhibitor, RO5444101 interacts primarily with the S2 pocket of the protein which is structurally unique in contrast to other variants of cathepsin. However, the molecular properties of RO5444101 can question its efficacy at the clinical level. In the present study, we have implemented a series of molecular modeling methods to screen the Maybridge library considering the pharmacophoric features of RO5444101 and other relevant inhibitors of CatS. Based on the priority list, eight hits were subjected to biological evaluation. Subsequently, KM07987 was found to be most potent, with the IC50 of <5 µM. Molecular dynamics simulations also relate to our experimental findings and propose the importance of CatS's S2 pocket, which primarily interacts with the inhibitors. Based on the S2 pocket interactions, structural modifications of the promising hits can further be translated into novel scaffolds for improved inhibition of CatS.


Subject(s)
Cathepsins , Molecular Dynamics Simulation , Cathepsins/metabolism , Humans
7.
Biosystems ; 175: 11-23, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30481546

ABSTRACT

Plant-insect interaction system is a widely studied model of the ecosystem. Numerical understanding of this counter system has developed from initial analogy based approach with a predator-prey model to its recent mathematical interpretation including plant immunity concept. In current work, we propose an extension to this model, including molecular interactions behind the plant defense system and its effect on ecological behaviour. Inspired from biomolecular interaction given by Louis and Shah in 2014, we propose here a mathematical model to depict molecular dependence and control of plant insect interaction system. Insect infestation mediated Botrytis Induced Kinase-1 (BIK1) induction resulted in inhibition of Phyto Alexin Deficient-4 (PAD4) protein. Lowered PAD4 triggers the plant defense mechanism, leading to degraded plant immune potential and thereby reducing the plant quality. We mathematically adapt these interactions to show their influence on plant-insect interaction system and hypothesize the significance of BIK1 inhibition leading to the improved plant quality. We implemented the plethora of computational modeling and all atom MD simulations to explain the Plant-Insect-PAD4-BIK1 interaction network and identify potential molecular mechanisms of plant improvement by BIK1 inhibition.


Subject(s)
Arabidopsis Proteins/chemistry , Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Carboxylic Ester Hydrolases/metabolism , Insecta/physiology , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Animals , Arabidopsis/immunology , Catalytic Domain , Gene Expression Regulation, Plant , Host-Pathogen Interactions , Plant Immunity , Protein Conformation , Signal Transduction
8.
ACS Nano ; 13(11): 12470-12486, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31693858

ABSTRACT

The development of effective therapies for stroke continues to face repeated translational failures. Brain endothelial cells form paracellular and transcellular barriers to many blood-borne therapies, and the development of efficient delivery strategies is highly warranted. Here, in a mouse model of stroke, we show selective recruitment of clinically used liposomes into the ischemic brain that correlates with biphasic blood brain barrier (BBB) breakdown. Intravenous administration of liposomes into mice exposed to transient middle cerebral artery occlusion took place at early (0.5 and 4 h) and delayed (24 and 48 h) time points, covering different phases of BBB disruption after stroke. Using a combination of in vivo real-time imaging and histological analysis we show that selective liposomal brain accumulation coincides with biphasic enhancement in transcellular transport followed by a delayed impairment to the paracellular barrier. This process precedes neurological damage in the acute phase and maintains long-term liposomal colocalization within the neurovascular unit, which could have great potential for neuroprotection. Levels of liposomal uptake by glial cells are similarly selectively enhanced in the ischemic region late after experimental stroke (2-3 days), highlighting their potential for blocking delayed inflammatory responses or shifting the polarization of microglia/macrophages toward brain repair. These findings demonstrate the capability of liposomes to maximize selective translocation into the brain after stroke and identify two windows for therapeutic manipulation. This emphasizes the benefits of selective drug delivery for efficient tailoring of stroke treatments.


Subject(s)
Blood-Brain Barrier/metabolism , Liposomes , Stroke/metabolism , Animals , Disease Models, Animal , Drug Delivery Systems , Infarction, Middle Cerebral Artery/metabolism , Liposomes/chemistry , Liposomes/pharmacokinetics , Liposomes/pharmacology , Male , Mice , Mice, Inbred C57BL , Transcytosis/drug effects
9.
J Mol Model ; 23(3): 92, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28236030

ABSTRACT

Cathepsin S has been demonstrated to play a crucial role in the remodeling of extracellular matrix proteins such as elastin and collagen, which in turn contribute to the structural integrity of the cardiovascular wall. Atherosclerotic lesions, aneurysm formation, plaque rupture, thrombosis, and calcification are some of the cardiovascular disorders related to cathepsin S. A highly selective inhibitor of human as well as animal cathepsin S, RO5444101, was recently reported to attenuate the progression of atherosclerotic lesions. Here, we attempted to gain insight into the molecular mechanism of action of RO5444101 on cathepsin S by performing molecular docking and molecular dynamics (MD) simulation studies. The results of our studies correlate well with relevant reported experimental data and potentially explain the selectivity of this inhibitor for cathepsin S rather than cathepsin L1/L, cathepsin L2/V, and cathepsin K, which share conserved catalytic sites and have sequence similarities of 49%, 50%, and 55%, respectively, with respect to cathepsin S. In contrast to those closely related cathepsins, 20 ns MD simulation data reveal that the overall interaction of cathepsin S with RO5444101 is more stable and involves more protein-molecule interactions than the interactions of the inhibitor with the other cathepsins. This study therefore considerably improves our understanding of the molecular mechanism responsible for cathepsin S inhibition and facilitates the identification of potential novel selective inhibitors of cathepsin S.


Subject(s)
Cardiovascular Diseases/drug therapy , Cathepsins/antagonists & inhibitors , Models, Molecular , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Cathepsins/chemistry , Humans , Macrophages/drug effects , Molecular Docking Simulation , Molecular Dynamics Simulation
10.
PLoS One ; 10(5): e0123996, 2015.
Article in English | MEDLINE | ID: mdl-25933436

ABSTRACT

Schistosomiasis is a major endemic disease known for excessive mortality and morbidity in developing countries. Because praziquantel is the only drug available for its treatment, the risk of drug resistance emphasizes the need to discover new drugs for this disease. Cathepsin SmCL1 is the critical target for drug design due to its essential role in the digestion of host proteins for growth and development of Schistosoma mansoni. Inhibiting the function of SmCL1 could control the wide spread of infections caused by S. mansoni in humans. With this objective, a homology modeling approach was used to obtain theoretical three-dimensional (3D) structure of SmCL1. In order to find the potential inhibitors of SmCL1, a plethora of in silico techniques were employed to screen non-peptide inhibitors against SmCL1 via structure-based drug discovery protocol. Receiver operating characteristic (ROC) curve analysis and molecular dynamics (MD) simulation were performed on the results of docked protein-ligand complexes to identify top ranking molecules against the modelled 3D structure of SmCL1. MD simulation results suggest the phytochemical Simalikalactone-D as a potential lead against SmCL1, whose pharmacophore model may be useful for future screening of potential drug molecules. To conclude, this is the first report to discuss the virtual screening of non-peptide inhibitors against SmCL1 of S. mansoni, with significant therapeutic potential. Results presented herein provide a valuable contribution to identify the significant leads and further derivatize them to suitable drug candidates for antischistosomal therapy.


Subject(s)
Anthelmintics/pharmacology , Cathepsins/antagonists & inhibitors , Computer Simulation , Drug Discovery , Helminth Proteins/antagonists & inhibitors , Protease Inhibitors/pharmacology , Schistosoma mansoni/enzymology , Algorithms , Amino Acid Sequence , Animals , Anthelmintics/chemistry , Catalytic Domain , Cathepsins/chemistry , Drug Evaluation, Preclinical , Helminth Proteins/chemistry , Hydrogen Bonding , Knowledge , Ligands , Molecular Dynamics Simulation , Molecular Sequence Data , Peptides/pharmacology , Protease Inhibitors/chemistry , ROC Curve , Reproducibility of Results , Schistosoma mansoni/drug effects , Sequence Alignment , Sequence Homology, Amino Acid , Static Electricity , Structure-Activity Relationship , Thermodynamics
SELECTION OF CITATIONS
SEARCH DETAIL