Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Front Microbiol ; 12: 766351, 2021.
Article in English | MEDLINE | ID: mdl-34925274

ABSTRACT

Early detection of asymptomatic cases through mass screening is essential to constrain the coronavirus disease 2019 (COVID-19) transmission. However, the existing diagnostic strategies are either resource-intensive, time-consuming, or less sensitive, which limits their use in the development of rapid mass screening strategies. There is a clear pressing need for simple, fast, sensitive, and economical diagnostic strategy for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) screening even in resource-limited settings. In the current work, we assessed the in silico feasibility of directly labeling virus surface proteins using fluorogenic molecules with aggregation-induced emission (AIE) property. Here, we present the results for binding of two such AIE probes, phosphonic acid derivative of tetraphenyl ethylene (TPE-P) and sulfonic acid derivative of tetraphenyl ethylene (TPE-S), to SARS-CoV-2 spike protein based on in silico docking studies. Our results show that both TPE-P and TPE-S bind to angiotensin converting enzyme 2 (ACE2)-binding, and N-terminal domains of SARS-CoV-2 spike protein. Molecular dynamic simulations have revealed specific nature of these interactions. We also show that TPE-P and TPE-S bind to hemagglutinin protein of influenza virus, but the interaction strength was found to be different. This difference in interaction strength may affect the emission spectrum of aforementioned AIE probes. Together, these results form a basis for the development of AIE-based diagnostics for differential detection of SARS-CoV-2 and influenza viruses. We believe that these in silico predictions certainly aid in differentially labeling of the both viruses toward the development of rapid detection by AIE probes.

2.
J Biomed Inform ; 85: 114-125, 2018 09.
Article in English | MEDLINE | ID: mdl-30092360

ABSTRACT

Molecular Property Diagnostic Suite - Diabetes Mellitus (MPDSDM) is a Galaxy-based, open source disease-specific web portal for diabetes. It consists of three modules namely (i) data library (ii) data processing and (iii) data analysis tools. The data library (target library and literature) module provide extensive and curated information about the genes involved in type 1 and type 2 diabetes onset and progression stage (available at http://www.mpds-diabetes.in). The database also contains information on drug targets, biomarkers, therapeutics and associated genes specific to type 1, and type 2 diabetes. A unique MPDS identification number has been assigned for each gene involved in diabetes mellitus and the corresponding card contains chromosomal data, gene information, protein UniProt ID, functional domains, druggability and related pathway information. One of the objectives of the web portal is to have an open source data repository that contains all information on diabetes and use this information for developing therapeutics to cure diabetes. We also make an attempt for computational drug repurposing for the validated diabetes targets. We performed virtual screening of 1455 FDA approved drugs on selected 20 type 1 and type 2 diabetes proteins using docking protocol and their biological activity was predicted using "PASS Online" server (http://www.way2drug.com/passonline) towards anti-diabetic activity, resulted in the identification of 41 drug molecules. Five drug molecules (which are earlier known for anti-malarial/microbial, anti-viral, anti-cancer, anti-pulmonary activities) were proposed to have a better repurposing potential for type 2 anti-diabetic activity and good binding affinity towards type 2 diabetes target proteins.


Subject(s)
Diabetes Mellitus/drug therapy , Diabetes Mellitus/genetics , Drug Discovery , Drug Repositioning , Computational Biology , Diabetes Mellitus/diagnosis , Drug Discovery/statistics & numerical data , Drug Evaluation, Preclinical , Drug Repositioning/statistics & numerical data , Humans , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Internet , Molecular Diagnostic Techniques/statistics & numerical data , Molecular Docking Simulation , User-Computer Interface
3.
Mol Biosyst ; 12(10): 3111-23, 2016 10 20.
Article in English | MEDLINE | ID: mdl-27470658

ABSTRACT

Helicobacter pylori is a Gram-negative bacterium that inhabits the human gastrointestinal tract, and some strains of this bacterium cause gastric ulcers and cancer. DNA methyltransferases (MTases) are promising drug targets for the treatment of cancer and other diseases that are also caused by epigenetic alternations of the genome. The C5-cytosine specific DNA methyltransferase from H. pylori (M. Hpy C5mC) catalyzes the transfer of the methyl group from the cofactor S-adenosyl-l-methionine (AdoMet) to the flipped cytosine of the substrate DNA. Herein we report the sequence analyses, 3-D structure modeling and molecular dynamics simulations of M. Hpy C5mC, when complexed with AdoMet as well as DNA. We analyzed the protein-DNA interactions prominently established by the flipped cytosine and the interactions between the protein and cofactor in the active site. We propose that the contacts made by cytosine O2 with Arg155 and Arg157, and the water-mediated interactions with cytosine N3 may be essential for the activity of methyl transfer as well as the deprotonation at the C5 position in our C5mC model. Specific recognition of DNA was mediated mainly by residues from Ser221-Arg229 and Ser243-Gln246 of the target recognition domain (TRD) and some residues of the loop Ser75-Lys83 from the large domain. These findings are further supported by alanine scanning mutagenesis studies. The results reported here explain the sequence, structure and binding features necessary for the recognition between the cofactor and the substrate by the key epigenetic enzyme, M. Hpy C5mC.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/chemistry , DNA/chemistry , Molecular Conformation , Molecular Dynamics Simulation , S-Adenosylmethionine/chemistry , Amino Acid Sequence , Binding Sites , Catalytic Domain , DNA/metabolism , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Helicobacter pylori/enzymology , Hydrogen Bonding , Macromolecular Substances/chemistry , Macromolecular Substances/metabolism , Mutation , Protein Binding , Protein Interaction Domains and Motifs , S-Adenosylmethionine/metabolism , Structure-Activity Relationship
4.
PLoS One ; 11(2): e0146786, 2016.
Article in English | MEDLINE | ID: mdl-26891364

ABSTRACT

The PE and PPE protein family are unique to mycobacteria. Though the complete genome sequences for over 500 M. tuberculosis strains and mycobacterial species are available, few PE and PPE proteins have been structurally and functionally characterized. We have therefore used bioinformatics tools to characterize the structure and function of these proteins. We selected representative members of the PE and PPE protein family by phylogeny analysis and using structure-based sequence annotation identified ten well-characterized protein domains of known function. Some of these domains were observed to be common to all mycobacterial species and some were species specific.


Subject(s)
Bacterial Proteins/genetics , Genome, Bacterial , Mycobacterium tuberculosis/genetics , Mycobacterium/genetics , Virulence Factors/genetics , Amino Acid Motifs , Bacterial Proteins/chemistry , Computational Biology , Models, Molecular , Molecular Sequence Data , Multigene Family , Mycobacterium/classification , Mycobacterium tuberculosis/classification , Phylogeny , Protein Structure, Tertiary , Sequence Alignment , Sequence Homology, Amino Acid , Species Specificity , Virulence Factors/chemistry
5.
J Biomol Struct Dyn ; 33(8): 1710-9, 2015.
Article in English | MEDLINE | ID: mdl-25245460

ABSTRACT

Human male germ cell-associated kinase (hMAK) is an androgen-inducible gene in prostate epithelial cells, and it acts as a coactivator of androgen receptor signaling in prostate cancer. The 3D structure of the hMAK kinase was modeled based on the crystal structure of CDK2 kinase using comparative modeling methods, and the ATP-binding site was characterized. We have collected five inhibitors of hMAK from the literature and docked into the ATP-binding site of the kinase domain. Solvated interaction energies (SIE) of inhibitor binding are calculated from the molecular dynamics simulations trajectories of protein-inhibitor complexes. The contribution from each active site residue in hMAK toward inhibitor binding revealed the nature and extent of interactions between inhibitors and individual residues. The main chain atoms of Met79 invariably form hydrogen bonds with all five inhibitors. The amino acids Leu7, Val15, and Leu129 stabilize the inhibitors via CH-pi interactions. The Asp140 in the active site and Glu77 in hinge region show characteristic hydrogen bonding interactions with inhibitors. From SIE, the residue-wise interactions revealed the nature of non-bonding contacts and modifications required to increase the inhibitor activity. Our work provides 3D model structure of hMAK and molecular basis for the mechanisms of hMAK inhibition at atomic level that aid in designing new potent inhibitors.


Subject(s)
Models, Molecular , Molecular Conformation , Protein Kinase Inhibitors/chemistry , Protein Kinases/chemistry , Binding Sites , Catalytic Domain , Humans , Male , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding , Protein Kinase Inhibitors/metabolism , Protein Kinases/metabolism
6.
Anticancer Agents Med Chem ; 14(9): 1237-48, 2014.
Article in English | MEDLINE | ID: mdl-25028149

ABSTRACT

Plants are fantastic sources for present day life saving drugs. Monocrotaline a natural ligand exhibits dose-dependent cytotoxicity with potent antineoplastic activity. This study was intended to disclose the therapeutic potential of monocrotaline against hepatocellular carcinoma. The in silico predictions have highlighted the antineoplastic potential, druglikeness and biodegradability of monocrotaline. The in silico docking study has provided an insight and evidence for the antineoplastic activity of monocrotaline against p53, HGF and TREM1 proteins which play a threatening role in causing hepatocellular carcinoma. The mode of action of monocrotaline was determined experimentally by in vitro techniques such as XTT assay, NRU assay and whole cell brine shrimp assay have further supported our in silico studies. The in vitro cytotoxicity of monocrotaline was proved at IC50 24.966 µg/mL and genotoxicity at 2 X IC50 against HepG2 cells. Further, the credible druglike properties with non-mutagenicity, non-toxic on mammalian fibroblast and the potential antineoplastic activity through in vitro experimental validations established monocrotaline as a novel scaffold for liver cancer with superior efficacy and lesser side effects.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/pathology , Cell Proliferation/drug effects , Liver Neoplasms/pathology , Monocrotaline/pharmacology , Animals , BALB 3T3 Cells , Computer Simulation , Hep G2 Cells , Hepatocyte Growth Factor/metabolism , Humans , Membrane Glycoproteins/metabolism , Mice , Molecular Docking Simulation , Receptors, Immunologic/metabolism , Triggering Receptor Expressed on Myeloid Cells-1 , Tumor Suppressor Protein p53/metabolism
7.
Biochim Biophys Acta ; 1840(9): 2685-94, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24972166

ABSTRACT

BACKGROUND: Transglycosylation (TG) activity is a property of glycosyl hydrolases (GHs) with which new glycosidic bonds are introduced between donor and acceptor sugar molecules. This special property of the GHs has potential to generate longer chain chitooligosaccharides (CHOS) that show elicitor activity in plants. We hypothesize that TG activity could be improved by retaining the substrate for a longer duration in the catalytic site. METHODS: Four variants of chitinase D from Serratia proteamaculans (SpChiD) i.e. G119S, G119W, W120A and G201W were analyzed in detail for improved TG activity using high performance liquid chromatography (HPLC) and high resolution mass spectrometry (HRMS). The results were strongly supported by 50ns molecular dynamics (MD) simulations and estimated solvated interaction energies (SIE). RESULTS: The mutant G119W lost much of both hydrolytic and TG activities, while the mutant G201W displayed increased TG. The trajectory of MD simulations of the mutant G119W showed that the indole rings of two adjacent Trp residues create a major hindrance for the DP4 movement towards the catalytic center. Increased van der Waals (vdW) and coulombic interactions between DP4 substrate and the Trp-201 resulted in enhanced TG activity with the mutant G201W. The average number of hydrogen bonds observed for the DP4 substrate was increased for the mutants G119W and G201W compared to SpChiD. CONCLUSION: The increase in TG activity could be due to partial blocking of product exit of SpChiD. GENERAL SIGNIFICANCE: This new approach can be used for generating mutants of GHs with improved TG activity to produce longer chain oligosaccharides.


Subject(s)
Bacterial Proteins/chemistry , Chitinases/chemistry , Molecular Dynamics Simulation , Mutagenesis , Mutation, Missense , Serratia/enzymology , Amino Acid Substitution , Bacterial Proteins/genetics , Chitinases/genetics , Oligosaccharides/chemistry , Protein Structure, Tertiary , Serratia/genetics , Substrate Specificity
8.
PLoS One ; 8(11): e78556, 2013.
Article in English | MEDLINE | ID: mdl-24236021

ABSTRACT

BCR-ABL kinase domain inhibition can be used to treat chronic myeloid leukemia. The inhibitors such as imatinib, dasatinib and nilotinib are effective drugs but are resistant to some BCR-ABL mutations. The pan-BCR-ABL kinase inhibitor ponatinib exhibits potent activity against native, T315I, and all other clinically relevant mutants, and showed better inhibition than the previously known inhibitors. We have studied the molecular dynamics simulations and calculated solvated interaction energies of native and fourteen mutant BCR-ABL kinases (M244V, G250E, Q252H, Y253F, Y253H, E255K, E255V, T315A, T315I, F317L, F317V, M351T, F359V and H396P) complexed with ponatinib. These studies revealed that the interactions between ponatinib and individual residues in BCR-ABL kinase are also affected due to the remote residue mutations. We report that some residues, Met244, Lys245, Gln252, Gly254, Leu370 and Leu298 do not undergo any conformational changes, while the fluctuations in residues from P-loop, ß3-, ß5- strands and αC- helix are mainly responsible for ponatinib binding to native and all mutant BCR-ABL kinases. Our work provides the molecular mechanisms of native and mutant BCR-ABL kinases inhibition by ponatinib at atomic level that has not been studied before.


Subject(s)
Antineoplastic Agents/chemistry , Fusion Proteins, bcr-abl/antagonists & inhibitors , Imidazoles/chemistry , Molecular Dynamics Simulation , Pyridazines/chemistry , Catalytic Domain , Fusion Proteins, bcr-abl/chemistry , Fusion Proteins, bcr-abl/genetics , Humans , Hydrogen Bonding , Mutation, Missense , Protein Binding , Protein Structure, Secondary , Solvents/chemistry , Thermodynamics , Water/chemistry
9.
Proc Natl Acad Sci U S A ; 110(51): 20467-72, 2013 Dec 17.
Article in English | MEDLINE | ID: mdl-24282299

ABSTRACT

Resistin, a cysteine-rich adipocytokine, proposed as a link between obesity and diabetes in mice, was shown as a proinflammatory molecule in humans. We earlier reported that human resistin (hRes), a trimer, was resistant to heat and urea denaturation, existed in an oligomeric polydispersed state, and showed a concentration-dependent conformational change. These properties and an intimate correlation of hRes expression with cellular stress prompted us to investigate hRes as a possible chaperone. Here, we show that recombinant human resistin was able to protect the heat-labile enzymes citrate synthase and Nde1 from thermal aggregation and inactivation and was able to refold and restore their enzymatic activities after heat/guanidinium chloride denaturation. Furthermore, recombinant human resistin could bind misfolded proteins only. Molecular dynamics-based association-dissociation kinetics of hRes subunits pointed to resistin being a molecular chaperone. Bis-ANS, which blocks surface hydrophobicity, abrogated the chaperone activity of hRes, establishing the importance of surface hydrophobicity for chaperone activity. Replacement of Phe49 with Tyr (F49YhRes), a critical residue within the hydrophobic patch of hRes, although it could prevent thermal aggregation of citrate synthase and Nde1, was unable to refold and restore their activities. Treatment of U937 cells with tunicamycin/thapsigargin resulted in reduced hRes secretion and concomitant localization in the endoplasmic reticulum. Escherichia coli transformants expressing hRes could be rescued from thermal stress, pointing to hRes's chaperone-like function in vivo. HeLa cells transfected with hRes showed protection from thapsigargin-induced apoptosis. In conclusion, hRes, an inflammatory protein, additionally exhibited chaperone-like properties, suggesting a possible link between inflammation and cellular stress.


Subject(s)
Cytokines/metabolism , Heat-Shock Response/physiology , Inflammation Mediators/metabolism , Microtubule-Associated Proteins/metabolism , Molecular Chaperones/metabolism , Resistin/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Cytokines/genetics , Enzyme Inhibitors/pharmacology , HeLa Cells , Heat-Shock Response/drug effects , Humans , Inflammation/genetics , Inflammation/metabolism , Mice , Microtubule-Associated Proteins/genetics , Molecular Chaperones/genetics , Resistin/genetics , Thapsigargin/pharmacology , Tunicamycin/pharmacology , U937 Cells
10.
Protein Pept Lett ; 20(11): 1246-56, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23848594

ABSTRACT

Aurora-A, B and C are non-receptor serine/threonine kinases in Homo sapiens. In spite of high similarity in their sequences, they possess distinct binding partners. These kinases play an important role in cell division and overexpressed in certain cancers. It has been demonstrated that Gly198 in Aurora-A kinase is responsible for its basal kinase activity, the mutation G198N transforms Aurora-A to Aurora-B like function and localization by binding to Inner centromere protein (INCENP). The molecular mechanisms, structural determinants and the binding energetics of the Aurora-A - INCENP complex owing to a single amino acid G198N mutation are not studied. Therefore, we have docked INCENP into human Aurora-A kinase, mutated Gly198 to Asn, Leu and Ala. The wild type and mutant Aurora-A - INCENP complexes were subjected to 40 ns molecular dynamics (MD) simulations. The Asn198 is located in the amphipathic cavity comprising Leu869(IN), Glu868(IN), Thr872(IN), Tyr197(AurA) and Tyr199(AurA) and the interactions mediated via hydrogen bonds are important to stabilize the Aurora-A(G198N) - INCENP complex. The fluctuations in the secondary structural elements and the solvent accessible surface area of all the four complexes during the MD simulations were studied. We calculated the binding free energy upon mutation in the three mutant complexes. The Aurora-A(G198N) - INCENP complex with hydrophilic amino acid mutation has the negative free energy of solvation indicating favorable interactions with INCENP. Our results provide the structural basis and energetics of the human Aurora-A(G198N) - INCENP complex.


Subject(s)
Aurora Kinase A/chemistry , Chromosomal Proteins, Non-Histone/chemistry , Mitosis , Protein Structure, Tertiary , Amino Acid Sequence , Aurora Kinase A/genetics , Cell Line , Chromosomal Proteins, Non-Histone/genetics , Energy Metabolism , Humans , Hydrogen Bonding , Models, Molecular , Molecular Dynamics Simulation , Mutation , Phosphorylation , Protein Binding , Sequence Homology, Amino Acid
11.
J Biol Chem ; 287(53): 44619-27, 2012 Dec 28.
Article in English | MEDLINE | ID: mdl-23115231

ABSTRACT

We describe the improvement of transglycosylation (TG) by chitinase D from Serratia proteamaculans (SpChiD). The SpChiD produced a smaller quantity of TG products for up to 90 min with 2 mm chitotetraose as the substrate and subsequently produced only hydrolytic products. Of the five residues targeted at the catalytic center, E159D resulted in substantial loss of both hydrolytic and TG activities. Y160A resulted in a product profile similar to SpChiD and a rapid turnover of substrate with slightly increased TG activity. The rest of the three mutants, M226A, Y228A, and R284A, displayed improved TG and decreased hydrolytic ability. Four of the five amino acid substitutions, F64W, F125A, G119S, and S116G, at the catalytic groove increased TG activity, whereas W120A completely lost the TG activity with a concomitant increase in hydrolysis. Mutation of Trp-247 at the solvent-accessible region significantly reduced the hydrolytic activity with increased TG activity. The mutants M226A, Y228A, F125A, S116G, F64W, G119S, R284A, and W247A accumulated approximately double the concentration of TG products like chitopentaose and chitohexaose, compared with SpChiD. The double mutant E159D/F64W regained the activity with accumulation of 6.0% chitopentaose at 6 h, similar to SpChiD at 30 min. Loss of chitobiase activity was unique to Y228A. Substitution of amino acids at the catalytic center and/or groove substantially improved the TG activity of SpChiD, both in terms of the quantity of TG products produced and the extended duration of TG activity.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Chitinases/chemistry , Chitinases/metabolism , Serratia/enzymology , Amino Acid Substitution , Bacterial Proteins/genetics , Chitinases/genetics , Glycosylation , Hydrolysis , Kinetics , Models, Molecular , Oligosaccharides/metabolism , Protein Binding , Serratia/chemistry , Serratia/genetics , Substrate Specificity
12.
Chem Biol Drug Des ; 79(1): 84-91, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21967481

ABSTRACT

MEK-1 and MEK-2 are dual-specificity kinases and important components in the mitogen-activated protein kinase pathway. These enzymes are crucial for normal cell survival and are also expressed in several types of cancers, making them important targets for drug design. We have applied an integrated in silico approach that combines comparative molecular field analysis, comparative molecular similarity indices analysis, and molecular docking to study the structural determinants for the recognition of substituted isothiazole analogs as allosteric inhibitors against MEK-1 kinase. The best 3D-QSAR models for comparative molecular field analysis and comparative molecular similarity indices analysis were selected based on statistical parameters. 3D contour maps suggested that bulky or long-chain substitutions at the X position on the core part decrease the inhibitory activity, and the presence of a hydrogen bond donor substitution enhances the activity. The bulky and electronegative substitutions at the Y position on the core part enhance the activity of the inhibitors. Molecular docking studies reveal a large and hydrophobic pocket that accommodates the Y substitution and a polar pocket that accommodates substitutions on the X position and forms hydrogen bonding interactions with MEK-1 kinase. The results of the 3D-QSAR analysis corroborate with the molecular docking results, and our findings will serve as a basis for further development of better allosteric inhibitors of MEK-1 kinase against several cancers.


Subject(s)
MAP Kinase Kinase Kinase 1/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Quantitative Structure-Activity Relationship , Thiazoles/chemistry , Allosteric Regulation , Computer Simulation , Enzyme Activation/drug effects , MAP Kinase Kinase Kinase 1/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Structure, Tertiary , Software
13.
J Mol Model ; 18(4): 1611-24, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21805127

ABSTRACT

The 3-D structure of the human mTOR kinase domain was modeled based on the crystal structure of PI3Kγ using comparative modeling methods, and the ATP-binding site of mTOR was characterized. The mTOR kinase 3-D model structure is similar to the structure of the PI3Kγ kinase domain, and exhibits great similarity to PI3Kγ at the active site of the kinase. Pharmacophore generation, the docking of mTOR inhibitors, and molecular dynamics (MD) simulations of mTOR-inhibitor docked complexes were carried out in this work. The best pharmacophore model generated from 27 ATP-competitive mTOR inhibitors comprised two hydrogen-bond acceptors, one aromatic ring, and one hydrophobic feature. These 27 inhibitors were docked into the ATP-binding site comprising the DFG motif, and the interactions in each protein-inhibitor complex were characterized. Mapping the pharmacophore model onto the docked inhibitors explained the specificity of the features of the pharmacophore and how they were arranged inside the active site of mTOR kinase. MD studies revealed important structural features, such as the large hydrophobic pocket "HP" and hydrophilic pocket "A1," and the solvent-exposed hydrophilic pocket "A2" at the active site of mTOR. Our results provide structural models of mTOR-inhibitor complexes and clues that should aid in the design of better mTOR kinase inhibitors.


Subject(s)
TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/chemistry , Catalytic Domain , Drug Design , Humans , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Molecular Dynamics Simulation , Phosphatidylinositol 3-Kinases/chemistry , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Protein Kinase Inhibitors/chemistry , Protein Structure, Quaternary , Quantitative Structure-Activity Relationship , TOR Serine-Threonine Kinases/metabolism
14.
Bioorg Med Chem Lett ; 21(23): 7219-23, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-22018459

ABSTRACT

The three dimensional-quantitative structure activity relationship (3D-QSAR) studies were performed on a series of falcipain-3 inhibitors using comparative molecular field analysis (CoMFA) and comparative molecular similarity indices analysis (CoMSIA) techniques. A training set containing 42 molecules served to establish the QSAR models. The optimum CoMFA and CoMSIA models obtained for the training set were statistically significant with cross-validated correlation coefficients r(cv)(2) (q(2)) of 0.549 and 0.608, and conventional correlation coefficients (r(2)) of 0.976 and 0.932, respectively. An independent test set of 12 molecules validated the external predictive power of both models with predicted correlation coefficients (r(pred)(2)) for CoMFA and CoMSIA as 0.697 and 0.509, respectively. The docking of inhibitors into falcipain-3 active site using GOLD software revealed the vital interactions and binding conformation of the inhibitors. The CoMFA and CoMSIA field contour maps agree well with the structural characteristics of the binding pocket of falcipain-3 active site, which suggests that the information rendered by 3D-QSAR models and the docking interactions can provide guidelines for the development of improved falcipain-3 inhibitors.


Subject(s)
Cysteine Endopeptidases/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Nitriles/chemistry , Pyrimidines/chemistry , Catalytic Domain , Enzyme Activation/drug effects , Models, Molecular , Nitriles/pharmacology , Pyrimidines/pharmacology , Quantitative Structure-Activity Relationship
15.
PLoS One ; 6(2): e16745, 2011 Feb 10.
Article in English | MEDLINE | ID: mdl-21347309

ABSTRACT

The PE and PPE proteins first reported in the genome sequence of Mycobacterium tuberculosis strain H37Rv are now identified in all mycobacterial species. The PE-PPE domain (Pfam ID: PF08237) is a 225 amino acid residue conserved region located towards the C-terminus of some PE and PPE proteins and hypothetical proteins. Our in-silico sequence analysis revealed that this domain is present in all Mycobacteria, some Rhodococcus and Nocardia farcinica genomes. This domain comprises a pentapeptide sequence motif GxSxG/S at the N-terminus and conserved amino acid residues Ser, Asp and His that constitute a catalytic triad characteristic of lipase, esterase and cutinase activity. The fold prediction and comparative modeling of the 3-D structure of the PE-PPE domain revealed a "serine α/ß hydrolase" structure with a central ß-sheet flanked by α-helices on either side. The structure comprises a lid insertion with a closed structure conformation and has a solvent inaccessible active site. The oxyanion hole that stabilizes the negative charge on the tetrahedral intermediate has been identified. Our findings add to the growing list of serine hydrolases in mycobacterium, which are essential for the maintenance of their impermeable cell wall and virulence. These results provide the directions for the design of experiments to establish the function of PE and PPE proteins.


Subject(s)
Catalytic Domain , Computational Biology , Hydrolases/chemistry , Hydrolases/metabolism , Mycobacterium tuberculosis/enzymology , Conserved Sequence , Models, Molecular
16.
Protein Pept Lett ; 18(6): 634-41, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21342099

ABSTRACT

The virulent form of malaria is caused by Plasmodium falciparum that infects red blood cells. In order to survive inside the host, the parasite remodels the infected erythrocytes by exporting more than 300 effector proteins outside the parasitophorous vacuole membrane into the cytosol. The main feature of all the export proteins is the presence of a pentapeptide sequence motif; RxLxE/Q/D. This sequence motif is hydrolysed between L-x and the proteins with the acetylated new N-terminus xE/Q/D are exported. The enzyme responsible for this hydrolysis is plasmepsin V which is one of the ten aspartic proteases in P. falciparum. In order to understand the structural rationale for the specificity of this protease towards cleavage of the above motif, we generated three-dimensional models of seven plasmepsins (I, V to X) for which experimental structures are not available and compared these along with the crystal structures of three P. falciparum plasmepsins (II to IV). The structure comparisons revealed the importance of Tyr13, Glu77 and Ala117 specific to plasmepsin V that facilitates the accommodation of arginine at P3 in the RxLxE/Q/D motif. Our analysis correlates the structure-function relationship of plasmepsin V.


Subject(s)
Arginine/metabolism , Aspartic Acid Endopeptidases/chemistry , Aspartic Acid Endopeptidases/metabolism , Plasmodium falciparum/enzymology , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Models, Molecular , Molecular Sequence Data , Mutation , Pepstatins/genetics , Pepstatins/metabolism , Protein Binding , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...