Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
PLoS Pathog ; 19(2): e1011196, 2023 02.
Article in English | MEDLINE | ID: mdl-36827451

ABSTRACT

The Omicron variant of SARS-CoV-2 is capable of infecting unvaccinated, vaccinated and previously-infected individuals due to its ability to evade neutralization by antibodies. With multiple sub-lineages of Omicron emerging in the last 12 months, there is inadequate information on the quantitative antibody response generated upon natural infection with Omicron variant and whether these antibodies offer cross-protection against other sub-lineages of Omicron variant. In this study, we characterized the growth kinetics of Kappa, Delta and Omicron variants of SARS-CoV-2 in Calu-3 cells. Relatively higher amounts infectious virus titers, cytopathic effect and disruption of epithelial barrier functions was observed with Delta variant whereas infection with Omicron sub-lineages led to a more robust induction of interferon pathway, lower level of virus replication and mild effect on epithelial barrier. The replication kinetics of BA.1, BA.2 and BA.2.75 sub-lineages of the Omicron variant were comparable in cell culture and natural infection in a subset of individuals led to a significant increase in binding and neutralizing antibodies to the Delta variant and all the three sub-lineages of Omicron but the level of neutralizing antibodies were lowest against the BA.2.75 variant. Finally, we show that Cu2+, Zn2+ and Fe2+ salts inhibited in vitro RdRp activity but only Cu2+ and Fe2+ inhibited both the Delta and Omicron variants in cell culture. Thus, our results suggest that high levels of interferons induced upon infection with Omicron variant may counter virus replication and spread. Waning neutralizing antibody titers rendered subjects susceptible to infection by Omicron variants and natural Omicron infection elicits neutralizing antibodies that can cross-react with other sub-lineages of Omicron and other variants of concern.


Subject(s)
COVID-19 , Humans , Broadly Neutralizing Antibodies , Kinetics , SARS-CoV-2/genetics , Antibodies, Neutralizing , Interferons/genetics , Antibodies, Viral
2.
J Biol Inorg Chem ; 26(1): 149-159, 2021 02.
Article in English | MEDLINE | ID: mdl-33427997

ABSTRACT

Lactoperoxidase, a heme-containing glycoprotein, catalyzes the oxidation of thiocyanate by hydrogen peroxide into hypothiocyanite which acts as an antibacterial agent. The prosthetic heme moiety is attached to the protein through two ester linkages via Glu258 and Asp108. In lactoperoxidase, the substrate-binding site is formed on the distal heme side. To study the effect of physiologically important potassium ion on the structure and function of lactoperoxidase, the fresh protein samples were isolated from yak (Bos grunniens) colostrum and purified to homogeneity. The biochemical studies with potassium fluoride showed a significant reduction in the catalytic activity. Lactoperoxidase was crystallized using 200 mM ammonium nitrate and 20% PEG-3350 at pH 6.0. The crystals of LPO were soaked in the solution of potassium fluoride and used for the X-ray intensity data collection. Structure determination at 2.20 Å resolution revealed the presence of a potassium ion in the distal heme cavity. Structure determination further revealed that the propionic chain attached to pyrrole ring C of the heme moiety, was disordered into two components each having an occupancy of 0.5. One component occupied a position similar to the normally observed position of propionic chain while the second component was found in the distal heme cavity. The potassium ion in the distal heme cavity formed five coordinate bonds with two oxygen atoms of propionic moiety, Nε2 atom of His109 and two oxygen atoms of water molecules. The presence of potassium ion in the distal heme cavity hampered the catalytic activity of lactoperoxidase.


Subject(s)
Lactoperoxidase/metabolism , Potassium/metabolism , Animals , Binding Sites , Biocatalysis , Calcium/chemistry , Calcium/metabolism , Cattle , Colostrum/enzymology , Crystallography, X-Ray , Heme/chemistry , Heme/metabolism , Hydrogen Peroxide/chemistry , Hydrogen Peroxide/metabolism , Lactoperoxidase/chemistry , Potassium/chemistry , Protein Binding
3.
Protein J ; 40(1): 8-18, 2021 02.
Article in English | MEDLINE | ID: mdl-33389415

ABSTRACT

Lactoperoxidase (LPO) is a heme containing oxido-reductase enzyme. It is secreted from mammary, salivary, lachrymal and mucosal glands. It catalyses the conversion of thiocyanate into hypothiocyanate and halides into hypohalides. LPO belongs to the superfamily of mammalian heme peroxidases which also includes myeloperoxidase (MPO), eosinophil peroxidase (EPO) and thyroid peroxidase (TPO). The heme prosthetic group is covalently linked in LPO through two ester bonds involving conserved residues Glu258 and Asp108. It was isolated from colostrum of yak (Bos grunniens), purified to homogeneity and crystallized using ammonium iodide as a precipitating agent. The crystals belonged to monoclinic space group P21 with cell dimensions of a = 53.91 Å, b = 78.98 Å, c = 67.82 Å and ß = 92.96°. The structure was determined at 1.55 Å resolution. This is the first structure of LPO from yak. Also, this is the highest resolution structure of LPO determined so far from any source. The structure determination revealed that three segments (Ser1-Cys15), (Thr117-Asn138) and (Cys167-Leu175) were disordered and formed one surface of LPO structure. In the substrate binding site, the iodide ions were observed in three subsites which are formed by (1) heme moiety and residues, Gln105, Asp108, His109, Phe113, Arg255, Glu258, Phe380 and Phe381, (2) residues, Asn230, Lys232, Pro236, Cys248, Phe254, Phe381 and Pro424 and (3) residues, Ser198, Leu199 and Arg202. The structure determination also revealed that the side chain of Phe254 was disordered. It was observed to adopt two conformations in the structures of LPO.


Subject(s)
Amino Acids/chemistry , Ammonium Compounds/chemistry , Heme/chemistry , Hydrogen Peroxide/chemistry , Lactoperoxidase/chemistry , Amino Acids/metabolism , Ammonium Compounds/metabolism , Animals , Binding Sites , Cattle , Colostrum/chemistry , Crystallization , Crystallography, X-Ray , Female , Gene Expression , Heme/metabolism , Hydrogen Peroxide/metabolism , Lactoperoxidase/genetics , Lactoperoxidase/metabolism , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Substrate Specificity
4.
Protein Pept Lett ; 28(4): 450-458, 2021.
Article in English | MEDLINE | ID: mdl-32798368

ABSTRACT

BACKGROUND: Type-III Pantothenate kinase from the multi drug resistant bacteria, Acinetobacter baumannii (AbPanK) catalyzes the first step of the essential Coenzyme A biosynthesis pathway. AbPanK is an attractive drug target against the bacteria since it is an essential enzyme and its structure is significantly different from the human PanK. METHODS: AbPanK was cloned, expressed, purified and crystallized. A good quality single crystal was used for X-ray intensity data collection. Dynamic light scattering was done for calculating the hydrodynamic radii and its oligomeric nature in the solution. Binding studies of this protein with its two substrates, Pantothenate and ATP were done using spectrofluorometer. RESULTS: Our results indicated that AbPanK shows a strong affinity with pantothenate with dissociation constant of 1.2 x 10- 8 M and moderate affinity towards ATP of 3.7x 10-3 M. This fact was further substantiated by the calculations of Km of both substrates using kinase assay kit. Dynamic light scattering studies have shown that it exists as homogenous solution with hydrodynamic radii corresponding to the molecular weight of 29.55 kDa. A low-resolution X-ray intensity data set was collected, which shows that AbPank crystallizes in P2 space group with cell dimensions of a= 165 Å, b= 260 Å, and, c= 197 Å and α= 90.0, ß= 113.60, γ= 90.0. DISCUSSION: Recombinant Pantothenate kinase from Acinetobacter baumannii was purified to homogeneity and crystallized. The enzyme exhibits very low sequence identity (28%) to other corresponding enzymes. CONCLUSION: The recombinant enzyme was active and its binding affinities with its substrates pantothenate and ATP have been studied. This information would be very useful while designing the inhibitors of this enzyme in order to fight bacterial infections associated to this pathogen.


Subject(s)
Acinetobacter baumannii/enzymology , Adenosine Triphosphate/chemistry , Bacterial Proteins/chemistry , Pantothenic Acid/chemistry , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Acinetobacter baumannii/genetics , Bacterial Proteins/genetics , Crystallography, X-Ray , Phosphotransferases (Alcohol Group Acceptor)/genetics , Protein Domains
5.
ACS Omega ; 5(51): 33151-33161, 2020 Dec 29.
Article in English | MEDLINE | ID: mdl-33398250

ABSTRACT

The recent pandemic caused by SARS-CoV-2 has led the world to a standstill, causing a medical and economic crisis worldwide. This crisis has triggered an urgent need to discover a possible treatment strategy against this novel virus using already-approved drugs. The main protease (Mpro) of this virus plays a critical role in cleaving the translated polypeptides that makes it a potential drug target against COVID-19. Taking advantage of the recently discovered three-dimensional structure of Mpro, we screened approved drugs from the Drug Bank to find a possible inhibitor against Mpro using computational methods and further validating them with biochemical studies. The docking and molecular dynamics study revealed that DB04983 (denufosol) showed the best glide docking score, -11.884 kcal/mol, and MM-PBSA binding free energy, -10.96 kcal/mol. Cobicistat, cangrelor (previous computational studies in our lab), and denufosol (current study) were tested for the in vitro inhibitory effects on Mpro. The IC50 values of these drugs were ∼6.7 µM, 0.9 mM, and 1.3 mM, respectively, while the values of dissociation constants calculated using surface plasmon resonance were ∼2.1 µM, 0.7 mM, and 1.4 mM, respectively. We found that cobicistat is the most efficient inhibitor of Mpro both in silico and in vitro. In conclusion, cobicistat, which is already an FDA-approved drug being used against HIV, may serve as a good inhibitor against the main protease of SARS-CoV-2 that, in turn, can help in combating COVID-19, and these results can also form the basis for the rational structure-based drug design against COVID-19.

6.
ACS Comb Sci ; 18(2): 100-16, 2016 Feb 08.
Article in English | MEDLINE | ID: mdl-26812086

ABSTRACT

Mycobacterium tuberculosis (Mtb) infections are causing serious health concerns worldwide. Antituberculosis drug resistance threatens the current therapies and causes further need to develop effective antituberculosis therapy. GlmU represents an interesting target for developing novel Mtb drug candidates. It is a bifunctional acetyltransferase/uridyltransferase enzyme that catalyzes the biosynthesis of UDP-N-acetyl-glucosamine (UDP-GlcNAc) from glucosamine-1-phosphate (GlcN-1-P). UDP-GlcNAc is a substrate for the biosynthesis of lipopolysaccharide and peptidoglycan that are constituents of the bacterial cell wall. In the current study, structure and ligand based computational models were developed and rationally applied to screen a drug-like compound repository of 20,000 compounds procured from ChemBridge DIVERSet database for the identification of probable inhibitors of Mtb GlmU. The in vitro evaluation of the in silico identified inhibitor candidates resulted in the identification of 15 inhibitory leads of this target. Literature search of these leads through SciFinder and their similarity analysis with the PubChem training data set (AID 1376) revealed the structural novelty of these hits with respect to Mtb GlmU. IC50 of the most potent identified inhibitory lead (5810599) was found to be 9.018 ± 0.04 µM. Molecular dynamics (MD) simulation of this inhibitory lead (5810599) in complex with protein affirms the stability of the lead within the binding pocket and also emphasizes on the key interactive residues for further designing. Binding site analysis of the acetyltransferase pocket with respect to the identified structural moieties provides a thorough analysis for carrying out the lead optimization studies.


Subject(s)
Antitubercular Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Combinatorial Chemistry Techniques , Computer Simulation , Multienzyme Complexes/antagonists & inhibitors , Mycobacterium tuberculosis/metabolism , Antitubercular Agents/chemistry , Bacterial Proteins/metabolism , Drug Discovery , Gene Expression Regulation, Bacterial , High-Throughput Screening Assays , Ligands , Models, Molecular , Multienzyme Complexes/metabolism , Mycobacterium tuberculosis/genetics , Protein Conformation , Structure-Activity Relationship
7.
J Biomol Screen ; 21(4): 342-53, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26762501

ABSTRACT

Secondary metabolite of Aspergillus terreus, terreic acid, is a reported potent antibacterial that was identified more than 60 years ago, but its cellular target(s) are still unknown. Here we screen its activity against the acetyltransferase domain of a bifunctional enzyme, Escherichia coli N-acetylglucosamine-1-phosphate-uridyltransferase/glucosamine-1-phosphate-acetyltransferase (GlmU). An absorbance-based assay was used to screen terreic acid against the acetyltransferase activity of E. coli GlmU. Terreic acid was found to inhibit the acetyltransferase domain of E. coli GlmU with an IC50 of 44.24 ± 1.85 µM. Mode of inhibition studies revealed that terreic acid was competitive with AcCoA and uncompetitive with GlcN-1-P. It also exhibited concentration-dependent killing of E. coli ATCC 25922 up to 4× minimum inhibitory concentration and inhibited the growth of biofilms generated by E. coli. Characterization of resistant mutants established mutation in the acetyltransferase domain of GlmU. Terreic acid was also found to be metabolically stable in the in vitro incubations with rat liver microsome in the presence of a NADPH regenerating system. The studies reported here suggest that terreic acid is a potent antimicrobial agent and support that E. coli GlmU acetyltransferase is a molecular target of terreic acid, resulting in its antibacterial activity.


Subject(s)
Anti-Bacterial Agents/pharmacology , Aspergillus/chemistry , Biofilms/drug effects , Enzyme Inhibitors/pharmacology , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli/drug effects , Multienzyme Complexes/antagonists & inhibitors , Acetyl Coenzyme A/chemistry , Acetyl Coenzyme A/metabolism , Animals , Anti-Bacterial Agents/isolation & purification , Aspergillus/metabolism , Binding, Competitive , Biofilms/growth & development , Biological Assay , Enzyme Inhibitors/isolation & purification , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gene Expression , Glucosamine/analogs & derivatives , Glucosamine/chemistry , Glucosamine/metabolism , Glucose-6-Phosphate/analogs & derivatives , Glucose-6-Phosphate/chemistry , Glucose-6-Phosphate/metabolism , Microbial Sensitivity Tests , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Molecular Docking Simulation , Multienzyme Complexes/chemistry , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Mutation , Protein Binding/drug effects , Protein Domains , Quinones/isolation & purification , Quinones/pharmacology , Rats , Secondary Metabolism
8.
Eur J Pharm Sci ; 83: 62-70, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26690048

ABSTRACT

In the past five years, an alarming increase in the number of patients with multidrug resistant tuberculosis (MDR TB) and extensively drug-resistant tuberculosis (XDR TB) has been reported, particularly in Eastern Europe, Asia and Southern Africa. Current situation has challenged the control and treatment of tuberculosis (TB) which sparked an emergent need to find new anti-tubercular agents with different chemical scaffolds and mechanisms of action. A very fruitful way to identify novel anti-tubercular agents is the development of compounds that target the enzymes essentially required for the biosynthesis and assembly of the mycobacterial cell wall. Biosynthesis of uridine diphospho-N-acetylglucosamine (UDP-GlcNAc) represents one such pathway. Enzymes involved in UDP-GlcNAc biosynthesis have been predicted to be essential for Mycobacterium tuberculosis growth in vitro. It is a key precursor molecule of M. tuberculosis cell wall, being situated at the branched point of two essential biosynthetic pathways, namely peptidoglycan and a disaccharide linker, D-N-GlcNAc-1-rhamnose. This article provides a comprehensive overview of the present knowledge on the enzymes catalyzing the particular steps of the pathway in M. tuberculosis, with special emphasis put on N-acetylglucosamine-1-phosphate uridyltransferase (GlmU), a bifunctional enzyme, which catalyzes the last two steps of this pathway. It also gives an insight into the present knowledge about the inhibitors reported against the enzymes, which could be further used as chemical scaffold for the discovery of more potent anti-TB compounds.


Subject(s)
Antitubercular Agents/pharmacology , Mycobacterium tuberculosis/drug effects , Nucleotidyltransferases/antagonists & inhibitors , Animals , Drug Design , Humans , Mycobacterium tuberculosis/enzymology , Nucleotidyltransferases/metabolism , Signal Transduction
9.
Appl Microbiol Biotechnol ; 100(7): 3071-85, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26563552

ABSTRACT

This study aims at identifying novel chemical scaffolds as inhibitors specific to the acetyltransferase domain of a bifunctional enzyme, Escherichia coli GlmU, involved in the cell wall biosynthesis of Gram-negative organisms. A two-pronged approach was used to screen a 50,000 small-molecule library. Using the first approach, the library was in silico screened by docking the library against acetyltransferase domain of E. coli GlmU studies. In the second approach, complete library was screened against Escherichia coli ATCC 25922 to identify the whole cell active compounds. Active compounds from both the screens were screened in a colorimetric absorbance-based assay to identify inhibitors of acetyltransferase domain of E. coli GlmU which resulted in the identification of 1 inhibitor out of 56 hits identified by in silico screening and 4 inhibitors out of 35 whole cell active compounds on Gram-negative bacteria with the most potent inhibitor showing IC50 of 1.40 ± 0.69 µM. Mode of inhibition studies revealed these inhibitors to be competitive with AcCoA and uncompetitive with GlcN-1-P. These selected inhibitors were also tested for their antibacterial and cytotoxic activities. Compounds 5175178 and 5215319 exhibited antibacterial activity that co-related with GlmU inhibition. These compounds, therefore, represent novel chemical scaffolds targeting acetyltransferase activity of E. coli GlmU.


Subject(s)
Anti-Bacterial Agents/pharmacology , Enzyme Inhibitors/pharmacology , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli/drug effects , Multienzyme Complexes/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Acetyl Coenzyme A/chemistry , Acetyl Coenzyme A/metabolism , Acetylglucosamine/analogs & derivatives , Acetylglucosamine/chemistry , Acetylglucosamine/metabolism , Anti-Bacterial Agents/chemistry , Binding, Competitive , Cell Wall/chemistry , Cell Wall/drug effects , Cell Wall/metabolism , Enzyme Inhibitors/chemistry , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gene Expression , High-Throughput Screening Assays , Kinetics , Microbial Sensitivity Tests , Molecular Docking Simulation , Multienzyme Complexes/chemistry , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Protein Binding , Small Molecule Libraries/chemistry , Structure-Activity Relationship
10.
Tuberculosis (Edinb) ; 95(6): 664-677, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26318557

ABSTRACT

N-acetylglucosamine-1-phosphate uridyltransferase (GlmU) is a pivotal bifunctional enzyme, its N and C terminal domains catalyzes uridyltransferase and acetyltransferase activities, respectively. Final product of GlmU catalyzed reaction, uridine-diphospho-N-acetylglucosamine (UDP-GlcNAc), acts as sugar donor providing GlcNAc residues in the synthesis of peptidoglycan and a disaccharide linker (D-N-GlcNAc-1-rhamnose), the key structural components of Mycobacterium tuberculosis (M. tuberculosis) cell wall. In the present study, we have searched new inhibitors against acetyltransferase activity of M. tuberculosis GlmU. A subset of 1607 synthetic compounds, selected through dual approach i.e., in-silico and whole cell screen against 20,000 compounds from ChemBridge library, was further screened using an in-vitro high throughput bioassay to identify inhibitors of acetyltransferase domain of M. tuberculosis GlmU. Four compounds were found to inhibit GlmU enzyme specific to acetyltransferase activity, with IC50 values ranging from 9 to 70 µM. Two compounds (6624116, 5655606) also exhibited whole cell activity against drug susceptible as well as drug resistant M. tuberculosis. These two compounds also exhibited increased anti-TB activity when tested in combination with rifampicin, isoniazid and ethambutol, however 5655606 was cytotoxic to eukaryotic cell line. These results demonstrate that identified chemical scaffolds can be used as inhibitors of M. tuberculosis cell wall enzyme after optimizations for future anti-TB drug development program.


Subject(s)
Antitubercular Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Drug Discovery/methods , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays , Multienzyme Complexes/antagonists & inhibitors , Mycobacterium tuberculosis/drug effects , Antitubercular Agents/chemistry , Antitubercular Agents/toxicity , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Resistance, Bacterial , Drug Therapy, Combination , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/toxicity , Glucosamine/analogs & derivatives , Glucosamine/metabolism , Glucosephosphates/metabolism , Hep G2 Cells , Humans , Kinetics , Microbial Sensitivity Tests , Molecular Docking Simulation , Molecular Structure , Molecular Targeted Therapy , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Mutation , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/growth & development , Structure-Activity Relationship
11.
Bioorg Med Chem Lett ; 23(24): 6842-6, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24206766

ABSTRACT

A series of 5-nitrofuran-triazole conjugates were synthesized and evaluated for their antimicrobial activity against both Gram-positive and Gram-negative bacterial strains. All the compounds exhibited promising inhibition towards Gram-positive pathogenic strains, while mild inhibitory effects were observed towards Gram-negative bacterial strains. Some of the compounds 8a, 8b, 8e, 8f, 8h are most active among the series exhibiting MIC value of 1.17 µg/ml against different bacterial strains. The bactericidal activity is found to be in accordance with the bacterial growth inhibition data. Compound 8e was found to be equipotent to the standard drug Ciprofloxacin displaying MBC value of 1.17 µg/ml against the bacterial strain Bacillus subtilis. The compounds have also demonstrated promising antibacterial activity against the resistant strain MRSA and were found to be effective inhibitors of biofilm formation. The compound 8b exhibited excellent anti-biofilm activity with IC50 value as low as 0.8 µg/ml. These conjugates were also screened for antitubercular activity against Mycobacterium tuberculosis H37Rv strain. Compound 8e showed promising antitubercular activity with MIC value of 0.25 µg/ml. Most of these compounds are less toxic to normal mammalian cells than the widely used antibacterial drug Ciprofloxacin.


Subject(s)
Antitubercular Agents/chemical synthesis , Antitubercular Agents/pharmacology , Mycobacterium tuberculosis/drug effects , Nitrofurans/chemistry , Triazoles/chemistry , Triazoles/pharmacology , Animals , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/toxicity , Cell Line , Cell Survival/drug effects , Chlorocebus aethiops , Crystallography, X-Ray , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Humans , Microbial Sensitivity Tests , Molecular Conformation , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...