Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Biomed Pharmacother ; 177: 117043, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38941896

ABSTRACT

This study investigated the chemical constituents, antioxidant potential, and in vitro and in silico antidiabetic activity of Gymnema sylvestre. Column chromatography and spectroscopic techniques identified twelve compounds from the methanol extract, including 4 sterols (1-4), 5 triterpenoids (5-9), and 3 flavonoids (10-12). The chemophenetic significance of all compounds was also investigated. The antioxidant capacity of the extract and compounds (1-4) was evaluated using FRAP and DPPH assays. The extract exhibited strong free radical scavenging activity (IC50 = 48.34 µg/mL), while compounds (1-4) displayed varying degrees of efficacy (IC50 = 98.30-286.13 µg/mL). The FRAP assay indicated significant reducing power for both extract and compounds (58.54, 47.61, 56.61, and 49.11 mg Eq.VitC/g for extract and compounds 1 & 2, 3, and 4, respectively). The antidiabetic potential was assessed through α-amylase and α-glucosidase enzyme inhibition assays. The crude extract demonstrated the most potent inhibition (IC50 = 218.46 and 57.42 µg/mL for α-glucosidase and α-amylase respectively) suggesting its potential for managing postprandial hyperglycaemia. In silico studies employed molecular docking and dynamics simulations to elucidate the interactions between identified compounds and α-amylase/α-glucosidase enzymes. The results revealed promising binding affinities between the compounds and target enzymes, with compound 6 demonstrating the highest predicted inhibitory activity with -10 kcal/mol and -9.1 kcal/mol for α-amylase and α-glucosidase, respectively. This study highlights the presence of diverse bioactive compounds in Gymnema sylvestre. The extract exhibits antioxidant properties and inhibits carbohydrate-digesting enzymes, suggesting its potential as a complementary therapeutic approach for managing hyperglycaemia associated with type 2 diabetes.

2.
Photochem Photobiol ; 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38943225

ABSTRACT

Elevated global pollution level is the prime reason that contributes to the onset of various harmful health diseases. The products of Biginelli reaction are enormously used in the pharmaceutical industry as they have antiviral, antibacterial, and calcium channel modulation abilities. This work reports a novel eosin Y sensitized boron graphitic carbon nitride (EY-Ben-g-C3N4) as a photocatalyst that efficiently produced 3,4-dihydropyrimidine-2-(1H)-one by the Biginelli reaction of benzaldehyde, urea, and methyl acetoacetate. The photocatalyst EY-Ben-g-C3N4 showed a successful generation of 3,4-dihydropyrimidine-2-(1H)-one (Biginelli product) in good yield via photocatalysis which is an eco-friendly method and has facile operational process. In addition to the production of Biginelli products, the photocatalyst also showed a remarkable NADH regeneration of 81.18%. The incorporation of g-C3N4 with boron helps increase the surface area and the incorporation of eosin Y which is an inexpensive and non-toxic dye, and in Ben-g-C3N4, enhanced the light-harvesting capacity of the photocatalyst. The production of 3,4-dihydropyrimidine-2-(1H)-one and NADH by the EY-Ben-g-C3N4 photocatalyst is attributed to the requisite band gap, high molar absorbance, low rate of charge recombination, and increased capacity of the photocatalyst to harvest solar light energy.

3.
Int J Biol Macromol ; 274(Pt 2): 133231, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38897495

ABSTRACT

Trypsin is a serine protease, an important digestive enzyme that digests the proteins in the small intestine. In the present study, we have investigated the interaction of safranal, a major saffron metabolite, with trypsin using spectroscopic and molecular docking analyses. Fluorescence emission spectra of trypsin were largely affected by the inner filter effect from safranal; that's why these were corrected using the standard procedure. The corrected fluorescence spectra have shown that the safranal quenched the intrinsic fluorescence of trypsin with a blue shift in the wavelength of emission maximum, which revealed that the microenvironment of the fluorophore became more hydrophobic. There was approximately 1: 1 fair binding between them, which increased with a rise in temperature. The interaction was favored, principally, by hydrophobic forces, and there was an efficient energy transfer from the fluorophore to the safranal. Synchronous fluorescence spectra suggested that the tryptophan residues were the major ones taking part in the fluorescence quenching of trypsin. Safranal also influenced the secondary structure of trypsin and caused partial unfolding. Molecular Docking and the Molecular Dynamics simulation of the free and complexed trypsin was also carried out. Safranal formed a stable, non-covalent complex within the S2'-S5' subsite. Moreover, two nearby tyrosine residues (Tyr39 and Tyr151) stabilized safranal through π-π interactions. Additionally, the presence of safranal led to changes in the protein flexibility and compactness, which could indicate changes in the surrounding of tryptophan residues, impacting their fluorescence. Furthermore, a loss in compactness is in line with the partial unfolding observed experimentally. Thus, both experimental and computational studies were in good agreement with each other.

4.
Heliyon ; 10(10): e30818, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38784535

ABSTRACT

Warfarin is a cardiovascular drug, used to treat or inhibit the coagulation of the blood. In this paper, we have studied the interaction of lysozyme with warfarin using several experimental (fluorescence, UV-visible and circular dichroism spectroscopies) and computational (molecular docking, molecular dynamics and DFT) approaches. Experimental studies have suggested that there was a strong interaction between lysozyme and warfarin. Inner filter effect played important role in fluorescence experimental data which show that the emission intensity of lysozyme decreased on the addition of warfarin, however, after inner filter effect correction the actual outcome turned out be the fluorescence enhancement. The extent of binding, increased with temperature rise. The interaction was primarily taken place via the dominance of hydrophobic forces. Small amount of warfarin didn't influence the secondary structure of lysozyme; however, the higher concentration of warfarin caused a decrease in the helicity of the protein and a consequent partial unfolding. Molecular docking studies were also performed which revealed that warfarin binds with lysozyme mainly with hydrophobic forces along with a significant contribution of hydrogen bonding. The flexibility of warfarin played important role in fitting the molecule into the binding pocket of lysozyme. Frontier molecular orbitals of warfarin, using DFT, in free as well as complexed form have also been calculated and discussed. Molecular dynamics simulations of unbound and warfarin bound lysozyme reveal a stable complex with slightly higher RMSD values in the presence of warfarin. Despite slightly increased RMSF values, the overall compactness and folding properties remain consistent, emphasizing strong binding towards lysozyme through the results obtained from intermolecular hydrogen bonding analysis. Essential dynamics analysis suggests warfarin induces slight structural changes without significantly altering the conformation, additionally supported by SASA patterns. Aside from the examination of global and essential motion, the MM/PBSA-based analysis of binding free energy elucidates the significant binding of warfarin to lysozyme, indicating a binding free energy of -13.3471 kcal/mol.

5.
J Fluoresc ; 2023 Sep 04.
Article in English | MEDLINE | ID: mdl-37665513

ABSTRACT

Interaction of diclofenac and indomethacin with lysozyme was studied using several spectroscopic and molecular docking methods. Difference UV-visible spectra showed that the absorption profile of lysozyme changed when both diclofenac and indomethacin were mixed with the former. The sequential addition of both drugs to the lysozyme solution caused the decrease of the intrinsic fluorescence of the latter, however, when the data were corrected for inner filter effect, an enhancement in the fluorescence of lysozyme was detected. Accordingly, the fluorescence enhancement data were analyzed using Benesi-Hildebrand equation. Both, diclofenac and indomethacin showed good interaction with lysozyme, although, the association constants of indomethacin were nearly two-fold higher as compared to that of diclofenac. The binding was slightly more spontaneous in case of indomethacin and the major forces involved in the binding of both drugs with lysozyme were hydrogen bonding and hydrophobic interactions. Secondary structural analysis revealed that both drugs partially unfolded lysozyme. Results obtained through molecular docking were also in good agreement with the experimental outcomes. Both, diclofenac and indomethacin, are bounded at the same site inside lysozyme which is located in the big hydrophobic cavity of the protein.

6.
Int J Biol Macromol ; 252: 126568, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37640184

ABSTRACT

The interaction of lysozyme with cefoperazone was studied by means of spectroscopic and computational approaches. The change in the UV-visible spectrum of lysozyme in presence of cefoperazone was an indication of the complex formation between them. Fluorescence spectroscopy suggested that there was a fair interaction between the protein and drug which was taken place via dynamic quenching mechanism and the binding ratio was approximately 1:1. The binding was energetically feasible and principally supported by the hydrophobic forces. CD spectroscopic studies have shown that cefoperazone induced the secondary structure of lysozyme by increasing the α-helical contents of the latter. In silico studies revealed that the large nonpolar cavity was the preferred binding site of cefoperazone within lysozyme and the interaction was taken place mainly through hydrophobic forces with small involvement of hydrogen bonding and electrostatic interactions which is in good agreement with the experimental analyses. Effect of paracetamol was also seen on the binding and it was found that paracetamol had a negative influence on the binding between cefoperazone and lysozyme.


Subject(s)
Acetaminophen , Cefoperazone , Cefoperazone/pharmacology , Acetaminophen/pharmacology , Circular Dichroism , Muramidase/chemistry , Cephalosporins , Molecular Docking Simulation , Thermodynamics , Binding Sites , Spectrometry, Fluorescence , Protein Binding
7.
Int J Biol Macromol ; 242(Pt 2): 124844, 2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37210056

ABSTRACT

This paper describes an inclusive biophysical study elucidating the interaction of therapeutic drug azithromycin (Azith) with hen egg white lysozyme (HEWL). Spectroscopic and computational tools have been employed to study the interaction of Azith with HEWL at pH 7.4. The fluorescence quenching constant values (Ksv) exhibited a decrease with the increase in temperature which revealed the occurrence of static quenching mechanism between Azith and HEWL. The thermodynamic data demonstrated that hydrophobic interactions were predominantly involved in the Azith-HEWL interaction. The negative value of standard Gibbs free energy (ΔG°) stated that the Azith-HEWL complex formed via spontaneous molecular interactions. The effect of sodium dodecyl sulfate (SDS) surfactant monomers on the binding propensity of Azith with HEWL was insignificant at lower concentrations however the binding significantly decreased at increased concentrations of the former. Far-UV CD data revealed alteration in the secondary structure of HEWL in the presence of Azith and the overall HEWL conformation changed. Molecular docking results revealed that the binding of Azith with HEWL takes place through hydrophobic interactions and hydrogen bonds.


Subject(s)
Azithromycin , Muramidase , Animals , Sodium Dodecyl Sulfate , Molecular Docking Simulation , Azithromycin/pharmacology , Protein Binding , Muramidase/chemistry , Chickens/metabolism
8.
Molecules ; 28(7)2023 Mar 27.
Article in English | MEDLINE | ID: mdl-37049745

ABSTRACT

The interaction of indomethacin with human serum albumin (HSA) has been studied here considering the primary and secondary binding sites. The Stern-Volmer plots were linear in the lower concentration range of indomethacin while a downward curvature was observed in the higher concentration range, suggesting the presence of more than one binding site for indomethacin inside HSA due to which the microenvironment of the fluorophore changed slightly and some of its fraction was not accessible to the quencher. The Stern-Volmer quenching constants (KSV) for the primary and secondary sites were calculated from the two linear portions of the Stern-Volmer plots. There was around a two-fold decrease in the quenching constants for the low-affinity site as compared to the primary binding site. The interaction takes place via a static quenching mechanism and the KSV decreases at both primary and secondary sites upon increasing the temperature. The binding constants were also evaluated, which show strong binding at the primary site and fair binding at the secondary site. The binding was thermodynamically favorable with the liberation of heat and the ordering of the system. In principle, hydrogen bonding and Van der Waals forces were involved in the binding at the primary site while the low-affinity site interacted through hydrophobic forces only. The competitive binding was also evaluated using warfarin, ibuprofen, hemin, and a warfarin + hemin combination as site markers. The binding profile remained unchanged in the presence of ibuprofen, whereas it decreased in the presence of both warfarin and hemin with a straight line in the Stern-Volmer plots. The reduction in the binding was at a maximum when both warfarin and hemin were present simultaneously with the downward curvature in the Stern-Volmer plots at higher concentrations of indomethacin. The secondary structure of HSA also changes slightly in the presence of higher concentrations of indomethacin. Molecular dynamics simulations were performed at the primary and secondary binding sites of HSA which are drug site 1 (located in the subdomain IIA of the protein) and the hemin binding site (located in subdomain IB), respectively. From the results obtained from molecular docking and MD simulation, the indomethacin molecule showed more binding affinity towards drug site 1 followed by the other two sites.


Subject(s)
Indomethacin , Serum Albumin, Human , Humans , Serum Albumin, Human/chemistry , Molecular Docking Simulation , Protein Binding , Ibuprofen , Warfarin , Hemin/metabolism , Binding Sites , Thermodynamics , Spectrometry, Fluorescence , Circular Dichroism
9.
Front Mol Biosci ; 10: 1327740, 2023.
Article in English | MEDLINE | ID: mdl-38187092

ABSTRACT

Digitoxin is a cardiac glycoside used to treat heart failure and heart arrhythmia. However, its therapeutic concentration range is very narrow. High doses of digitoxin are associated with severe side effects; therefore, it is necessary to develop the delivery system which can control the plasma levels of it. In this context, the binding of lysozyme, an important protein having many applications, with digitoxin has been studied to see the ability of the former as a carrier. The studies were carried out using both experimental and computational methods. The intrinsic fluorescence of lysozyme increased on the addition of digitoxin. Fluorescence results suggested that there was a strong interaction between lysozyme and digitoxin which was favored, mainly, by hydrophobic forces. Further, digitoxin affected the secondary structure of lysozyme slightly by causing the partial unfolding of lysozyme. The preferred binding site of digitoxin within lysozyme was the large cavity of the protein. Molecular docking studies also established the principal role of hydrophobic forces in the binding with a significant support of hydrogen bonding. Frontier molecular orbitals of free digitoxin and in complexation with lysozyme were also computed and discussed. The findings from molecular dynamics simulation studies elucidate that, when contrasted with the first and third conformations of the digitoxin-bound lysozyme complex, the second conformation promotes structural stability, reduces flexibility, and enhances the compactness and folding properties of lysozyme. The overall study shows that lysozyme could act as a potential carrier for digitoxin in pharmaceutical formulations.

10.
Int J Mol Sci ; 23(8)2022 Apr 07.
Article in English | MEDLINE | ID: mdl-35456897

ABSTRACT

The interaction of the important plasma protein, human serum albumin (HSA), with two monoterpenes found in cumin oil, i.e., cuminaldehyde (4-isopropylbenzaldehyde) and cuminol (4-isopropylbenzyl alcohol), was studied in this paper. Both experimental and computational methods were utilized to understand the mechanism of binding. The UV absorption profile of HSA changes in the presence of both cuminaldehyde and cuminol, due to the interaction between HSA with both monoterpenes. The intrinsic fluorescence intensity of HSA was also quenched on the sequential addition of both ligands, due to change in the microenvironment of the fluorophore present in the former. Quenching of HSA by cuminaldehyde was much higher in comparison to that in the presence of cuminol. Fluorescence quenching data were analyzed using modified Stern-Volmer and Lineweaver-Burk methods, which suggested that the binding mechanism was of a static type for both ligands. In both cases, the binding was favored by the domination of hydrophobic as well as hydrogen bonding/Van der Waals forces. Both ligands partially unfolded the secondary structure of HSA, although the effect of cuminaldehyde was more pronounced, as compared to cuminol. The preferred binding site of cuminaldehyde and cuminol inside HSA was also the same; namely, drug binding site 1, located in subdomain IIA. The study showed that cuminaldehyde binds strongly with albumin as compared to its alcohol counterpart, which is due to the more hydrophobic nature of the former.


Subject(s)
Cuminum , Serum Albumin, Human , Aldehydes , Binding Sites , Circular Dichroism , Humans , Ligands , Molecular Docking Simulation , Monoterpenes , Protein Binding , Serum Albumin, Human/chemistry , Spectrometry, Fluorescence , Thermodynamics
11.
Molecules ; 27(5)2022 Mar 01.
Article in English | MEDLINE | ID: mdl-35268736

ABSTRACT

The interaction of common anticancer drug gemcitabine with human serum albumin (HSA) has been studied in detail. The effect of an omnipresent nonsteroidal anti-inflammatory drug ibuprofen was also seen on the binding of HSA and gemcitabine. A slight hyperchromic shift in the difference UV-visible absorption spectra of HSA on the addition of gemcitabine gave a primary idea of the possible complex formation between them. The inner filter effect, which happens due to the significant absorbance of the ligand at the excitation and/or emission wavelengths, played an important role in the observed fluorescence quenching of HSA by gemcitabine that can be understood by comparing the observed and corrected fluorescence intensities obtained at λex = 280 nm and 295 nm. Gemcitabine showed weak interaction with HSA, which took place via a dynamic quenching mechanism with 1:1 cooperative binding between them. Secondary structural analysis, based on circular dichroism (CD) spectroscopy, showed that low concentrations of gemcitabine did not affect the native structure of protein; however, higher concentrations affected it slightly with partial unfolding. For understanding the binding site of gemcitabine within HSA, both experimental (using site markers, warfarin and ibuprofen) as well as computational methods were employed, which revealed that the gemcitabine binding site is located between the interface of subdomain IIA and IIB within the close proximity of the warfarin site (drug site 1). The effect of ibuprofen on the binding was further elaborated because of the possibility of its coexistence with gemcitabine in the prescription given to the cancer patients, and it was noticed that, ibuprofen, even present in high amounts, did not affect the binding efficacy of gemcitabine with HSA. DFT analyses of various conformers of gemcitabine obtained from its docking with various structures of HSA (free and bounded with site markers), show that the stability of the gemcitabine molecule increased slightly after binding with ibuprofen-complexed HSA. Both experimental as well as computational results were in good agreement with each other.


Subject(s)
Ibuprofen
12.
J Biomol Struct Dyn ; 40(22): 12404-12412, 2022.
Article in English | MEDLINE | ID: mdl-34488560

ABSTRACT

Cuminol (4-Isopropylbenzyl alcohol), found in the essential oils of several plant sources, is an important constituent of several cosmetics formulations. The interaction of cuminol with model plasma protein bovine serum albumin was studied in this paper. The experimental studies were mainly carried out using fluorescence spectrophotometry aided with UV visible and CD spectroscopies. Intrinsic fluorescence measurements showed that there was a weak binding between cuminol and BSA. The mechanism of binding involved static quenching with around 1:1 binding. The binding was chiefly supported by hydrophobic forces although a little contribution of hydrogen bonding was also found in the interaction and the values of enthalpy change were negative with positive entropy change. The secondary structure of BSA didn't change significantly in presence of low concentrations of cuminol, however, partial unfolding of the former taken place when the concentration of the latter increased. Molecular docking analyses showed cuminol binds at the intersection of subdomains IIA and IIIA, i.e. its binding site is in between Sudlow sites I and II. Molecular dynamics simulations results have shown that BSA forms a stable complex with cuminol and the structure of the former didn't change much in presence of later. Communicated by Ramaswamy H. Sarma.


Subject(s)
Molecular Dynamics Simulation , Serum Albumin, Bovine , Molecular Docking Simulation , Serum Albumin, Bovine/chemistry , Protein Binding , Spectrophotometry, Ultraviolet , Spectroscopy, Fourier Transform Infrared , Spectrometry, Fluorescence/methods , Binding Sites , Thermodynamics , Circular Dichroism
13.
J Biomol Struct Dyn ; 40(10): 4558-4569, 2022 07.
Article in English | MEDLINE | ID: mdl-33331234

ABSTRACT

In an attempt to identify suitable nano-carriers for drug delivery, natural drug umbelliferone was chosen to synthesize new modulated nanoconjugate of umbelliferone cobalt oxide with cobalt (II) nitrate in one pot assembly in the presence of tannic acid. The synthesized nanoconjugate drug (NCD) was then loaded on graphene oxide (GO) as drug carrier by simple ultrasonication method and thoroughly characterized by various spectroscopic techniques (FT-IR, SEM, TEM, XRD, EPR and thermogravimetric analysis) which revealed the successful loading of the nanoconjugate drug on GO. The UV-visible, fluorescence and electrochemical studies suggested that strong π-π stacking interactions exist between nanoconjugate drug and GO. The binding studies of NCD-GO with ct-DNA were performed by various optical and biophysical methods viz., UV-visible, fluorescence, circular dichroism (CD) and cyclic voltammetry (CV) which indicated electrostatic mode of binding towards the ct-DNA. Furthermore, condensate of nanoconjugate drug-loaded GO (NCD-GO) with ct-DNA was prepared and analyzed by scanning electron microscopy (SEM) which revealed that the interaction of NCD-GO with ct-DNA had occurred. Cleavage activity of NCD-GO with pBR322 was evaluated by gel electrophoresis and it was found that NCD-GO cleave DNA through hydrolytic pathway involving hydroxyl radical (OH). The cytotoxicity of NCD-GO was evaluated against human liver carcinoma (Huh-7), prostate cancer (Du-145) cell lines along with normal cell line (PNT 2). The results obtained showed selective cytotoxic activity of NCD-GO against Du-145 cell lines. The intracellular uptake was visualized by confocal microscopy which revealed the significant cellular uptake and internalization of nanoparticles by cells. Moreover, the adsorption of cobalt oxide umbelliferone on GO was studied by density functional theory. The process of adsorption was found exothermic in nature and the optimized geometry structure is quite stable. Communicated by Ramaswamy H. Sarma.


Subject(s)
Antineoplastic Agents , Drug Carriers , Graphite , Noncommunicable Diseases , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cobalt/chemistry , DNA/chemistry , Graphite/chemistry , Humans , Male , Nanoconjugates , Oxides , Spectroscopy, Fourier Transform Infrared , Umbelliferones/pharmacology
14.
J Biomol Struct Dyn ; 40(19): 9144-9157, 2022.
Article in English | MEDLINE | ID: mdl-33998966

ABSTRACT

This study reports the experimental and computational investigation on the binding of a common anticancer drug, gemcitabine, with the model plasma protein, bovine serum albumin (BSA). Several experimental and computational methods, such as intrinsic and synchronous fluorescence, UV-visible, and circular dichroism spectroscopies, consensus molecular docking and molecular dynamics simulation have been employed to elucidate the binding mechanism. Gemcitabine altered the UV-visible spectrum of BSA, which is a clear indication of the complex formation between them. The visual inspection of observed fluorescence quenching results at λex = 280 nm and 295 nm has shown the substantial involvement of tyrosine residue, even larger than tryptophan. However, after the correction of inner filter effect of the observed data, it became clear that tyrosine has a negligible role in quenching. A 20-fold decrease in quenching constant was found in the corrected data, as compared to the observed data at λex = 280 nm. There was a 1:1 weak binding between BSA and gemcitabine accompanied by dynamic quenching. The secondary structure of BSA remained almost intact in the presence of gemcitabine. The primary binding site of gemcitabine inside BSA was the drug binding site 2 or DS II, which is located in the subdomain 3 A. MD Simulation results suggested that gemcitabine doesn't affect or deviate the structure of BSA upon interaction throughout 100 ns time period. The dominating intermolecular forces were hydrophobic forces and hydrogen bonding. A small change in the frontier molecular orbitals of gemcitabine was also observed after its binding with BSA.Communicated by Ramaswamy H. Sarma.


Subject(s)
Antineoplastic Agents , Serum Albumin, Bovine , Serum Albumin, Bovine/chemistry , Molecular Docking Simulation , Gemcitabine , Circular Dichroism , Spectrometry, Fluorescence/methods , Binding Sites , Tyrosine/metabolism , Protein Binding , Thermodynamics , Spectrophotometry, Ultraviolet/methods
15.
Molecules ; 26(21)2021 Nov 02.
Article in English | MEDLINE | ID: mdl-34771048

ABSTRACT

Lysozyme is often used as a model protein to study interaction with drug molecules and to understand biological processes which help in illuminating the therapeutic effectiveness of the drug. In the present work, in vitro interaction studies of 1-{(2-hydroxyethyl)amino}-2-amino-1,2-dideoxy-d-glucose triphenyl tin (IV) (GATPT) complex with lysozyme were carried out by employing various biophysical methods such as absorption, fluorescence, and circular dichroism (CD) spectroscopies. The experimental results revealed efficient binding affinity of GATPT with lysozyme with intrinsic binding (Kb) and binding constant (K) values in the order of 105 M-1. The number of binding sites and thermodynamic parameters ΔG, ΔH, and ΔS at four different temperatures were also calculated and the interaction of GATPT with lysozyme was found to be enthalpy and entropy driven. The CD spectra revealed alterations in the population of α-helical content within the secondary structure of lysozyme in presence of GATPT complex. The morphological analysis of the complex with lysozyme and lysozyme-DNA condensates was carried out by employing confocal and SEM studies. Furthermore, the molecular docking studies confirmed the interaction of GATPT within the larger hydrophobic pocket of the lysozyme via several non-covalent interactions.


Subject(s)
Aminoglycosides/chemistry , Carrier Proteins/chemistry , Molecular Docking Simulation , Molecular Dynamics Simulation , Organometallic Compounds/chemistry , Organotin Compounds/chemistry , Spectrum Analysis , Binding Sites , DNA/chemistry , Kinetics , Molecular Conformation , Muramidase/chemistry , Protein Binding , Thermodynamics
16.
Int J Biol Macromol ; 182: 993-1002, 2021 Jul 01.
Article in English | MEDLINE | ID: mdl-33857514

ABSTRACT

Herein we have studied the noncovalent molecular interactions between hen egg white lysozyme (HEWL) and the commonly employed antineoplastic drug gemcitabine through the cumulative implementation of spectroscopic techniques and in silico approaches. The formation of a complex between HEWL and gemcitabine was made evident by the differences between the UV-visible spectra of the protein and protein-gemcitabine complex. Fluorescence quenching of HEWL by gemcitabine was hardly detectable at room temperature, but it became prominent at higher temperatures. Very low values for the bimolecular quenching constant and the non-reciprocal dependence of quenching on temperature indicated that dynamic quenching was taking place. Analysis of experimental data indicated that the interaction was dominated by hydrophobic forces, while the results of a computational investigation suggested the concomitant contribution of hydrogen bonding. Gemcitabine binding induced modifications of the secondary structure of HEWL by slightly increasing the α-helical content of the protein. Finally, gemcitabine binding site was inferred to be located in HEWL big hydrophobic cavity.


Subject(s)
Antineoplastic Agents/chemistry , Deoxycytidine/analogs & derivatives , Molecular Docking Simulation , Muramidase/chemistry , Antineoplastic Agents/pharmacology , Binding Sites , Deoxycytidine/chemistry , Deoxycytidine/pharmacology , Muramidase/metabolism , Protein Binding , Gemcitabine
17.
ACS Omega ; 5(16): 9131-9141, 2020 Apr 28.
Article in English | MEDLINE | ID: mdl-32363265

ABSTRACT

Owing to the various beneficial properties of the popular spice saffron, the interaction of safranal, a secondary metabolite of the former, with hen egg white lysozyme was investigated. The formation of a complex was evidenced by UV-visible spectroscopy. Fluorescence quenching experiments were also performed to understand the binding mechanism and to evaluate the forces involved in binding. The strong absorption of safranal in the range of excitation and emission wavelengths of lysozyme fluorescence required the correction of the inner filter effect for fluorescence spectra to obtain the apparent extent of binding. There was a considerable difference between the observed spectra and corrected spectra, and a similar observation was found in the case of synchronous fluorescence spectra. From the analysis of quenching data, it was found that the mechanism involved in quenching was static with 1:1 binding between them. The interaction was found to be driven, mainly, by hydrophobic forces and hydrogen bonding. Safranal had negligible impact on the secondary structure of lysozyme. The interaction was also studied by molecular docking, and the results were in good agreement with the results obtained experimentally. The binding site of safranal was in the big hydrophobic cavity of lysozyme. The amino acids involved in the interaction were Asp52, Ile58, Gln57, Asn59, Trp62, Trp63, Trp108, Ile98, Asp101, and Ala107.

18.
RSC Adv ; 10(22): 13126-13138, 2020 Mar 30.
Article in English | MEDLINE | ID: mdl-35492119

ABSTRACT

Nano cobalt and porous zinc-cobalt oxide particles were synthesized using the concept of coordination compounds of the type [M(ii)L,L'] (where M(ii) = Co(ii) & Zn(ii) L= 4-hydroxy benzaldehyde and L' = piperazine) and were thoroughly characterized. Because the precursors are coordination compounds possessing specific geometry in the crystal lattice, uniform and appropriately sized homo- and heterometallic nanocrystals of Co3O4 and ZnO·Co3O4 were obtained after a thermal process. The homo and hetero composite particles were characterized by transmission electron microscopy (TEM), scanning electron microscopy (SEM), energy dispersive X-ray analysis (EDX), X-ray diffraction (XRD), FT IR spectroscopy and electrochemistry. The paramagnetic chemical shift of the methyl protons in DMSO due to the nanoparticles was studied by NMR spectroscopy, which indicated that the cobalt particles were ferromagnetic. The structural design modification and surface area of Co3O4 was improved by adding the ZnO component. DFT calculations were done to validate the nano structure. Supercapacitance ability of the nanoparticles was studied by cyclic voltammetry, and electrochemical calculations were performed to determine the microelectronic characteristics of the material. The specific capacitance was estimated at 207.3 and 51.1 F g-1 for the ZnO·Co3O4 and Co3O4 electrodes, respectively. Clearly, ZnO·Co3O4 exhibited a much higher specific capacitance than the Co3O4 nanocrystal, which was attributed to better conductivity and higher surface area. The capacitance activity showed multifold enhancement due to the porous nature of Zn oxide in the heterometallic nano ZnO·Co3O4 composite.

19.
Spectrochim Acta A Mol Biomol Spectrosc ; 220: 117101, 2019 Sep 05.
Article in English | MEDLINE | ID: mdl-31150923

ABSTRACT

In this work, a new method for the preparation of ZnO hexagonal nanocrystals by using Sn(IV) as a catalyst was established, which resulted in tranformation of Porous to nanorod-like structures of ZnO. X-ray diffraction (XRD), Energy Dispersion X-ray analysis (EDX), and FT-IR measurements showed that all ZnO nanostructures were of hexagonal phase structure. Transmission electron microscopy (TEM) and scanning electron microscopic (FESEM) studies revealed that morphology of porous-like ZnO (100-200 nm) was converted into nanorod-like (length ~2 µm, diameter ~80 nm) structures upon addition of Sn(IV) as a catalyst. Spectroscopic studies demonstrated that the Zinc(II) compound yields high-quality porous ZnO which upon addition of Sn(IV) catalyst changes into crystalline hexagonal nanorods. The band gap of ZnO nanoparticles calculated employing UV spectrum was found to be 3.31 eV. Moreover, the photocatalytic degradation of methylene blue (MB) under UV light irradiation was performed, which confirmed higher photodegradation of hexagonal ZnO than porous ZnO nanostructures. Furthermore, DFT/TDDFT calculations of MB dye and the expected photodegradation product were also assessed, which were consistent with the kinetic studies. Additionally, zeta potential of the ZnO nanoparticles was measured in the dispersion medium of SDS surfactant which supported high stability of particles are in solution.

20.
Int J Biol Macromol ; 127: 529-535, 2019 Apr 15.
Article in English | MEDLINE | ID: mdl-30654036

ABSTRACT

Anti-amyloidogenic activity of safranal towards induced HSA amyloids has been observed using a variety of techniques including fluorescence, UV-visible, CD, DLS and microscopies. The HSA solution was pre-incubated at 65 °C for 120 h and, in between, the growth of amyloid fibrils, using ThT aggregation kinetics, was monitored at different time intervals. It was found that the amyloid fibril formation of HSA diminishes in presence of safranal and the inhibition was concentration dependent. The surface hydrophobicity of HSA amyloid fibrils also decreased in presence of safranal. The increased CR binding of HSA fibrils also decreased and high concentration of safranal causes the CR binding to resemble like that of native HSA. Both RLS and turbidity intensities were also in inverse relation to the safranal concentration. Safranal also has a good impact to protect the secondary structure of incubated HSA. From the electron microscopy it was seen that the fibrillar network of HSA amyloids gradually vanishes as the concentration of safranal increased. The largely decreased population of HSA aggregates in safranal containing solution as compared to the one without it also suggests the inhibition of formation of large fibrillar aggregates.


Subject(s)
Amyloid/chemistry , Crocus/chemistry , Cyclohexenes/chemistry , Serum Albumin, Human/chemistry , Terpenes/chemistry , Amyloid/ultrastructure , Humans , Hydrophobic and Hydrophilic Interactions , Protein Structure, Secondary
SELECTION OF CITATIONS
SEARCH DETAIL
...