Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-38899750

ABSTRACT

The salts bis(2-amino-3-methylpyridinium) fumarate dihydrate, 2C6H9N2+·C4H2O22-·2H2O (I), and 2-amino-3-methylpyridinium 5-chlorosalicylate, C6H9N2+·C7H4ClO3- (II), were synthesized from 2-amino-3-methylpyridine with fumaric acid and 5-chlorosalicylic acid, respectively. The crystal structures of these salts were characterized by single-crystal X-ray diffraction, revealing protonation in I and II by the transfer of a H atom from the acid to the pyridine base. In the crystals of both I and II, N-H...O interactions form an R22(8) ring motif. Hirshfeld surface analysis distinguishes the interactions present in the crystal structures of I and II, and the two-dimensional (2D) fingerprint plot analysis shows the percentage contribution of each type of interaction in the crystal packing. The volumes of the crystal voids of I (39.65 Å3) and II (118.10 Å3) have been calculated and reveal that the crystal of I is more mechanically stable than II. Frontier molecular orbital (FMO) analysis predicts that the band gap energy of II (2.6577 eV) is lower compared to I (4.0035 eV). The Quantum Theory of Atoms In Molecules (QTAIM) analysis shows that the pyridinium-carboxylate N-H...O interaction present in I is stronger than the other interactions, whereas in II, the hydroxy-carboxylate O-H...O interaction is stronger than the pyridinium-carboxylate N-H...O interaction; the bond dissociation energies also confirm these results. The positive Laplacian [∇2ρ(r) > 0] of these interactions shows that the interactions are of the closed shell type. An in-silico ADME (Absorption, Distribution, Metabolism and Excretion) study predicts that both salts will exhibit good pharmacokinetic properties and druglikeness.

2.
Acta Crystallogr C Struct Chem ; 80(Pt 4): 115-122, 2024 04 01.
Article in English | MEDLINE | ID: mdl-38502537

ABSTRACT

Acridines are a class of bioactive agents which exhibit high biological stability and the ability to intercalate with DNA; they have a wide range of applications. Pyridine derivatives have a wide range of biological activities. To enhance the properties of acridine and 2-amino-3-methylpyridine as the active pharmaceutical ingredient (API), 4-nitrobenzoic acid was chosen as a coformer. In the present study, a mixture of acridine and 4-nitrobenzoic acid forms the salt acridinium 4-nitrobenzoate, C13H10N+·C7H4NO4- (I), whereas a mixture of 2-amino-3-methylpyridine and 4-nitrobenzoic acid forms the salt 2-amino-3-methylpyridinium 4-nitrobenzoate, C6H9N2+·C7H4NO4- (II). In both salts, protonation takes place at the ring N atom. The crystal structure of both salts is predominantly governed by hydrogen-bond interactions. In salt I, C-H...O and N-H...O interactions form an infinite chain in the crystal, whereas in salt II, intermolecular N-H...O interactions form an eight-membered R22(8) ring motif. A theoretical charge-density analysis reveals the charge-density distribution of the inter- and intramolecular interactions of both salts. An in-silico ADME analysis predicts the druglikeness properties of both salts and the results confirm that both salts are potential drug candidates with good bioavailability scores and there is no violation of the Lipinski rules, which supports the druglikeness properties of both salts. However, although both salts exhibit drug-like properties, salt I has higher gastrointestinal absorption than salt II and hence it may be considered a potential drug candidate.


Subject(s)
Aminopyridines , Nitrobenzoates , Picolines , Salts , Crystallography, X-Ray , Salts/chemistry , Hydrogen Bonding , Nitrobenzoates/chemistry , Models, Theoretical , Acridines
3.
Comput Biol Chem ; 107: 107964, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37820470

ABSTRACT

Diabetes mellitus Type 2 (DM2T) is a rapidly expanding metabolic endocrine disorder worldwide. It is caused due to inadequate insulin secretion by pancreatic beta cells as well as development of insulin resistance. This study aimed to investigate the anti-α-glucosidase, insulin stabilization effect, and non-cytotoxic nature of Gymnema latifolium leaf aqueous extract (GLAE). FTIR analysis revealed the functional groups of compounds present in GLAE. Through LC/ESI-MS/MS analysis, about 12 compounds which belongs to different classes, triterpene glycosides, flavonoids, phenolics, stilbene glycosides and chlorophenolic glycosides were identified. GLAE showed in vitro antioxidant activity. GLAE stabilized insulin by increasing its α-helical content. GLAE inhibited the mammalian α-glucosidase (IC50 = 144 µg/mL) activity through competitive mode (Ki = 61.30 µg/mL). GLAE did not affect the viability of normal cell line (Vero cell line) which shows its non-toxic nature. Molecular docking of phytocompounds identified in GLAE was done with human α-glucosidase and insulin. The top 2 compounds [Gymnema saponin V (GSV) and quercetin 3-(2-galloylglucoside) (QGG) with α-glucosidase; GSV and Z)-resveratrol 3,4'-diglucoside (RDG) with human insulin] with low binding free energy were subjected to 100 ns molecular dynamics simulation to ascertain the stable binding of ligand with protein. The MM/GBSA analysis revealed binding free energy of GSV/α-glucosidase and QGG /α-glucosidase to be - 20.9935 and, - 30.9461 kcal/mol, respectively. Altogether GLAE is valuable source of anti-α-glucosidase inhibitors and insulin stabilizing compounds, suggesting potential lead for further exploration as complementary medicine against DM2T.


Subject(s)
Gymnema , Insulins , Animals , Humans , alpha-Glucosidases/metabolism , Glycosides/analysis , Insulins/analysis , Molecular Docking Simulation , Phytochemicals/pharmacology , Plant Extracts/chemistry , Plant Leaves/chemistry , Tandem Mass Spectrometry
4.
J Cell Biochem ; 124(8): 1115-1134, 2023 08.
Article in English | MEDLINE | ID: mdl-37435893

ABSTRACT

Currently, no approved drug is available as a causative agent of coronavirus disease 2019 (COVID-19) except for some repurposed drugs. The first structure of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was reported in late 2019, based on that some vaccines and repurposed drugs were approved to prevent people from COVID-19 during the pandemic situation. Since then, new types of variants emerged and notably, the receptor binding domain (RBD) adopted different binding modes with angiotensin-converting enzyme 2 (ACE2); this made significant changes in the progression of COVID-19. Some of the new variants are highly infectious spreading fast and dangerous. The present study is focused on understanding the binding mode of the RBD of different mutated SARS-CoV-2 variants of concern (alpha to omicron) with the human ACE2 using molecular dynamics simulation. Notably, some variants adopted a new binding mode of RBD with ACE2 and formed different interactions, which is unlike the wild type; this was confirmed from the comparison of interaction between RBD-ACE2 of all variants with its wild-type structure. Binding energy values confirm that some mutated variants exhibit high binding affinity. These findings demonstrate that the variations in the sequence of SARS-CoV-2 S-protein altered the binding mode of RBD; this may be the reason that the virus has high transmissibility and causes new infections. This in-silico study on mutated variants of SARS-CoV-2 RBD with ACE2 insights into their binding mode, binding affinity, and stability. This information may help to understand the RBD-ACE2 binding domains, which allows for designing newer drugs and vaccines.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Angiotensin-Converting Enzyme 2/genetics , Molecular Dynamics Simulation , Mutation , Protein Binding , SARS-CoV-2/genetics
5.
J Biomol Struct Dyn ; : 1-14, 2023 Jun 10.
Article in English | MEDLINE | ID: mdl-37301607

ABSTRACT

Since 2019 the SARS-CoV-2 and its variants caused COVID-19, such incidents brought the world in pandemic situation. This happened due to furious mutations in SARS-CoV-2, in which some variants had high transmissibility and infective, this led the virus emerged as virulent and worsened the COVID-19 situation. Among the variants, P323L is one of the important mutants of RdRp in SARS-CoV-2. To inhibit the erroneous function of this mutated RdRp, we have screened 943 molecules against the P323L mutated RdRp with the criteria that the molecules with 90% similar to the structure of remdesivir (control drug) resulted nine molecules. Further, these molecules were evaluated by induced fit docking (IFD) identified two molecules (M2 & M4) which are forming strong intermolecular interactions with the key residues of mutated RdRp and has high binding affinity. Docking score of the M2 and M4 molecules with mutated RdRp are -9.24 and -11.87 kcal/mol, respectively. Further, to understand the intermolecular interactions, conformational stability, the molecular dynamics simulation and binding free energy calculations were performed. The binding free energy values of M2 and M4 molecules with the P323L mutated RdRp complexes are -81.60 and -83.07 kcal/mol, respectively. The results of this in silico study confirm that M4 is a potential molecule; hence, it may be considered as the potential inhibitor of P323L mutated RdRp to treat COVID-19 after clinical investigation.Communicated by Ramaswamy H. Sarma.

6.
J Biomol Struct Dyn ; 41(23): 13752-13765, 2023.
Article in English | MEDLINE | ID: mdl-36905654

ABSTRACT

Lawsonia inermis (henna) has been used in traditional medicine throughout the world and biological property of its flower has been least explored. In the present study, the phytochemical characterization and biological activity (in vitro radical scavenging activity, anti-alpha glucosidase and anti-acetylcholinesterase) of aqueous extract prepared from henna flower (HFAE) was carried out by both Qualitative and quantitative phytochemical analysis and Fourier-transform infrared spectroscopy revealed the functional group of the phytoconstituents such as phenolics, flavonoids, saponin, tannins and glycosides. The phytochemicals present in HFAE was preliminary identified by liquid chromatography/electrospray ionization tandem mass spectrometry. The HFAE showed potent in vitro antioxidant activity and the HFAE inhibited mammalian α-glucosidase (IC50 = 129.1 ± 5.3 µg/ml; Ki = 38.92 µg/ml) and acetylcholinesterase (AChE; IC50 = 137.77 ± 3.5 µg/ml; Ki = 35.71 µg/ml) activity by competitive manner. In silico molecular docking analysis revealed the interaction of active constituents identified in HFAE with human α-glucosidase and AChE. Molecular dynamics simulation for 100 ns showed the stable binding of top two ligand/enzyme complexes with lowest binding energy such as 1,2,3,6-Tetrakis-O-galloyl-beta-D-glucose (TGBG)/human α-glucosidase, Kaempferol 3-glucoside-7-rhamnoside (KGR)/α-glucosidase, agrimonolide 6-O-ß-D-glucopyranoside (AMLG)/human AChE and KGR/AChE. Through MM/GBSA analysis, the binding energy for TGBG/human α-glucosidase, KGR/α-glucosidase, AMLG/human AChE and KGR/AChE was found to be -46.3216, -28.5772, -45.0077 and -47.0956 kcal/mol, respectively. Altogether, HFAE showed an excellent antioxidant, anti-alpha glucosidase and anti-AChE activity under in vitro. This study suggest HFAE with remarkable biological activities could be further explored for therapeutics against type 2 diabetes and diabetes-associated cognitive decline.Communicated by Ramaswamy H. Sarma.


Subject(s)
Diabetes Mellitus, Type 2 , Lawsonia Plant , Animals , Humans , alpha-Glucosidases/metabolism , Plant Extracts/pharmacology , Plant Extracts/chemistry , Molecular Dynamics Simulation , Lawsonia Plant/metabolism , Molecular Docking Simulation , Acetylcholinesterase/metabolism , Flowers/chemistry , Flowers/metabolism , Phytochemicals/pharmacology , Phytochemicals/chemistry , Antioxidants/pharmacology , Antioxidants/chemistry , Mammals/metabolism
7.
J Biomol Struct Dyn ; 41(4): 1351-1365, 2023 03.
Article in English | MEDLINE | ID: mdl-34974819

ABSTRACT

The p300 histone acetyltransferase (HAT) enzyme acetylates the lysine residue of histone promotes the transcription reaction. The abnormal function of p300 HAT enzyme causes various diseases such as Cancer, Asthma, Alzheimer, Diabetics, and AIDS. In the recent years, several studies have been conducted to design potential drug to inhibit this enzyme. Recently, an in vitro study has been performed on the synthetic molecules PU139 and PU141 to inhibit the p300 HAT enzyme. The present study aims to understand the binding affinity, intermolecular interactions, conformational stability and binding energy of PU139 and PU141 molecules in the active site of p300 HAT enzyme from the in silico studies. The molecular docking and molecular dynamics (MD) simulations were carried out for both ligands with the p300 HAT enzyme. The molecular docking and MD simulations reveals that both molecules forms expected interactions with the catalytic site key residues of p300 enzyme. The MD simulation shows the maximum RMSD value for the PU141 is 2.3 Å, whereas for PU139 is 3.3 Å; these low RMSD values indicate that both molecules are highly stable in the active site of p300. The calculated binding free energy of PU141 (-20.62 kcal/mol) is higher than the molecule PU139 (-17.67 kcal/mol). Among the results, PU141 shows the high binding affinity with p300 while comparing with PU139. The results of this in-silico study coupled with the findings reported in the in vitro study confirm that PU141 may be suitable for clinical study.Communicated by Ramaswamy H. Sarma.


Subject(s)
Histone Acetyltransferases , Molecular Dynamics Simulation , Molecular Docking Simulation , Histone Acetyltransferases/chemistry , Catalytic Domain , Histones/metabolism
8.
J Biomol Struct Dyn ; 41(20): 10396-10410, 2023 12.
Article in English | MEDLINE | ID: mdl-36510678

ABSTRACT

In this COVID-19 pandemic situation, an appropriate drug is urgent to fight against this infectious disease to save lives and prevent mortality. Repurposed drugs and vaccines are the immediate solutions for this medical emergency until discover a new drug to treat this disease. As of now, no specific drug is available to cure this disease completely. Several drug targets were identified in SARS-CoV-2, in which RdRp protein is one of the potential targets to inhibit this virus infection. In-Silico studies plays a vital role to understand the binding nature of the drugs at the atomic level against the disease targets. The present study explores the binding mechanism of reported 53 nucleoside and non-nucleoside RdRp inhibitors and Ivermectin which are in clinical trials. These molecules were screened by molecular docking simulation; in which, the molecules are showing high binding affinity and forming interactions with the key amino acids of active site of RdRp protein are chosen for molecular dynamics simulation (MD) and binding free energy analysis. The results of molecular docking and MD simulation studies reveal that IDX184 is a stable molecule and forms strong interactions with the key amino acids and shows high binding affinity towards RdRp. Hence, IDX184 may also be considered as a potential inhibitor of RdRp after clinical study.Communicated by Ramaswamy H. Sarma.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Molecular Dynamics Simulation , Nucleosides/pharmacology , Molecular Docking Simulation , Pandemics , Amino Acids , RNA-Dependent RNA Polymerase , Antiviral Agents/pharmacology
9.
J Mol Struct ; 1266: 133483, 2022 Oct 15.
Article in English | MEDLINE | ID: mdl-35692554

ABSTRACT

A novel drug to treat SARS-CoV-2 infections and hydroxyl chloroquine analogue, (E)-2,6-bis(4-chlorophenyl)-3-methyl-4-(2-(2,4,6-trichlorophenyl)hydrazono)piperidine (BCMTP) compound has been synthesized in one pot reaction. The novel compound BCMTP has been characterized by FT-IR, 1H-NMR, 13C-NMR and single-crystal X-ray diffraction patterns. Crystal packing is stabilized by C8-H8A•••Cl10i, C41-H41•••Cl1ii and N1-H1A•••Cl6iii intermolecular hydrogen bonds. From the geometrical parameters, it is observed that the piperidine ring adopts chair conformation. Hirshfeld surface analysis was carried out to quantify the interactions and an interaction energy analysis was done to study the interactions between pairs of molecules. The geometrical structure was optimized by density functional theory (DFT) method at B3LYP/6-31G (d, p) as the basic set. The smaller binding energy value provides the higher reactivity of BCMTP compound than hydroxyl chloroquine and was corrected by high electrophilic and low nucleophilic reactions. The stability and charge delocalization of the molecule were also considered by natural bond orbital (NBO) analysis. The HOMO-LUMO energies describe the charge transfer which takes place within the molecule. Molecular electrostatic potential has also been analysed. Molecular docking studies are implemented to analyse the binding energy of the BCMTP compound against standard drugs such as the crystal structure of ADP ribose phosphatase of NSP3 from SARS-CoV-2 in complex with MES and SARS-CoV-2 main protease with an unliganded active site (2019-nCoV, corona virus disease 2019, COVID-19) and found to be considered having better antiviral agents. Molecular dynamics simulation was performed for COVID-19 main protease (Mpro: 6WCF/6Y84) to understand the elements governing the inhibitory effect and the stability of interaction under dynamic conditions.

10.
Molecules ; 27(12)2022 Jun 20.
Article in English | MEDLINE | ID: mdl-35745085

ABSTRACT

The high expression of 17ß-hydroxysteroid dehydrogenase type 1 (17ß-HSD1) mRNA has been found in breast cancer tissues and endometriosis. The current research focuses on preparing a range of organic molecules as 17ß-HSD1 inhibitors. Among them, the derivatives of hydroxyphenyl naphthol steroidomimetics are reported as one of the potential groups of inhibitors for treating estrogen-dependent disorders. Looking at the recent trends in drug design, many halogen-based drugs have been approved by the FDA in the last few years. Here, we propose sixteen potential hydroxyphenyl naphthol steroidomimetics-based inhibitors through halogen substitution. Our Frontier Molecular Orbitals (FMO) analysis reveals that the halogen atom significantly lowers the Lowest Unoccupied Molecular Orbital (LUMO) level, and iodine shows an excellent capability to reduce the LUMO in particular. Tri-halogen substitution shows more chemical reactivity via a reduced HOMO-LUMO gap. Furthermore, the computed DFT descriptors highlight the structure-property relationship towards their binding ability to the 17ß-HSD1 protein. We analyze the nature of different noncovalent interactions between these molecules and the 17ß-HSD1 using molecular docking analysis. The halogen-derived molecules showed binding energy ranging from -10.26 to -11.94 kcal/mol. Furthermore, the molecular dynamics (MD) simulations show that the newly proposed compounds provide good stability with 17ß-HSD1. The information obtained from this investigation will advance our knowledge of the 17ß-HSD1 inhibitors and offer clues to developing new 17ß-HSD1 inhibitors for future applications.


Subject(s)
Halogens , Molecular Dynamics Simulation , 17-Hydroxysteroid Dehydrogenases , Enzyme Inhibitors/pharmacology , Female , Humans , Molecular Docking Simulation , Naphthols , Structure-Activity Relationship
11.
Acta Crystallogr C Struct Chem ; 78(Pt 3): 181-191, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35245215

ABSTRACT

The proton-transfer complexes 2-amino-4-methoxy-6-methylpyrimidinium (2A4M6MP) 4-aminosalicylate (4AMSA), C6H10N3O+·C7H6NO3-, I, and 5-chlorosalicylate (5ClSA), C6H10N3O+·C7H4ClO3-, II, were synthesized by slow evaporation and crystallized. The crystal structures of both I and II were determined by single-crystal X-ray structure analysis. The crystal structures of both salts exhibit O-H...O, N-H...O, N-H...N and C-H...O interactions in their crystals. The 4AMSA and 5ClSA anions in combination with the 2A4M6MP cations form distinct synthons, which are represented by the graph-set notations R22(8), R42(8) and R22(8). Furthermore, the ΔpKa values were calculated and clearly demonstrate that 2A4M6MP is a good salt former when combined with carboxylic acids. Hirshfeld surface analysis was used to quantify the weak and strong interactions in the solid state, and energy framework calculations showed the stability of the hydrogen-bonding interactions. QTAIM (quantum theory of atoms in molecules) analysis revealed the nature of the chemical bonding in I and II, and the charge-density distribution in the intermolecular interactions in the crystal structures.


Subject(s)
Quantum Theory , Salts , Crystallography, X-Ray , Hydrogen Bonding , Salicylates
12.
Heliyon ; 7(4): e06684, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33898820

ABSTRACT

The reduced level of dopamine at midbrain (substantia nigra) leads to Parkinson disease by the influence of monoamine oxidation process of monoamine oxidase B (MAO-B) enzyme. This disease mostly affects the aged people. Reports outline that the naringenin molecule acts as an inhibitor of MAO-B enzyme and it potentially prevents the development of PD. To elucidate the binding mechanism of naringenin with MAO-B, we performed the molecular docking, QM/MM and molecular dynamics (MD) simulations. The molecular docking results confirm that the naringenin strongly binds with the substrate binding site of MAO-B enzyme (-12.0 kcal/mol). The low values of RMSD, RMSF and Rg indicate that the naringenin - MAO-B complex is stable over the entire period of MD simulation. Naringenin forms strong interaction with the orient keeper residue Tyr326 and other binding site residues Leu171, Glu206 and these interactions were maintained throughout the MD simulation. It is also important to block the function of MAO-B enzyme. The QM/MM study coupled with the charge density analysis reveals the charge density distribution and the strength of intermolecular interactions of naringenin-MAO-B complex. The above results suggest that this molecule is a potential inhibitor of MAO-B enzyme.

13.
Appl Biochem Biotechnol ; 193(6): 1909-1923, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33512650

ABSTRACT

The unprecedented coronavirus SARS-CoV-2 outbreak at Wuhan, China, caused acute respiratory infection to humans. There is no precise vaccine/therapeutic agents available to combat the COVID-19 disease. Some repurposed drugs are saving the life of diseased, but the complete cure is relatively less. Several drug targets have been reported to inhibit the SARS-CoV-2 virus infection, in that TMPRSS2 (transmembrane protease serine 2) is one of the potential targets; inhibiting this protease stops the virus entry into the host human cell. Camostat mesylate, nafamostat, and leupeptin are the drugs, in which the first two drugs are being used for COVID-19 and leupeptin also tested. To consider these drugs as the repurposed drug for COVID-19, it is essential to understand their binding affinity and stability with TMPRSS2. In the present study, we performed the molecular docking and molecular dynamics (MD) simulation of these molecules with the TMPRSS2. The docking study reveals that leupeptin molecule strongly binds with TMPRSS2 protein than the other two drug molecules. The RMSD and RMSF values of MD simulation confirm that leupeptin and the amino acids of TMPRSS2 are very stable than the other two molecules. Furthermore, leupeptin forms interactions with the key amino acids of TMPRSS2 and the same have been maintained during the MD simulations. This structural and dynamical information is useful to evaluate these drugs to be used as repurposed drugs, however, the strong binding profile of leupeptin with TMPRSS2, suggests, it may be considered as a repurposed drug for COVID-19 disease after clinical trial.


Subject(s)
Antiviral Agents/pharmacology , Benzamidines/therapeutic use , COVID-19 Drug Treatment , Drug Repositioning , Esters/therapeutic use , Guanidines/therapeutic use , Leupeptins/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Serine Endopeptidases/metabolism , Antiviral Agents/therapeutic use , Benzamidines/pharmacology , COVID-19/virology , Esters/pharmacology , Guanidines/pharmacology , Humans , Protein Binding , SARS-CoV-2/drug effects
14.
Acta Crystallogr E Crystallogr Commun ; 76(Pt 9): 1535-1538, 2020 Sep 01.
Article in English | MEDLINE | ID: mdl-32939314

ABSTRACT

In the crystals of the title compound, C5H7N2 +·CNS-·C5H6N2, the components are linked by three N-H⋯N and two N-H⋯S hydrogen bonds, resulting in two inter-penetrating three-dimensional networks. Hirshfeld surface analysis shows that the most important contributions to the crystal packing are from H⋯H (36.6%), C⋯H/H⋯C (20.4%), S⋯H/H⋯S (19.7%) and N⋯H/H⋯N (13.4%) inter-actions.

15.
Int J Biol Macromol ; 148: 696-703, 2020 Apr 01.
Article in English | MEDLINE | ID: mdl-31954795

ABSTRACT

The isolation and purification of active components from the brown algae Sargassum.wightii is highly limited. In the present study, fucoxanthin was purified from S. wightii using simple methods. Ethyl acetate fraction obtained by Soxhlet extraction contained high concentration of fucoxanthin. Fucoxanthin-rich fraction was further subjected to open silica column chromatography and thin layer chromatography to obtain purified fucoxanthin. Purified fucoxanthin showed in vitro antioxidant activity. Fucoxanthin showed inhibition of angiotensin I-converting enzyme (ACE) with half maximal inhibitory value of 822.64 ± 17.69 µM. Kinetic analysis revealed mixed non-competitive inhibition with inhibitory constant of 600 µM for fucoxanthin against ACE. Molecular docking analysis showed the interaction of fucoxanthin with amino acids and zinc ion present in the active site of the human ACE. Molecular dynamics analysis demonstrated the stability of the fucoxanthin and ACE complex in in silico. These results show that S. wightii may be used as food ingredient to overcome hypertension.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/chemistry , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Peptidyl-Dipeptidase A/metabolism , Sargassum/chemistry , Xanthophylls/chemistry , Xanthophylls/pharmacology , Animals , Antioxidants/chemistry , Antioxidants/pharmacology , Computer Simulation , Kinetics , Molecular Docking Simulation , Rabbits
16.
J Biomol Struct Dyn ; 38(1): 219-235, 2020 01.
Article in English | MEDLINE | ID: mdl-31038398

ABSTRACT

The resistance to the endocrine therapy of breast cancer leads to the emergence of new class of drugs that downregulates the estrogen receptor action known as selective estrogen receptor downregulators (SERDs). The first approved SERD is fluvestrant; after this, there are several downregulators evolved and are in clinical trials, in which the brilanestrant (BRI) molecule shows nM range of binding affinity and efficacy. In the present study, to understand the binding nature of BRI molecule in the active site of ERα, the molecular docking analysis has been performed. Further, the QM/MM calculations were performed for the BRI-ERα complex to analyze the charge density distribution of intermolecular interactions. The molecular dynamics (MD) simulation was employed to understand the stability and binding mechanism of BRI molecule through root-mean-square deviation (RMSD), root-mean-square fluctuation (RMSF) and binding free energy calculations. From the MD simulation trajectory analysis, the alterations of Helix12 conformation and the key residue (Lys529), which is responsible for the ERα downregulation, have been identified. Further, the interaction between the H3 and H12 regions was identified for the antagonism of BRI molecule. The current study led us to understand the binding mechanism, antagonism and downregulation of BRI molecule, and this knowledge is essential to design novel SERDs for the treatment of endocrine-resistant positive breast cancer.Communicated by Ramaswamy H. Sarma.


Subject(s)
Antineoplastic Agents/chemistry , Estrogen Receptor alpha/chemistry , Molecular Docking Simulation , Molecular Dynamics Simulation , Quantum Theory , Algorithms , Antineoplastic Agents/pharmacology , Binding Sites , Catalytic Domain , Estrogen Receptor alpha/metabolism , Hydrogen Bonding , Ligands , Molecular Conformation , Protein Binding
17.
J Cell Biochem ; 121(2): 1759-1777, 2020 02.
Article in English | MEDLINE | ID: mdl-31633226

ABSTRACT

The chromatin modification is regulated by the histone acetyltransferase (HAT) and histone deacetyltransferase (HDAC) enzymes; abnormal function of these enzymes leads to several malignant diseases. The inhibition of these enzymes using natural ligand molecules is an emerging technique to cure these diseases. The in vitro analysis of natural molecules, venenatine, spinosine, palmatine and taxodione are giving the best inhibition rate against p300 HAT enzyme. However, the detailed understanding of binding and the stability of these molecules with p300 HAT is not yet known. The aim of the present study is focused to determine the binding strength of the molecules from molecular dynamics simulation analysis. The docking analysis confirms that, the venenatine (-6.97 kcal/mol - conformer 8), spinosine (-6.52 kcal/mol conformer -10), palmatine (-5.72 kcal/mol conformer-3) and taxodione (-4.99 kcal/mol conformer-4) molecules form strong hydrogen bonding interactions with the key amino acid residues (Arg1410, Thr1411 and Trp1466) present in the active site of p300. In the molecular dynamics (MD) simulation, the spinosine retain these key interactions with the active site amino acid residues (Arg1410, Thr1411, and Trp1466) than venenatine and are stable throughout the simulation. The RMSD value of spinosine (0.5 to 1.3 Å) and venenatine (0.3 to 1.3 Å) are almost equal during the MD simulation. However, during the MD simulation, the intermolecular interaction between venenatine and the active site amino acid residues (Arg1410, Thr1411, and Trp1466) decreased on comparing with the spinosine-p300 interaction. The binding free energy of the spinosine (-15.30 kcal/mol) is relatively higher than the venenatine (-11.8 kcal/mol); this increment is attributed to the strong hydrogen bonding interactions of spinosine molecule with the active site amino acid residues of p300.


Subject(s)
Alkaloids/metabolism , Berberine Alkaloids/metabolism , Molecular Dynamics Simulation , p300-CBP Transcription Factors/chemistry , p300-CBP Transcription Factors/metabolism , Alkaloids/chemistry , Berberine Alkaloids/chemistry , Catalytic Domain , Crystallography, X-Ray , Enzyme Stability , Humans , Models, Molecular
18.
J Biomol Struct Dyn ; 38(18): 5307-5319, 2020 Nov.
Article in English | MEDLINE | ID: mdl-31779524

ABSTRACT

The low level of dopamine at substantia nigra (mid-brain) has been considered to be one of the reasons for Parkinson's disease (PD). This dopamine deficit is due to the influence of Catechol-O-Methyltransferase (COMT). A recent report outline that the flavonoid family of molecules are able to inhibit the COMT enzyme. To identify a potential molecule from the flavonoid family, we performed molecular screening over a group of flavonoid molecules using glide method. Among the screened molecules, morin molecule shows, relatively larger binding affinity (-7.90 kcal/mol) towards COMT enzyme. Further, an Induced Fit Docking (IFD) has been carried out for morin with COMT enzyme; the corresponding docking score value is -8.17 kcal/mol. To understand the conformational flexibility of morin in the active site of COMT, its conformation has been compared with the corresponding gas phase conformation. Further, molecular dynamics (MD) simulation has been performed to understand the dynamical behavior and the stability of morin molecule in the active site of COMT enzyme. The morin strongly binds with the catalytic triad and gatekeeper residues and these interactions have been maintained during the 50 ns MD simulation. Notably, the O(1) atom of morin forms interaction with Glu198, Mg ion and catalytic residue Asn169; in which, Glu198 is more stable during the simulation. The O(11) atom blocks the ionization process through the interaction with Lys143. Both of these interactions are essential to inhibit the enzymatic function of COMT enzyme. The binding free energy study shows morin molecule exhibit good binding towards COMT enzyme.Communicated by Ramaswamy H. Sarma.


Subject(s)
Catechol O-Methyltransferase , Flavonoids , Molecular Dynamics Simulation , Catalytic Domain , Catechol O-Methyltransferase Inhibitors , Molecular Docking Simulation
19.
J Comput Chem ; 41(2): 97-109, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31602686

ABSTRACT

The selective estrogen receptor downregulators (SERDs) are the new emerging class of drugs that are used for the treatment of endocrine resistance breast cancer. Elacestrant (ELA) is a new SERD, currently it is in phase II clinical trial. To understand the ELA-ERα interactions, the molecular docking analysis has been carried out. The ELA molecule binds with the helices H3, H5, H6, and H11 and forms important intermolecular interactions. In addition to this, the tetrahydronapthalene and phenyl rings of ELA are forming T-shaped π···π interactions with the Phe404 and Trp383 residues. Further to understand the stability and flexibility of ELA molecule in the active site of wild and mutated L536S ERα, 100ns molecular dynamics (MD) simulation was performed for both complexes. Interestingly, the MD analysis of wild complex revealed an interaction between ELA and the Asn532 of H11, which is an essential interaction for the downregulation/degradation of ERα, whereas this interaction is not observed in the mutated complex. The drug binding mechanism and H12 dynamics have been elucidated from the analysis of hydrogen bonding interactions and the secondary structure analysis. To explore the binding affinity of ELA molecule, the binding free energy and normal mode analyses were carried out. The per residue decomposition analysis also performed, which shows the contribution of individual amino acids. The principal component analysis and residue interaction network analysis were used to identify the modifications and the interaction between the residues. From the results of different analysis, the inhibition mechanism and downregulation of ERα-ELA complex has been investigated. © 2019 Wiley Periodicals, Inc.


Subject(s)
Estrogen Receptor alpha/chemistry , Molecular Dynamics Simulation , Tetrahydronaphthalenes/chemistry , Thermodynamics , Binding Sites , Down-Regulation , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Humans , Molecular Docking Simulation , Mutation
20.
Int J Biol Macromol ; 140: 1147-1157, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-31442505

ABSTRACT

In Alzheimer's disease (AD) and diabetes-associated cognitive decline, the acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) activity is increased. AChE exists as different globular molecular forms: tetramer (G4), dimer (G2) and monomer (G1). In adult brain, G4 form is abundant however in AD, the ratio of lower molecular forms (G1) to G4 form increased. Hence, the present study delineated the inhibition of novel astaxanthin-s-allyl cysteine (AST-SAC) against BChE and various molecular forms of AChE. Cobra venom, human erythrocyte and Electrophorus electricus was used as source of G1, G2 and G4 form of AChE. AST-SAC showed inhibition against G1 (IC50 = 0.72 µM, competitive, Ki = 0.66 µM), G2 (IC50 = 0.65 µM, mixed, Ki = 0.50 µM) and G4 (IC50 = 0.67 µM, competitive, Ki = 0.67 µM) form of AChE. AST-SAC inhibited human brain AChE (IC50 = 0.84 µM, competitive, Ki = 0.53 µM) and human serum BChE (IC50 = 0.80 µM, competitive, Ki = 0.58 µM). In silico analysis revealed the interaction of AST-SAC with the amino acids present in peripheral anionic and catalytic site of human AChE and BChE. Molecular dynamics simulation confirmed the stable interaction of AST-SAC in the active site gorge of AChE and BChE.


Subject(s)
Acetylcholinesterase/metabolism , Butyrylcholinesterase/metabolism , Cholinesterase Inhibitors/pharmacology , Cysteine/analogs & derivatives , Acetylcholinesterase/chemistry , Animals , Brain/enzymology , Butyrylcholinesterase/blood , Butyrylcholinesterase/chemistry , Cholinesterase Inhibitors/chemistry , Computer Simulation , Cysteine/chemistry , Cysteine/pharmacology , Humans , Molecular Dynamics Simulation , Xanthophylls/chemistry , Xanthophylls/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...