Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Biomol Struct Dyn ; 41(9): 4024-4039, 2023 06.
Article in English | MEDLINE | ID: mdl-35403561

ABSTRACT

Chebulinic acid (CHN) and chebulagic acid (CHG) have been known for centuries for their anti-cancer, anti-diabetes, HIV and anti-inflammatory properties. In this study, the interaction of these phytochemicals CHN/CHG, with the two major transport proteins for various drugs, human serum albumin (HSA) and α-1-acid glycoprotein (AGP), was unraveled by using several spectroscopic techniques and computational methods. The binding of CHN/CHG quenches the HSA/AGP fluorescence intensities, and also these phytochemicals are bound strongly to HSA/AGP proteins. An apparent decrease in fluorescence intensities of CHN/CHG-HSA and CHN/CHG-AGP complex showed the static mode of fluorescence quenching. Furthermore, the intrinsic fluorescence and using site-specific markers ibuprofen competing with these molecules, thereby replacing it in the binding site of subdomain IIIA. The computational methods substantiated the experimental findings, revealing that CHN interacted with Lys414A, Glu492A, Glu492A and Lys413A residues of subdomain IIIA of HSA and for CHG showed the interaction with Lys545A and Lys413A residues of subdomain IIIA of HSA. Fluorescence and surface plasmon resonance data unveiled a previously unreported binding event between CHN/CHG and HSA; the determined binding affinities of both compounds were slightly higher for HSA than AGP. A change in functionality of protein confirmed the esterase-like activity of HSA in the presence of CHG/CHN upon binding with CHG/CHN. Displacement and circular dichroism (CD) experiments analysis showed that the two CHN/CHG and binding specifically to IIIA subdomain on HSA results in the conformational changes in the HSA. Thus, CD revealed a few conformational changes in HSA due to CHN/CHG. The binding of these two phytochemicals to the plasma proteins would give a path to develop new inspired drug molecules for chronic diseases.Communicated by Ramaswamy H. Sarma.


Subject(s)
Serum Albumin, Human , Humans , Molecular Docking Simulation , Protein Binding , Spectrometry, Fluorescence , Thermodynamics , Serum Albumin, Human/chemistry , Binding Sites , Circular Dichroism
2.
J Biomol Struct Dyn ; 40(22): 12135-12147, 2022.
Article in English | MEDLINE | ID: mdl-34463217

ABSTRACT

In the present study, we have analyzed the interaction of a phytochemical, stigmasterol (Stig), with human serum albumin (HSA) under physiological conditions using fluorescence quenching, circular dichroism and molecular modeling methods. Cytotoxic studies with Stig in mouse macrophages (RAW 246.7) and HeLa cell lines showed anti-inflammatory and anti-cancer properties. Further, the intrinsic fluorescence of HSA was quenched by Stig, which was considered a static quenching mechanism. The site-specific marker experiments revealed that Stig binds to the IIIA subdomain of HSA with a binding constant of KStig=1.8 ± 0.03 × 105 M-1 and free energy of -7.26 ± 0.031 Kcal/mol. The secondary structure of HSA was partially unfolded after binding of Stig, which indicates an alteration in the microenvironment of the protein binding site. Molecular docking experiments found that Stig binds strongly with HSA at the IIIA domain of the hydrophobic pocket with one hydrogen bond. The rigidity of the protein-Stig complex and free energies were analyzed by molecular dynamic simulation (MDS) for 100 ns, where the HSA-Stig was stabilized after 40 ns. MDS studies revealed that HSA does not significantly change the secondary structure when it binds with Stig, which is in agreement with the circular dichroism data. Overall, the results obtained gave qualitative and quantitative insight into the binding interaction between HSA and Stig, which is essential in understanding the latter as a therapeutic molecule.Communicated by Ramaswamy H. Sarma.


Subject(s)
Molecular Dynamics Simulation , Serum Albumin, Human , Animals , Mice , Humans , Serum Albumin, Human/chemistry , Stigmasterol/pharmacology , Molecular Docking Simulation , HeLa Cells , Spectrometry, Fluorescence , Thermodynamics , Protein Binding , Binding Sites , Circular Dichroism
3.
J Pharm Biomed Anal ; 209: 114478, 2022 Feb 05.
Article in English | MEDLINE | ID: mdl-34894460

ABSTRACT

Bacosine (BAC) is a natural product isolated from a herb and used in the Ayurvedic system of medicine. It is reported to have a wide array of biological activities, which has generated interest in its therapeutic potential. To better understand how BAC may operate as a potential anti-cancer therapeutic, we examined its anti-cancer properties in the human breast cancer cell line, MCF-7. In order to get an idea of how it may behave in vivo, we also evaluated its interaction with human serum albumin (HSA) and α-1-acid glycoprotein (AGP) using fluorescence spectroscopy and in silico molecular modelling. Based on our in vitro studies, we found that BAC inhibited MCF-7 cell growth in a dose-dependent manner with an IC50 value of 9 µM. In addition, the intrinsic fluorescence of HSA and AGP was quenched by BAC, consistent with a static quenching mechanism. Fluorescence emission spectroscopy revealed a binding of 2.97 ± 0.01 × 104 M-1 for HSA-BAC which corresponded to a free energy change of - 6.07 kcal/mol at 25 °C. In addition, we found that BAC had a binding constant of 1.8 ± 0.02 × 103 M-1 to AGP which corresponded to a change in free energy - 4.42 kcal/mol at 25 °C. We also identified the site of BAC binding to the HSA protein using the site-specific marker, phenylbutazone, along with molecular docking studies. Circular dichroism spectra revealed partial changes in the secondary structure of HSA in the presence of BAC suggesting direct interactions. Molecular dynamics simulations demonstrated that the HSA-BAC complex reaches an equilibration state at around 4 ns, suggesting that the HSA-BAC complex is quite stable. Our results provide evidence that serum proteins can act as a carrier protein for BAC, potentially impacting its development as an anti-cancer agent.


Subject(s)
Orosomucoid , Serum Albumin , Binding Sites , Circular Dichroism , Humans , Molecular Docking Simulation , Orosomucoid/metabolism , Protein Binding , Serum Albumin/metabolism , Serum Albumin, Human , Spectrometry, Fluorescence , Thermodynamics , Triterpenes
4.
J Biomol Struct Dyn ; 38(5): 1539-1550, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31057091

ABSTRACT

In this study, forskolin-loaded human serum albumin nanoparticles (FR-HSANPs) were successfully prepared by incorporation and affinity-binding methods. FR-HSANPs were characterized by transmission electron microscope that most of them are circular in shape and size is around 340 nm. The drug loading was more than 88% and further sustained release profiles were observed as it is 77.5% in 24 h time. Additionally, the cytotoxicity results with HepG2 cells indicated that FR-HSANPs showed significantly higher cytotoxicity and lower cell viability as compared to free forskolin (FR). Furthermore, to understand the binding mechanism of human serum albumin (HSA) with forskolin resulted from fluorescence quenching as a static mechanism and the binding constant is 6.26 ± 0.1 × 104 M-1, indicating a strong binding affinity. Further, association and dissociation kinetics of forskolin-HSA was calculated from surface plasmon resonance spectroscopy and the binding constant found to be Kforskolin = 3.4 ± 0.24 × 104 M-1 and also fast dissociation was observed. Further, we used circular dichroism and molecular dynamics simulations to elucidate the possible structural changes including local conformational changes and rigidity of the residues of both HSA and HSA-forskolin complexes.Communicated by Ramaswamy H. Sarma.


Subject(s)
Nanoparticles , Serum Albumin, Human , Binding Sites , Circular Dichroism , Colforsin/pharmacology , Humans , Protein Binding , Serum Albumin, Human/metabolism , Spectrometry, Fluorescence , Thermodynamics
5.
Bioorg Chem ; 88: 102960, 2019 07.
Article in English | MEDLINE | ID: mdl-31102808

ABSTRACT

In a search for novel multifunctional anti-Alzheimer agents, a congeneric set of seventeen flavone-8-acrylamide derivatives (8a─q) were synthesized and evaluated for their cholinesterase inhibitory, antioxidant, neuroprotective and modulation of Aß aggregation activities. The target compounds showed effective and selective inhibitory activity against the AChE over BuChE. In addition, the target compounds also showed moderate anti-oxidant activity and strong neuroprotective capacities, and accelerated dosage-dependently the Aß aggregation. Also, we presented here a complete study on the interaction of 8a, 8d, 8e, 8h and 8i with AChE. Through fluorescence emission studies, the binding sites number found to be 1, binding constants were calculated as 2.04 × 104, 2.22 × 104, 1.18 × 104, 9.8 × 103 and 3.2 × 104 M-1 and free energy change as -5.83, -5.91, -5.51, -5.41 and -6.12 kcal M-1 at 25 °C which were well agreed with the computational calculations indicating a strong binding affinity of flavones and AChE. Furthermore, the CD studies revealed that the secondary structure of AChE became partly unfolded upon binding with 8a, 8d, 8e, 8h and 8i.


Subject(s)
Acetylcholinesterase/metabolism , Acrylamide/pharmacology , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/antagonists & inhibitors , Cholinesterase Inhibitors/pharmacology , Flavones/pharmacology , Neuroprotective Agents/pharmacology , Acrylamide/chemical synthesis , Acrylamide/chemistry , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Binding Sites/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/chemistry , Dose-Response Relationship, Drug , Flavones/chemical synthesis , Flavones/chemistry , Humans , Hydrogen Peroxide/antagonists & inhibitors , Hydrogen Peroxide/pharmacology , Molecular Structure , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry , Protein Aggregates/drug effects , Structure-Activity Relationship , Thermodynamics
6.
Mol Pharm ; 15(6): 2206-2223, 2018 06 04.
Article in English | MEDLINE | ID: mdl-29745222

ABSTRACT

In line with the modern multi-target-directed ligand paradigm of Alzheimer's disease (AD), a series of 19 compounds composed of flavone and cyanoacetamide groups have been synthesized and evaluated as multifunctional agents against AD. Biological evaluation demonstrated that compounds 7j, 7n, 7o, 7r, and 7s exhibited excellent inhibitory potency (AChE, IC50 of 0.271 ± 0.012 to 1.006 ± 0.075 µM) and good selectivity toward acetylcholinesterase, significant antioxidant activity, good modulation effects on self-induced Aß aggregation, low cytotoxicity, and neuroprotection in human neuroblastoma SK-N-SH cells. Further, an inclusive study on the interaction of 7j, 7n, 7o, 7r, and 7s with AChE at physiological pH 7.2 using fluorescence, circular dichroism, and molecular docking methods suggested that these derivatives bind strongly to the peripheral anionic site of AChE mostly through hydrophobic interactions. Overall, the multifunctional profiles and strong AChE binding affinity highlight these compounds as promising prototypes for further pursuit of innovative multifunctional drugs for AD.


Subject(s)
Acetylcholinesterase/metabolism , Alzheimer Disease/drug therapy , Drug Design , Neuroprotective Agents/pharmacology , Acetylcholinesterase/chemistry , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Antioxidants/chemistry , Antioxidants/pharmacology , Antioxidants/therapeutic use , Cell Line, Tumor , Cholinergic Agents/chemistry , Cholinergic Agents/pharmacology , Cholinergic Agents/therapeutic use , Drug Evaluation, Preclinical , Enzyme Assays , Flavones/chemistry , Humans , Molecular Docking Simulation , Neuroprotective Agents/chemistry , Neuroprotective Agents/therapeutic use , Nitriles/chemistry , Protein Aggregates/drug effects , Protein Binding
9.
J Photochem Photobiol B ; 160: 248-59, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27130964

ABSTRACT

Here, we present the inclusive binding mode of phytochemical embelin, an anticancer drug with human serum albumin (HSA) established under physiological condition. Also, to understand the pharmacological role of embelin molecule, here, we have studied the anti-cancer activity of embelin on human cervical cancer cell line (HeLa cell line), which revealed that embelin showed dose dependent inhibition in the growth of cancer cells and also induces 26.3% of apoptosis at an IC50 value of 29µM. Further, embelin was titrated with HSA and the fluorescence emission quenching of HSA due to the formation of the HSA-embelin complex was observed. The binding constant of this complex is 5.9±.01×10(4)M(-1) and the number of bound embelin molecules is approximately 1.0. Consequently, molecular displacement and computational docking experiments show that the embelin is binding to subdomain IB to HSA. Further evidence from microTOF-Q mass spectrometry showed an increase in mass from 66,563Da to 66,857Da observed for free HSA and HSA+embelin complex, signifying that there is robust binding of embelin with HSA. In addition, the variations of HSA secondary structural elements in presence of embelin were confirmed by circular dichroism which indicates partial unfolding of protein. Furthermore, the transmission electron micrographs established that complex formation leads to aggregation of HSA plus embelin. Molecular dynamics simulations revealed that the stability of the HSA-embelin complexes and results suggests that at around 3500ps the complex reaches equilibration state which clearly contributes to the understanding of the stability of the HSA-embelin complexes.


Subject(s)
Benzoquinones/metabolism , Serum Albumin/metabolism , Apoptosis/drug effects , Benzoquinones/chemistry , Benzoquinones/toxicity , Binding Sites , Circular Dichroism , HeLa Cells , Humans , Microscopy, Electron, Transmission , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding , Protein Stability , Protein Structure, Tertiary , Serum Albumin/chemistry , Spectrometry, Fluorescence , Spectrometry, Mass, Electrospray Ionization , Thermodynamics
10.
Mol Biosyst ; 11(4): 1172-83, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25710711

ABSTRACT

Lupeol, a triterpene, possesses beneficial effects like anti-inflammatory and anti-cancer properties. Binding of lupeol and its derivative (phytochemicals) to plasma proteins such as human serum albumin (HSA) and α-1-acid glycoprotein (AGP) is a major determinant in the disposition of drugs. Cytotoxic studies with mouse macrophages (RAW 246.7) and HeLa cell lines revealed anti-inflammatory and anti-cancer properties for both lupeol and lupeol derivative. Both molecules reduced the expression of pro-inflammatory cytokines in LPS induced macrophages. Further, apoptosis was observed in HeLa cell lines when they were incubated with these molecules for 24 h. The fluorescence quenching of HSA was observed upon titration with different concentrations of lupeol and lupeol derivative; their binding constants were found to be 3 ± 0.01 × 10(4) M(-1) and 6.2 ± 0.02 × 10(4) M(-1), with binding free energies of -6.59 kcal M(-1) and -7.2 kcal M(-1). With AGP, however, the lupeol and lupeol derivative showed binding constants of 0.9 ± 0.02 × 10(3) M(-1) and 2.7 ± 0.01 × 10(3) M(-1), with free energies of -4.6 kcal M(-1) and -5.1 kcal M(-1) respectively. Molecular displacement studies based on competition with site I-binding phenylbutazone (which binds site I of HSA) and ibuprofen (which binds site II) suggest that lupeol binds site II and the lupeol derivative site I. Molecular docking studies also confirmed that lupeol binds to the IIIA and the lupeol derivative to the IIA domain of HSA. Secondary structure changes were observed upon formation of HSA-lupeol/lupeol derivative complexes by circular dichroism spectroscopy. Molecular dynamics simulations support greater stability of HSA-lupeol and HSA-lupeol derivative complexes compared to that of HSA alone.


Subject(s)
Blood Proteins/chemistry , Blood Proteins/metabolism , Pentacyclic Triterpenes/chemistry , Pentacyclic Triterpenes/metabolism , Animals , Apoptosis/drug effects , Cell Line , Cell Survival/drug effects , Circular Dichroism , Cytokines/analysis , Cytokines/metabolism , HeLa Cells , Humans , Mice , Molecular Docking Simulation , Molecular Dynamics Simulation , Pentacyclic Triterpenes/toxicity , Protein Binding , Spectrometry, Fluorescence
11.
J Biomol Struct Dyn ; 33(6): 1336-51, 2015.
Article in English | MEDLINE | ID: mdl-25054206

ABSTRACT

Human serum albumin (HSA) and α-1-acid glycoprotein (AGP) (acute phase protein) are the plasma proteins in blood system which transports many drugs. To understand the pharmacological importance of piperine molecule, here, we studied the anti-inflammatory activity of piperine on mouse macrophages (RAW 264.7) cell lines, which reveals that piperine caused an increase in inhibition growth of inflammated macrophages. Further, the fluorescence maximum quenching of proteins were observed upon binding of piperine to HSA and AGP through a static quenching mechanism. The binding constants obtained from fluorescence emission were found to be K(piperine) = 5.7 ± .2 × 10(5) M(-1) and K(piperine) = 9.3± .25 × 10(4) M(-1) which correspond to the free energy of -7.8 and -6.71 kcal M(-1)at 25 °C for HSA and AGP, respectively. Further, circular dichrosim studies revealed that there is a marginal change in the secondary structural content of HSA due to partial destabilization of HSA-piperine complexes. Consequently, inference drawn from the site-specific markers (phenylbutazone, site I marker) studies to identify the binding site of HSA noticed that piperine binds at site I (IIA), which was further authenticated by molecular docking and molecular dynamic (MD) studies. The binding constants and free energy corresponding to experimental and computational analysis suggest that there are hydrophobic and hydrophilic interactions when piperine binds to HSA. Additionally, the MD studies have showed that HSA-piperine complex reaches equilibration state at around 3 ns, which prove that the HSA-piperine complex is stable in nature.


Subject(s)
Alkaloids/chemistry , Benzodioxoles/chemistry , Orosomucoid/chemistry , Piperidines/chemistry , Polyunsaturated Alkamides/chemistry , Serum Albumin/chemistry , Algorithms , Alkaloids/metabolism , Alkaloids/pharmacology , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/pharmacology , Benzodioxoles/metabolism , Benzodioxoles/pharmacology , Binding Sites , Cell Line , Circular Dichroism , Humans , Hydrophobic and Hydrophilic Interactions , Macrophages/drug effects , Macrophages/metabolism , Mice , Models, Theoretical , Molecular Docking Simulation , Molecular Dynamics Simulation , Orosomucoid/metabolism , Piperidines/metabolism , Piperidines/pharmacology , Polyunsaturated Alkamides/metabolism , Polyunsaturated Alkamides/pharmacology , Protein Binding , Protein Conformation , Protein Stability , Protein Structure, Secondary , Serum Albumin/metabolism , Thermodynamics
12.
Mol Biosyst ; 10(12): 3101-10, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25209359

ABSTRACT

L-Dopa has been used to increase dopamine concentrations in the treatment of Parkinson's disease and dopamine-responsive dystonia. The binding interaction between L-dopa (phytochemical) and human serum albumin (HSA) under simulated physiological conditions was investigated by spectroscopic and molecular modeling methods. The results revealed that L-dopa caused fluorescence emission quenching of HSA through a static quenching procedure and the binding constant obtained was 2.3 ± 0.01 × 10(4) M(-1), which is corresponding to -5.9 kcal M(-1) of free energy at 25 °C. Interestingly, L-dopa is not binding to the α-1-acidglycoprotein, which is also a plasma protein and an acute phase protein. Furthermore, circular dichroism results confirm that in the presence of L-dopa the secondary structure of HSA is altered due to partial unfolding of the protein. Importantly, the displacement experiment with site specific probes, phenylbutazone (site I) and ibuprofen (site II), depicts that L-dopa binds particularly to site II of HSA. In addition, the molecular modeling results also confirmed that L-dopa is binding to the subdomain IIIA of HSA and is stabilized by hydrogen bonds and hydrophilic forces. Additionally, the molecular dynamic simulation studies showed that the HSA-L-dopa complex reaches an equilibration state at around 2 ns, which indicates that the HSA-L-dopa complex is very stable. These results provided valuable information of pharmacological mechanisms of L-dopa under in vivo conditions and play a pivotal role in the development of L-dopa-inspired drugs.


Subject(s)
Levodopa/chemistry , Serum Albumin/chemistry , Binding Sites , Circular Dichroism , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Molecular Dynamics Simulation , Protein Binding , Protein Structure, Secondary , Spectrometry, Fluorescence
13.
Mol Pharm ; 11(4): 1117-31, 2014 Apr 07.
Article in English | MEDLINE | ID: mdl-24495045

ABSTRACT

Human serum albumin (HSA) is one of the most widely studied proteins and is an important plasma protein responsible for binding and transport of many exogenous and endogenous drugs. Coumarin derivatives play a critical role as anticancer, antidiabetic, anticoagulant, and analgesic agents. Here we have studied the cytotoxic activity of 7-hydroxycoumarin derivatives (7HC-1, 7HC-2, and 7HC-3) on mouse macrophage (RAW 264.7) cell lines. These studies revealed that 7-hydroxycoumarin derivatives caused an increased inhibition in growth of inflamed macrophages in a concentration-dependent manner with an IC50 of 78, 63, and 50 µM. Further studies, using fluorescence, circular dichroism spectroscopy, molecular docking, and molecular dynamics methods, show binding of 7HC (umbelliferone) derivatives with HSA at physiological pH 7.2. The binding constant of 7HC derivatives with HSA obtained from fluorescence emission was found to be K7HC-1 = 4.6 ± 0.01 × 10(4) M(-1), K7HC-2 = 1.3 ± 0.01 × 10(4) M(-1), and K7HC-3 = 7.9 ± 0.01 × 10(4) M(-1) which corresponds to -6.34 kcal/mol, -5.58 kcal/mol, and -6.65 kcal/mol of free energy. In contrast, the binding of these coumarin derivatives (7HC-1, 7HC-2, and 7HC-3) was almost negligible with α-1-glycoprotein (AGP). Circular dichroism (CD) studies revealed a decreased α-helix content with an increase in the ß-sheets and random coils in HSA upon interaction with coumarin derivatives, suggesting a partial unfolding of the HSA secondary structure. Site probe studies with phenylbutazone (Site I) and ibuprofen (Site II) indicated that 7HC derivatives specifically bind to sub domains IIIA and IIIB of HSA which is further corroborated by molecular dynamics and docking studies suggesting that binding is specific in nature. The values of free energies and binding constants coincide for both experimental and in silico analysis and suggest that there are hydrophobic interactions when coumarin derivatives bind to HSA. Molecular dynamics studies showed that the HSA-coumarin complex reaches an equilibration state at around 3.5 ns which indicates that the HSA-coumarin complexes were stable. Thus these interactions play a central role in development of coumarin derivative-inspired drugs.


Subject(s)
Coumarins/pharmacology , Molecular Dynamics Simulation , Serum Albumin/metabolism , Umbelliferones/pharmacology , Animals , Cells, Cultured , Circular Dichroism , Coumarins/chemistry , Coumarins/metabolism , Humans , Mice , Molecular Docking Simulation , Protein Binding , Serum Albumin/chemistry , Spectrometry, Fluorescence , Thermodynamics
SELECTION OF CITATIONS
SEARCH DETAIL
...