1
|
Đurđević Đelmaš A, Šeba T, Gligorijević N, Pavlović M, Gruden M, Nikolić M, Milcic K, Milčić M. Perfluoroalkyl acids interact with major human blood protein fibrinogen: Experimental and computation study. Int J Biol Macromol 2025; 306:141425. [PMID: 40010474 DOI: 10.1016/j.ijbiomac.2025.141425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/19/2025] [Accepted: 02/22/2025] [Indexed: 02/28/2025]
Abstract
PFAS (per- and polyfluorinated alkyl substances) are synthetic compounds prized for their stability across various industries, but they pose an increasing threat to the environment and human health. Following the regulation of long-chain PFAS, short-chain and ultra-short-chain molecules have been introduced as substitutes, yet their bioaccumulation potential remains poorly understood. In this study, we combined experimental (intrinsic fluorescence, microscale thermophoresis, clotting assays) and computational approaches to investigate how trifluoroacetic acid, perfluorobutanoic acid, and perfluorooctanoic acid bind to fibrinogen, a key human blood protein. All tested perfluoroalkyl acids (PFAAs) exhibited moderate binding affinity (Kd in the 10-4-10-5 M range), yet circular dichroism and fibrin clot formation assays revealed no functional impairment of fibrinogen. Molecular docking indicated distinct, chain-length-specific binding sites, suggesting multiple routes for PFAAs to interact with fibrinogen without disrupting its primary biological role. These findings challenge the assumption that short-chain PFAS are less bioaccumulative and underscore the need for further research into their long-term health impacts, particularly given their widespread presence in the environment and potential accumulation in human blood.
Collapse
Affiliation(s)
| | - Tino Šeba
- Department of General and Inorganic Chemistry, Faculty of Pharmacy and Biochemistry, University of Zagreb, 10000 Zagreb, Croatia
| | - Nikola Gligorijević
- University of Belgrade, Institute of Chemistry, Technology and Metallurgy, National Institute of the Republic of Serbia, Studentski trg 12-16, 11000 Belgrade, Serbia
| | - Marko Pavlović
- School of Engineering and Applied Sciences, Harvard University, 11 Oxford Street, 02138 Cambridge, MA, USA; BioSense Institute, University of Novi Sad, Dr Zorana Djindjica 1, Novi Sad 21000, Serbia
| | - Maja Gruden
- University of Belgrade - Faculty of Chemistry, Studentski trg 12-16, 11000 Belgrade, Serbia
| | - Milan Nikolić
- University of Belgrade - Faculty of Chemistry, Studentski trg 12-16, 11000 Belgrade, Serbia
| | - Karla Milcic
- University of Belgrade - Faculty of Chemistry, Studentski trg 12-16, 11000 Belgrade, Serbia; School of Engineering and Applied Sciences, Harvard University, 11 Oxford Street, 02138 Cambridge, MA, USA; BioSense Institute, University of Novi Sad, Dr Zorana Djindjica 1, Novi Sad 21000, Serbia.
| | - Miloš Milčić
- University of Belgrade - Faculty of Chemistry, Studentski trg 12-16, 11000 Belgrade, Serbia.
| |
Collapse
|
2
|
Ali MS, Al-Lohedan HA, Bhati R, Muthukumaran J. Interaction of the lysozyme with anticoagulant drug warfarin: Spectroscopic and computational analyses. Heliyon 2024; 10:e30818. [PMID: 38784535 PMCID: PMC11112289 DOI: 10.1016/j.heliyon.2024.e30818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 05/02/2024] [Accepted: 05/06/2024] [Indexed: 05/25/2024] Open
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.
Collapse
Affiliation(s)
- Mohd Sajid Ali
- Department of Chemistry, College of Science, King Saud University, P.O. Box-2455, Riyadh, 11451, Saudi Arabia
| | - Hamad A. Al-Lohedan
- Department of Chemistry, College of Science, King Saud University, P.O. Box-2455, Riyadh, 11451, Saudi Arabia
| | - Rittik Bhati
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, India
| | - Jayaraman Muthukumaran
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, India
| |
Collapse
|
3
|
Jana G, Sing S, Das A, Basu A. Interaction of food colorant indigo carmine with human and bovine serum albumins: A multispectroscopic, calorimetric, and theoretical investigation. Int J Biol Macromol 2024; 259:129143. [PMID: 38176484 DOI: 10.1016/j.ijbiomac.2023.129143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/21/2023] [Accepted: 12/28/2023] [Indexed: 01/06/2024]
Abstract
In this work we have studied the interaction of the food dye Indigo-Carmine (IndC) with the most studied model transport proteins i.e. human and bovine serum albumin (HSA & BSA). A multispectroscopic approach was used to analyze the details of the binding process. The intrinsic fluorescence of both the albumins was significantly quenched by IndC and the quenching was both static and dynamic in nature with the former being dominant. The HSA-lndC and BSA-IndC distance after complexation was determined by Förster resonance energy transfer (FRET) method which suggested efficient energy transfer from the albumins to IndC. Thermodynamics of serum protein-IndC complexation was estimated by isothermal titration calorimetry (ITC) which revealed that the binding was enthalpy driven. Circular dichroism (CD) and FTIR spectroscopy revealed that the binding of IndC induced secondary structural changes in both the serum proteins. Synchronous and 3D fluorescence spectroscopy revealed that the binding interaction caused microenvironmental changes of protein fluorophores. Molecular docking analysis suggested that hydrogen bonding and hydrophobic interactions are the major forces involved in the complexation process.
Collapse
Affiliation(s)
- Gouranga Jana
- Department of Chemistry, Vidyasagar University, Midnapore 721 102, India
| | - Shukdeb Sing
- Department of Chemistry, Vidyasagar University, Midnapore 721 102, India
| | - Arindam Das
- Department of Chemistry, Vidyasagar University, Midnapore 721 102, India
| | - Anirban Basu
- Department of Chemistry, Vidyasagar University, Midnapore 721 102, India.
| |
Collapse
|
4
|
Ali MS, Waseem M, Subbarao N, Alahamed AN, Al-Lohedan HA. Probing the interaction of cephalosporin antibiotic "cefoperazone" with lysozyme using spectroscopic and in silico methods: Effect of paracetamol on binding. Int J Biol Macromol 2023; 252:126568. [PMID: 37640184 DOI: 10.1016/j.ijbiomac.2023.126568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 08/16/2023] [Accepted: 08/25/2023] [Indexed: 08/31/2023]
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.
Collapse
Affiliation(s)
- Mohd Sajid Ali
- Surfactant Research Chair, Department of Chemistry, College of Science, King Saud University, P.O. Box-2455, Riyadh 11451, Saudi Arabia.
| | - Mohd Waseem
- School of Computational and Integrative Science, Jawaharlal Nehru University, New Delhi, India
| | - Naidu Subbarao
- School of Computational and Integrative Science, Jawaharlal Nehru University, New Delhi, India
| | - Abdullah Nasser Alahamed
- Surfactant Research Chair, Department of Chemistry, College of Science, King Saud University, P.O. Box-2455, Riyadh 11451, Saudi Arabia
| | - Hamad A Al-Lohedan
- Surfactant Research Chair, Department of Chemistry, College of Science, King Saud University, P.O. Box-2455, Riyadh 11451, Saudi Arabia
| |
Collapse
|
5
|
Jiang SL, Hu ZY, Wang WJ, Hu L, Li L, Kou SB, Shi JH. Investigation on the binding behavior of human α1-acid glycoprotein with Janus Kinase inhibitor baricitinib: Multi-spectroscopic and molecular simulation methodologies. Int J Biol Macromol 2023:125096. [PMID: 37285878 DOI: 10.1016/j.ijbiomac.2023.125096] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 06/09/2023]
Abstract
Baricitinib is a Janus Kinase (JAK) inhibitor that is primarily used to treat moderately to severely active rheumatoid arthritis in adults and has recently been reported for the treatment of patients with severe COVID-19. This paper describes the investigation of the binding behavior of baricitinib to human α1-acid glycoprotein (HAG) employing a variety of spectroscopic techniques, molecular docking and dynamics simulations. Baricitinib can quench the fluorescence from amino acids in HAG through a mix of dynamic and static quenching, according to steady-state fluorescence and UV spectra observations, but it is mainly static quenching at low concentration. The binding constant (Kb) of baricitinib to HAG at 298 K was at the level of 104 M-1, indicating a moderate affinity of baricitinib to HAG. Hydrogen bonding and hydrophobic interactions conducted the main effect, according to thermodynamic characteristics, competition studies between ANS and sucrose, and molecular dynamics simulations. For the change in HAG conformation, the results of multiple spectra showed that baricitinib was able to alter the secondary structure of HAG as well as increase the polarity of the microenvironment around the Trp amino acid. Furthermore, the binding behavior of baricitinib to HAG was investigated by molecular docking and molecular dynamics simulations, which validated experimental results. Also explored is the influence of K+, Co2+, Ni2+, Ca2+, Fe3+, Zn2+, Mg2+ and Cu2+plasma on binding affinity.
Collapse
Affiliation(s)
- Shao-Liang Jiang
- College of Pharmaceutic Science, Zhejiang University of Technology, Hangzhou 310032, China.
| | - Zhe-Ying Hu
- College of Pharmaceutic Science, Zhejiang University of Technology, Hangzhou 310032, China
| | - Wan-Jun Wang
- College of Pharmaceutic Science, Zhejiang University of Technology, Hangzhou 310032, China
| | - Lu Hu
- College of Pharmaceutic Science, Zhejiang University of Technology, Hangzhou 310032, China
| | - Li Li
- College of Pharmaceutic Science, Zhejiang University of Technology, Hangzhou 310032, China
| | - Song-Bo Kou
- College of Pharmaceutic Science, Zhejiang University of Technology, Hangzhou 310032, China
| | - Jie-Hua Shi
- College of Pharmaceutic Science, Zhejiang University of Technology, Hangzhou 310032, China.
| |
Collapse
|
6
|
Nedić O, Penezić A, Minić S, Radomirović M, Nikolić M, Ćirković Veličković T, Gligorijević N. Food Antioxidants and Their Interaction with Human Proteins. Antioxidants (Basel) 2023; 12:antiox12040815. [PMID: 37107190 PMCID: PMC10135064 DOI: 10.3390/antiox12040815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/22/2023] [Accepted: 03/25/2023] [Indexed: 03/29/2023] Open
Abstract
Common to all biological systems and living organisms are molecular interactions, which may lead to specific physiological events. Most often, a cascade of events occurs, establishing an equilibrium between possibly competing and/or synergistic processes. Biochemical pathways that sustain life depend on multiple intrinsic and extrinsic factors contributing to aging and/or diseases. This article deals with food antioxidants and human proteins from the circulation, their interaction, their effect on the structure, properties, and function of antioxidant-bound proteins, and the possible impact of complex formation on antioxidants. An overview of studies examining interactions between individual antioxidant compounds and major blood proteins is presented with findings. Investigating antioxidant/protein interactions at the level of the human organism and determining antioxidant distribution between proteins and involvement in the particular physiological role is a very complex and challenging task. However, by knowing the role of a particular protein in certain pathology or aging, and the effect exerted by a particular antioxidant bound to it, it is possible to recommend specific food intake or resistance to it to improve the condition or slow down the process.
Collapse
Affiliation(s)
- Olgica Nedić
- Institute for the Application of Nuclear Energy, Department for Metabolism, University of Belgrade, Banatska 31b, 11080 Belgrade, Serbia
- Correspondence:
| | - Ana Penezić
- Institute for the Application of Nuclear Energy, Department for Metabolism, University of Belgrade, Banatska 31b, 11080 Belgrade, Serbia
| | - Simeon Minić
- Center of Excellence for Molecular Food Sciences, Department of Biochemistry, Faculty of Chemistry, University of Belgrade, 11000 Belgrade, Serbia
| | - Mirjana Radomirović
- Center of Excellence for Molecular Food Sciences, Department of Biochemistry, Faculty of Chemistry, University of Belgrade, 11000 Belgrade, Serbia
| | - Milan Nikolić
- Center of Excellence for Molecular Food Sciences, Department of Biochemistry, Faculty of Chemistry, University of Belgrade, 11000 Belgrade, Serbia
| | - Tanja Ćirković Veličković
- Center of Excellence for Molecular Food Sciences, Department of Biochemistry, Faculty of Chemistry, University of Belgrade, 11000 Belgrade, Serbia
- Serbian Academy of Sciences and Arts, Knez Mihailova 35, 11000 Belgrade, Serbia
| | - Nikola Gligorijević
- Institute for the Application of Nuclear Energy, Department for Metabolism, University of Belgrade, Banatska 31b, 11080 Belgrade, Serbia
| |
Collapse
|
7
|
Ali MS, Muthukumaran J, Jain M, Tariq M, Al-Lohedan HA, Al-Sanea ASS. Detailed Experimental and In Silico Investigation of Indomethacin Binding with Human Serum Albumin Considering Primary and Secondary Binding Sites. Molecules 2023; 28:molecules28072979. [PMID: 37049745 PMCID: PMC10095894 DOI: 10.3390/molecules28072979] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/19/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
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.
Collapse
Affiliation(s)
- Mohd Sajid Ali
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
- Correspondence:
| | - Jayaraman Muthukumaran
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, India
| | - Monika Jain
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, India
| | - Mohammad Tariq
- LAQV-REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal
| | - Hamad A. Al-Lohedan
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Abdullah Saad S. Al-Sanea
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| |
Collapse
|
8
|
Li X, Han L, Song Z, Xu R, Wang L. Comparative study on the interaction between transferrin and flavonols: Experimental and computational modeling approaches. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2023; 288:122128. [PMID: 36455462 DOI: 10.1016/j.saa.2022.122128] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/27/2022] [Accepted: 11/14/2022] [Indexed: 06/17/2023]
Abstract
Transferrin is the indispensable component in the body fluids and has been explored as a potential drug carrier for target drugs to cancer cells. Flavonols are widely distributed in plants and shown a wide range of biological activities. In the present study, the interaction between flavonols (including galangin, kaempferol, quercetin, and myricetin) and transferrin under physiological conditions was investigated by using experimental as well as computational approaches. Fluorescence data reveal that the fluorescence quenching mechanism of transferrin by flavonols is static quenching. Transferrin has moderate affinity with flavonols, and the binding constants (Ka) are 103-104 L/mol. In addition, there are two different binding sites for the interaction between kaempferol and transferrin. Thermodynamic parameter analysis shows that the interaction of flavonols and transferrin is synergistically driven by enthalpy and entropy. Hydrophobic interaction, electrostatic force and hydrogen bonds are the main force types. Synchronous fluorescence spectroscopy shows that flavonols decrease the hydrophobicity of the microenvironment around tryptophan (Trp) and have no effect on the microenvironment around tyrosine (Tyr). UV-vis and CD spectra show that the interaction between transferrin and flavonols leads to the loosening and unfolding of transferrin backbone. The increase of β-sheet is accompanied by the decrease of α-helix and β-turn. The specific binding sites of flavonols to transferrin are confirmed by molecular docking. Molecular dynamic simulation suggests that the transferrin-flavonols docked complex is stable throughout the simulation trajectory.
Collapse
Affiliation(s)
- Xiangrong Li
- Department of Medical Chemistry, Key Laboratory of Medical Molecular Probes, School of Basic Medicine, Xinxiang Medical University, Xinxiang, Henan 453003, PR China.
| | - Linyu Han
- Grade 2020, Clinical Medicine, School of Basic Medicine, Xinxiang Medical University, Xinxiang, Henan 453003, PR China
| | - Zhizhi Song
- Grade 2020, Clinical Medicine, School of Basic Medicine, Xinxiang Medical University, Xinxiang, Henan 453003, PR China
| | - Ruonan Xu
- Department of Medical Chemistry, Key Laboratory of Medical Molecular Probes, School of Basic Medicine, Xinxiang Medical University, Xinxiang, Henan 453003, PR China
| | - Lixia Wang
- College of Science, Henan Agricultural University, Zhengzhou, Henan 450002, PR China
| |
Collapse
|
9
|
Ali MS, Rehman MT, Al-Lohedan HA, AlAjmi MF. Exploration of the binding between cuminol and bovine serum albumin through spectroscopic, molecular docking and molecular dynamics methods. J Biomol Struct Dyn 2022; 40:12404-12412. [PMID: 34488560 DOI: 10.1080/07391102.2021.1971560] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
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.
Collapse
Affiliation(s)
- Mohd Sajid Ali
- Surfactant Research Chair, Department of Chemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Md Tabish Rehman
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Hamad A Al-Lohedan
- Surfactant Research Chair, Department of Chemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Mohamed Fahad AlAjmi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
10
|
Spectroscopic and Molecular Docking Investigation on the Interaction of Cumin Components with Plasma Protein: Assessment of the Comparative Interactions of Aldehyde and Alcohol with Human Serum Albumin. Int J Mol Sci 2022; 23:ijms23084078. [PMID: 35456897 PMCID: PMC9028573 DOI: 10.3390/ijms23084078] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/03/2022] [Accepted: 04/04/2022] [Indexed: 01/27/2023] Open
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.
Collapse
|
11
|
Ali MS, Al-Lohedan HA. Experimental and Computational Investigation on the Interaction of Anticancer Drug Gemcitabine with Human Plasma Protein: Effect of Copresence of Ibuprofen on the Binding. Molecules 2022; 27:1635. [PMID: 35268736 PMCID: PMC8912049 DOI: 10.3390/molecules27051635] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/25/2022] [Accepted: 02/26/2022] [Indexed: 12/24/2022] Open
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.
Collapse
Affiliation(s)
- Mohd Sajid Ali
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia;
| | | |
Collapse
|
12
|
Experimental and in silico investigation on the interaction of indomethacin with bovine serum albumin: Effect of sodium dodecyl sulfate surfactant monomers on the binding. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2021.116858] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
13
|
Liao X, Zhu C, Huang D, Wen X, Zhang SL, Shen Y. Profiling the interaction of a novel toxic pyruvate dehydrogenase kinase inhibitor with human serum albumin. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2021; 256:119733. [PMID: 33827040 DOI: 10.1016/j.saa.2021.119733] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 03/16/2021] [Accepted: 03/18/2021] [Indexed: 06/12/2023]
Abstract
To discover novel pyruvate dehydrogenase kinase (PDK) inhibitors, a new compound 2,2-dichloro-1-(4-((4-isopropylphenyl)amino)-3-nitrophenyl)ethan-1-one, namely XB-1 was identified, which inhibited PDK activity with a half maximal inhibitory concentration (IC50) value of 337.0 nM, and reduced A549 cell proliferation with a half maximal effective concentration (EC50) value of 330.0 nM. However, the compound appears to exhibit a negligible selectivity between cancer cell and normal one, indicating a potential toxicity existed for the compound. Herein, the interaction of the toxic XB-1 to human serum albumin (HSA) was firstly explored by spectroscopic approaches with the aim to reduce/avoid the toxicity of PDK inhibitors in the next hit-to-lead campaign. In detail, it was found that the XB-1 could effectively bind to HSA mainly via hydrogen bond interaction in PBS buffer (pH = 7.4, 10.0 mM), resulting in the formation of HSA-XB-1 complex. The negative value of ΔG showed that the binding of XB-1 to HSA is a spontaneous process. The result from site-selective binding assay suggested that the XB-1 bound to the site I of HSA by competing with warfarin, which was perfect in agreement with the molecular docking method. The results of this paper may offer a valuable theoretical basis to study the toxicity of biofunctional molecules and may offer thoughts about how to avoid/reduce toxicity for a small molecule.
Collapse
Affiliation(s)
- Xianjiu Liao
- West Guangxi Key Laboratory for Prevention and Treatment of High-Incidence Diseases, Youjiang Medical University for Nationalities, Baise 533000, China
| | - Chunlei Zhu
- School of Food & Biological Engineering, Hefei University of Technology, Hefei 230009, China
| | - Ding Huang
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China
| | - Xiaoqing Wen
- West Guangxi Key Laboratory for Prevention and Treatment of High-Incidence Diseases, Youjiang Medical University for Nationalities, Baise 533000, China
| | - Shao-Lin Zhang
- School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, China.
| | - Yizhong Shen
- School of Food & Biological Engineering, Hefei University of Technology, Hefei 230009, China.
| |
Collapse
|
14
|
Ali MS, Muthukumaran J, Jain M, Al-Lohedan HA, Farah MA, Alsowilem OI. Experimental and computational investigation on the binding of anticancer drug gemcitabine with bovine serum albumin. J Biomol Struct Dyn 2021; 40:9144-9157. [PMID: 33998966 DOI: 10.1080/07391102.2021.1924270] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
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.
Collapse
Affiliation(s)
- Mohd Sajid Ali
- Surfactant Research Chair, Department of Chemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Jayaraman Muthukumaran
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, India
| | - Monika Jain
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, India
| | - Hamad A Al-Lohedan
- Surfactant Research Chair, Department of Chemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - M Abul Farah
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Osama Ibrahim Alsowilem
- Surfactant Research Chair, Department of Chemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
15
|
Ali MS, Waseem M, Subbarao N, Al-Lohedan HA. Noncovalent molecular interactions between antineoplastic drug gemcitabine and a carrier protein identified through spectroscopic and in silico methods. Int J Biol Macromol 2021; 182:993-1002. [PMID: 33857514 DOI: 10.1016/j.ijbiomac.2021.04.049] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/31/2021] [Accepted: 04/07/2021] [Indexed: 12/11/2022]
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.
Collapse
Affiliation(s)
- Mohd Sajid Ali
- Department of Chemistry, King Saud University, P.O. Box-2455, Riyadh 11451, Saudi Arabia.
| | - Mohd Waseem
- School of Computational and Integrative Science, Jawaharlal Nehru University, New Delhi, India
| | - Naidu Subbarao
- School of Computational and Integrative Science, Jawaharlal Nehru University, New Delhi, India
| | - Hamad A Al-Lohedan
- Department of Chemistry, King Saud University, P.O. Box-2455, Riyadh 11451, Saudi Arabia
| |
Collapse
|
16
|
Ali MS, Waseem M, Subbarao N, Al-Lohedan HA. Dynamic interaction between lysozyme and ceftazidime: Experimental and molecular simulation approaches. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2021.115412] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
17
|
Li B, Sun Y, Lu J, Peng X. Investigation on the binding interaction of rhodamine B with human serum albumin: effect of metal ions. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART. B, PESTICIDES, FOOD CONTAMINANTS, AND AGRICULTURAL WASTES 2021; 56:259-271. [PMID: 33459173 DOI: 10.1080/03601234.2021.1873030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The binding of rhodamine B (RB) to human serum albumin (HSA) in the absence and presence of Cu2+ or Fe3+ under simulated physiological conditions was studied by using various biophysical methods for the first time. The results showed that the interaction between HSA and RB could spontaneously result in the formation of HSA-RB complex (namely, static quenching mechanism) through hydrophobic interactions and hydrogen bonds irrespective of the absence or presence of metal ions. The presence of metal ions led to the reduction of binding affinity of RB to HSA compared with no metal ions, which might result from the conformational change of HSA caused by the binding of metal ions. Furthermore, the analysis of UV-vis absorption, circular dichroism, synchronous fluorescence and three-dimensional fluorescence experiments demonstrated that the addition of RB induced conformational and microenvironmental changes of HSA without and with metal ions. In short, this work will be helpful to in-depth understand the transport mechanism and biological effect of RB and the effect of metal ions on the interaction of HSA-RB in vivo.
Collapse
Affiliation(s)
- Baicun Li
- College of Life Science and Food Engineering, Shaanxi Xueqian Normal University, Xi'an, Shaanxi, People's Republic of China
| | - Yinhe Sun
- Tianjin Institute of Metrological Supervision and Testing, Tianjin, People's Republic of China
| | - Jing Lu
- Xi'an Modern Chemistry Research Institute, Xi'an, Shaanxi, People's Republic of China
| | - Xin Peng
- School of Life Sciences, Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, Tianjin University, Tianjin, People's Republic of China
| |
Collapse
|
18
|
Konar M, Sahoo H. Tyrosine mediated conformational change in bone morphogenetic protein – 2: Biophysical implications of protein – phytoestrogen interaction. Int J Biol Macromol 2020; 150:727-736. [DOI: 10.1016/j.ijbiomac.2020.02.113] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 02/10/2020] [Accepted: 02/11/2020] [Indexed: 01/18/2023]
|
19
|
Ali MS, Al-Lohedan HA. Spectroscopic and Molecular Docking Investigation on the Noncovalent Interaction of Lysozyme with Saffron Constituent "Safranal". ACS OMEGA 2020; 5:9131-9141. [PMID: 32363265 PMCID: PMC7191604 DOI: 10.1021/acsomega.9b04291] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 03/26/2020] [Indexed: 05/05/2023]
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.
Collapse
|
20
|
Experimental and computational investigation on the molecular interactions of safranal with bovine serum albumin: Binding and anti-amyloidogenic efficacy of ligand. J Mol Liq 2019. [DOI: 10.1016/j.molliq.2019.01.034] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
21
|
Akram M, Ansari F. Biophysical investigation of the interaction between cationic biodegradable C m-E2O-C m gemini surfactants and porcine serum albumin (PSA). SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2019; 206:520-528. [PMID: 30176428 DOI: 10.1016/j.saa.2018.08.043] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/14/2018] [Accepted: 08/24/2018] [Indexed: 06/08/2023]
Abstract
The interaction of porcine serum albumin (PSA) with biodegradable, less cytotoxic Cm-E2O-Cm gemini surfactants was monitored using state-of-the-art techniques. The temperature variation fluorescence experiments were used to derive the thermodynamic parameters of non-covalent interaction in PSA-Cm-E2O-Cm gemini systems, which indicate an exothermic and a hydrogen bonding/Van der Waals force predominated binding process. Synchronous fluorescence spectra indicate that tryptophan fluorescence gets more quenched than the tyrosine fluorescence. The pyrene micropolarity assay signifies that pyrene is subjected to mild micropolarity changes. UV absorption spectra verify the ground state complexation between PSA and Cm-E2O-Cm geminis. Far-UV CD spectra reveal negligible changes in secondary structure with respect to PSA in its native state. These results indicate that the cationic Cm-E2O-Cm geminis, at lower concentrations, substantially bind to PSA but do not disrupt its secondary structure. These observations are favorable for the potential utilization of the concerned geminis in the field of drug delivery, especially in self-emulsifying drug delivery systems.
Collapse
Affiliation(s)
- Mohd Akram
- Department of Chemistry, Aligarh Muslim University, Aligarh 202002, India.
| | - Farah Ansari
- Department of Chemistry, Aligarh Muslim University, Aligarh 202002, India
| |
Collapse
|
22
|
Ronzetti M, Baljinnyam B, Yasgar A, Simeonov A. Testing for drug-human serum albumin binding using fluorescent probes and other methods. Expert Opin Drug Discov 2018; 13:1005-1014. [PMID: 30320522 PMCID: PMC11369766 DOI: 10.1080/17460441.2018.1534824] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
INTRODUCTION Drug plasma protein binding remains highly relevant to research and drug development, making the assessment and profiling of compound affinity to plasma proteins essential to drug discovery efforts. Although there are a number of fully-characterized methods, they lack the throughput to handle large numbers of compounds. As the evaluation of adsorption, distribution, metabolism, and excretion is addressed earlier in the drug development timeline, the need for higher-throughput methods has grown. Areas Covered: This review will highlight recent developments on methods for profiling drug plasma binding, with an emphasis on fluorescent probes and emerging high-throughput methodologies. Expert Opinion: There have been a number of high-throughput assays developed in recent years to meet the scaled up demands for compound profiling. Ultimately, the selection of assay technology relies on a number of factors, such as capabilities of the laboratory and the breadth and amount of data required. Fluorescent probe displacement assays are highly flexible and amenable to high-throughput screening, easily scaling up to handle large compound libraries. Recent developments in fluorescence technologies, such as homogenous time-resolved fluorescence and probes utilizing the aggregation-induced emission effect, have improved the sensitivity of these assays. Other technologies, such as microscale thermophoresis and quantitative structure-activity relationship modeling, are gaining popularity as alternative techniques for drug plasma protein binding characterization.
Collapse
Affiliation(s)
- Michael Ronzetti
- a National Center for Advancing Translational Sciences , National Institutes of Health , Rockville , Maryland , USA
| | - Bolormaa Baljinnyam
- a National Center for Advancing Translational Sciences , National Institutes of Health , Rockville , Maryland , USA
| | - Adam Yasgar
- a National Center for Advancing Translational Sciences , National Institutes of Health , Rockville , Maryland , USA
| | - Anton Simeonov
- a National Center for Advancing Translational Sciences , National Institutes of Health , Rockville , Maryland , USA
| |
Collapse
|
23
|
Ali MS, Al-Lohedan HA. Spectroscopic and computational evaluation on the binding of safranal with human serum albumin: Role of inner filter effect in fluorescence spectral correction. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2018; 203:434-442. [PMID: 29894957 DOI: 10.1016/j.saa.2018.05.102] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 05/21/2018] [Accepted: 05/27/2018] [Indexed: 05/21/2023]
Abstract
For determining the pharmacological properties of medicinal compounds, their binding with serum albumins is very crucial. Herein, we have selected safranal, a major constituent of saffron which is known to retain a number of medicinal properties including antioxidant, anti-inflammatory, tumoricidal, anti-genotoxic, and anti-aging activities; and studied its mechanism of binding with human serum albumin at physiological pH using various spectroscopic methods along with computational approach using molecular docking. A change in the difference UV-visible spectrum of HSA in presence of safranal was found which is due to the complex formation. Owing to the strong absorption of safranal at the fluorescence excitation wavelength of HSA (295 nm) and in the whole range of emission, the fluorescence spectra of HSA in presence of safranal were corrected for the inner filter effect. After the correction the spectra were free from the safranal absorption effect and it was found that addition of safranal causes the quenching of HSA fluorescence and a blue shift of the emission maximum which are attributed to the binding of safranal to the protein and dominance of hydrophobic forces in the interaction, respectively. It was evident from the comparison of observed and corrected fluorescence spectra that before correction there was a large red shift while after correction appearance of blue shift was occurred. The involvement of hydrophobic interaction was also found from the extrinsic fluorescence measurements using ANS dye as well as from the analyzed thermodynamic parameters. Safranal was found to partially induce the secondary structure of HSA as construed from the CD measurements. The size of the HSA was also decreased as evident from the DLS and RLS measurements. Both site marker studies and molecular docking simulations suggested that the primary binding site of the safranal in the HSA is Sudlow's site 1 located in the subdomain IIA. Hydrophobic interaction provides the major contribution to the binding forces along with a little amount of hydrogen bonding.
Collapse
Affiliation(s)
- Mohd Sajid Ali
- Surfactant Research Chair, Department of Chemistry, College of Science, King Saud University, P.O. Box-2455, Riyadh 11451, Saudi Arabia.
| | - Hamad A Al-Lohedan
- Surfactant Research Chair, Department of Chemistry, College of Science, King Saud University, P.O. Box-2455, Riyadh 11451, Saudi Arabia
| |
Collapse
|
24
|
Zhuo W, Peng X, Lin X. Insights into the interaction mechanism between tiagabine hydrochloride and two serum albumins. RSC Adv 2018; 8:24953-24960. [PMID: 35542170 PMCID: PMC9082336 DOI: 10.1039/c8ra04153a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 06/29/2018] [Indexed: 12/03/2022] Open
Abstract
Tiagabine hydrochloride (TGB) is a widely used anticonvulsive drug for the treatment of epilepsy. To better understand the interactions of TGB with plasma proteins, human serum albumin (HSA) and bovine serum albumin (BSA) were selected as model proteins. TGB slightly increased thermal stability of the proteins as confirmed by VP-capillary differential scanning calorimetric (DSC) measurements. Isothermal titration calorimeter (ITC) results showed that TGB could be combined with HSA and BSA moderately, which was also corroborated by fluorescence analysis. Besides, the thermodynamic parameters (ΔH > 0, ΔS > 0) indicated that hydrophobic forces played a major role in the formulation of TGB-HSA and TGB-BSA complexes. Moreover, the main binding sites of TGB to HSA and BSA were also examined by classical fluorescent probe (dansylsarcosine and dansylamide) experiments, showing that TGB and dansylsarcosine competitively interacted with HSA and BSA at the same binding sites. Additionally, TGB had no obvious effect on the conformation change of HSA and BSA as indicated by spectroscopic analyses. This study provides useful information about the interaction mechanism of TGB and serum albumins, which could help to better utilize TGB in biomedical field.
Collapse
Affiliation(s)
- Weiling Zhuo
- School of Chemical Engineering, Sichuan University Chengdu 610065 People's Republic of China +86 028 85990908 +86 02885990908
| | - Xilin Peng
- Sinopharm Chuankang Pharmaceutical Co., Ltd. Chengdu 611731 People's Republic of China
| | - Xiang Lin
- School of Chemical Engineering, Sichuan University Chengdu 610065 People's Republic of China +86 028 85990908 +86 02885990908
| |
Collapse
|