1
|
Zhang W, Gao L, Zhang S, Luo J, Yu R, Li X, Lu Z, Chi B, Guo Y, Tuo X. Structural and functional alterations of human serum albumin (HSA) induced by TBBPS-BME: A spectroscopic, computational, and molecular dynamics study. Chem Biol Interact 2025; 413:111498. [PMID: 40174686 DOI: 10.1016/j.cbi.2025.111498] [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: 01/02/2025] [Revised: 03/18/2025] [Accepted: 03/29/2025] [Indexed: 04/04/2025]
Abstract
Tetrabromobisphenol S Dimethyl Ether (TBBPS-BME) serves as a substitute for Tetrabromobisphenol A (TBBPA). It exhibits a greater potential for bioaccumulation, potentially posing substantial risks to environmental health and human safety. This research utilized a variety of spectroscopic and computational modeling methods to investigate structural and functional alterations of human serum albumin (HSA) induced by TBBPS-BME. The findings revealed that TBBPS-BME induces fluorescence quenching in HSA by binding to site I. The predominant force in this binding process is hydrophobic interaction. The binding constant for this complex is 2.394 ± 0.032 × 104 M-1 at 298 K, suggesting that they can form complex in vivo. The interaction with TBBPS-BME causes structural alterations in HSA, leading to a decrease in α-helix proportion and an overall enhancement of protein structural flexibility. The esterase-like function of HSA was also impacted by the presence of TBBPS-BME. Computational simulation tests indicate that this may be through interactions with residues Lys199 and Lys195. Molecular dynamics simulations further validated the formation of stable TBBPS-BME-HSA binary complexes, highlighting the critical role of hydrogen bonds in this steadiness. Alanine scanning mutation analysis revealed that Trp214, Phe211, Arg218, Lys199, and His242 are necessary for the assembly of the TBBPS-BME-HSA complex. Overall, this study provides an exhaustive examination of the binding mechanisms between TBBPS-BME and HSA, elucidating the underlying health hazards that may arise from exposure to TBBPS-BME.
Collapse
Affiliation(s)
- Wei Zhang
- Jiangxi Key Laboratory of Flood and Drought Disaster Defense, Jiangxi Academy of Water Science and Engineering, Nanchang, 330029, China; Jiangxi Provincial Eco-hydraulic Technology Innovation Center of Poyang Lake Basin, Jiangxi Academy of Water Science and Engineering, Nanchang, 330029, China; Research Center for Water Resources and Ecological Environment of Poyang Lake, The Ministry of Water Resources of the People's Republic of China, Nanchang, 330029, China
| | - Linna Gao
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Shuyuan Zhang
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Jiaqing Luo
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Ruoxuan Yu
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Xiting Li
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Zhili Lu
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Baozhu Chi
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Ying Guo
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, 330031, Jiangxi, China.
| | - Xun Tuo
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, 330031, Jiangxi, China.
| |
Collapse
|
2
|
Nazari M, Emamzadeh R, Masoudi-Khoram N, Nazari M. Immobilization of albumin binding domain (ABD) on Sepharose 4B and magnetic particle for efficient single-step purification of human serum albumin. J Chromatogr B Analyt Technol Biomed Life Sci 2025; 1261:124655. [PMID: 40403581 DOI: 10.1016/j.jchromb.2025.124655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 05/04/2025] [Accepted: 05/13/2025] [Indexed: 05/24/2025]
Abstract
Human serum albumin (HSA) is an important protein in plasma with various biological functions in the human body. Due to its unique features in the binding and transfer of ligands and pharmaceutical molecules, HSA is extensively used in therapeutics and pharmaceutical approaches. Commercial albumin is produced by a multi-step process of plasma fractionation. However, this traditional method has some limitations such as risk of contamination, low quality, and quantity of the purified final protein. In this study, we developed two affinity chromatography platforms for the purification of human serum albumin. The recombinant albumin-binding domain (ABD) was expressed and purified using molecular biology techniques. Two types of commercial beads-Cyanogen bromide-activated Sepharose 4B and amine-functionalized magnetic particles-were then functionalized with the recombinant ABD. Protein purification using chromatography columns demonstrated that HSA can be purified to 95 % purity in a single step. Circular dichroism (CD) spectroscopy revealed structural similarities in HSA purified through affinity chromatography and fractionation using the Cohen method. Furthermore, the study of aspirin binding to HSA demonstrated that proteins purified via affinity chromatography and those fractionated by the Cohen method exhibited identical drug-binding affinities. The results of this study may have important implications for the clinical purification of human serum albumin.
Collapse
Affiliation(s)
| | - Rahman Emamzadeh
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Nastaran Masoudi-Khoram
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | - Mahboobeh Nazari
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran; Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran.
| |
Collapse
|
3
|
Yang X, Li T, Chen X, Zhang H, Liu C, Tao C, Nie H. Tetraphenylethylene-indole as a novel fluorescent probe for selective and sensitive detection of human serum albumin (HSA) in biological matrices and monitoring of HSA purity and degradation. Talanta 2025; 286:127471. [PMID: 39736207 DOI: 10.1016/j.talanta.2024.127471] [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: 11/04/2024] [Revised: 12/09/2024] [Accepted: 12/26/2024] [Indexed: 01/01/2025]
Abstract
Human serum albumin (HSA) levels in serum and urine is a crucial biomarker for diagnosing liver and kidney diseases. HSA is used to treat various disorders in clinical practice and as an excipient in the production of vaccine or protein drug, ensuring its purity essential for patient safety. However, selective and sensitive detection of HSA remains challenging due to its structural similarity with bovine serum albumin (BSA) and the inherent complexity of biological matrices. This study presents a novel application of the tetraphenylethylene-indole (TPE-indo) fluorophore for the identification and quantification of HSA. The findings demonstrate that TPE-indo binds specifically to HSA in a 1:1 M ratio, thereby triggering its aggregation-induced emission (AIE) mechanism and producing a selective, sensitive, and rapid "turn-on" fluorescence response. The fluorescence intensity of TPE-indo exhibited minimal interference from proteins, amino acids, sugars, ions, and urine metabolites, and demonstrated a linear correlation with HSA concentration up to 60 μg/mL, with a limit of detection of 0.30 μg/mL. Furthermore, TPE-indo displays a markedly enhanced response to HSA in comparison to BSA, which can be ascribed to the distinct binding modes between TPE-indo and these two proteins. TPE-indo can be used to quantify HSA in serum, grade proteinuria samples, detect BSA adulteration in HSA samples, and real-time monitor HSA degradation processes. This study not only advances the development of efficient HSA detection methods but also highlights the significance of TPE-indo as a versatile tool for bioanalysis and clinical diagnosis.
Collapse
Affiliation(s)
- Xueping Yang
- Key Laboratory of Public Health Safety of Hebei Province, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, School of Public Health, Hebei University, Baoding, 071002, PR China
| | - Taoran Li
- Key Laboratory of Public Health Safety of Hebei Province, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, School of Public Health, Hebei University, Baoding, 071002, PR China
| | - Xu Chen
- Key Laboratory of Public Health Safety of Hebei Province, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, School of Public Health, Hebei University, Baoding, 071002, PR China
| | - Huan Zhang
- Key Laboratory of Public Health Safety of Hebei Province, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, School of Public Health, Hebei University, Baoding, 071002, PR China
| | - Chao Liu
- Department of Statistics, Hebei University, Baoding, 071002, PR China.
| | - Chenchuang Tao
- Key Laboratory of Public Health Safety of Hebei Province, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, School of Public Health, Hebei University, Baoding, 071002, PR China
| | - Hailiang Nie
- Key Laboratory of Public Health Safety of Hebei Province, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, School of Public Health, Hebei University, Baoding, 071002, PR China
| |
Collapse
|
4
|
Sookai S, Akerman M, Færch M, Sayed Y, Munro OQ. Cytotoxic pyrrole-based gold(III) chelates target human topoisomerase II as dual-mode inhibitors and interact with human serum albumin. Eur J Med Chem 2025; 287:117330. [PMID: 39904146 DOI: 10.1016/j.ejmech.2025.117330] [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: 12/23/2024] [Revised: 01/24/2025] [Accepted: 01/24/2025] [Indexed: 02/06/2025]
Abstract
Topoisomerase IIα (Top II) is a critical enzyme that resolves DNA topology during transcription and replication. Inhibitors of Top II are used as anticancer agents and are classified as interfacial poisons (IFPs) or catalytic inhibitors (CICs). Here, we report a novel class of cytotoxic, stable cationic gold(III) Schiff base chelates (AuL1, AuL2, and AuL3) with DNA-intercalating properties. In the NCI-60 screen, AuL1 and AuL3 exhibited potent cytotoxicity (mean GI50 values of 11 (7) μM and 14 (9) μM, respectively), whereas AuL2 showed minimal cytotoxicity. Cluster analysis aligned AuL1 and AuL3 with the Top II poison etoposide. Mechanistic studies revealed that AuL1 acts as an IFP at concentrations between 0.5 and 50 μM and as a CIC at concentrations between 50 and 500 μM. Further investigations demonstrated that all three gold(III) chelates bind to and intercalate DNA, the main substrate for Top II. Finally, binding studies with human serum albumin (HSA) indicated that the chelates have moderate affinity for the protein. Thermodynamic analysis indicates entropically driven binding, with minimal structural disruption observed via UV-CD spectroscopy. These findings highlight the dual mode Top II inhibition mechanism delineated for the gold(III) chelates and their favourable pharmacodynamic interactions with HSA, underscoring their potential as promising anticancer agents.
Collapse
Affiliation(s)
- Sheldon Sookai
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand, PO WITS 2050, Johannesburg, South Africa.
| | - Matthew Akerman
- School of Chemistry, University of KwaZulu-Natal, Pietermaritzburg, 3201, South Africa
| | - Mia Færch
- Protein Structure-Function Research Unit, School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg, 2050, South Africa
| | - Yasien Sayed
- Protein Structure-Function Research Unit, School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg, 2050, South Africa
| | - Orde Q Munro
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand, PO WITS 2050, Johannesburg, South Africa; School of Chemistry, University of Leeds, Woodhouse Lane, LS2 9JT, UK
| |
Collapse
|
5
|
Shahabadi N, Marzbani A, Hadidi S, Mardani Z. Spectroscopic and molecular docking investigation on the interaction of a water-soluble Cu(II) complex containing diethanolamine and dipicolinic acid ligands with human serum albumin. J Biomol Struct Dyn 2025; 43:3366-3374. [PMID: 38147399 DOI: 10.1080/07391102.2023.2297812] [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: 01/29/2023] [Accepted: 12/15/2023] [Indexed: 12/28/2023]
Abstract
Under physiological conditions, spectroscopic techniques as well as molecular docking simulation have been used to investigate the binding interaction mechanism between Cu(II) complex containing Pyridine-2,6-dicarboxylic acid (PDCA) and Diethanolamine (DEA) ligands, [Cu(DEA)(PDCA)] and human serum albumin (HSA). UV spectral changes of protein in the presence of the Cu(II) complex suggested the formation of a Protein-Cu(II) complex conjugate with specific new structure. The Cu(II) complex quenches the intrinsic fluorescence of the HSA via a static mechanism in which van der Waals interactions along with hydrogen bonds are fundamental binding forces. Displacement experiments performed by warfarin and ibuprofen site probes predict that the Cu(II) complex is located in subdomain IIA, Sudlow site 1 of HSA. Molecular docking results showed close resemblance with experimental data.
Collapse
Affiliation(s)
- Nahid Shahabadi
- Department of Inorganic Chemistry, Faculty of Chemistry, Razi University, Kermanshah, Iran
| | - Azadeh Marzbani
- Department of Inorganic Chemistry, Faculty of Chemistry, Razi University, Kermanshah, Iran
| | - Saba Hadidi
- Department of Inorganic Chemistry, Faculty of Chemistry, Razi University, Kermanshah, Iran
| | - Zahra Mardani
- Inorganic Chemistry Department, Faculty of Chemistry, Urmia University, Urmia, Iran
| |
Collapse
|
6
|
Zheng Y, Duan XY, Wang X, Wang XF, Liu B. Insight into the effect of ZIF-8 on the interaction between drugs and protein/cell. Int J Biol Macromol 2025; 294:139530. [PMID: 39761886 DOI: 10.1016/j.ijbiomac.2025.139530] [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: 09/03/2024] [Revised: 12/17/2024] [Accepted: 01/03/2025] [Indexed: 02/20/2025]
Abstract
Understanding the impact of nanomaterials on drug-protein/cell interactions is crucial for comprehending their in vivo biological effects. We investigated the impact of zeolitic imidazolate framework (ZIF)-8 on the interaction between curcumin (Cur) and human serum albumin (HSA) using various spectroscopic techniques and molecular docking. Additionally, we examined its effect on drug-cell interaction using HepG2 cells and Escherichia coli (E. coli). The UV-vis spectra and fluorescence results demonstrated the occurrence of an interaction between Cur-HSA and ZIF-8, potentially resulting in the formation of ground-state complexes. ZIF-8 did not alter the static quenching mechanism, interaction force type, and binding stoichiometry between Cur and HSA, but it induced subtle changes in the secondary structure and esterase activity of HSA. Cur predominantly binds in the site I of HSA. Molecular docking analysis confirmed the results. The incorporation of ZIF-8 enhanced the antitumor activity of Cur-HSA and the antibacterial efficacy of ciprofloxacin (CIP)-HSA, while concurrently enhancing the uptake of CIP by E. coli. These results indicate that the influence of ZIF-8 on drug-protein interactions may consequently exert a significant impact on drug efficacy.
Collapse
Affiliation(s)
- Ying Zheng
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xin-Yue Duan
- School of Pharmaceutical Sciences, Liaoning University, Shenyang 110036, China
| | - Xin Wang
- School of Pharmaceutical Sciences, Liaoning University, Shenyang 110036, China; Shenyang Key Laboratory for Causes and Drug Discovery of Chronic Diseases, Liaoning University, Shenyang 110036, China
| | - Xiao-Fang Wang
- School of Pharmaceutical Sciences, Liaoning University, Shenyang 110036, China.
| | - Bin Liu
- School of Pharmaceutical Sciences, Liaoning University, Shenyang 110036, China.
| |
Collapse
|
7
|
Otero-Luis I, Martínez-Rodrigo A, Cavero-Redondo I, Moreno-Herráiz N, López-López S, Saz-Lara A. Comparative effect of oral drugs in improving spasticity of different etiology: a network meta-analysis. Postgrad Med J 2025; 101:212-218. [PMID: 39348794 DOI: 10.1093/postmj/qgae126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 08/09/2024] [Accepted: 09/06/2024] [Indexed: 10/02/2024]
Abstract
INTRODUCTION Spasticity, a prevalent manifestation of various neurological conditions, significantly impacts the quality of life of patients. Research on the effects of oral drugs on spasticity has produced controversial results. Thus, the aim of this network meta-analysis was to compare the efficacy of oral drugs for improving spasticity in patients with different etiologies. METHODS We searched four different databases from their inception to 30 November 2023. A network meta-analysis using a frequentist perspective was conducted to assess the effects of different oral drugs on spasticity, evaluated by the modified Ashworth scale. RESULTS Our findings showed that, in a frequentist network meta-analysis, eperisone, diazepam, and baclofen had significantly greater spasticity, as measured by the modified Ashworth scale, than did the placebo (MD: -0.80; 95% CIs: -1.42, -0.18; MD: -0.68; 95% CIs: -1.28, -0.09; MD: -0.58; 95% CIs: -1.11, -0.06, respectively). CONCLUSION In summary, our study confirmed that eperisone, diazepam, and baclofen could be effective approaches for reducing spasticity of different etiologies and could be useful approaches for improving patient quality of life. Key messages What is already known on this topic: The impact of oral drugs, such as baclofen, gabapentin, tizanidine, and dantrolene, in the treatment of spasticity has been documented. What this study adds: This study determines which of the oral drugs aimed at treating spasticity is the most effective across different etiologies. How this study might affect research, practice, or policy: This study suggests tailored treatment strategies for spasticity based on its etiology.
Collapse
Affiliation(s)
- Iris Otero-Luis
- CarVasCare Research Group (2023-GRIN-34459), Facultad de Enfermería de Cuenca, Universidad de Castilla-La Mancha, 16002 Cuenca, Spain
| | | | - Iván Cavero-Redondo
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca 3460000, Chile
| | - Nerea Moreno-Herráiz
- CarVasCare Research Group (2023-GRIN-34459), Facultad de Enfermería de Cuenca, Universidad de Castilla-La Mancha, 16002 Cuenca, Spain
| | - Samuel López-López
- Castilla-La Mancha Health Services, SESCAM, Hospital of Cuenca, C/Hermandad de Donantes de Sangre, 1, 16002, Cuenca, Spain
| | - Alicia Saz-Lara
- CarVasCare Research Group (2023-GRIN-34459), Facultad de Enfermería de Cuenca, Universidad de Castilla-La Mancha, 16002 Cuenca, Spain
| |
Collapse
|
8
|
Kenéz B, Koplányi G, Decsi B, Molnár Z, Horváth P, Katona G, Balogh GT, Balogh-Weiser D. Development of a Novel Human Serum Albumin-Based Tool for Effective Drug Discovery: The Investigation of Protein Quality and Immobilization. J Med Chem 2025; 68:2840-2848. [PMID: 39813641 PMCID: PMC11831590 DOI: 10.1021/acs.jmedchem.4c02136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/30/2024] [Accepted: 01/03/2025] [Indexed: 01/18/2025]
Abstract
The binding ability of human serum albumin (HSA) on active pharmaceutical ingredients (APIs) is one of the most important parameters in the early stages of drug discovery. In this study, an immobilized HSA-based tool was developed for the rapid and easy in vitro screening of API binding. The work explored the serious incompleteness in the identification of HSA used for in vitro screening published in the last five years. To mitigate this problem, a comprehensive analysis and immobilization studies were performed on the most used HSA types. Serious differences in the colloidal stability of HSAs and their API binding ability on a selected set of APIs were observed. HSAs were immobilized on magnetic nanoparticles with glutardialdehyde (GDA) or cyclohexyl-diglycidyl ether (CDGE) linkers, which have never been used for HSA immobilization before. The HSA-MNP-CDGE complexes achieved a higher immobilization yield and preserved API binding ability; however, the esterase-like enzymatic activity of HSA reduced significantly.
Collapse
Affiliation(s)
- Balázs Kenéz
- Department
of Organic Chemistry and Technology, Budapest
University of Technology and Economics, Műegyetem rkp. 3, H-1111Budapest, Hungary
| | - Gábor Koplányi
- Department
of Organic Chemistry and Technology, Budapest
University of Technology and Economics, Műegyetem rkp. 3, H-1111Budapest, Hungary
| | - Balázs Decsi
- Department
of Chemical and Environmental Process Engineering, Budapest University of Technology and Economics, Műegyetem rkp. 3, H-1111Budapest, Hungary
| | - Zsófia Molnár
- Department
of Organic Chemistry and Technology, Budapest
University of Technology and Economics, Műegyetem rkp. 3, H-1111Budapest, Hungary
| | - Péter Horváth
- Department
of Pharmaceutical Chemistry, Semmelweis
University, Hőgyes E. Street 7−9, H-1092 Budapest, Hungary
| | - Gábor Katona
- Institute
of Pharmaceutical Technology and Regulatory Affairs, Faculty of Pharmacy, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary
| | - György T. Balogh
- Department
of Chemical and Environmental Process Engineering, Budapest University of Technology and Economics, Műegyetem rkp. 3, H-1111Budapest, Hungary
- Department
of Pharmaceutical Chemistry, Semmelweis
University, Hőgyes E. Street 7−9, H-1092 Budapest, Hungary
- Center
for Pharmacology and Drug Research & Development, Semmelweis University, Üllői Street 26, H-1085Budapest, Hungary
| | - Diána Balogh-Weiser
- Department
of Organic Chemistry and Technology, Budapest
University of Technology and Economics, Műegyetem rkp. 3, H-1111Budapest, Hungary
- Department
of Physical Chemistry and Materials Science, Budapest University of Technology and Economics, Műegyetem rkp. 3, H-1111Budapest, Hungary
| |
Collapse
|
9
|
Tong W, Liu X, Yang Y, Wang Y, Huang Z, Fan H. Molecular and technical aspects on the interaction of bovine serum albumin with pyrazine derivatives: From molecular docking to spectroscopy study. J Food Sci 2025; 90:e70017. [PMID: 39898927 DOI: 10.1111/1750-3841.70017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 12/22/2024] [Accepted: 01/09/2025] [Indexed: 02/04/2025]
Abstract
In order to better understand the transport and action mechanism of flavor substance and proteins in human body, the interaction mechanism between pyrazine derivatives and bovine serum albumin (BSA) was studied by molecular dynamics simulation and a series of spectroscopic methods. In molecular docking, it was observed that the small molecules were surrounded by hydrophobic amino acid residues of the protein, and the main amino acid residues formed π-π interaction and hydrogen bond interaction with BSA. The results of fluorescence emission spectroscopy combined with thermodynamic analysis showed that static quenching was the main mechanism of the interaction between three pyrazine derivatives and BSA, which was dominated by hydrophobic interaction. Synchronous fluorescence spectroscopy and three-dimensional fluorescence spectroscopy combined with molecular dynamics simulation proved that the pyrazine derivatives changed the conformation of BSA. In summary, pyrazine derivatives can interact with BSA, and the complexation of the complex changes its spatial conformation. The research in this paper has positive significance for understanding the binding, transport, and metabolism of pyrazine compounds in the process of blood circulation and provides key data for the metabolism of pyrazine compounds in vivo. PRACTICAL APPLICATION: The interaction of pyrazine derivatives-BSA is studied by multi-spectra and MD. The fluorescence quenching of pyrazine derivatives-BSA is static quenching. The main force between pyrazine derivatives and BSA is hydrophobic force. There is only one site of association between pyrazine derivatives and BSA. Pyrazine derivatives have effects on conformation of BSA.
Collapse
Affiliation(s)
- Wenghua Tong
- School of Biological Engineering, Sichuan University of Science and Engineering, Yibin, China
- Liquor Brewing Biotechnology and Application Key Laboratory of Sichuan Province, Yibin, China
| | - XingYan Liu
- School of Biological Engineering, Sichuan University of Science and Engineering, Yibin, China
| | - Ying Yang
- School of Biological Engineering, Sichuan University of Science and Engineering, Yibin, China
| | - Yan Wang
- School of Biological Engineering, Sichuan University of Science and Engineering, Yibin, China
| | | | - Hongjun Fan
- Luzhou Guozhirongyao Liquor Co., Ltd., Luzhou, China
| |
Collapse
|
10
|
Tan X, Wang Y, Long L, Chen H, Qu L, Cao X, Li H, Chen Z, Luo S, Shi C. A theranostic photosensitizer-conjugated albumin co-loading with resiquimod for cancer-targeted imaging and robust photo-immunotherapy. Pharmacol Res 2024; 210:107489. [PMID: 39510147 DOI: 10.1016/j.phrs.2024.107489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 11/15/2024]
Abstract
Cancer immunotherapy remains a low immune response rate in clinic because of dominant immunosuppressive tumor microenvironment (TME) and lack of effective drug to specifically remodel the TME. In this work, we introduced a tumor-seeking human serum albumin (HSA) based delivery platform by covalently conjugating with a tumor-targeting near-infrared (NIR) photosensitizer (IR-DBI) and non-covalently loading of immune modulator Resiquimod (R848). HSA exhibited tumor-preferential accumulation after covalent conjugation with IR-DBI. Meanwhile, HSA restricted the rotation of IR-DBI, narrowed the HOMO-LUMO energy gap, significantly enhanced fluorescent intensity and dual-modal phototherapy (PTT/PDT). The enhanced phototherapeutic effect further induced robust ICD effect. More importantly, non-covalent loading of R848 could be released from HSA at tumor sites by laser irradiation-induced heat. The in-situ release of R848 in TME efficiently promoted the maturation of DC cells and repolarized M2 macrophages to M1 macrophages. Consequently, robust photo-induced antitumor immunity was triggered in the different mice models bearing primary and distant tumors or lung metastasis, which was further enhanced by combining with CTLA-4 blockade therapy. Taken together, this work may present a versatile albumin composite which exhibits tumor-preferential accumulation and imaging-guided PDT/PTT/immunotherapy.
Collapse
Affiliation(s)
- Xu Tan
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma and Chemical Poisoning, College of Preventive Medicine, Army Medical University, Gaotanyan Street 30, Chongqing 400038, China
| | - Yu Wang
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma and Chemical Poisoning, College of Preventive Medicine, Army Medical University, Gaotanyan Street 30, Chongqing 400038, China
| | - Lei Long
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma and Chemical Poisoning, College of Preventive Medicine, Army Medical University, Gaotanyan Street 30, Chongqing 400038, China
| | - Hongdan Chen
- Department of Breast and Thyroid Surgery, Chongqing General Hospital, Xingguang Road 118, Chongqing 401121, China
| | - Langfan Qu
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma and Chemical Poisoning, College of Preventive Medicine, Army Medical University, Gaotanyan Street 30, Chongqing 400038, China
| | - Xiaohui Cao
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma and Chemical Poisoning, College of Preventive Medicine, Army Medical University, Gaotanyan Street 30, Chongqing 400038, China
| | - Huijuan Li
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma and Chemical Poisoning, College of Preventive Medicine, Army Medical University, Gaotanyan Street 30, Chongqing 400038, China
| | - Zelin Chen
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma and Chemical Poisoning, College of Preventive Medicine, Army Medical University, Gaotanyan Street 30, Chongqing 400038, China.
| | - Shenglin Luo
- Institute of Combined Injury, State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Gaotanyan Street 30, Chongqing 400038, China.
| | - Chunmeng Shi
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma and Chemical Poisoning, College of Preventive Medicine, Army Medical University, Gaotanyan Street 30, Chongqing 400038, China.
| |
Collapse
|
11
|
Bora B, Das N, Bera A, Upadhyay A, Goswami TK. Fluorinated High-Valent Sn(IV) Porphyrins Show Remarkable Photodynamic Activity in Cancer Cells. ChemMedChem 2024; 19:e202400376. [PMID: 39017962 DOI: 10.1002/cmdc.202400376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 07/18/2024]
Abstract
In recent years, Sn(IV) porphyrins have proven to be excellent choice as photosensitizers for photodynamic therapy. This work reports the synthesis, characterization and photodynamic activity of four high-valent fluorinated Sn(IV) porphyrins having different numbers of F-atoms in the peripheral of meso-phenyl groups viz. (Dichloro)meso-tetrakis(4-fluorophenylporphyrinato)stannic(IV), [Sn(IV)FTPP(Cl)2] or Sn1; (Dichloro)meso-tetrakis(2,4-difluorophenylporphyrinato)stannic(IV), [Sn(IV)2,4-FTPP(Cl)2] or Sn2; (Dichloro)meso-tetrakis(2,6-difluorophenylporphyrinato)stannic(IV), [Sn(IV)2,6-FTPP(Cl)2] or Sn3 and (Dichloro)meso-tetrakis(4-trifluoromethylphenylporphyrinato)stannic(IV), [Sn(IV)CF3TPP(Cl)2] or Sn4. The solid-state structure of Sn1 has been determined by single crystal X-ray diffraction analysis. The increasing number of F-atoms attached to the meso-phenyl positions of the porphyrin framework results in increase of their lipophilicity, singlet oxygen quantum yield (ΦΔ) and photocytotoxicity in A549 (human lung adenocarcinoma cells), MCF-7 and MDA-MB-231 (human breast adenocarcinoma) cells. Sn4 predominantly localize in the mitochondria of A549 cells. The light-induced cell death by the Sn(IV) porphyrins in A549 cells occur primarily via apoptosis.
Collapse
Affiliation(s)
- Bidisha Bora
- Department of Chemistry, Gauhati University, Guwahati, Assam, 781014, India
| | - Namisha Das
- Department of Chemistry, Gauhati University, Guwahati, Assam, 781014, India
| | - Arpan Bera
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Aarti Upadhyay
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Tridib K Goswami
- Department of Chemistry, Gauhati University, Guwahati, Assam, 781014, India
| |
Collapse
|
12
|
Das R, Dash PP, Bishoyi AK, Mohanty P, Mishra L, Prusty L, Sahoo CR, Padhy RN, Mishra M, Sahoo H, Sahoo SK, Sethi SK, Jali BR. Antibacterial and cytotoxicity studies of pyrrolo-based organic scaffolds and their binding interaction with bovine serum albumin. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:8725-8743. [PMID: 38829386 DOI: 10.1007/s00210-024-03187-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 05/24/2024] [Indexed: 06/05/2024]
Abstract
Two pyrrolo-based compounds, 1H-pyrrolo[3,2-b]pyridine-3-carboxylic acid (L1) and 1H-pyrrolo[3,2-c]pyridine-4-carboxylic acid (L2), were employed for the detection of bovine serum albumin (BSA) by UV-Vis and fluorescence spectroscopic methods in phosphate buffer solution (pH = 7). In the presence of L1 and L2, the fluorescence emission of BSA at 340 nm was quenched and concomitantly a red-shifted emission band appeared at 420 nm (L1)/450 nm (L2). The fluorescence spectral changes indicate the protein-ligand complex formation between BSA and L1/L2. An isothermal titration calorimetry (ITC) experiment was conducted to determine the binding ability between BSA and L1/L2. The binding constants are found to be 4.45 ± 0.22 × 104 M-1 for L1 and 2.29 ± 0.11 × 104 M-1 for L2, respectively. The thermodynamic parameters were calculated from ITC measurements (i.e. ∆rH = -40 ± 2 kcal/mol, ∆rG = -4.57 ± 0.22 kcal/mol and -T∆rS = 35.4 ± 1.77 kcal/mol), which indicated that the protein-ligand complex formation between L1/L2 with BSA is mainly due to the electrostatic interactions. The protein-ligand interactions were studied by performing molecular docking. Further, the antibacterial assay of L1 and L2 was conducted against gram-positive and gram-negative bacterial strains in an effort to address the difficulties caused by the co-occurrence of antimicrobial and multidrug-resistant bacteria. E. coli and S. aureus were significantly inhibited by L1 and L2. The L1 exhibits 13, 12 and 15 mm, whereas L2 exhibits a 2, 3 and 5 mm zone of inhibition against S. aureus, S. pyogenes and E. coli, respectively. In silico molecular docking of L1 and L2 was performed with bacterial DNA gyrase to establish the intermolecular interactions. Finally, the in vitro cytotoxicity activities of the ligands L1 and L2 have been carried out using drosophila.
Collapse
Affiliation(s)
- Rosalin Das
- School of Biotechnology, Gangadhar Meher University, Sambalpur, 768001, Odisha, India
| | - Pragyan P Dash
- Department of Chemistry, Veer Surendra Sai University of Technology, Burla, Sambalpur, 768018, Odisha, India
| | - Ajit K Bishoyi
- Central Research Laboratory, Institute of Medical Sciences and SUM Hospital, Siksha 'O' Anusandhan Deemed University, Bhubaneswar, 751003, Odisha, India
| | - Patitapaban Mohanty
- Department of Chemistry, Veer Surendra Sai University of Technology, Burla, Sambalpur, 768018, Odisha, India
| | - Lokanath Mishra
- Department of Life Science, National Institute of Technology, Rourkela, 769008, India
| | - Laxmipriya Prusty
- Department of Chemistry, National Institute of Technology, Rourkela, 769008, India
| | - Chita R Sahoo
- Central Research Laboratory, Institute of Medical Sciences and SUM Hospital, Siksha 'O' Anusandhan Deemed University, Bhubaneswar, 751003, Odisha, India
| | - Rabindra N Padhy
- Central Research Laboratory, Institute of Medical Sciences and SUM Hospital, Siksha 'O' Anusandhan Deemed University, Bhubaneswar, 751003, Odisha, India
| | - Monalisa Mishra
- Department of Life Science, National Institute of Technology, Rourkela, 769008, India
| | - Harekrushna Sahoo
- Department of Chemistry, National Institute of Technology, Rourkela, 769008, India
| | - Suban K Sahoo
- Department of Chemistry, SV National Institute of Technology, Surat, 395007, Gujarat, India
| | - Santosh K Sethi
- School of Biotechnology, Gangadhar Meher University, Sambalpur, 768001, Odisha, India.
| | - Bigyan R Jali
- Department of Chemistry, Veer Surendra Sai University of Technology, Burla, Sambalpur, 768018, Odisha, India.
| |
Collapse
|
13
|
Li W, Sun L, Yang X, Peng C, Hua R, Zhu M. Enantioselective effects of chiral profenofos on the conformation for human serum albumin. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2024; 205:106159. [PMID: 39477612 DOI: 10.1016/j.pestbp.2024.106159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/23/2024] [Accepted: 09/28/2024] [Indexed: 11/07/2024]
Abstract
Profenofos, as a typical chiral organophosphorus pesticide, can cause various environmental problems and even endanger human health when used in excess. The toxicity of chiral profenofos was investigated through multispectral analysis, molecular docking, and density functional theory (DFT), employing human serum albumin (HSA) as the model protein. Fluorescence titration and lifetime measurements demonstrated that the interaction between chiral profenofos and HSA involves static quenching. Chiral profenofos forms a 1:1 complex with HSA at site II (subdomain IIIA), primarily driven by hydrophobic interactions and hydrogen bonds. Notably, the binding efficacy diminishes as temperature increases. Spectroscopic analyses confirm that chiral profenofos alters the microenvironment and structure of HSA, with the R-enantiomer exerting a greater impact than the S-enantiomer. Consequently, the toxicological implications of the R-profenofos is significantly more pronounced. Investigating the molecular-level toxic effects of chiral pesticides enhances the thoroughness of pesticide assessments, aids in understanding their distribution, metabolism, and associated risks, and facilitates the development of mitigation strategies.
Collapse
Affiliation(s)
- Wenze Li
- School of Chemical and Environmental Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, China
| | - Long Sun
- School of Chemical and Environmental Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, China
| | - Xiaofan Yang
- School of Chemical and Environmental Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, China
| | - Changsheng Peng
- School of Chemical and Environmental Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, China
| | - Rimao Hua
- Key Laboratory of Agri-Food Safety of Anhui Province, School of Resources and Environment, Anhui Agricultural University, No. 130 Changjiang West Road, Hefei 230036, China
| | - Meiqing Zhu
- School of Chemical and Environmental Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, China.
| |
Collapse
|
14
|
Gonzalez-Posada AH, Mesa M, Sierra L, Lopez B. Interactions of human serum albumin with phosphate and Tris buffers: impact on paclitaxel binding and nanoparticles self-assembly. J Microencapsul 2024; 41:564-575. [PMID: 39268923 DOI: 10.1080/02652048.2024.2389135] [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: 04/16/2024] [Accepted: 08/01/2024] [Indexed: 09/15/2024]
Abstract
AIM To investigate the conformational changes in human serum albumin (HSA) caused by chemical (CD) and thermal denaturation (TD) at pH 7.4 and 9.9, crucial for designing controlled drug delivery systems with paclitaxel (PTX). METHODS Experimental and computational methods, including differential scanning calorimetry (DSC), UV-Vis and intrinsic fluorescence spectroscopy, mean diameter, polydispersity index (PDI), ζ-potential, encapsulation efficiency (EE), in vitro release and protein docking studies were conducted to study the HSA denaturation and nanoparticles (NPs) preparation. RESULTS TD at pH 7.4 produced smaller NPs (287.1 ± 12.9 nm) than CD at pH 7.4 with NPs (584.2 ± 47.7 nm). TD at pH 9.9 exhibited high EE (97.3 ± 0.2%w/w) with rapid PTX release (50% within 1h), whereas at pH 7.4 (96.4 ± 2.1%w/w), release only 40%. ζ-potentials were around -30 mV. CONCLUSION Buffer type and pH significantly influence NP properties. TD in PBS at pH 7.4, provided optimal conditions for a stable and efficient drug delivery system.
Collapse
Affiliation(s)
- A H Gonzalez-Posada
- Materials Science Group, Institute of Chemistry, University of Antioquia, Medellín, Colombia
| | - M Mesa
- Materials Science Group, Institute of Chemistry, University of Antioquia, Medellín, Colombia
| | - L Sierra
- Materials Science Group, Institute of Chemistry, University of Antioquia, Medellín, Colombia
| | - B Lopez
- Materials Science Group, Institute of Chemistry, University of Antioquia, Medellín, Colombia
| |
Collapse
|
15
|
Jouyaeian P, Kamkar-Vatanparast M, Tehranian-Torghabeh F, Hoseinpoor S, Saberi MR, Chamani J. New perspective into the interaction behavior explore of Nano-berberine with alpha-lactalbumin in the presence of beta-lactoglobulin: Multi-spectroscopic and molecular dynamic investigations. J Mol Struct 2024; 1316:139020. [DOI: 10.1016/j.molstruc.2024.139020] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
16
|
Parsadanyan MA, Shahinyan MA, Mikaelyan MS, Grigoryan SV, Poghosyan GH, Vardevanyan PO. Influence of millimeter range electromagnetic waves on bovine serum albumin interaction with acridine orange. Electromagn Biol Med 2024; 43:246-255. [PMID: 39068541 DOI: 10.1080/15368378.2024.2383683] [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: 04/03/2024] [Accepted: 07/17/2024] [Indexed: 07/30/2024]
Abstract
The effect of non-ionizing millimeter range electromagnetic waves (MM EMW) (30-300 GHz) on the bovine serum albumin (BSA) interaction peculiarities with acridine orange (AO) has been studied in vitro. The frequencies 41.8 and 50.3 GHz were chosen, since the first one is nonresonant frequency for the water, while the second one is resonant for water. The binding constant and number of binding sites were calculated at both irradiation presence and absence. AO was revealed to bind to BSA, while after the protein irradiation the interaction force strengthens. However, it was also shown that there are differences of the interaction parameters while irradiating by 41.8 or 50.3 GHz. AO binds to BSA, irradiated by MM EMW with the frequency 41.8 GHz much more weaker, than to that, irradiated by MM EMW with the frequency 50.3 GHz.
Collapse
Affiliation(s)
- M A Parsadanyan
- Research Institute of Biology, Faculty of Biology, Yerevan State University, Yerevan, Armenia
| | - M A Shahinyan
- Research Institute of Biology, Faculty of Biology, Yerevan State University, Yerevan, Armenia
| | - M S Mikaelyan
- Research Institute of Biology, Faculty of Biology, Yerevan State University, Yerevan, Armenia
| | - S V Grigoryan
- Research Institute of Biology, Faculty of Biology, Yerevan State University, Yerevan, Armenia
| | - G H Poghosyan
- Research Institute of Biology, Faculty of Biology, Yerevan State University, Yerevan, Armenia
| | - P O Vardevanyan
- Research Institute of Biology, Faculty of Biology, Yerevan State University, Yerevan, Armenia
| |
Collapse
|
17
|
Eslami Moghadam M, Tavakoli Hafshajani K, Sohrabi N, Rezaeisadat M, Oftadeh M. Platinum (II) complex of isopentyl glycine ligand: DNA binding, molecular dynamic, and anticancer activity against breast cancer. J Biomol Struct Dyn 2024; 42:8229-8241. [PMID: 37578043 DOI: 10.1080/07391102.2023.2246564] [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: 03/30/2023] [Accepted: 07/29/2023] [Indexed: 08/15/2023]
Abstract
In this paper, we performed thorough experimental and theoretical calculations to examine the interaction between Pt derivative, as an anticancer, and ct-DNA. The mode of DNA binding with [Pt(NH3)2(Isopentylgly)]NO3, where Isopentylgly is Isopentyl glycine, was evaluated by various spectroscopic methods, docking, and molecular dynamics simulation studies. UV-Vis and fluorescence spectroscopic titration results and CD spectra of DNA-drug showed this interaction is via groove binding. Also, thermal stability studies or DNA melting temperature changes (ΔTm), as well as the quenching emissions monitoring proved it. Also, the thermodynamic parameter and binding constant displayed that complex-DNA formation is a spontaneous process, and H-binding and also groove binding were found to be the main forces. Theoretical studies stated [Pt(NH3)2(Isopentylgly)]NO3-DNA formation occurs on C-G center on DNA, along with rising DNA-compound stability. IC50 value against the human breast cell line probably is due to the Isopentyl glycine ligand in the structure of the Pt compound, and it was obtained more than cisplatin and less than carboplatin against the MCF7 cell.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
| | | | - Nasrin Sohrabi
- Department of Chemistry, Payame Noor University (PNU), Tehran, Iran
| | | | - Mohsen Oftadeh
- Department of Chemistry, Payame Noor University (PNU), Tehran, Iran
| |
Collapse
|
18
|
de Barros MC, de Oliveira APS, dos Santos FG, Silva FAC, Menezes TM, Seabra GDM, Yoneda JS, Coelho LCBB, Macedo MLR, Napoleão TH, Lima TDA, Neves JL, Paiva PMG. Carbohydrate-Binding Mechanism of the Coagulant Lectin from Moringa oleifera Seeds (cMoL) Is Related to the Dimeric Protein Structure. Molecules 2024; 29:4615. [PMID: 39407546 PMCID: PMC11477877 DOI: 10.3390/molecules29194615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/21/2024] [Accepted: 09/03/2024] [Indexed: 10/19/2024] Open
Abstract
This study characterized the binding mechanisms of the lectin cMoL (from Moringa oleifera seeds) to carbohydrates using spectroscopy and molecular dynamics (MD). The interaction with carbohydrates was studied by evaluating lectin fluorescence emission after titration with glucose or galactose (2.0-11 mM). The Stern-Volmer constant (Ksv), binding constant (Ka), Gibbs free energy (∆G), and Hill coefficient were calculated. After the urea-induced denaturation of cMoL, evaluations were performed using fluorescence spectroscopy, circular dichroism (CD), and hemagglutinating activity (HA) evaluations. The MD simulations were performed using the Amber 20 package. The decrease in Ksv revealed that cMoL interacts with carbohydrates via a static mechanism. The cMoL bound carbohydrates spontaneously (ΔG < 0) and presented a Ka on the order of 102, with high selectivity for glucose. Protein-ligand complexes were stabilized by hydrogen bonds and hydrophobic interactions. The Hill parameter (h~2) indicated that the binding occurs through the cMoL dimer. The loss of HA at urea concentrations at which the fluorescence and CD spectra indicated protein monomerization confirmed these results. The MD simulations revealed that glucose bound to the large cavity formed between the monomers. In conclusion, the biotechnological application of cMoL lectin requires specific methods or media to improve its dimeric protein structure.
Collapse
Affiliation(s)
- Matheus Cavalcanti de Barros
- Departamento de Bioquímica, Universidade Federal de Pernambuco, Recife 50670-901, PE, Brazil; (M.C.d.B.); (L.C.B.B.C.); (T.H.N.); (T.d.A.L.); (P.M.G.P.)
| | - Ana Patrícia Silva de Oliveira
- Departamento de Bioquímica, Universidade Federal de Pernambuco, Recife 50670-901, PE, Brazil; (M.C.d.B.); (L.C.B.B.C.); (T.H.N.); (T.d.A.L.); (P.M.G.P.)
| | - Franciane Gonçalves dos Santos
- Departamento de Química Fundamental, Universidade Federal de Pernambuco, Recife 50670-901, PE, Brazil; (F.G.d.S.); (T.M.M.); (J.L.N.)
| | | | - Thais Meira Menezes
- Departamento de Química Fundamental, Universidade Federal de Pernambuco, Recife 50670-901, PE, Brazil; (F.G.d.S.); (T.M.M.); (J.L.N.)
| | - Gustavo de Miranda Seabra
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development, College of Pharmacy, University of Florida, Gainseville, FL 32611, USA;
| | - Juliana Sakamoto Yoneda
- Laboratório Nacional de Luz Síncrotron, Centro Nacional de Pesquisa em Energia e Materiais, Campinas 13083-100, SP, Brazil;
| | | | - Maria Lígia Rodrigues Macedo
- Departamento de Tecnologia de Alimentos e da Saúde, Faculdade de Ciências Farmacêuticas, Alimentos e 22 Nutrição, Universidade Federal do Mato Grosso do Sul, Campo Grande 79070-900, MS, Brazil;
| | - Thiago Henrique Napoleão
- Departamento de Bioquímica, Universidade Federal de Pernambuco, Recife 50670-901, PE, Brazil; (M.C.d.B.); (L.C.B.B.C.); (T.H.N.); (T.d.A.L.); (P.M.G.P.)
| | - Thâmarah de Albuquerque Lima
- Departamento de Bioquímica, Universidade Federal de Pernambuco, Recife 50670-901, PE, Brazil; (M.C.d.B.); (L.C.B.B.C.); (T.H.N.); (T.d.A.L.); (P.M.G.P.)
| | - Jorge Luiz Neves
- Departamento de Química Fundamental, Universidade Federal de Pernambuco, Recife 50670-901, PE, Brazil; (F.G.d.S.); (T.M.M.); (J.L.N.)
| | - Patrícia Maria Guedes Paiva
- Departamento de Bioquímica, Universidade Federal de Pernambuco, Recife 50670-901, PE, Brazil; (M.C.d.B.); (L.C.B.B.C.); (T.H.N.); (T.d.A.L.); (P.M.G.P.)
| |
Collapse
|
19
|
Khatun R, Dolai M, Sasmal M, Katarkar A, Islam ASM, Yasmin N, Maryum S, Haribabu J, Ali M. Small molecule interactions with biomacromolecules: selective sensing of human serum albumin by a hexanuclear manganese complex - photophysical and biological studies. J Mater Chem B 2024; 12:9408-9419. [PMID: 39192836 DOI: 10.1039/d4tb00712c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
A covalently bonded hexanuclear neutral complex, [Mn6(μ3-O)2(3-MeO-salox)6(OAc)2(H2O)4] (1), has been synthesized and characterized by single crystal X-ray diffraction analysis along with IR and HRMS studies. Complex 1 has been found to selectively interact with human serum albumin (HSA), a model transport protein. The interaction of 1 with HSA was investigated by monitoring the change in the absorbance value of HSA at λ = 280 nm with increasing concentration of 1. Likewise, fluorescence titrations were carried out under two conditions: (i) titration of a 5 μM solution of complex 1 with the gradual addition of HSA, showing a ∼9-fold fluorescence intensity enhancement at 424 nm, upon excitation at 300 nm; and (ii) upon excitation at 295 nm, titration of 5 μM HSA solution with the incremental addition of complex 1, showing a quenching of fluorescence intensity at 334 nm, with simultaneous development of a new emission band at 424 nm. A linear form of the Stern-Volmer equation gives KSV = 9.77 × 104 M-1 and the Benesi-Hildebrand plot yields the binding constant as KBH = 1.98 × 105 M-1 at 298 K. The thermodynamic parameters, ΔS°, ΔH°, and ΔG°, were estimated by using the van't Hoff relationship which infer the major contribution of hydrophobic interactions between HSA and 1. It was observed that quenching of HSA emission arises mainly through a dynamic quenching mechanism as indicated by the dependence of average lifetime 〈τ〉 on the concentration of 1. The changes in the CD (circular dichroism) spectral pattern of HSA in the presence of 1 clearly establish the variation of HSA secondary structure on interaction with 1. The most probable interaction region in HSA for 1 was determined from molecular docking studies which establish the preferential trapping of 1 in the subdomain IIA of site I in HSA and substantiated by the results of site-specific marker studies. Complex 1 was further evaluated for its antiproliferative effects in lung cancer A549 cells, which strictly inhibits the growth of the cells in both 2D and 3D mammospheres, indicating its potential application as an anticancer drug.
Collapse
Affiliation(s)
- Rousunara Khatun
- Department of Chemistry, Jadavpur University, 188, Raja S. C. Mullick Road, Kolkata 700 032, India.
- Aliah University, ll-A/27, Action Area II, Newtown, Action Area II, Kolkata, West Bengal 700160, India
| | - Malay Dolai
- Department of Chemistry, Prabhat Kumar College, Purba Medinipur 721404, India
| | - Mihir Sasmal
- Department of Chemistry, Jadavpur University, 188, Raja S. C. Mullick Road, Kolkata 700 032, India.
| | - Atul Katarkar
- Department of Biochemistry, University of Lausanne, 1066 Epalinges, Switzerland
| | - Abu Saleh Musha Islam
- School of Chemical Sciences, Indian Association for the Cultivation of Science, & 2B Raja S.C. Mullick Road, Kolkata 700032, India
| | - Nasima Yasmin
- Aliah University, ll-A/27, Action Area II, Newtown, Action Area II, Kolkata, West Bengal 700160, India
| | - Sana Maryum
- Department of Chemistry, Jadavpur University, 188, Raja S. C. Mullick Road, Kolkata 700 032, India.
| | - Jebiti Haribabu
- Facultad de Medicina, Universidad de Atacama, Los Carreras 1579, 1532502 Copiapo, Chile
- Chennai Institute of Technology (CIT), Chennai 600069, India
| | - Mahammad Ali
- Department of Chemistry, Jadavpur University, 188, Raja S. C. Mullick Road, Kolkata 700 032, India.
| |
Collapse
|
20
|
Hu ZY, Wu M, Wang WJ, Jiang SL, Shi JH. Exploring the binding behaviors between nisoldipine and bovine serum albumin as a model protein by the aid of multi-spectroscopic approaches and in silico. J Biomol Struct Dyn 2024; 42:6108-6118. [PMID: 37403263 DOI: 10.1080/07391102.2023.2232027] [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: 03/24/2023] [Accepted: 06/25/2023] [Indexed: 07/06/2023]
Abstract
Bovine serum albumin (BSA), a model protein was used to evaluate the binding behavior of nisoldipine and human serum albumin by a series of experiments and in silico in this article. The outcomes suggested that nisoldipine and BSA formed the nisoldipine-BSA complex with a molar ratio of 1:1, caused the fluorescence quenching of BSA, which quenching mechanism was attributable to static quenching. The binding constant of the nisoldipine-BSA complex was (1.3-3.0) × 104 M-1 at 298-310 K, indicating that nisoldipine on BSA protein had a moderate affinity. During the complexation of nisoldipine with BSA, nisoldipine can spontaneously insert into the site II (subdomain III A) of BSA and the distance of energy transfer from donor group in protein to acceptor group in nisoldipine was 3.21 nm, which led to the change in the hydrophobicity of the microenvironment surrounding Trp residues and in the secondary structure of BSA. Additionally, the findings also confirmed that the hydrogen bond and van der Waals force were responsible for forming the nisoldipine-BSA complex and the complexation process was a spontaneous exothermic process.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Zhe-Ying Hu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Meng Wu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Wan-Jun Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Shao-Liang Jiang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Jie-Hua Shi
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| |
Collapse
|
21
|
Ning H, Liu WL, Li QY, Liu YY, Huang ST, Liu HB, Tang AX. Substrate Characterization for Hydrolysis of Multiple Types of Aromatic Esters by Promiscuous Aminopeptidases. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024. [PMID: 39021280 DOI: 10.1021/acs.jafc.4c02053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Synthetic aromatic esters, widely employed in agriculture, food, and chemical industries, have become emerging environmental pollutants due to their strong hydrophobicity and poor bioavailability. This study attempted to address this issue by extracellularly expressing the promiscuous aminopeptidase (Aps) from Pseudomonas aeruginosa GF31 in B. subtilis, achieving an impressive enzyme activity of 13.7 U/mg. Notably, we have demonstrated, for the first time, the Aps-mediated degradation of diverse aromatic esters, including but not limited to pyrethroids, phthalates, and parabens. A biochemical characterization of Aps reveals its esterase properties and a broader spectrum of substrate profiles. The degradation rates of p-nitrobenzene esters (p-NB) with different side chain structures vary under the action of Aps, showing a preference for substrates with relatively longer alkyl side chains. The structure-dependent degradability aligns well with the binding energies between Aps and p-NB. Molecular docking and enzyme-substrate interaction elucidate that hydrogen bonding, hydrophobic interactions, and π-π stacking collectively stabilize the enzyme-substrate conformation, promoting substrate hydrolysis. These findings provide new insights into the enzymatic degradation of aromatic ester pollutants, laying a foundation for the further development and modification of promiscuous enzymes.
Collapse
Affiliation(s)
- Hang Ning
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning 530004, Guangxi, People's Republic of China
| | - Wen-Long Liu
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning 530004, Guangxi, People's Republic of China
| | - Qing-Yun Li
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning 530004, Guangxi, People's Republic of China
- Key Laboratory of Guangxi Biorefinery, Nanning 530003, People's Republic of China
| | - You-Yan Liu
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning 530004, Guangxi, People's Republic of China
- Key Laboratory of Guangxi Biorefinery, Nanning 530003, People's Republic of China
| | - Shi-Ting Huang
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning 530004, Guangxi, People's Republic of China
| | - Hai-Bo Liu
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning 530004, Guangxi, People's Republic of China
| | - Ai-Xing Tang
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning 530004, Guangxi, People's Republic of China
- Key Laboratory of Guangxi Biorefinery, Nanning 530003, People's Republic of China
| |
Collapse
|
22
|
Cho YJ, Kim H, Lim SI. Preserved structure and function of human serum albumin self-folded in the oxidative cytoplasm of Escherichia coli. J Biotechnol 2024; 390:62-70. [PMID: 38761885 DOI: 10.1016/j.jbiotec.2024.05.005] [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: 02/01/2024] [Revised: 04/11/2024] [Accepted: 05/07/2024] [Indexed: 05/20/2024]
Abstract
Human serum albumin (HSA), a polypeptide featuring 17 disulfide bonds, acts as a crucial transport protein in human blood plasma. Its extended circulation half-life, mediated by FcRn (neonatal Fc receptor)-facilitated recycling, positions HSA as an excellent carrier for long-acting drug delivery. However, the conventional method of obtaining HSA from human blood faces limitations due to availability and potential contamination risks, such as blood-borne diseases. This study introduced SHuffle, an oxidative Escherichia coli (E. coli) expression system, for the production of recombinant HSA (rHSA) that spontaneously self-folds into its native conformation. This system ensures precise disulfide bond formation and correct folding of cysteine-rich rHSA, eliminating the need for chaperone co-expression or domain fusion of a folding enhancer. The purified rHSA underwent thorough physicochemical characterization, including mass spectrometry, circular dichroism spectroscopy, intrinsic fluorescence spectroscopy, esterase-like activity assay, and size exclusion chromatography, to assess critical quality attributes. Importantly, rHSA maintained native binding affinity to FcRn and the albumin-binding domain. Collectively, our analyses demonstrated a high comparability between rHSA and plasma-derived HSA. The expression of rHSA in E. coli with an oxidizing cytosol provides a secure and cost-effective approach, enhancing the potential of rHSA for diverse medical applications.
Collapse
Affiliation(s)
- Yong Joon Cho
- Department of Chemical Engineering, Pukyong National University, Yongso-ro 45, Nam-gu, Busan, Republic of Korea
| | - Hyunji Kim
- Department of Chemical Engineering, Pukyong National University, Yongso-ro 45, Nam-gu, Busan, Republic of Korea
| | - Sung In Lim
- Department of Chemical Engineering, Pukyong National University, Yongso-ro 45, Nam-gu, Busan, Republic of Korea.
| |
Collapse
|
23
|
H Krishne D, Sharma K, Reddy AJ, Koppal VV. Exploring the molecular binding mechanism of 6-fluoro, 4-hydroxy, 2- methyl quinoline with TiO 2 nanoparticles: A spectroscopic, thermodynamic, and insights into the solvatochromic effect. J Fluoresc 2024:10.1007/s10895-024-03829-z. [PMID: 38958908 DOI: 10.1007/s10895-024-03829-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 06/24/2024] [Indexed: 07/04/2024]
Abstract
This study investigates the interaction between titanium oxide nanoparticles (TiO2 NPs) and the heterocyclic fluorophore 6-fluoro,4-hydroxy,2-methylquinoline (6-FHMQ), aiming to understand fluorescence quenching mechanisms and thermodynamic characteristics. Spectroscopic techniques including spectrofluorometry (FL) and spectrophotometry (UV-Vis) were used, with a lifetime decay (τ) of 0.18 ns for 6-FHMQ measured using time correlated single photon counting (TCSPC). The interaction between 6-FHMQ and TiO2 NPs revealed a mix of static and dynamic fluorescence quenching mechanisms, with increasing quenching constants (Ksv) and a higher bimolecular quenching rate constant (Kq). The dynamic nature was highlighted by a temperature-dependent increase in binding sites from 1 to ~ 2. Spontaneous complexation was affirmed by negative change in free energy (ΔG), with negative change in enthalpy (ΔH) and a positive change in entropy (ΔS) values indicating favorable electrostatic and ionic interactions. The impact of varying TiO2 NP concentrations on 6-FHMQ absorption was analyzed using the Benesi-Hildbrand equation, with a quantum yield of 0.61 determined. By forster resonance energy transfer (FRET) theory, the proximity between 6-FHMQ and TiO2 NPs was found to be less than 70 Å. Ground and excited state dipole moments of 6-FHMQ in different solvents were calculated to demonstrate solvent sensing ability and charge transfer properties. Ultimately, this study serves as a testament to the power of scientific innovation in the realms of drug delivery and tissue engineering.
Collapse
Affiliation(s)
- Deepa H Krishne
- Department of Physics, M S Ramaiah Institute of Technology (Affiliated to VTU), Bangalore, 560054, Karnataka, India.
| | - Kalpana Sharma
- Department of Physics, M S Ramaiah Institute of Technology (Affiliated to VTU), Bangalore, 560054, Karnataka, India
| | - A Jagannatha Reddy
- Department of Physics, M S Ramaiah Institute of Technology (Affiliated to VTU), Bangalore, 560054, Karnataka, India
| | - V V Koppal
- Department of Physics, KLE Technological University, Hubli, 580031, Karnataka, India
| |
Collapse
|
24
|
Paliwal H, Kaewpaiboon S, Ali Khumaini Mudhar Bintang M, Srichana T. Interaction studies of cannabidiol with human serum albumin by surface plasmon resonance, spectroscopy, and molecular docking. J Biomol Struct Dyn 2024; 42:5147-5158. [PMID: 37434318 DOI: 10.1080/07391102.2023.2234494] [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: 03/24/2023] [Accepted: 06/08/2023] [Indexed: 07/13/2023]
Abstract
The binding interaction of cannabidiol (CBD) and human serum albumin (HSA) under physiological blood pH conditions (pH 7.4) was conducted by surface plasmon resonance (SPR), fluorescence spectroscopy, UV-Visible spectrophotometry, and molecular docking. The responses from SPR measurement increased with the increase in CBD concentration until equilibrium was reached at the equilibrium dissociation constant (KD) of 9.8 × 10-4 M. The results from fluorescence and UV-Visible spectroscopy showed that CBD bound to HSA at one site in a spontaneous manner to form protein-CBD complexes. The quenching process involved both static and dynamic mechanisms while the static mechanism contributed predominantly to the binding between CBD and albumin. The binding constants estimated from the fluorescence studies were from 0.16 × 103 to 8.10 × 103 M-1, which were calculated at different temperature conditions using Stern-Volmer plots. The thermodynamic parameters demonstrated that the binding interaction was a spontaneous reaction as Gibbs free energy had negative values (ΔG = -12.57 to -23.20 kJ.mol-1). Positive ΔH and ΔS values (ΔH = 2.46 × 105 J.mol-1 and ΔS = 869.81 J.mol-1K-1) indicated that the hydrophobic force was the major binding interaction. Finally, confirmation of the type and extent of interaction was provided using UV-spectroscopy and molecular docking studies. The outcomes of this study are expected to serve as a platform to conduct future studies on binding interactions and toxicological research of CBD.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Himanshu Paliwal
- Drug Delivery System Excellence Center, Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| | - Sunisa Kaewpaiboon
- Drug Delivery System Excellence Center, Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| | - Muhammad Ali Khumaini Mudhar Bintang
- Drug Delivery System Excellence Center, Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| | - Teerapol Srichana
- Drug Delivery System Excellence Center, Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| |
Collapse
|
25
|
Yang H, Ji X, Wang H, Yang R, Ma J. Mechanism understanding of PIKfyve inhibitor YM201636 with human serum albumin: Insights from molecular modeling and multiple spectroscopic techniques. LUMINESCENCE 2024; 39:e4838. [PMID: 39051537 DOI: 10.1002/bio.4838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 07/04/2024] [Accepted: 07/07/2024] [Indexed: 07/27/2024]
Abstract
YM201636 is the potent PIKfyve inhibitor that is being actively investigated for liver cancer efficacy. In this study, computer simulations and experiments were conducted to investigate the interaction mechanism between YM201636 and the transport protein HSA. Results indicated that YM201636 is stably bound between the subdomains IIA and IIIA of HSA, supported by site marker displacement experiments. YM201636 quenched the endogenous fluorescence of HSA by static quenching since a decrease in quenching constants was observed from 7.74 to 2.39 × 104 M-1. UV-vis and time-resolved fluorescence spectroscopy confirmed the YM201636-HSA complex formation and this binding followed a static mechanism. Thermodynamic parameters ΔG, ΔH, and ΔS obtained negative values suggesting the binding was a spontaneous process driven by Van der Waals interactions and hydrogen binding. Binding constants ranged between 5.71 and 0.33 × 104 M-1, which demonstrated a moderately strong affinity of YM201636 to HSA. CD, synchronous, and 3D fluorescence spectroscopy revealed that YM201636 showed a slight change in secondary structure. The increase of Kapp and a decrease of PSH with YM201636 addition showed that YM201636 changed the surface hydrophobicity of HSA. The research provides reasonable models helping us further understand the transportation and distribution of YM201636 when it absorbs into the blood circulatory system.
Collapse
Affiliation(s)
- Hongqin Yang
- College of Life Sciences, Northwest Normal University, Lanzhou, China
| | - Xinzhu Ji
- College of Life Sciences, Northwest Normal University, Lanzhou, China
| | - Huiling Wang
- College of Life Sciences, Northwest Normal University, Lanzhou, China
| | - Ruijing Yang
- College of Life Sciences, Northwest Normal University, Lanzhou, China
| | - Junyi Ma
- College of Life Sciences, Northwest Normal University, Lanzhou, China
| |
Collapse
|
26
|
Zhang Z, Dai L, Yang K, Luo J, Zhang Y, Ding P, Tian J, Tuo X, Chi B. Molecular insight on the binding of halogenated organic phosphate esters to human serum albumin and its effect on cytotoxicity of halogenated organic phosphate esters. Int J Biol Macromol 2024; 270:132383. [PMID: 38754667 DOI: 10.1016/j.ijbiomac.2024.132383] [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: 03/21/2024] [Revised: 04/25/2024] [Accepted: 05/13/2024] [Indexed: 05/18/2024]
Abstract
Halogenated Organic Phosphate Esters (OPEs) are commonly found in plasticizers and flame retardants. However, they are one kind of persistent contaminants that can pose a significant threat to human health and ecosystem as new environmental estrogen. In this study, two representative halogenated OPEs, tris(1,3-dichloro-2-propyl) phosphate (TDCP) and tris(2,3-dibromopropyl) phosphate (TDBP), were selected as experimental subjects to investigate their interaction with human serum albumin (HSA). Despite having similar structures, the two ligands exhibited contrasting effects on enzyme activity of HSA, TDCP inhibiting enzyme activity and TDBP activating it. Furthermore, both TDCP and TDBP could bind to HSA at site I, interacted with Arg222 and other residues, and made the conformation of HSA unfolded. Thermodynamic parameters indicated the main driving forces between TDBP and HSA were hydrogen bonding and van der Waals forces, while TDCP was mainly hydrophobic force. Molecular simulations found that more hydrogen bonds of HSA-TDBP formed during the binding process, and the larger charge area of TDBP than TDCP could partially account for the differences observed in their binding abilities to HSA. Notably, the cytotoxicity of TDBP/TDCP was inversely proportional to their binding ability to HSA, implying a new method for determining the cytotoxicity of halogenated OPEs in vitro.
Collapse
Affiliation(s)
- Zihang Zhang
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Lulu Dai
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Kaiyu Yang
- School of Pharmacy, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Jiaqing Luo
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Yue Zhang
- School of Pharmacy, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Pei Ding
- School of Pharmacy, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Jianwen Tian
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Xun Tuo
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang 330031, Jiangxi, China.
| | - Baozhu Chi
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang 330031, Jiangxi, China.
| |
Collapse
|
27
|
Brahma D, Sarangi AN, Kaushik R, Gupta AN. Oxidative stress induced conformational changes of human serum albumin. Phys Chem Chem Phys 2024; 26:8528-8538. [PMID: 38411624 DOI: 10.1039/d4cp00059e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Oxidative stress, generated by reactive oxygen species (ROS), is responsible for the loss of structure and functionality of proteins and is associated with several aging-related diseases. Here, we report an in vitro study to gauge the effect of ROS on the structural rearrangement of human serum albumin (HSA), a plasma protein, through metal-catalyzed oxidation (MCO) at physiological temperature through various biophysical techniques like UV-vis absorption, circular dichroism (CD), differential scanning calorimetry (DSC), MALDI-TOF, FTIR, and Raman spectroscopy. The UV-vis spectra of oxidized HSA show an early blueshift, signifying the unfolding of the protein because of ROS followed by the broadening of the absorption peak at a longer time. The DSC data corroborate the observation, revealing an exothermic transition for the oxidized sample at a longer time, suggesting in situ aggregation. The CD and FTIR spectra indicate the associated secondary structural changes occurring with time, depicting the variation of the helical content of HSA. The amide-III analysis of Raman data also complements the structural changes, and MALDI-TOF data show the mass distribution with time. Overall, this work might help determine the effect of oxidation on the biological activity of serum albumin as it can impact the physiological properties of HSA.
Collapse
Affiliation(s)
- Debdip Brahma
- Biophysics and Soft Matter Laboratory, Department of Physics, Indian Institute of Technology, Kharagpur, 721302, India.
| | - Akshay Narayan Sarangi
- Biophysics and Soft Matter Laboratory, Department of Physics, Indian Institute of Technology, Kharagpur, 721302, India.
| | - Rupal Kaushik
- Biophysics and Soft Matter Laboratory, Department of Physics, Indian Institute of Technology, Kharagpur, 721302, India.
| | - Amar Nath Gupta
- Biophysics and Soft Matter Laboratory, Department of Physics, Indian Institute of Technology, Kharagpur, 721302, India.
| |
Collapse
|
28
|
Abubakar M, Mohamad SB, Abd Halim AA, Tayyab S. Unveiling the molecular interaction of hepatitis B virus inhibitor, entecavir with human serum albumin through computational, microscopic and spectroscopic approaches. J Biomol Struct Dyn 2024:1-14. [PMID: 38315445 DOI: 10.1080/07391102.2024.2311331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 01/23/2024] [Indexed: 02/07/2024]
Abstract
Molecular docking, molecular dynamics (MD) simulation, atomic force microscopy (AFM) and multi-spectroscopic techniques were selected to unveil the molecular association between the hepatitis B virus (HBV) inhibitor, entecavir (ETR), and the major blood plasma transporter, human serum albumin (HSA). The entire docking and simulation analyses recognized ETR binding to subdomain IIA (Site I) of HSA through hydrogen bonds, hydrophobic and van der Waals forces while maintaining the complex's stability throughout the 100 ns. A gradual lessening in the Stern-Volmer quenching constant (K sv ) with rising temperatures registered ETR-induced quenching of HBV fluorescence as static quenching, thus advising complexation between ETR and HSA. The further advocation of this conclusion was seen from a larger value of the biomolecular quenching rate constant ((kq ) > 1010 M-1s-1), changes in the spectra (UV-Vis absorption) of HSA following ETR inclusion and ETR-induced swelling of HSA in the AFM results. The ETR appeared to bind to HSA with moderate affinity (K a = 1.87 - 1.19 × 10 4 M-1) at 290, 300 and 310 K. Significant alterations in the protein's secondary and tertiary structures, including changes in the protein's Tyr/Trp microenvironment, were also detected by circular dichroism and three-dimensional fluorescence spectra when the protein was bound to ETR. The findings of the drug displacement study backed the docking results of Site I as ETR's preferred binding site in HSA.
Collapse
Affiliation(s)
- Mujaheed Abubakar
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Saharuddin B Mohamad
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Adyani Azizah Abd Halim
- Department of Oral and Craniofacial Sciences, Faculty of Dentistry, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Saad Tayyab
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia
| |
Collapse
|
29
|
Pani BSUL, Chandrasekaran N. Adsorption of clarithromycin on polystyrene nanoplastics surface and its combined adverse effect on serum albumin. Colloids Surf B Biointerfaces 2024; 234:113673. [PMID: 38086277 DOI: 10.1016/j.colsurfb.2023.113673] [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: 09/19/2023] [Revised: 11/20/2023] [Accepted: 11/24/2023] [Indexed: 02/09/2024]
Abstract
Emerging contaminants, such as antibiotics and nanoplastics, have garnered significant attention due to their potential adverse effects on diverse ecosystems. Antibiotic adsorption on the surface of nanoplastics potentially facilitates their long-range transport, leading to the synergistic effects of the complex. This research aims to examine the adsorption behavior of clarithromycin binding with polystyrene nanoplastics surface as well as their interaction between drug adsorbed polystyrene nanoplastics with serum albumin. Different spectroscopic methods were used to find out the interaction between clarithromycin and nanoplastics, under stimulated physiological conditions UV-vis spectroscopy showed a maximum of 22.8% percentage of the drug adsorbed with the polystyrene nanoplastics surface after 6 h of incubation. The fluorescence spectroscopic results demonstrated that the fluorescence intensity of serum albumin was quenched by the clarithromycin-polystyrene nanoplastics (CLA-PSNP) complex through static quenching. We calculated the number of binding stoichiometry, binding constants, and thermodynamic parameters. This study revealed that the CLA-PSNP binds to serum albumin spontaneously and its hydrophobic interactions played a significant role. The conformational changes in the structure of serum albumin were revealed from the findings of synchronous fluorescence spectra, CD spectra, and 3D fluorescence spectra, leading to the disturbance in functional activity. This study focuses valuable insights into the intermolecular interactions between clarithromycin-adsorbed polystyrene nanoplastics and serum albumin and its potential molecular-level biological toxicity.
Collapse
|
30
|
Burra VLSP, Sahoo PS, Dhankhar A, Jhajj J, Kasamuthu PS, K SSVK, Macha SKR. Understanding the structural basis of the binding specificity of c-di-AMP to M. smegmatis RecA using computational biology approach. J Biomol Struct Dyn 2024; 42:2043-2057. [PMID: 38093709 DOI: 10.1080/07391102.2023.2227709] [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: 12/15/2022] [Accepted: 04/09/2023] [Indexed: 02/21/2024]
Abstract
Mycobacterium tuberculosis RecA (MtRecA), a protein involved in DNA repair, homologous recombination and SOS pathway, contributes to the development of multidrug resistance. ATP binding-site in RecA has been a drug target to disable RecA dependent DNA repair. For the first time, experiments have shown the existence and binding of c-di-AMP to a novel allosteric site in the C-terminal-Domain (CTD) of Mycobacterium smegmatis RecA (MsRecA), a close homolog of MtRecA. In addition, it was observed that the c-di-AMP was not binding to Escherichia coli RecA (EcRecA). This article analyses the possible interactions of the three RecA homologs with the various c-di-AMP conformations to gain insights into the structural basis of the natural preference of c-di-AMP to MsRecA and not to EcRecA, using the structural biology tools. The comparative analysis, based on amino acid composition, homology, motifs, residue types, docking, molecular dynamics simulations and binding free energy calculations, indeed, conclusively indicates strong binding of c-di-AMP to MsRecA. Having very similar results as MsRecA, it is highly plausible for c-di-AMP to strongly bind MtRecA as well. These insights from the in-silico studies adds a new therapeutic approach against TB through design and development of novel allosteric inhibitors for the first time against MtRecA.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- V L S Prasad Burra
- Centre for Advanced Research and Innovation in Structural Biology of Diseases, K L E F (Deemed to be) University, Vaddeswaram, Andhra Pradesh, India
| | - Partha Sarathi Sahoo
- Centre for Advanced Research and Innovation in Structural Biology of Diseases, K L E F (Deemed to be) University, Vaddeswaram, Andhra Pradesh, India
| | - Amit Dhankhar
- Centre for Advanced Research and Innovation in Structural Biology of Diseases, K L E F (Deemed to be) University, Vaddeswaram, Andhra Pradesh, India
| | - Jatinder Jhajj
- Centre for Advanced Research and Innovation in Structural Biology of Diseases, K L E F (Deemed to be) University, Vaddeswaram, Andhra Pradesh, India
| | - Prasanna Sudharson Kasamuthu
- Centre for Advanced Research and Innovation in Structural Biology of Diseases, K L E F (Deemed to be) University, Vaddeswaram, Andhra Pradesh, India
| | - S S V Kiran K
- Centre for Advanced Research and Innovation in Structural Biology of Diseases, K L E F (Deemed to be) University, Vaddeswaram, Andhra Pradesh, India
| | - Samuel Krupa Rakshan Macha
- Centre for Advanced Research and Innovation in Structural Biology of Diseases, K L E F (Deemed to be) University, Vaddeswaram, Andhra Pradesh, India
| |
Collapse
|
31
|
Yang Y, Wang S, Liu X, Zhang W, Tong W, Luo H, Zhao L. Interactions of ferulic acid and ferulic acid methyl ester with endogenous proteins: Determination using the multi-methods. Heliyon 2024; 10:e24605. [PMID: 38312678 PMCID: PMC10835327 DOI: 10.1016/j.heliyon.2024.e24605] [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: 07/20/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 02/06/2024] Open
Abstract
Ferulic acid (FA) and ferulic acid methyl ester (FAM) are important phenolic compounds in Baijiu. In this study, the interaction of FA and FAM with human serum albumin (HSA) and lysozyme (LZM) was investigated using multispectral methods and molecular dynamics simulation. FA and FAM could interact with HSA and LZM, changing the conformation and hydrophilicity of the protein. The quenching mechanisms of FA-HSA, FA-LZM, FAM-HSA, and FAM-LZM were all static-quenching. In the FA-HSA, FAM-HSA, and FA-LZM systems, the interaction forces were mainly hydrophobic interactions and hydrogen bonding. In the FAM-LZM system, the interaction forces were mainly hydrophobic interactions, hydrogen bonding, and van der Waals force. Common metal ions such as K+, Ca2+, Cu2+, Mg2+, and Mn2+ could affect the binding ability of FA and FAM to HSA and LZM. Moreover, FA and FAM could increase the stability of HSA and LZM, and the protein bound to FA/FAM was more stable than the free protein. FA and FAM had varying degrees of impact on the physiological activities of HSA and LZM. This study provides relevant information on the interactions and metabolic mechanisms of FA and its derivatives with endogenous proteins.
Collapse
Affiliation(s)
- Ying Yang
- School of Biological Engineering, Sichuan University of Science and Engineering, Yibin, 644000, China
| | - Shuqin Wang
- School of Biological Engineering, Sichuan University of Science and Engineering, Yibin, 644000, China
| | - Xingyan Liu
- School of Biological Engineering, Sichuan University of Science and Engineering, Yibin, 644000, China
| | - Wenbin Zhang
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, 510000, China
| | - Wenhua Tong
- School of Biological Engineering, Sichuan University of Science and Engineering, Yibin, 644000, China
- Key Laboratory of Brewing Biotechnology and Application, Yibin, 644000, China
| | - Huibo Luo
- School of Biological Engineering, Sichuan University of Science and Engineering, Yibin, 644000, China
- Key Laboratory of Brewing Biotechnology and Application, Yibin, 644000, China
| | - Liming Zhao
- East China University of Science and Technology, Shanghai, 200000, China
| |
Collapse
|
32
|
Qu L, Li T, Cun S, Zheng X, Xiang M, Dong Y, Ji X, Bian L, Li Q, Zhao X. A chromatographic method for determining the interaction between a drug and two target proteins by fabricating a dual-heterogeneous surface. J Chromatogr A 2024; 1715:464606. [PMID: 38154257 DOI: 10.1016/j.chroma.2023.464606] [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: 10/29/2023] [Revised: 12/21/2023] [Accepted: 12/22/2023] [Indexed: 12/30/2023]
Abstract
Characterization of the drug-target interactions is pivotal throughout the whole procedure of drug development. Most of the current assays, particularly, chromatographic methods lack the capacity to reveal drug adsorption on the muti-target surface. To this end, we derived a reliable and workable mathematical equation for revealing drug bindings to dual targets on the heterogeneous surface starting from the mass balance equation. The derivatization relied on the correlation of drug injection amounts with their retention factors. Experimental validation was performed by determining the binding parameters of three canonical drugs on a heterogeneous surface, which was fabricated by fusing angiotensin receptor type I and type II receptors (AT1R and AT2R) at the terminuses of circularly permuted HaloTag (cpHaloTag) and immobilizing the whole fusion protein onto 6-bromohexanoic acid modified silica gel. We proved that immobilized AT1R-cpHalo-AT2R maintained the original ligand- and antibody-binding activities of the two receptors in three weeks. The association constants of valsartan, candesartan, and telmisartan to AT1R were (6.26±0.14) × 105, (9.66±0.71) × 105, and (3.17±0.03) × 105 L/mol. In the same column, their association constants to AT2R were (1.25±0.04) × 104, (2.30±0.08) × 104, and (8.51±0.06) × 103 L/mol. The patterns of the association constants to AT1R/AT2R (candesartan>valsartan>telmisartan) were in good line with the data by performing nonlinear chromatography on control columns containing immobilized AT1R or AT2R alone. This provided proof of the fact that the derivatization allowed the determination of drug bindings on the heterogeneous surface with the utilization of a single series of injections and linear regression. We reasoned that is simple enough to model the bindings of drug adsorption on commercially available adsorbents in fundamental or industrial fields, thus having the potential to become a universal method for analyzing the bindings of a drug to the heterogeneous surface containing multiple targets.
Collapse
Affiliation(s)
- Lejing Qu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Ting Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Sidi Cun
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Xinxin Zheng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Mingjuan Xiang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Yuxuan Dong
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Xu Ji
- Engineering Research Center of Tibetan Medicine Detection Technology, Ministry of Education, Xizang Minzu University, Xianyang 712082, China
| | - Liujiao Bian
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Qian Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, China.
| | - Xinfeng Zhao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, China
| |
Collapse
|
33
|
Xu S, Luo W, Zhu M, Zhao L, Gao L, Liang H, Zhang Z, Yang F. Human Serum Albumin-Platinum(II) Agent Nanoparticles Inhibit Tumor Growth Through Multimodal Action Against the Tumor Microenvironment. Mol Pharm 2024; 21:346-357. [PMID: 38015620 DOI: 10.1021/acs.molpharmaceut.3c00881] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2023]
Abstract
To overcome the limitations of traditional platinum (Pt)-based drugs and further improve the targeting ability and therapeutic efficacy in vivo, we proposed to design a human serum albumin (HSA)-Pt agent complex nanoparticle (NP) for cancer treatment by multimodal action against the tumor microenvironment. We not only synthesized a series of Pt(II) di-2-pyridone thiosemicarbazone compounds and obtained a Pt(II) agent [Pt(Dp44mT)Cl] with significant anticancer activity but also successfully constructed a novel HSA-Pt(Dp44mT) complex nanoparticle delivery system. The structure of the HSA-Pt(Dp44mT) complex revealed that Pt(Dp44mT)Cl binds to the IIA subdomain of HSA and coordinates with His-242. The HSA-His242-Pt-Dp44mT NPs had an obvious effect on the inhibition of tumor growth, which was superior to that of Dp44mT and Pt(Dp44mT)Cl, and they had almost no toxicity. In addition, the HSA-His242-Pt-Dp44mT NPs were found to kill cancer cells by inducing apoptosis, autophagy, and inhibiting angiogenesis.
Collapse
Affiliation(s)
- Shihang Xu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, P. R. China
| | - Weicong Luo
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, P. R. China
| | - Minghui Zhu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, P. R. China
| | - Lei Zhao
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, P. R. China
| | - Lijuan Gao
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, P. R. China
| | - Hong Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, P. R. China
| | - Zhenlei Zhang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, P. R. China
| | - Feng Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, P. R. China
| |
Collapse
|
34
|
Meng X, Nan G, Du Y, Zhao H, Zheng H, Lin R, Yang G. Comparing the interactions of nitrendipine with lysozyme or human serum albumin and the effects of vitamin C and naringin on these interactions by spectroscopy and molecular docking methods. LUMINESCENCE 2024; 39:e4618. [PMID: 37937696 DOI: 10.1002/bio.4618] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 09/30/2023] [Accepted: 10/16/2023] [Indexed: 11/09/2023]
Abstract
The interactions between drugs and proteins play a pivotal role in determining the pharmacological effects and disposition of drugs within the human body. This study focuses on exploring the interaction between nitrendipine and lysozyme/human serum albumin. Spectroscopic analysis indicated a compound static quenching, indicative of the formation of stable complexes between the drug and proteins. The addition of vitamin C or naringin resulted in a decrease of the binding constant between nitrendipine and lysozyme/human serum albumin. The presence of these compounds may disrupt the interactions between the drug and proteins, potentially leading to an increased concentration of free nitrendipine in the bloodstream. Nitrendipine binds more easily to human serum albumin at 310 K, and human serum albumin has an average binding site ratio with nitrendipine approximately 0.1 higher than that with lysozyme. Vitamin C has a greater impact on the binding constant of nitrendipine to human serum albumin and lysozyme. Compared to the binary system of proteins with the drug, the ternary system with the addition of vitamin C at 310 K reduces the binding constants of lysozyme and human serum albumin by 85%. In conclusion, this study explores the significance of considering drug-protein interactions in understanding drug behavior and potential drug-food interactions.
Collapse
Affiliation(s)
- Xianxin Meng
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Guanjun Nan
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yan Du
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Hongwen Zhao
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Hongxia Zheng
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Rong Lin
- School of Basic Medical Sciences, Xian Jiaotong University, Xi'an, Shaanxi, China
| | - Guangde Yang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
35
|
Vinod LA, Rajendran D, Shivashankar M, Chandrasekaran N. Surface interaction of vancomycin with polystyrene microplastics and its effect on human serum albumin. Int J Biol Macromol 2024; 256:128491. [PMID: 38043666 DOI: 10.1016/j.ijbiomac.2023.128491] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 11/20/2023] [Accepted: 11/27/2023] [Indexed: 12/05/2023]
Abstract
Microplastics have a well-documented ability to adsorb various chemicals and contaminants found in the environment. By similar mechanisms, when medicines are stored in plastic packaging, the leaching of plastics into the contents poses the risk of possible toxicity and decreased drug efficacy. The work thus examines the presence of two categories of anthropogenic materials - microplastics (MPs) and medications - with their possible combined effects and fate in biological systems. A study on the kinetics and isotherm of the adsorption of vancomycin hydrochloride on the surface of polystyrene microspheres is performed, and the best-fitting models are obtained respectively as the pseudo-second-order model and the Temkin isotherm. Further, the interaction of each of, the drug, MPs and drug-adsorbed MPs with human serum albumin (HSA), the model protein chosen to validate the potential toxicity in humans, is determined by fluorescence spectroscopy. A thermodynamic analysis of this protein-ligand interaction shows that the process is spontaneous, endothermic and entropically favoured, and that hydrophobic forces operate between the interacting species. An unfolding of HSA is observed, disrupting its functions like the esterase activity. Competitive binding experiments with Warfarin and Ibuprofen as specific site markers on HSA reveal that all the studied ligands bind non-specifically to HSA.
Collapse
Affiliation(s)
- Lydia Ann Vinod
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Durgalakshmi Rajendran
- Centre for Nanobiotechnology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Murugesh Shivashankar
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Natarajan Chandrasekaran
- Centre for Nanobiotechnology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India.
| |
Collapse
|
36
|
Akawa OB, Okunlola FO, Alahmdi MI, Abo-Dya NE, Sidhom PA, Ibrahim MAA, Shibl MF, Khan S, Soliman MES. Multi-cavity molecular descriptor interconnections: Enhanced protocol for prediction of serum albumin drug binding. Eur J Pharm Biopharm 2024; 194:9-19. [PMID: 37984594 DOI: 10.1016/j.ejpb.2023.11.003] [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: 09/01/2023] [Revised: 11/03/2023] [Accepted: 11/03/2023] [Indexed: 11/22/2023]
Abstract
The role of human serum albumin (HSA) in the transport of molecules predicates its involvement in the determination of drug distribution and metabolism. Optimization of ADME properties are analogous to HSA binding thus this is imperative to the drug discovery process. Currently, various in silico predictive tools exist to complement the drug discovery process, however, the prediction of possible ligand-binding sites on HSA has posed several challenges. Herein, we present a strong and deeper-than-surface case for the prediction of HSA-ligand binding sites using multi-cavity molecular descriptors by exploiting all experimentally available and crystallized HSA-bound drugs. Unlike previously proposed models found in literature, we established an in-depth correlation between the physicochemical properties of available crystallized HSA-bound drugs and different HSA binding site characteristics to precisely predict the binding sites of investigational molecules. Molecular descriptors such as the number of hydrogen bond donors (nHD), number of heteroatoms (nHet), topological polar surface area (TPSA), molecular weight (MW), and distribution coefficient (LogD) were correlated against HSA binding site characteristics, including hydrophobicity, hydrophilicity, enclosure, exposure, contact, site volume, and donor/acceptor ratio. Molecular descriptors nHD, TPSA, LogD, nHet, and MW were found to possess the most inherent capacities providing baseline information for the prediction of serum albumin binding site. We believe that these associations may form the bedrock for establishing a solid correlation between the physicochemical properties and Albumin binding site architecture. Information presented in this report would serve as critical in provisions of rational drug designing as well as drug delivery, bioavailability, and pharmacokinetics.
Collapse
Affiliation(s)
- Oluwole B Akawa
- Molecular Bio-computational and Drug Design Laboratory, School of Health Sciences, University of KwaZulu Natal, Westville Campus, Durban 4001, South Africa
| | - Felix O Okunlola
- Molecular Bio-computational and Drug Design Laboratory, School of Health Sciences, University of KwaZulu Natal, Westville Campus, Durban 4001, South Africa
| | - Mohammed Issa Alahmdi
- Faculty of Science, Department of Chemistry, University of Tabuk, Tabuk 7149, Saudi Arabia
| | - Nader E Abo-Dya
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tabuk University, Tabuk 71491, Saudi Arabia; Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Peter A Sidhom
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| | - Mahmoud A A Ibrahim
- Molecular Bio-computational and Drug Design Laboratory, School of Health Sciences, University of KwaZulu Natal, Westville Campus, Durban 4001, South Africa; Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia 61519
| | - Mohamed F Shibl
- Renewable Energy Program, Center for Sustainable Development, College of Arts and Sciences, Qatar University, 2713 Doha, Qatar
| | - Shahzeb Khan
- Centre for Pharmaceutical Engineering Science, Faculty of life Science, School of Pharmacy and Medical Sciences, University of Bradford UK, West Yorkshire, BD7 1DP, UK
| | - Mahmoud E S Soliman
- Molecular Bio-computational and Drug Design Laboratory, School of Health Sciences, University of KwaZulu Natal, Westville Campus, Durban 4001, South Africa.
| |
Collapse
|
37
|
Wang H, Rao P, Qiu Y, Xiang L. Interaction mechanism between hydroxychloroquine sulfate and collagen: Insights from multi-spectroscopy, molecular docking, and molecular dynamic simulation methods. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2023; 303:123155. [PMID: 37480720 DOI: 10.1016/j.saa.2023.123155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 07/04/2023] [Accepted: 07/12/2023] [Indexed: 07/24/2023]
Abstract
Hydroxychloroquine sulfate (HCQ) can be used to treat various connective tissue diseases. Collagen, which is not only an important drug delivery carrier but also the main component in the connective tissue, is the focus of this study. Here, the interaction mechanism of HCQ with collagen was investigated through various spectroscopic and computational methods. It is found that HCQ binds to collagen spontaneously, primarily via hydrophobic interactions and some hydrogen bonds. The findings of X-ray diffraction (XRD), Fourier transform infrared spectroscopy (FTIR) and scanning electron microscopy (SEM) verified that formation of HCQ-collagen complex and the amorphous structure, secondary structures, and microstructure of collagen were changed after HCQ binding. A decrease in the relaxation time of free water was observed in the collagen system when HCQ was added. Molecular docking demonstrated that HCQ was almost buried in the cavity of collagen via some hydrophobic interactions with one hydrogen bond, which conforms to the findings of the fluorescence and FTIR analyses. Molecular dynamic (MD) simulations further revealed the structural change information in the docking process. Hopefully, the information generated in this study can provide some useful insights for the research on the pharmacological mechanisms of HCQ in the treatment of the connective tissue diseases and the application of collagen as a drug carrier.
Collapse
Affiliation(s)
- Hailin Wang
- College of Food and Bioengineering, Fujian Polytechnic Normal University, Fuqing, Fujian, China; Food Nutrition Science Centre, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, China
| | - Pingfan Rao
- Food Nutrition Science Centre, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, China
| | - Yunjie Qiu
- College of Food and Bioengineering, Fujian Polytechnic Normal University, Fuqing, Fujian, China
| | - Leiwen Xiang
- College of Food and Bioengineering, Fujian Polytechnic Normal University, Fuqing, Fujian, China.
| |
Collapse
|
38
|
Meng S, Yu Q, Li M, Liu X, Zhao X, Wu K, Wang Q, Liu Y, Wu Y, Gong Z. Unveiling the molecular interactions between alkyl imidazolium ionic liquids and human serum albumin: Implications for toxicological significance. Chem Biol Interact 2023; 386:110762. [PMID: 37844773 DOI: 10.1016/j.cbi.2023.110762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/02/2023] [Accepted: 10/09/2023] [Indexed: 10/18/2023]
Abstract
Alkyl imidazolium-based ionic liquids (ILs) are promising for diverse industrial applications; however, their growing prevalence has raised concerns regarding human exposure and potential health implications. A critical aspect to be clarified to address the adverse health effects associated with ILs exposure is their binding mode to human serum albumin (HSA). In this study, we delved into the binding interactions between three alkyl imidazolium ILs (1-hexyl-3-methyl-imidazolium (C6[MIM]), 1-ethyl-3-methyl-imidazolium chloride (C8[MIM]) and 1-decyl-3-methyl-imidazolium (C10[MIM]) and human serum albumins (HSAs) using a comprehensive approach encompassing molecular docking and multi-spectroscopy (UV-visible, Fluorescence, Circular Dichroism, FTIR). Furthermore, for the first time, we developed an ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) approach time to quantify plasma protein binding rates. Our results revealed that the ILs primarily bind to the hydrophobic cavity of HSA through hydrogen bonding and van der Waals forces, forming stable complexes via static quenching. This affected HSA's secondary structure, reducing α-helical content, particularly around specific residues. Equilibrium dialysis and ultrafiltration coupled with UPLC-MS/MS analysis showed modest plasma protein binding rates (17.84%-31.85%) for the three ILs, with no significant influence from alkyl chain effects or concentration relationship. Lower plasma protein binding rates can affect bioavailability and distribution of ILs, potentially influencing their toxicity. These findings provide critical insights into the potential toxicological implications at the molecular level, thereby contributing to continuous efforts to evaluate the risk profiles and ensure the safe utilization of these compounds.
Collapse
Affiliation(s)
- Shizhen Meng
- College of Food Science and Engineering, Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Qingqing Yu
- College of Food Science and Engineering, Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Ming Li
- College of Food Science and Engineering, Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Xin Liu
- College of Food Science and Engineering, Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan Polytechnic University, Wuhan, 430023, China.
| | - Xiaole Zhao
- College of Food Science and Engineering, Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Kejia Wu
- Wuxi School of Medicine, Jiangnan University, Jiangsu, China
| | - Qiao Wang
- College of Food Science and Engineering, Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Yan Liu
- College of Food Science and Engineering, Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Yongning Wu
- College of Food Science and Engineering, Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan Polytechnic University, Wuhan, 430023, China; NHC Key Laboratory of Food Safety Risk Assessment, Food Safety Research Unit (2019RU014) of Chinese Academy of Medical Science, China National Center for Food Safety Risk Assessment, Beijing, 100021, China
| | - Zhiyong Gong
- College of Food Science and Engineering, Key Laboratory for Deep Processing of Major Grain and Oil, Ministry of Education, Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan Polytechnic University, Wuhan, 430023, China
| |
Collapse
|
39
|
Rahman S, Iram S, Rehman MT, Hussain A, Jan AT, Kim J. Study of Amiloride Binding to Human Serum Albumin: Insights from Thermodynamic, Spectroscopic, and Molecular Docking Investigations. Molecules 2023; 28:7688. [PMID: 38067419 PMCID: PMC10707572 DOI: 10.3390/molecules28237688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/10/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
This study was undertaken to investigate the interaction between the sodium channel blocker amiloride (AML) and human serum albumin (HSA). A combination of multi-spectroscopic techniques and computational methods were employed to identify the AML binding site on HSA and the forces responsible for the formation of the HSA-AML complex. Our findings revealed that AML specifically binds to Sudlow's site II, located in subdomain IIIA of HSA, and that the complex formed is stabilized using van der Waals hydrogen-bonding and hydrophobic interactions. FRET analysis showed that the distance between AML and Trp214 was optimal for efficient quenching. UV-Vis spectroscopy and circular dichroism indicated minor changes in the structure of HSA after AML binding, and molecular dynamics simulations (MDS) conducted over 100 ns provided additional evidence of stable HSA-AML-complex formation. This study enhances understanding of the interaction between AML and HSA and the mechanism responsible.
Collapse
Affiliation(s)
- Safikur Rahman
- Munshi Singh College, BR Ambedkar Bihar University, Muzaffarpur 845401, India;
| | - Sana Iram
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 712-749, Republic of Korea;
| | - Md Tabish Rehman
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (M.T.R.); (A.H.)
| | - Afzal Hussain
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (M.T.R.); (A.H.)
| | - Arif Tasleem Jan
- School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri 185234, India;
| | - Jihoe Kim
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 712-749, Republic of Korea;
| |
Collapse
|
40
|
Messina GML, Campione P, Marletta G. Building Surfaces with Controlled Site-Density of Anchored Human Serum Albumin. ACS APPLIED BIO MATERIALS 2023; 6:4952-4960. [PMID: 37902234 DOI: 10.1021/acsabm.3c00647] [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] [Indexed: 10/31/2023]
Abstract
Stable and uniform layers of protein molecules at the surface are important to build passive devices as well as active constructs for smart biointerfaces for a large number of biomedical applications. In this context, a strategy to build-up surfaces able to anchor protein molecules on specific and controlled surface sites has been developed. Human serum albumin (HSA) has been chosen as a model protein due to its important antithrombogenic properties and its features in cell response highly valuable for in vivo devices. Uniform self-assembled monolayers of 2,2':6'2″-terpyridines (SAM), whose sites were further employed to chelate copper and iron ions, forming SAM-Cu(II) and SAM-Fe(II) complexes, have been developed. The effect of two metal cations on the physicochemical features of SAM, including thickness, Young's modulus, and tip-monolayer adhesion factors, has been investigated. Protein adsorption at different concentrations showed that the copper ion-templated surfaces exhibit highly specific mass uptake, kinetic behavior, and recognition and anchoring of HSA molecules owing to the coordination sphere of the different cations. The results pave the way to the development of a more general strategy to obtain ordered and density-tuned arrays of specific metal cations, which in turn would drive the anchoring of precise proteins for different biological functions.
Collapse
Affiliation(s)
- Grazia M L Messina
- Laboratory for Molecular Surfaces and Nanotechnology (LAMSUN), Department of Chemical Sciences, University of Catania and CSGI, Viale A. Doria 6, 95125 Catania, Italy
| | - Paola Campione
- Laboratory for Molecular Surfaces and Nanotechnology (LAMSUN), Department of Chemical Sciences, University of Catania and CSGI, Viale A. Doria 6, 95125 Catania, Italy
| | - Giovanni Marletta
- Laboratory for Molecular Surfaces and Nanotechnology (LAMSUN), Department of Chemical Sciences, University of Catania and CSGI, Viale A. Doria 6, 95125 Catania, Italy
| |
Collapse
|
41
|
Lakis R, Sauvage FL, Pinault E, Marquet P, Saint-Marcoux F, El Balkhi S. Semi-synthetic human albumin isoforms: Production, structure, binding capacities and influence on a routine laboratory test. Int J Biol Macromol 2023; 250:126239. [PMID: 37572814 DOI: 10.1016/j.ijbiomac.2023.126239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/02/2023] [Accepted: 08/07/2023] [Indexed: 08/14/2023]
Abstract
Human Serum Albumin (HSA) undergoes Post-Translational-Modifications (PTMs) leading to isoforms affecting its oncotic and non-oncotic properties. HSA is comprised of several isoforms whose abundance may vary with pathologies such as diabetes, kidney and liver diseases. Studying their impact separately may help to understand their sources and potential pathogenicity and further their evaluation as biomarkers. The present study examined semi-synthetic HSA isoforms to investigate independently their structure by means of advanced mass spectrometry techniques (LC-TOF-MS and ICP-MS), influence on the HSA binding/antioxidant activities using a binding capacity test, and potential impact on albumin quantification by a routine immunoturbidimetric assay. Applying different chemical reactions to a commercial HSA solution, we obtained different solutions enriched up to 53 % of native HSA, 78 % of acetylated HSA, 71 % of cysteinylated HSA, 94 % of oxidized HSA, 58 % of nitrosylated HSA and 96 % of glycated HSA, respectively. Moreover, the semi-synthetic isoforms showed differently altered binding capacities for a panel of ligands (Cu, Cd, Au, Ds and L-T4). Furthermore, immunoturbidimetry was found to be insensitive to the presence and abundance of the different isoforms. The fully characterized semi synthetic HSA isoforms obtained should be useful to further investigate their pathogenicity and potential roles as biomarkers.
Collapse
Affiliation(s)
- Roy Lakis
- P&T, UMR1248, University of Limoges, National Institute for Health and Medical Research (INSERM), Limoges, France
| | - François-Ludovic Sauvage
- P&T, UMR1248, University of Limoges, National Institute for Health and Medical Research (INSERM), Limoges, France
| | - Emilie Pinault
- P&T, UMR1248, University of Limoges, National Institute for Health and Medical Research (INSERM), Limoges, France
| | - Pierre Marquet
- P&T, UMR1248, University of Limoges, National Institute for Health and Medical Research (INSERM), Limoges, France; Department of Pharmacology, Toxicology and Pharmacovigilance, CHU Limoges, Limoges, France
| | - Franck Saint-Marcoux
- P&T, UMR1248, University of Limoges, National Institute for Health and Medical Research (INSERM), Limoges, France; Department of Pharmacology, Toxicology and Pharmacovigilance, CHU Limoges, Limoges, France
| | - Souleiman El Balkhi
- P&T, UMR1248, University of Limoges, National Institute for Health and Medical Research (INSERM), Limoges, France; Department of Pharmacology, Toxicology and Pharmacovigilance, CHU Limoges, Limoges, France.
| |
Collapse
|
42
|
Cavalieri G, Cilurzo G, Pettorosso L, Mansueto A, Laurini E, Pricl S. Biophysical and docking study on the interaction of anticancer drugs encorafenib and binimetinib with human serum albumin. Eur J Pharm Sci 2023; 189:106550. [PMID: 37527692 DOI: 10.1016/j.ejps.2023.106550] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/21/2023] [Accepted: 07/28/2023] [Indexed: 08/03/2023]
Abstract
The utilization of BRAF and MEK inhibitors in combination therapy has demonstrated superior outcomes in the treatment of melanoma as compared to monotherapy. In the present scenario, the combination therapy of Encorafenib (ENC), a BRAF inhibitor, and Binimetinib (BINI), a MEK inhibitor, has been identified as one of the most efficacious treatment modalities for this malignancy. Investigations of protein binding, particularly with human serum albumin (HSA), are essential to understand drug performance and enhance therapeutic outcomes. The investigation of the interplay between small molecule drugs and HSA is of paramount importance, given that such interactions can exert a substantial influence on the pharmacokinetics of these therapeutic agents. The present study aims to bridge these lacunae by implementing a comprehensive approach that integrates fluorescence spectroscopy (FS), isothermal titration calorimetry (ITC), far-ultraviolet circular dichroism (far-UV CD), and molecular simulations. Through analysis of the fluorescence quenching of HSA at three distinct temperatures, it was ascertained that the association constants for the complexes formed between drugs and HSA were of the magnitude of 104 M-1. This suggests that the interactions between the compounds and albumin were moderate and comparable. Simultaneously, the investigation of fluorescence indicated a contrasting binding mechanism for the two inhibitors: ENC predominantly binds to HSA through enthalpic interaction, while BINI/HSA is stabilized by entropic contributions. The data obtained was confirmed through experimental procedures conducted using the ITC method. The results of ligand-competitive displacement experiments indicate that ENC and BINI can bind to HSA within subdomain IIA, specifically Sudlow site I. However, far-UV CD studies show that there are no notable alterations in the structure of HSA upon binding with either of the two inhibitors. Ultimately, the results were supported by computational molecular analysis, which identified the key interactions that contribute to the stabilization of the two ligand/HSA complexes.
Collapse
Affiliation(s)
- Gabriele Cavalieri
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTS), DEA, University of Trieste, Piazzale Europa 1, 34127 Trieste, Italy
| | - Giulia Cilurzo
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTS), DEA, University of Trieste, Piazzale Europa 1, 34127 Trieste, Italy
| | - Lorenzo Pettorosso
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTS), DEA, University of Trieste, Piazzale Europa 1, 34127 Trieste, Italy
| | - Andrea Mansueto
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTS), DEA, University of Trieste, Piazzale Europa 1, 34127 Trieste, Italy
| | - Erik Laurini
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTS), DEA, University of Trieste, Piazzale Europa 1, 34127 Trieste, Italy.
| | - Sabrina Pricl
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTS), DEA, University of Trieste, Piazzale Europa 1, 34127 Trieste, Italy; Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, ul. Pomorska 141/143, 90-236 Łódź, Poland
| |
Collapse
|
43
|
Kowalska D, Dołżonek J, Żamojć K, Samsonov SA, Maszota-Zieleniak M, Makowska J, Stepnowski P, Białk-Bielińska A, Wyrzykowski D. Insights into the interaction of human serum albumin with ionic liquids - Thermodynamic, spectroscopic and molecular modelling studies. Int J Biol Macromol 2023; 249:125883. [PMID: 37499721 DOI: 10.1016/j.ijbiomac.2023.125883] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/12/2023] [Accepted: 07/17/2023] [Indexed: 07/29/2023]
Abstract
Human serum albumin (HSA) effectively binds different types of low-molecular-weight compounds and thus enables their distribution in living organisms. Recently, it has been reported that the protein-ligand interactions play a crucial role in bioaccumulation processes and provide an important sorption phase, especially for ionogenic compounds. Therefore, the binding interactions of such compounds with proteins are the subject of an ongoing interest in environmental and life sciences. In this paper, the influence of some counter-ions, namely [B(CN)4]- and [C(CN)3]- on the affinity of the [IM1-12]+ towards HSA has been investigated and discussed based on experimental methods (isothermal titration calorimetry and steady-state fluorescence spectroscopy) and molecular dynamics-based computational approaches. Furthermore, the thermal stability of the resulting HSA/ligand complexes was assessed using DSC and CD spectroscopy. As an outcome of the work, it has been ascertained that the protein is able to bind simultaneously the ligands under study but in different regions of HSA. Thus, the presence in the system of [IM1-12]+ does not disturb the binding of [C(CN)3]- and [B(CN)4]-. The presented results provide important information on the presence of globular proteins and some ionogenic compounds in the distribution and bioaccumulation of ILs in the environment and living organisms.
Collapse
Affiliation(s)
- Dorota Kowalska
- Department of Environmental Analysis, Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308 Gdańsk, Poland
| | - Joanna Dołżonek
- Department of Environmental Analysis, Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308 Gdańsk, Poland.
| | - Krzysztof Żamojć
- Department of General and Inorganic Chemistry, Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308 Gdańsk, Poland
| | - Sergey A Samsonov
- Department of Theoretical Chemistry, Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308 Gdańsk, Poland
| | - Martyna Maszota-Zieleniak
- Department of Theoretical Chemistry, Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308 Gdańsk, Poland
| | - Joanna Makowska
- Department of General and Inorganic Chemistry, Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308 Gdańsk, Poland
| | - Piotr Stepnowski
- Department of Environmental Analysis, Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308 Gdańsk, Poland
| | - Anna Białk-Bielińska
- Department of Environmental Analysis, Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308 Gdańsk, Poland
| | - Dariusz Wyrzykowski
- Department of General and Inorganic Chemistry, Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, 80-308 Gdańsk, Poland
| |
Collapse
|
44
|
Göktürk T, Sakallı Çetin E, Hökelek T, Pekel H, Şensoy Ö, Aksu EN, Güp R. Synthesis, Structural Investigations, DNA/BSA Interactions, Molecular Docking Studies, and Anticancer Activity of a New 1,4-Disubstituted 1,2,3-Triazole Derivative. ACS OMEGA 2023; 8:31839-31856. [PMID: 37692230 PMCID: PMC10483525 DOI: 10.1021/acsomega.3c03355] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 08/09/2023] [Indexed: 09/12/2023]
Abstract
We report herein a new 1,2,3-triazole derivative, namely, 4-((1-(3,4-dichlorophenyl)-1H-1,2,3-triazol-4-yl)methoxy)-2-hydroxybenzaldehyde, which was synthesized by copper(I)-catalyzed azide-alkyne cycloaddition (CuAAC). The structure of the compound was analyzed using Fourier transform infrared spectroscopy (FTIR), 1H NMR, 13C NMR, UV-vis, and elemental analyses. Moreover, X-ray crystallography studies demonstrated that the compound adapted a monoclinic crystal system with the P21/c space group. The dominant interactions formed in the crystal packing were found to be hydrogen bonding and van der Waals interactions according to Hirshfeld surface (HS) analysis. The volume of the crystal voids and the percentage of free spaces in the unit cell were calculated as 152.10 Å3 and 9.80%, respectively. The evaluation of energy frameworks showed that stabilization of the compound was dominated by dispersion energy contributions. Both in vitro and in silico investigations on the DNA/bovine serum albumin (BSA) binding activity of the compound showed that the CT-DNA binding activity of the compound was mediated via intercalation and BSA binding activity was mediated via both polar and hydrophobic interactions. The anticancer activity of the compound was also tested by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay using human cell lines including MDA-MB-231, LNCaP, Caco-2, and HEK-293. The compound exhibited more cytotoxic activity than cisplatin and etoposide on Caco-2 cancer cell lines with an IC50 value of 16.63 ± 0.27 μM after 48 h. Annexin V suggests the induction of cell death by apoptosis. Compound 3 significantly increased the loss of mitochondrial membrane potential (MMP) levels in Caco-2 cells, and the reactive oxygen species (ROS) assay proved that compound 3 could induce apoptosis by ROS generation.
Collapse
Affiliation(s)
- Tolga Göktürk
- Department
of Chemistry, Muğla Sıtkı
Koçman University, 48000 Muğla, Türkiye
| | - Esin Sakallı Çetin
- Department
of Medical Biology, Muğla Sıtkı
Koçman University, 48000 Muğla, Türkiye
| | - Tuncer Hökelek
- Department
of Physics, Hacettepe University, 06800 Ankara, Türkiye
| | - Hanife Pekel
- Department
of Pharmacy Services, Vocational School of Health Services, Istanbul Medipol University, 34810 Istanbul, Türkiye
| | - Özge Şensoy
- Department
of Computer Engineering, Istanbul Medipol
University, 34000 Istanbul, Türkiye
| | - Ebru Nur Aksu
- Department
of Medical Biology, Muğla Sıtkı
Koçman University, 48000 Muğla, Türkiye
| | - Ramazan Güp
- Department
of Chemistry, Muğla Sıtkı
Koçman University, 48000 Muğla, Türkiye
| |
Collapse
|
45
|
Hadidi S. A binuclear Cu(I)-phosphine complex as a specific HSA site I binder: synthesis, X-ray structure determination, and a comprehensive HSA interaction analysis. J Biomol Struct Dyn 2023; 41:7616-7626. [PMID: 36120938 DOI: 10.1080/07391102.2022.2123401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 09/04/2022] [Indexed: 10/14/2022]
Abstract
In this research, we present a method for synthesis and a detailed description of geometry characterization of a novel binuclear Cu(I) phosphine complex, along with analysis of its interaction with HSA using spectroscopic and simulation methods. The Cu atoms are coordinated in a tetrahedral geometry, which results in coordination by two nitrogen atoms from the N,N'-(ethane-1,2-diyl)bis(1-(pyridin-2-yl)methanimine ligand (L), a chloride, and a PPh3. The complex binding constant to HSA in a biochemical environment was determined to be ∼106, which is indicative of a strong interaction. The fluorescence of HSA is significantly quenched by binding to the complex via a static mechanism, whereas the microenvironment of the tryptophan residue remains unchanged. A spontaneous binding process was indicated by a negative value for ΔG. Thermodynamic signatures reflect the dominance of hydrophobic forces during the interaction. The site marker competitive experiment combined with docking simulation analysis revealed the closeness position of the complex binding site to warfarin location in specific ligand site I of HSA. The information generated in the present study would be valuable to understand the interaction mechanistic and pharmacological behavior of Cu(I) complexes.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Saba Hadidi
- Department of Inorganic Chemistry, Faculty of Chemistry, Razi University, Kermanshah, Iran
| |
Collapse
|
46
|
Mishra V, Pathak AK, Bandyopadhyay T. Binding of human serum albumin with uranyl ion at various pH: an all atom molecular dynamics study. J Biomol Struct Dyn 2023; 41:7318-7328. [PMID: 36099177 DOI: 10.1080/07391102.2022.2120080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 08/26/2022] [Indexed: 10/14/2022]
Abstract
Uranium is routinely handled in various stages of nuclear fuel cycle and its association with human serum albumin (HSA) has been reported in literature, however, their binding characteristics still remains obscure. The present study aims to understand interaction of uranium with HSA by employing all atom molecular dynamics simulation of the HSA-metal ion complex. His67, His247 and Asp249 residues constitute the major binding site of HSA, which capture the uranyl ion (UO22+). A total of six sets of initial coordinates are used for Zn2+-HSA and UO22+-HSA system at pH = 4, 7.4 and 9, respectively. Enhance sampling method, namely, well-tempered meta-dynamics (WT-MtD) is employed to study the binding and un-binding processes of UO22+ and Zn2+ ions. Potential of mean force (PMF) profiles are generated for all the six sets of complexes from the converged WT-MtD run. Various basins and barriers are observed along the (un)binding pathways. Hydrogen bond dynamics and short-range Coulomb interactions are evaluated from the equilibrium run at each basins and barriers for both the ions at all pH values. The binding of UO22+ ion with HSA is the result of the dynamical balance between UO22+-HSA and UO22+-water short range Coulomb interactions. Zn2+ ion interact more strongly than UO22+ at all pH through short range Coulomb interactions. PMF values further concludes that UO22+ cannot associate to the Zn2+ bound HSA protein but can be captured by free HSA at all pH values i.e. endosomal, alkaline and physiological pH.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Vijayakriti Mishra
- Radiation Safety Systems Division, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Arup Kumar Pathak
- Homi Bhabha National Institute, Mumbai, India
- Chemistry Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Tusar Bandyopadhyay
- Homi Bhabha National Institute, Mumbai, India
- Chemistry Division, Bhabha Atomic Research Centre, Mumbai, India
| |
Collapse
|
47
|
Duman B, Erkmen C, Zahirul Kabir M, Ching Yi L, Mohamad SB, Uslu B. In vitro interactions of two pesticides, propazine and quinoxyfen with bovine serum albumin: Spectrofluorometric and molecular docking investigations. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2023; 300:122907. [PMID: 37257323 DOI: 10.1016/j.saa.2023.122907] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 05/15/2023] [Accepted: 05/19/2023] [Indexed: 06/02/2023]
Abstract
Binding mechanisms of two selected pesticides, propazine (PRO) and quinoxyfen (QUI) with bovine serum albumin (BSA) was examined using fluorescence, absorption and molecular docking methods. Intrinsic fluorescence of BSA was quenched in the presence of both PRO and QUI. The quenching was ascertained to be conversely linked to temperature, which suggested the contribution of static quenching process in the PRO-BSA and QUI-BSA complex formations. This results were validated by the enhancement in absorption spectrum of BSA upon binding with PRO and QUI. Binding constant values (Kf = 9.55-0.60 × 10-3 M-1 for PRO-BSA system; Kf = 7.08-5.01 × 102 M-1 for QUI-BSA system) and number of binding site (n) values for the PRO-BSA and QUI-BSA systems at different temperatures affirmed a weak binding strength with a set of equivalent binding sites on BSA. Thermodynamic data obtained for both the PRO-BSA and QUI-BSA interactions predicted that the association process was spontaneous and non-covalent contacts such as hydrophobic interactions, van der Waals forces and hydrogen bonds participated in the binding reactions. This result was further supported by the molecular docking assessments. Three-dimensional spectral results revealed the microenvironmental alterations near tryptophan (Trp) and tyrosine (Tyr) residues in BSA by the addition of PRO and QUI. The docking analysis demonstrated the binding pattern for the PRO-BSA and QUI-BSA systems and disclosed the preferred binding site of both PRO and QUI as site I (subdomain IIA) of BSA.
Collapse
Affiliation(s)
- Bahadir Duman
- Ankara University, Faculty of Pharmacy, Department of Analytical Chemistry, 06560 Ankara, Türkiye; Ankara University, The Graduate School of Health Sciences, 06110 Ankara, Türkiye
| | - Cem Erkmen
- Ankara University, Faculty of Pharmacy, Department of Analytical Chemistry, 06560 Ankara, Türkiye
| | - Md Zahirul Kabir
- Ankara University, Faculty of Pharmacy, Department of Analytical Chemistry, 06560 Ankara, Türkiye
| | - Lim Ching Yi
- Faculty of Science, Bioinformatics Programme, Institute of Biological Sciences, University of Malaya, Kuala Lumpur, Malaysia
| | - Saharuddin B Mohamad
- Faculty of Science, Bioinformatics Programme, Institute of Biological Sciences, University of Malaya, Kuala Lumpur, Malaysia; Centre of Research for Computational Sciences and Informatics for Biology, Bioindustry, Environment, Agriculture and Healthcare, University of Malaya, Kuala Lumpur, Malaysia
| | - Bengi Uslu
- Ankara University, Faculty of Pharmacy, Department of Analytical Chemistry, 06560 Ankara, Türkiye.
| |
Collapse
|
48
|
Tavakoli Hafshajani K, Sohrabi N, Eslami Moghadam M, Oftadeh M. Spectroscopy and molecular dynamic study of the interaction of calf thymus DNA by anticancer Pt complex with butyl glycine ligand. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2023; 299:122826. [PMID: 37216815 DOI: 10.1016/j.saa.2023.122826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 04/29/2023] [Accepted: 05/04/2023] [Indexed: 05/24/2023]
Abstract
Despite the past few decades since the discovery of anticancer drugs, there is still no definitive treatment for its treatment. Cisplatin is a chemotherapy medication used to treat some cancers. In this research, the DNA binding affinity of Pt complex with butyl glycine ligand was studied by various spectroscopy methods and simulation studies. Fluorescence and UV-Vis spectroscopic data showed groove binding in ct-DNA-[Pt(NH3)2(butylgly)]NO3 complex formation by the spontaneous process. The results were also confirmed by small changes in CD spectra and thermal study (Tm), as well as the quenching emission of [Pt(NH3)2(butylgly)]NO3 complex on DNA. Finally, thermodynamic and binding parameters displayed that hydrophobic forces are the main forces. Based on docking simulation, [Pt(NH3)2(butylgly)]NO3 could bind to DNA and via minor groove binding on C-G center on DNA, formed a stable DNA complex.
Collapse
Affiliation(s)
| | - Nasrin Sohrabi
- Department of Chemistry, Payame Noor University (PNU), P.O.Box 19395-4697, Tehran, Iran.
| | | | - Mohsen Oftadeh
- Department of Chemistry, Payame Noor University (PNU), P.O.Box 19395-4697, Tehran, Iran
| |
Collapse
|
49
|
Yin J, Liu K, Yuan S, Guo Y, Yu H, Cheng Y, Xie Y, Qian H, Yao W. Carbon dots in breadcrumbs: Effect of frying on them and interaction with human serum albumin. Food Chem 2023; 424:136371. [PMID: 37210845 DOI: 10.1016/j.foodchem.2023.136371] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 04/25/2023] [Accepted: 05/11/2023] [Indexed: 05/23/2023]
Abstract
This research confirmed the existence of carbon dots (CDs) in breadcrumbs before frying, and CDs could be significantly affected by frying. The content of CDs increased from 0.013 ± 0.002% to 1.029 ± 0.002%, and the fluorescence quantum yield increased from 1.82 ± 0.01% to 3.16 ± 0.002% after frying at 180℃ for 5 min. The size reduced from 3.32 ± 0.71 nm to 2.67 ± 0.48 nm, and the content of N increased from 1.58% to 2.53%. In addition, the interaction of the CDs and human serum albumin (HSA) through electrostatic and hydrophobic induces the increase of α-helix structure and the change of the amino acid microenvironment of HSA. CDs corona, which may have physiological significance, was found through the transmission electron microscope.
Collapse
Affiliation(s)
- Jie Yin
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, China; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, China
| | - Kunfeng Liu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, China; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, China
| | - Shaofeng Yuan
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, China; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, China
| | - Yahui Guo
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, China; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, China
| | - Hang Yu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, China; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, China
| | - Yuliang Cheng
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, China; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, China
| | - Yunfei Xie
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, China; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, China
| | - He Qian
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, China; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, China
| | - Weirong Yao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, China; School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, China; Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, Jiangnan University, China.
| |
Collapse
|
50
|
Kubczak M, Grodzicka M, Michlewska S, Karimov M, Ewe A, Aigner A, Bryszewska M, Ionov M. The effect of novel tyrosine-modified polyethyleneimines on human albumin structure - Thermodynamic and spectroscopic study. Colloids Surf B Biointerfaces 2023; 227:113359. [PMID: 37209597 DOI: 10.1016/j.colsurfb.2023.113359] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/10/2023] [Accepted: 05/16/2023] [Indexed: 05/22/2023]
Abstract
The interaction of proteins with nanoparticle components are crucial for the evaluation of nanoparticle function, toxicity and biodistribution. Polyethyleneimines (PEIs) with defined tyrosine modifications are a class of novel polymers designed for improved siRNA delivery. Their interactions with biomacromolecules are still poorly described. This paper analyzes the interaction of different tyrosine-modified PEIs with human serum albumin as the most abundant serum protein. The ability of tyrosine modified, linear or branched PEIs to bind human serum albumin (HSA) was analyzed and further characterized. The interaction with hydrophobic parts of protein were studied using 1- nilinonaphthalene-8-sulfonic acid (ANS) and changes in the HSA secondary structure were evaluated using circular dichroism (CD). Complex formation and sizes were studied by transmission electron microscopy (TEM) and dynamic light scattering methods (DLS). We demonstrate that tyrosine modified PEIs are able to bind human serum albumin. Based on thermodynamic studies, van der Waals interaction, H-bonding and hydrophobic interactions are determined as main molecular forces involved in complex formation. Analysis of secondary structures revealed that the polymers decreased α-helix content, while increasing levels of randomly folded structures. Complex formation was confirmed by TEM and DLS. These findings are crucial for understanding polymer-protein interactions and the properties of nanoparticles.
Collapse
Affiliation(s)
- Małgorzata Kubczak
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Poland, PomorskaStr. 141/143, 90-236 Lodz, Poland.
| | - Marika Grodzicka
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Poland, PomorskaStr. 141/143, 90-236 Lodz, Poland; BioMedChem Doctoral School of the UL and Lodz Institutes of the Polish Academy of Science, Banacha 12/16, 90-237 Lodz, Poland
| | - Sylwia Michlewska
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Poland, PomorskaStr. 141/143, 90-236 Lodz, Poland; Laboratory of Microscopic Imaging and Specialized Biological Techniques, Faculty of Biology and Environmental Protection, University of Lodz, Poland, Banacha 12/16, 90-237 Lodz, Poland
| | - Michael Karimov
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, Faculty of Medicine, Leipzig University, Germany, Härtelstrasse 16-18, 04107 Leipzig, Germany
| | - Alexander Ewe
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, Faculty of Medicine, Leipzig University, Germany, Härtelstrasse 16-18, 04107 Leipzig, Germany
| | - Achim Aigner
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, Faculty of Medicine, Leipzig University, Germany, Härtelstrasse 16-18, 04107 Leipzig, Germany
| | - Maria Bryszewska
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Poland, PomorskaStr. 141/143, 90-236 Lodz, Poland
| | - Maksim Ionov
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Poland, PomorskaStr. 141/143, 90-236 Lodz, Poland
| |
Collapse
|