1
|
Woodson ME, Walden HF, Mottaleb MA, Makri M, Prifti GM, Moianos D, Pardali V, Zoidis G, Tavis JE. Efficacy and in vitro pharmacological assessment of novel N-hydroxypyridinediones as hepatitis B virus ribonuclease H inhibitors. Antimicrob Agents Chemother 2025; 69:e0145524. [PMID: 39601549 PMCID: PMC11784145 DOI: 10.1128/aac.01455-24] [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: 09/26/2024] [Accepted: 10/23/2024] [Indexed: 11/29/2024] Open
Abstract
We previously reported N-hydroxypyridinedione (HPD) compounds with mid-nanomolar efficacy and selectivity indexes around 300 against hepatitis B virus (HBV) replication. However, they lack pharmacological evaluation. Here, we report in vitro anti-HBV efficacy, cytotoxicity, and pharmacological characterization of 29 novel HPDs within seven subgroups. The best two compounds had EC50s of 61 and 190 nM and selectivity indexes of 526 and 1,071. Compounds with one halogen on the major R group were most effective and compounds with large ether R groups were most cytotoxic. Compounds were not cytotoxic in primary human hepatocytes. All compounds were freely soluble in pHs reflecting plasma (7.4) and the gastrointestinal tract (5 and 6.5). Almost all highly soluble compounds were passively permeable at pH 5.0 and 7.4. Only 2 of 11 compounds tested were likely to be effluxed by p-glycoprotein. The most potent HPDs inhibited HBV replication over human ribonuclease H1 activity by 10-fold. Four of 19 compounds inhibited CYP2D6 >50%, but their CYP2D6 IC50s were >8× higher than their anti-HBV EC50. No compound substantially inhibited CYP3A4. Thirteen of 15 compounds had human microsomal half-lives >30 min with medium to low rates of intrinsic clearance. Eleven of 12 compounds bound plasma proteins by ≥80%; however, effects against HBV replication for only one would likely be physiologically relevant. These results identify two lead candidate HPDs with pharmacological characteristics resembling commercially available drugs that are suitable for in vivo pharmacological and efficacy studies.
Collapse
Affiliation(s)
- Molly E. Woodson
- Department of Molecular Microbiology and Immunology, St. Louis University School of Medicine, St. Louis, Missouri, USA
- Institute for Drug and Biotherapeutic Innovation, St. Louis University, St. Louis, Missouri, USA
| | - Holly F. Walden
- Department of Molecular Microbiology and Immunology, St. Louis University School of Medicine, St. Louis, Missouri, USA
- Institute for Drug and Biotherapeutic Innovation, St. Louis University, St. Louis, Missouri, USA
| | - M. Abdul Mottaleb
- Institute for Drug and Biotherapeutic Innovation, St. Louis University, St. Louis, Missouri, USA
| | - Maria Makri
- Division of Pharmaceutical Chemistry, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, Athens, Greece
| | - Georgia-Myrto Prifti
- Division of Pharmaceutical Chemistry, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, Athens, Greece
| | - Dimitrios Moianos
- Division of Pharmaceutical Chemistry, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, Athens, Greece
| | - Vasiliki Pardali
- Division of Pharmaceutical Chemistry, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, Athens, Greece
| | - Grigoris Zoidis
- Division of Pharmaceutical Chemistry, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, Athens, Greece
| | - John E. Tavis
- Department of Molecular Microbiology and Immunology, St. Louis University School of Medicine, St. Louis, Missouri, USA
- Institute for Drug and Biotherapeutic Innovation, St. Louis University, St. Louis, Missouri, USA
| |
Collapse
|
2
|
Bao Y, Wang Y, Liu H, Lan J, Li Z, Zong W, Zhao Z. Co-Existing Nanoplastics Further Exacerbates the Effects of Triclosan on the Physiological Functions of Human Serum Albumin. Life (Basel) 2025; 15:112. [PMID: 39860052 PMCID: PMC11766571 DOI: 10.3390/life15010112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/09/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
The potential health risks posed by the coexistence of nanoplastics (NPs) and triclosan (TCS) have garnered significant attention. However, the effects and underlying mechanisms of NPs and TCS on key functional proteins at the molecular level remain poorly understood. This study reports the effect of polystyrene nanoplastics (PSNPs) on the binding of TCS to human serum albumin (HSA) using multispectral methods and molecular simulation systems. The experimental results show that TCS significantly inhibits HSA esterase activity, with exacerbating inhibition in the presence of PSNPs, which is attributed to the alteration of HSA conformation and microenvironment of the amino acid residues induced by PSNPs. Molecular docking and site marker competitive studies indicate that TCS predominantly binds to site I of subdomain Sudlow II and the presence of PSNPs does not affect the binding sites. Spectra analyses indicate that the quenching mechanism between TCS and HSA belongs to the static quenching type and the presence of PSNPs does not change the fluorescence quenching type. The HSA fluorescence quenching and the conformational alterations induced by TCS are further enhanced in the presence of PSNPs, indicating that PSNPs enhance the binding of TCS to HSA by making TCS more accessible to the binding sites. This study provides valuable information about the toxicity of PSNPs and TCS in case of co-exposure.
Collapse
Affiliation(s)
- Yan Bao
- Shandong Key Laboratory of Water Pollution Control and Resource Reuse, School of Environmental Science and Engineering, Shandong University, Qingdao 266237, China
- Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Shandong Energy Institute, Qingdao New Energy Shandong Laboratory, Qingdao 266101, China
| | - Yaoyao Wang
- School of Environment and Geography, Qingdao University, Qingdao 266071, China
| | - Hongbin Liu
- School of Environment and Geography, Qingdao University, Qingdao 266071, China
| | - Jing Lan
- School of Environment and Geography, Qingdao University, Qingdao 266071, China
| | - Zhicai Li
- Anqiu Branch of Weifang Municipal Bureau of Ecology and Environment, Weifang 262199, China
| | - Wansong Zong
- College of Geography and Environment, Shandong Normal University, Jinan 250014, China
| | - Zongshan Zhao
- School of Environment and Geography, Qingdao University, Qingdao 266071, China
| |
Collapse
|
3
|
Coppinger C, Anderson PL. Considerations for drug-drug interactions between long-acting antiretrovirals and immunosuppressants for solid organ transplantation. Expert Opin Drug Metab Toxicol 2025:1-4. [PMID: 39757464 DOI: 10.1080/17425255.2024.2448970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 12/30/2024] [Indexed: 01/07/2025]
Affiliation(s)
- Corwin Coppinger
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Peter L Anderson
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
4
|
Ivanov I, Manolov S, Bojilov D, Stremski Y, Marc G, Statkova-Abeghe S, Oniga S, Oniga O, Nedialkov P. Synthesis of Novel Benzothiazole-Profen Hybrid Amides as Potential NSAID Candidates. Molecules 2024; 30:107. [PMID: 39795166 PMCID: PMC11721736 DOI: 10.3390/molecules30010107] [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: 11/30/2024] [Revised: 12/15/2024] [Accepted: 12/29/2024] [Indexed: 01/13/2025] Open
Abstract
Herein, we report the synthesis of a series of new compounds by combining 2-aminobenzothiazole with various profens. The compounds were characterized using techniques such as 1H- and 13C-NMR, FT-IR spectrometry, and high-resolution mass spectrometry (HRMS), with detailed HRMS analysis conducted for each molecule. Their biological activities were tested in vitro, revealing significant anti-inflammatory and antioxidant effects, comparable to those of standard reference compounds. Lipophilicity was experimentally determined through partition coefficient (RM) measurements. To understand their binding affinity, molecular docking studies were perfsormed to analyze interactions with human serum albumin (HSA). The stability of these predicted complexes was further evaluated through molecular dynamics simulations. The results highlight the compounds' promising biological activity and strong affinity for HSA. The new hybrid molecule between 2-ABT and ketoprofen 3b demonstrates significant promise based on the experimental data and is further supported by in silico calculations. Compound 3b exhibits the best hydrogen peroxide scavenging activity among the tested compounds, with an IC50 of 60.24 μg/mL. Furthermore, 3b also displays superior anti-inflammatory activity, with an IC50 of 54.64 μg/mL, making it more effective than the standard ibuprofen (76.05 μg/mL).
Collapse
Affiliation(s)
- Iliyan Ivanov
- Department of Organic Chemistry, Faculty of Chemistry, University of Plovdiv, 24 “Tsar Assen” Str., 4000 Plovdiv, Bulgaria; (S.M.); (D.B.); (Y.S.); (S.S.-A.)
| | - Stanimir Manolov
- Department of Organic Chemistry, Faculty of Chemistry, University of Plovdiv, 24 “Tsar Assen” Str., 4000 Plovdiv, Bulgaria; (S.M.); (D.B.); (Y.S.); (S.S.-A.)
| | - Dimitar Bojilov
- Department of Organic Chemistry, Faculty of Chemistry, University of Plovdiv, 24 “Tsar Assen” Str., 4000 Plovdiv, Bulgaria; (S.M.); (D.B.); (Y.S.); (S.S.-A.)
| | - Yordan Stremski
- Department of Organic Chemistry, Faculty of Chemistry, University of Plovdiv, 24 “Tsar Assen” Str., 4000 Plovdiv, Bulgaria; (S.M.); (D.B.); (Y.S.); (S.S.-A.)
| | - Gabriel Marc
- Department of Organic Chemistry, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 41 Victor Babeș Street, RO-400010 Cluj-Napoca, Romania;
| | - Stela Statkova-Abeghe
- Department of Organic Chemistry, Faculty of Chemistry, University of Plovdiv, 24 “Tsar Assen” Str., 4000 Plovdiv, Bulgaria; (S.M.); (D.B.); (Y.S.); (S.S.-A.)
| | - Smaranda Oniga
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 41 Victor Babeș Street, RO-400010 Cluj-Napoca, Romania;
| | - Ovidiu Oniga
- Department of Therapeutic Chemistry, “Iuliu Hațieganu” University of Medicine and Pharmacy, 12 Ion Creangă Street, RO-400010 Cluj-Napoca, Romania;
| | - Paraskev Nedialkov
- Department of Pharmacognosy, Faculty of Pharmacy, Medical University of Sofia, 2 Dunav Street, 1000 Sofia, Bulgaria;
| |
Collapse
|
5
|
Shamsi M, Al-Asbahy WM, Al-Areqi HQN, Alzowahi FAM. Probing the Biomolecular Interactions of DNA/HSA with the New Sn(IV) Complex and Computational Perspectives: Design, Synthesis, Characterization, Anticancer Activity, and Molecular Modeling Approach. J Med Chem 2024; 67:21841-21858. [PMID: 39661984 DOI: 10.1021/acs.jmedchem.4c01306] [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: 12/13/2024]
Abstract
The ligands 2,2'-bipyridyl and indole-3-carboxylic acid were used to create a Sn(IV) complex, which was then synthesized and carefully characterized using elemental analysis and spectroscopic techniques (UV-vis, IR, 1H, 13C, and 119Sn NMR, and ESI-MS) and RXPD. Utilizing biophysical techniques such as UV-vis, fluorescence titrations, circular dichroism, FTIR (for HSA), and cleavage activity (for DNA), in vitro binding studies of Sn(IV) complex and DNA/HSA were satisfied with the strong electrostatic binding interaction of the Sn(IV) complex via the phosphate backbone of the DNA helix as well as in the subdomain IIA of HSA. The observed trend in the binding interactions and computational studies of the Sn(IV) complex was attributed to the nature of the ligands bound to the Sn(IV) center that influences their in vitro activities. The Sn(IV) complex showed sufficient effectiveness to be considered a viable candidate for the creation of anticancer medications.
Collapse
Affiliation(s)
- Manal Shamsi
- Department of Pharmacy, Faculty of Medical Sciences, National University (Ibb Branch), Ibb 46654, Yemen
| | - Waddhaah M Al-Asbahy
- Department of Chemistry, Faculty of Applied Sciences, Taiz University, Taiz 6803, Yemen
| | - Hakim Q N Al-Areqi
- Department of Physics, Faculty of Applied Sciences, Taiz University, Taiz 6803, Yemen
| | - Fahad A M Alzowahi
- Department of Pharmacy, Faculty of Medical Sciences, National University (Ibb Branch), Ibb 46654, Yemen
| |
Collapse
|
6
|
Fernández-Sainz J, Herrera-Ochoa D, Pacheco-Liñán PJ, Darder M, Albaladejo J, Bravo I, Garzón-Ruiz A. Spectroscopic study on volasertib: Highly stable complexes with albumin and encapsulation into alginate/montmorillonite bionanocomposites. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 322:124823. [PMID: 39033609 DOI: 10.1016/j.saa.2024.124823] [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: 02/26/2024] [Revised: 07/05/2024] [Accepted: 07/13/2024] [Indexed: 07/23/2024]
Abstract
In the present work, we study different physicochemical properties related to LADME processes of volasertib, a Polo-like kinase 1 inhibitor in advanced clinical trials. Firstly, the protonation equilibria, the extent of ionization at the physiological pH and pKa values of this drug are studied combining spectroscopic techniques and computational calculations. Secondly, the binding process of volasertib to the human serum albumin (HSA) protein is analyzed by fluorescence spectroscopy. We report a high binding constant to HSA (Ka = 4.10 × 106 M-1) and their pharmacokinetic implications are discussed accordingly. The negative enthalpy and entropy (ΔH0 = -54.49 kJ/mol; ΔS0 = -58.90 J K-1 mol-1) determined for the binding process suggests the implication of hydrogen bonds and van der Waals interactions in the formation of the HSA-volasertib complex. Additionally, volasertib is encapsulated in an alginate/montmorillonite bionanocomposite as a proof of concept for an oral delivery nanocarrier. The physical properties of that nanocomposite as well as volasertib delivery kinetics are analyzed.
Collapse
Affiliation(s)
- Jesús Fernández-Sainz
- Departamento de Química Física, Facultad de Farmacia, Universidad de Castilla-La Mancha, Av. Dr. José María Sánchez Ibáñez, s/n, 02071 Albacete, Spain
| | - Diego Herrera-Ochoa
- Departamento de Química Física, Facultad de Farmacia, Universidad de Castilla-La Mancha, Av. Dr. José María Sánchez Ibáñez, s/n, 02071 Albacete, Spain
| | - Pedro J Pacheco-Liñán
- Departamento de Química Física, Facultad de Farmacia, Universidad de Castilla-La Mancha, Av. Dr. José María Sánchez Ibáñez, s/n, 02071 Albacete, Spain
| | - Margarita Darder
- Instituto de Ciencia de Materiales de Madrid (ICMM), CSIC, Cantoblanco, 28049 Madrid, Spain
| | - José Albaladejo
- Departamento de Química Física, Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, Avenida Camilo José Cela, 10, 13071 Ciudad Real, Spain
| | - Iván Bravo
- Departamento de Química Física, Facultad de Farmacia, Universidad de Castilla-La Mancha, Av. Dr. José María Sánchez Ibáñez, s/n, 02071 Albacete, Spain
| | - Andrés Garzón-Ruiz
- Departamento de Química Física, Facultad de Farmacia, Universidad de Castilla-La Mancha, Av. Dr. José María Sánchez Ibáñez, s/n, 02071 Albacete, Spain.
| |
Collapse
|
7
|
Zhang SH, Zeng N, Xu JZ, Liu CQ, Xu MY, Sun JX, An Y, Zhong XY, Miao LT, Wang SG, Xia QD. Recent breakthroughs in innovative elements, multidimensional enhancements, derived technologies, and novel applications of PROTACs. Biomed Pharmacother 2024; 180:117584. [PMID: 39427546 DOI: 10.1016/j.biopha.2024.117584] [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: 08/06/2024] [Revised: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 10/22/2024] Open
Abstract
Proteolysis Targeting Chimera (PROTAC) is an emerging and evolving technology based on targeted protein degradation (TPD). Small molecule PROTACs have shown great efficacy in degrading disease-specific proteins in preclinical and clinical studies, but also showed various limitations. In recent years, new technologies and advances in TPD have provided additional optimized strategies based on conventional PROTACs that can overcome the shortcomings of conventional PROTACs in terms of undruggable targets, bioavailability, tissue-specificity, spatiotemporal control, and degradation scope. In addition, some designs of special targeting chimeras and applications based on multidisciplinary science have shed light on novel therapeutic modalities and drug design. However, each improvement has its own advantages, disadvantages and application conditions. In this review, we summarize the exploration of PROTAC elements, depict a landscape of improvements and derived concepts of PROTACs, and expect to provide perspectives for technological innovations, combinations and applications in future targeting chimera design.
Collapse
Affiliation(s)
- Si-Han Zhang
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China
| | - Na Zeng
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China
| | - Jin-Zhou Xu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China
| | - Chen-Qian Liu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China
| | - Meng-Yao Xu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China
| | - Jian-Xuan Sun
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China
| | - Ye An
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China
| | - Xing-Yu Zhong
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China
| | - Lin-Tao Miao
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China
| | - Shao-Gang Wang
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China.
| | - Qi-Dong Xia
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China.
| |
Collapse
|
8
|
Catalano C, Lucier KW, To D, Senko S, Tran NL, Farwell AC, Silva SM, Dip PV, Poweleit N, Scapin G. The CryoEM structure of human serum albumin in complex with ligands. J Struct Biol 2024; 216:108105. [PMID: 38852682 DOI: 10.1016/j.jsb.2024.108105] [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: 03/29/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 06/11/2024]
Abstract
Human serum albumin (HSA) is the most prevalent plasma protein in the human body, accounting for 60 % of the total plasma protein. HSA plays a major pharmacokinetic function, serving as a facilitator in the distribution of endobiotics and xenobiotics within the organism. In this paper we report the cryoEM structures of HSA in the apo form and in complex with two ligands (salicylic acid and teniposide) at a resolution of 3.5, 3.7 and 3.4 Å, respectively. We expand upon previously published work and further demonstrate that sub-4 Å maps of ∼60 kDa proteins can be routinely obtained using a 200 kV microscope, employing standard workflows. Most importantly, these maps allowed for the identification of small molecule ligands, emphasizing the practical applicability of this methodology and providing a starting point for subsequent computational modeling and in silico optimization.
Collapse
Affiliation(s)
- Claudio Catalano
- NanoImaging Services, 4940 Carroll Canyon Road, Suite 115, San Diego, CA 92121, USA.
| | - Kyle W Lucier
- NanoImaging Services, 4940 Carroll Canyon Road, Suite 115, San Diego, CA 92121, USA
| | - Dennis To
- NanoImaging Services, 4940 Carroll Canyon Road, Suite 115, San Diego, CA 92121, USA
| | - Skerdi Senko
- NanoImaging Services, 4940 Carroll Canyon Road, Suite 115, San Diego, CA 92121, USA
| | - Nhi L Tran
- NanoImaging Services, 4940 Carroll Canyon Road, Suite 115, San Diego, CA 92121, USA
| | - Ashlyn C Farwell
- NanoImaging Services, 4940 Carroll Canyon Road, Suite 115, San Diego, CA 92121, USA
| | - Sabrina M Silva
- NanoImaging Services, 4940 Carroll Canyon Road, Suite 115, San Diego, CA 92121, USA
| | - Phat V Dip
- NanoImaging Services, 4940 Carroll Canyon Road, Suite 115, San Diego, CA 92121, USA
| | - Nicole Poweleit
- NanoImaging Services, 4940 Carroll Canyon Road, Suite 115, San Diego, CA 92121, USA
| | - Giovanna Scapin
- NanoImaging Services, 4940 Carroll Canyon Road, Suite 115, San Diego, CA 92121, USA
| |
Collapse
|
9
|
Guzzi R, Bartucci R. Thermal effects and drugs competition on the palmitate binding capacity of human serum albumin. Biochem Biophys Res Commun 2024; 722:150168. [PMID: 38797156 DOI: 10.1016/j.bbrc.2024.150168] [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: 03/14/2024] [Revised: 05/17/2024] [Accepted: 05/22/2024] [Indexed: 05/29/2024]
Abstract
Human serum albumin (HSA) is the most abundant plasma protein of the circulatory system. It is a multidomain, multifunctional protein that, combining diverse affinities and wide specificity, binds, stores, and transports a variety of biological compounds, pharmacores, and fatty acids. HSA is finding increasing uses in drug-delivery due to its ability to carry functionalized ligands and prodrugs. All this raises the question of competition for binding sites occupancy in case of multiple ligands, which in turn influences the protein structure/dynamic/function relationship and also has an impact on the biomedical applications. In this work, the effects of interactive binding of palmitic acid (PA), warfarin (War) and ibuprofen (Ibu) on the thermal stability of HSA were studied using DSC, ATR-FTIR, and EPR. PA is a high-affinity physiological ligand, while the two drugs are widely used for their anticoagulant (War) and anti-inflammatory (Ibu) efficacy, and are exogenous compounds that accommodate in the deputed drug site DS1 and DS2, respectively overlapping with some of the fatty acid binding sites. The results indicate that HSA acquires the highest thermal stability when it is fully saturated with PA. The binding of this physiological ligand does not hamper the binding of War or Ibu to the native state of the protein. In addition, the three ligands bind simultaneously, suggesting a synergic cooperative influence due to allosteric effects. The increased thermal stability subsequent to binary and multiple ligands binding moderates protein aggregation propensity and restricts protein dynamics. The biophysics findings provide interesting features about protein stability, aggregation, and dynamics in interaction with multiple ligands and are relevant in drug-delivery.
Collapse
Affiliation(s)
- Rita Guzzi
- Department of Physics, Molecular Biophysics Laboratory, University of Calabria, 87036, Rende, Italy; CNR-NANOTEC, Department of Physics, University of Calabria, 87036, Rende, Italy.
| | - Rosa Bartucci
- Department of Physics, Molecular Biophysics Laboratory, University of Calabria, 87036, Rende, Italy
| |
Collapse
|
10
|
Bisaria I, Chauhan C, Muthu SA, Parvez S, Ahmad B. The effect of chrysin binding on the conformational dynamics and unfolding pathway of human serum albumin. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 316:124332. [PMID: 38676982 DOI: 10.1016/j.saa.2024.124332] [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: 11/29/2023] [Revised: 03/20/2024] [Accepted: 04/22/2024] [Indexed: 04/29/2024]
Abstract
Studies on the interactions between ligands and proteins provide insights into how a possible medication alters the structures and activities of the target or carrier proteins. The natural flavonoid aglycone Chrysin (CHR) has demonstrated anti-inflammatory, antioxidant, antiapoptotic, neuroprotective, and antineoplastic effects, both in vitro and in vivo. In this work, we investigated the impact of CHR binding on the as-yet-unexplored conformation, dynamics, and unfolding mechanism of human serum albumin (HSA). We determined CHR binding to HSA domain-II with the association constant (Ka) of 2.70 ± 0.21 × 105 M-1. The urea-induced sequential unfolding mechanism of HSA was used to elucidate the debatable binding location of CHR. CHR binding induced both secondary and tertiary structural alterations in the protein as studied by far-UV circular dichroism and intrinsic fluorescence spectroscopy. Red edge excitation shift (REES) indicated a decrease in conformational dynamics of the protein on the complex formation. This suggested an ordered compact and spatial arrangement of the CHR-boundmolecule. The binding of CHR was found to significantly modulate the urea-induced unfolding pathway of HSA. Urea-induced unfolding pathway of HSA became a two-state process (N-U) from a three-state process (N-I-U). The interaction of CHR is found to increase the thermal stability of the protein by ∼4 °C. This study focuses on the fundamental sciences and demonstrates how prospective medication compounds can alter the dynamics and stability of protein structure.
Collapse
Affiliation(s)
- Ishita Bisaria
- Protein Assembly Laboratory, Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Chanchal Chauhan
- Protein Assembly Laboratory, Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India; Department of Molecular Medicine, School of Interdisciplinary Studies, Jamia Hamdard, New Delhi 110062, India
| | - Shivani A Muthu
- Protein Assembly Laboratory, Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India; Department of Molecular Medicine, School of Interdisciplinary Studies, Jamia Hamdard, New Delhi 110062, India
| | - Suhel Parvez
- Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Basir Ahmad
- Protein Assembly Laboratory, Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
11
|
Yang Y, Wei S, Tian H, Cheng J, Zhong Y, Zhong X, Huang D, Jiang C, Ke X. Adverse event profile of memantine and donepezil combination therapy: a real-world pharmacovigilance analysis based on FDA adverse event reporting system (FAERS) data from 2004 to 2023. Front Pharmacol 2024; 15:1439115. [PMID: 39101151 PMCID: PMC11294921 DOI: 10.3389/fphar.2024.1439115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 06/24/2024] [Indexed: 08/06/2024] Open
Abstract
Background Donepezil in combination with memantine is a widely used clinical therapy for moderate to severe dementia. However, real-world population data on the long-term safety of donepezil in combination with memantine are incomplete and variable. Therefore, the aim of this study was to analyze the adverse events (AEs) of donepezil in combination with memantine according to US Food and Drug Administration Adverse Event Reporting System (FAERS) data to provide evidence for the safety monitoring of this therapy. Methods We retrospectively analyzed reports of AEs associated with the combination of donepezil and memantine from 2004 to 2023 extracted from the FAERS database. Whether there was a significant association between donepezil and memantine combination therapy and AEs was assessed using four disproportionality analysis methods, namely, the reporting odds ratio, proportional reporting ratio, Bayesian confidence propagation neural network, and multi-item gamma Poisson shrinker methods. To further investigate potential safety issues, we also analyzed differences and similarities in the time of onset and incidence of AEs stratified by sex and differences and similarities in the incidence of AEs stratified by age. Results Of the 2,400 adverse drug reaction (ADR) reports in which the combination of donepezil and memantine was the primary suspected drug, most of the affected patients were female (54.96%) and older than 65 years of age (79.08%). We identified 22 different system organ classes covering 100 AEs, including some common AEs such as dizziness and electrocardiogram PR prolongation; fall, pleurothotonus and myoclonus were AEs that were not listed on the drug label. Moreover, we obtained 88 reports of AEs in men and 100 reports of AEs in women; somnolence was a common AE in both men and women and was more common in women, whereas pleurothotonus was a more common AE in men. In addition, we analyzed 12 AEs in patients younger than 18 years, 16 in patients between 18 and 65 years, and 113 in patients older than 65 years. The three age groups had distinctive AEs, but lethargy was the common AE among all age groups. Finally, the median time to AE onset was 19 days in all cases. In both men and women, most AEs occurred within a month of starting donepezil plus memantine, but some continued after a year of treatment. Conclusion Our study identified potential and new AEs of donepezil in combination with memantine; some of these AEs were the same as in the specification, and some of the AE signals were not shown in the specification. In addition, there were sex and age differences in some of the AEs. Therefore, our findings may provide valuable insights for further studies on the safety of donepezil and memantine combination therapy, which are expected to contribute to the safe use of this therapy in clinical practice.
Collapse
Affiliation(s)
- Yihan Yang
- The Institution of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Sheng Wei
- Department of General Practice, The Second Affiliated Hospital of Wannan Medical College, Anhui, China
| | - Huan Tian
- Department of Rehabilitation Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- School of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jing Cheng
- The First Clinical Medical College, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yue Zhong
- Department of Rehabilitation Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiaoling Zhong
- Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Dunbing Huang
- Department of Rehabilitation Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Cai Jiang
- Rehabilitation Medicine Center, Fujian Provincial Hospital, Fuzhou, China
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| | - Xiaohua Ke
- Department of Rehabilitation Medicine, Shanghai Fourth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
12
|
Holowinski P, Dybowski MP. Determination of 3- and 4-chloromethcathinone interactions with plasma proteins: study involving analytical and theoretical methods. Forensic Toxicol 2024; 42:111-124. [PMID: 38108940 PMCID: PMC11269353 DOI: 10.1007/s11419-023-00677-7] [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: 05/14/2023] [Accepted: 09/27/2023] [Indexed: 12/19/2023]
Abstract
PURPOSE The purpose of this paper was to determine 3- and 4-chloromethcathinone (3- and 4-CMC) binding degree and possible binding interaction modes with human serum albumin (HSA) using analytical and theoretical methods. METHODS Experimental determination of 3- and 4-CMC binding degree with HSA was performed using gas chromatography-tandem mass spectrometry preceded by the equilibrium dialysis (ED) and ultrafiltration (UF). Nuclear magnetic resonance (NMR) spectroscopy was used to determine 3- and 4-CMC epitope-binding maps and possible binding sites in HSA. The molecular docking and molecular dynamics were employed to obtain detailed information about binding modes of 3- and 4-CMC enantiomers in HSA. RESULTS As follows from the presented data, the degree of binding of 3- and 4-CMC is at a similar level of approx. 80%. This indicates a relatively strong binding of CMC to plasma proteins. The model studies employing the NMR spectroscopy and molecular simulations indicate that both CMCs bind to HSA. The whole 3- and 4-CMC molecules are embedded in the binding sites, with aromatic moieties being in the closest contact with the HSA residues. Moreover, conducted experiments show that Sudlow site II is the main binding center for 3- and 4-CMC and Sudlow site I acts as the secondary binding site. CONCLUSIONS Although many studies describe pharmacological and toxicological properties of synthetic cathinones (SC), the data taking SCs binding in plasma into consideration are scarce. To our knowledge, this is the first report presenting comprehensive experimental and theoretical characterization of 3- and 4-CMC binding with plasma proteins.
Collapse
Affiliation(s)
- Piotr Holowinski
- Faculty of Chemistry, Department of Chromatography, Institute of Chemical Sciences, Maria Curie Sklodowska University in Lublin, 20-031, Lublin, Poland
| | - Michal P Dybowski
- Faculty of Chemistry, Department of Chromatography, Institute of Chemical Sciences, Maria Curie Sklodowska University in Lublin, 20-031, Lublin, Poland.
| |
Collapse
|
13
|
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
|
14
|
Simko P, Leskanicova A, Suvakova-Nunhart M, Koval J, Zidekova N, Karasova M, Majerova P, Verboova L, Blicharova A, Kertys M, Barvik I, Kovac A, Kiskova T. The First In Vivo Study Shows That Gyrophoric Acid Changes Behavior of Healthy Laboratory Rats. Int J Mol Sci 2024; 25:6782. [PMID: 38928485 PMCID: PMC11203575 DOI: 10.3390/ijms25126782] [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: 04/12/2024] [Revised: 06/17/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Gyrophoric acid (GA), a lichen secondary metabolite, has attracted more attention during the last years because of its potential biological effects. Until now, its effect in vivo has not yet been demonstrated. The aim of our study was to evaluate the basic physicochemical and pharmacokinetic properties of GA, which are directly associated with its biological activities. The stability of the GA in various pH was assessed by conducting repeated UV-VIS spectral measurements. Microsomal stability in rat liver microsomes was performed using Ultra-Performance LC/MS. Binding to human serum albumin (HSA) was assessed using synchronous fluorescence spectra, and molecular docking analysis was used to reveal the binding site of GA to HSA. In the in vivo experiment, 24 Sprague-Dawley rats (Velaz, Únetice, Czech Republic) were used. The animals were divided as follows. The first group (n = 6) included healthy males as control intact rats (♂INT), and the second group (n = 6) included healthy females as controls (♀INT). Groups three and four (♂GA/n = 6 and ♀GA/n = 6) consisted of animals with daily administered GA (10 mg/kg body weight) in an ethanol-water solution per os for a one-month period. We found that GA remained stable under various pH and temperature conditions. It bonded to human serum albumin with the binding constant 1.788 × 106 dm3mol-1 to reach the target tissue via this mechanism. In vivo, GA did not influence body mass gain, food, or fluid intake during the experiment. No liver toxicity was observed. However, GA increased the rearing frequency in behavioral tests (p < 0.01) and center crossings in the elevated plus-maze (p < 0.01 and p < 0.001, respectively). In addition, the time spent in the open arm was prolonged (p < 0.01 and p < 0.001, respectively). Notably, GA was able to pass through the blood-brain barrier, indicating its ability to permeate into the brain and to stimulate neurogenesis in the hilus and subgranular zone of the hippocampus. These observations highlight the potential role of GA in influencing brain function and neurogenesis.
Collapse
Affiliation(s)
- Patrik Simko
- Institute of Biology and Ecology, Faculty of Science, Pavol Jozef Šafárik University in Kosice, 040 01 Kosice, Slovakia; (P.S.); (A.L.); (J.K.)
| | - Andrea Leskanicova
- Institute of Biology and Ecology, Faculty of Science, Pavol Jozef Šafárik University in Kosice, 040 01 Kosice, Slovakia; (P.S.); (A.L.); (J.K.)
| | - Maria Suvakova-Nunhart
- Institute of Chemistry, Faculty of Science, Pavol Jozef Šafárik University in Kosice, 040 01 Kosice, Slovakia;
| | - Jan Koval
- Institute of Biology and Ecology, Faculty of Science, Pavol Jozef Šafárik University in Kosice, 040 01 Kosice, Slovakia; (P.S.); (A.L.); (J.K.)
| | - Nela Zidekova
- Biomedical Center Martin (BioMed), Jessenius Faculty of Medicine in Martin, Comenius University, 841 99 Bratislava, Slovakia; (N.Z.); (M.K.)
| | - Martina Karasova
- Small Animal Clinic, University of Veterinary Medicine and Pharmacy in Kosic, 041 81 Kosice, Slovakia;
| | - Petra Majerova
- Institute of Neuroimmunology, Slovak Academy of Sciences, 831 01 Bratislava, Slovakia; (P.M.); (A.K.)
| | - Ludmila Verboova
- Institute of Pathology, Faculty of Medicine, Pavol Jozef Šafárik University in Kosice, 040 01 Kosice, Slovakia; (L.V.); (A.B.)
| | - Alzbeta Blicharova
- Institute of Pathology, Faculty of Medicine, Pavol Jozef Šafárik University in Kosice, 040 01 Kosice, Slovakia; (L.V.); (A.B.)
| | - Martin Kertys
- Biomedical Center Martin (BioMed), Jessenius Faculty of Medicine in Martin, Comenius University, 841 99 Bratislava, Slovakia; (N.Z.); (M.K.)
| | - Ivan Barvik
- Institute of Physics, Faculty of Mathematics and Physics, Charles University, 110 00 Prague, Czech Republic;
| | - Andrej Kovac
- Institute of Neuroimmunology, Slovak Academy of Sciences, 831 01 Bratislava, Slovakia; (P.M.); (A.K.)
| | - Terezia Kiskova
- Institute of Biology and Ecology, Faculty of Science, Pavol Jozef Šafárik University in Kosice, 040 01 Kosice, Slovakia; (P.S.); (A.L.); (J.K.)
| |
Collapse
|
15
|
Babayeva M, Srdanovic I. Non-linear plasma protein binding of cannabidiol. J Cannabis Res 2024; 6:27. [PMID: 38902820 PMCID: PMC11191238 DOI: 10.1186/s42238-024-00238-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 05/27/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND Cannabidiol is highly bound to plasma proteins. Changes in its protein binding can lead to altered unbound plasma concentrations and result in alteration of pharmacological activity of cannabidiol-containing medications. This research has assessed non-linearity of cannabidiol plasma protein binding and the potential effect of tizoxanide on the binding. METHOD Cannabidiol protein binding was evaluated by ultrafiltration technique. Human plasma was spiked with cannabidiol stock solution to produce samples of various concentrations. For interaction study potential interactant tizoxanide was added in each sample. All samples were processed through Amicon Micropartition system and analyzed by HPLC. RESULTS The study has detected cannabidiol binding to borosilicate glass (9%) and polyethylene plastics (15%). In the interaction study the mean protein unbound fraction of cannabidiol was 0.05 (5%), indicating no binding interaction between cannabidiol and tizoxanide since cannabidiol unbound fraction without tizoxanide was also 5%. The cannabidiol fraction unbound was more than 2-fold greater at high concentrations compared to low concentrations. CONCLUSION a). At high concentrations cannabidiol plasma protein binding is non-linear. The non-linearity can affect elimination and medicinal effect of cannabidiol drugs. b). Borosilicate and polyethylene containers should be avoided in formulation, packing and administration of cannabidiol-containing medicines to guarantee correct doses. c). Cannabidiol medications can be co-administered with tizoxanide without caution.
Collapse
Affiliation(s)
- Mariana Babayeva
- Department of Biomedical and Pharmaceutical Sciences, Touro College of Pharmacy, 3 Times Square, New York, NY, 10036, USA.
| | - Iva Srdanovic
- Department of Biomedical and Pharmaceutical Sciences, Touro College of Pharmacy, 3 Times Square, New York, NY, 10036, USA
| |
Collapse
|
16
|
Pineda-Alemán R, Cabarcas-Herrera C, Alviz-Amador A, Galindo-Murillo R, Pérez-Gonzalez H, Rodríguez-Cavallo E, Méndez-Cuadro D. Molecular dynamics of structural effects of reactive carbonyl species derivate of lipid peroxidation on bovine serum albumin. Biochim Biophys Acta Gen Subj 2024; 1868:130613. [PMID: 38593934 DOI: 10.1016/j.bbagen.2024.130613] [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: 07/03/2023] [Revised: 03/09/2024] [Accepted: 04/04/2024] [Indexed: 04/11/2024]
Abstract
BACKGROUND Serum albumin is the most abundant protein in the Mammalia blood plasma at where plays a decisive role in the transport wide variety of hydrophobic ligands. BSA undergoes oxidative modifications like the carbonylation by the reactive carbonyl species (RCSs) 4-hydroxy-2-nonenal (HNE), 4 hydroxy-2-hexenal (HHE), malondialdehyde (MDA) and 4-oxo-2-nonenal (ONE), among others. The structural and functional changes induced by protein carbonylation have been associated with the advancement of neurodegenerative, cardiovascular, metabolic and cancer diseases. METHODS To elucidate structural effects of protein carbonylation with RCSs on BSA, parameters for six new non-standard amino acids were designated and molecular dynamics simulations of its mono‑carbonylated-BSA systems were conducted in the AMBER force field. Trajectories were evaluated by RMSD, RMSF, PCA, RoG and SASA analysis. RESULTS An increase in the conformational instability for all proteins modified with local changes were observed, without significant changes on the BSA global three-dimensional folding. A more relaxed compaction level and major solvent accessible surface area for modified systems was found. Four regions of high molecular fluctuation were identified in all modified systems, being the subdomains IA and IIIB those with the most remarkable local conformational changes. Regarding essential modes of domain movements, it was evidenced that the most representatives were those related to IA subdomain, while IIIB subdomain presented discrete changes. CONCLUSIONS RCSs induces local structural changes on mono‑carbonylated BSA. Also, this study extends our knowledge on how carbonylation by RCSs induce structural effects on proteins.
Collapse
Affiliation(s)
- Rafael Pineda-Alemán
- Analytical Chemistry and Biomedicine Group, Medicine Faculty, University of Cartagena, Cartagena, Colombia
| | - Camila Cabarcas-Herrera
- Analytical Chemistry and Biomedicine Group, Exact and Natural Sciences Faculty, University of Cartagena, Cartagena, Colombia
| | - Antistio Alviz-Amador
- Analytical Chemistry and Biomedicine Group, Pharmaceutical Sciences Faculty, University of Cartagena, Cartagena, Colombia
| | | | - Humberto Pérez-Gonzalez
- Department of Mathematics, Exact and Natural Sciences Faculty, University of Cartagena, Cartagena, Colombia
| | - Erika Rodríguez-Cavallo
- Analytical Chemistry and Biomedicine Group, Pharmaceutical Sciences Faculty, University of Cartagena, Cartagena, Colombia
| | - Darío Méndez-Cuadro
- Analytical Chemistry and Biomedicine Group, Exact and Natural Sciences Faculty, University of Cartagena, Cartagena, Colombia.
| |
Collapse
|
17
|
Song Y, Bienvenu LA, Bongcaron V, Prijaya SA, Maluenda AC, Walsh APG, McFayden JD, Pietersz GA, Peter K, Wang X. Platelet-targeted thromboprophylaxis with a human serum albumin fusion drug: Preventing thrombosis and reducing cardiac ischemia/reperfusion injurywithout bleeding complications. Theranostics 2024; 14:3267-3281. [PMID: 38855181 PMCID: PMC11155409 DOI: 10.7150/thno.97517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 05/11/2024] [Indexed: 06/11/2024] Open
Abstract
Background: Myocardial infarction (MI) as a consequence of atherosclerosis-associated acute thrombosis is a leading cause of death and disability globally. Antiplatelet and anticoagulant drugs are standard therapies in preventing and treating MI. However, all clinically used drugs are associated with bleeding complications, which ultimately limits their use in patients with a high risk of bleeding. We have developed a new recombinant drug, targ-HSA-TAP, that combines targeting and specific inhibition of activated platelets as well as anticoagulation. This drug is designed and tested for a prolonged circulating half-life, enabling unique thromboprophylaxis without bleeding complications. Methods: Targ-HSA-TAP combines a single-chain antibody (scFv) that targets activated glycoprotein IIb/IIIa on activated platelets, human serum albumin (HSA) for prolonged circulation, and tick anticoagulant peptide (TAP) for coagulation FX inhibition. A non-binding scFv is employed as a non-targeting control (non-targ-HSA-TAP). Its efficacy was investigated in vivo using murine models of acute thrombosis and cardiac ischemia-reperfusion (I/R) injury. Results: Our experiments confirmed the targeting specificity of targ-HSA-TAP to activated platelets and demonstrated effective prevention of platelet aggregation and thrombus formation, as well as FXa inhibition in vitro. Thromboprophylactic administration of targ-HSA-TAP subcutaneously in mice prevented occlusion of the carotid artery after ferric chloride injury as compared to non-targ-HSA-TAP and PBS-control treated mice. By comparing the therapeutic outcomes between targ-TAP and targ-HSA-TAP, we demonstrate the significant improvements brought by the HSA fusion in extending the drug's half-life and enhancing its therapeutic window for up to 16 h post-administration. Importantly, tail bleeding time was not prolonged with targ-HSA-TAP in contrast to the clinically used anticoagulant enoxaparin. Furthermore, in a murine model of cardiac I/R injury, mice administered targ-HSA-TAP 10 h before injury demonstrated preserved cardiac function, with significantly higher ejection fraction and fractional shortening, as compared to the non-targ-HSA-TAP and PBS control groups. Advanced strain analysis revealed reduced myocardial deformation and histology confirmed a reduced infarct size in targ-HSA-TAP treated mice compared to control groups. Conclusion: The inclusion of HSA represents a significant advancement in the design of targeted therapeutic agents for thromboprophylaxis. Our activated platelet-targeted targ-HSA-TAP is a highly effective antithrombotic drug with both anticoagulant and antiplatelet effects while retaining normal hemostasis. The long half-life of targ-HSA-TAP provides the unique opportunity to use this antithrombotic drug for more effective, long-lasting and safer anti-thrombotic prophylaxis. In cases where MI occurs, this prophylactic strategy reduces thrombus burden and effectively reduces cardiac I/R injury.
Collapse
Affiliation(s)
- Yuyang Song
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Cardiometabolic Health, University of Melbourne, VIC, Australia
| | - Laura A. Bienvenu
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Cardiometabolic Health, University of Melbourne, VIC, Australia
- Baker Department of Cardiovascular Research, Translational and Implementation, La Trobe University, Melbourne, Australia
| | - Viktoria Bongcaron
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Cardiometabolic Health, University of Melbourne, VIC, Australia
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Shania A. Prijaya
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Cardiometabolic Health, University of Melbourne, VIC, Australia
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Ana C. Maluenda
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Aidan P. G. Walsh
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - James D. McFayden
- Department of Cardiometabolic Health, University of Melbourne, VIC, Australia
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Geoffrey A. Pietersz
- Department of Cardiometabolic Health, University of Melbourne, VIC, Australia
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Karlheinz Peter
- Department of Cardiometabolic Health, University of Melbourne, VIC, Australia
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Medicine, Monash University, Melbourne, VIC, Australia
| | - Xiaowei Wang
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Cardiometabolic Health, University of Melbourne, VIC, Australia
- Baker Department of Cardiovascular Research, Translational and Implementation, La Trobe University, Melbourne, Australia
- Department of Medicine, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
18
|
Cavalieri G, Marson D, Giurgevich N, Valeri R, Felluga F, Laurini E, Pricl S. Molecular Ballet: Investigating the Complex Interaction between Self-Assembling Dendrimers and Human Serum Albumin via Computational and Experimental Methods. Pharmaceutics 2024; 16:533. [PMID: 38675194 PMCID: PMC11054399 DOI: 10.3390/pharmaceutics16040533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Dendrimers, intricate macromolecules with highly branched nanostructures, offer unique attributes including precise control over size, shape, and functionality, making them promising candidates for a wide range of biomedical applications. The exploration of their interaction with biological environments, particularly human serum albumin (HSA), holds significant importance for biomedical utilization. In this study, the interaction between HSA and a recently developed self-assembling amphiphilic dendrimer (AD) was investigated using various experimental techniques. Fluorescence spectroscopy and isothermal titration calorimetry revealed moderate interactions between the protein and the AD nanomicelles (NMs), primarily attributed to favorable enthalpic contributions arising from electrostatic interactions and hydrogen bonding. Structural analysis indicated minimal changes in HSA upon complexation with the AD NMs, which was further supported by computational simulations demonstrating stable interactions at the atomistic level. These findings provide valuable insights into the binding mechanisms and thermodynamic parameters governing HSA/AD NM interactions, thereby contributing to the understanding of their potential biomedical applications.
Collapse
Affiliation(s)
- Gabriele Cavalieri
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTS), DEA, University of Trieste, Piazzale Europa 1, 34127 Trieste, Italy; (G.C.); (D.M.); (N.G.); (R.V.); (S.P.)
| | - Domenico Marson
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTS), DEA, University of Trieste, Piazzale Europa 1, 34127 Trieste, Italy; (G.C.); (D.M.); (N.G.); (R.V.); (S.P.)
| | - Nicoletta Giurgevich
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTS), DEA, University of Trieste, Piazzale Europa 1, 34127 Trieste, Italy; (G.C.); (D.M.); (N.G.); (R.V.); (S.P.)
| | - Rachele Valeri
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTS), DEA, University of Trieste, Piazzale Europa 1, 34127 Trieste, Italy; (G.C.); (D.M.); (N.G.); (R.V.); (S.P.)
| | - Fulvia Felluga
- Department of Chemical and Pharmaceutical Sciences, DSCF, University of Trieste, Via Giorgieri 1, 34127 Trieste, Italy;
| | - Erik Laurini
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTS), DEA, University of Trieste, Piazzale Europa 1, 34127 Trieste, Italy; (G.C.); (D.M.); (N.G.); (R.V.); (S.P.)
| | - Sabrina Pricl
- Molecular Biology and Nanotechnology Laboratory (MolBNL@UniTS), DEA, University of Trieste, Piazzale Europa 1, 34127 Trieste, Italy; (G.C.); (D.M.); (N.G.); (R.V.); (S.P.)
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, ul. Pomorska 141/143, 90-236 Łódź, Poland
| |
Collapse
|
19
|
Mukai R, Okuyama H, Uchimura M, Sakao K, Matsuhiro M, Ikeda-Imafuku M, Ishima Y, Nishikawa M, Ikushiro S, Tai A. The binding selectivity of quercetin and its structure-related polyphenols to human serum albumin using a fluorescent dye cocktail for multiplex drug-site mapping. Bioorg Chem 2024; 145:107184. [PMID: 38364549 DOI: 10.1016/j.bioorg.2024.107184] [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: 12/18/2023] [Revised: 01/25/2024] [Accepted: 02/04/2024] [Indexed: 02/18/2024]
Abstract
Human serum albumin (HSA) is a serum protein that carries flavonoids in blood circulation. In this report, the binding selectivity and strength of interactions to HSA-binding sites (sites I or II) by flavonoids were evaluated using competition experiments and the specific fluorescent dyes, dansylamide and BD140. Most tested flavonoids bound site I preferentially, with the binding strength dependent on the mother structure in the order flavonol > flavone > flavanone > flavan 3-ols. Glycosylation or glucuronidation reduced the binding of quercetin to site I of HSA, whereas sulfation increased binding. Quercetin 7-sulfate showed the strongest binding and molecular docking simulations supported this observation. Prenylation at any position or glucuronidation and sulfation at the C-4' or C-7 position of quercetin facilitated stronger binding to site II. The binding affinity of flavonoids toward site I correlated with the partition coefficient value (logP), whereas no corresponding correlation was observed for site II.
Collapse
Affiliation(s)
- Rie Mukai
- Department of Food Science, Graduate School of Technology, Industrial and Social Sciences, Tokushima University, 2-1, Minamijosan-jima, Tokushima 770-8513, Japan.
| | - Hitomi Okuyama
- Department of Food Science, Graduate School of Technology, Industrial and Social Sciences, Tokushima University, 2-1, Minamijosan-jima, Tokushima 770-8513, Japan
| | - Miku Uchimura
- Department of Food Science and Technology, Graduate School of Agriculture, Kagoshima University, 1-21-24, Korimoto, Kagoshima 890-0065, Japan.
| | - Kozue Sakao
- Department of Food Science and Technology, Graduate School of Agriculture, Kagoshima University, 1-21-24, Korimoto, Kagoshima 890-0065, Japan.
| | - Miyu Matsuhiro
- Department of Food Science, Graduate School of Technology, Industrial and Social Sciences, Tokushima University, 2-1, Minamijosan-jima, Tokushima 770-8513, Japan.
| | - Mayumi Ikeda-Imafuku
- Department of Physical Pharmaceutics, School of Pharmaceutical Sciences, Wakayama Medical University, 25-1 Shichiban-cho, Wakayama 640-8156, Japan.
| | - Yu Ishima
- Laboratory of Biopharmaceutics, Kyoto Pharmaceutical University, 5 Nakauchi-cho, Misasagi, Yamashina-ku, Kyoto 607-8414, Japan.
| | - Miyu Nishikawa
- Department of Biotechnology, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan.
| | - Shinichi Ikushiro
- Department of Biotechnology, Faculty of Engineering, Toyama Prefectural University, 5180 Kurokawa, Imizu, Toyama 939-0398, Japan.
| | - Akihiro Tai
- Department of Food Science, Graduate School of Technology, Industrial and Social Sciences, Tokushima University, 2-1, Minamijosan-jima, Tokushima 770-8513, Japan.
| |
Collapse
|
20
|
Liu Y, Yuan Z, Zhao P, Li C, Qin L, Zhao T, Zhu X, Feng S. Studies on the binding of wedelolactone to human serum albumin with multi-spectroscopic analysis, molecular docking and molecular dynamic simulation. Biophys Chem 2024; 307:107198. [PMID: 38359582 DOI: 10.1016/j.bpc.2024.107198] [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: 12/14/2023] [Accepted: 02/02/2024] [Indexed: 02/17/2024]
Abstract
Wedelolactone (WEL) is a small molecule compound isolated from Eclipta prostrate L., which has been reported to possess various biological activities such as anti-hepatotoxicity, anti-hypertension, anti-tumour, anti-phospholipase A2 and detoxification activity against snake venom. In the present study, we investigated the interaction of WEL with human serum albumin (HSA) using simultaneous fluorescence, UV-visible spectroscopy, 3D fluorescence spectroscopy, Fourier transform infrared spectroscopy (FTIR), molecular docking technique and molecular dynamics simulation. We found that the interaction between HSA and WEL can exhibit a static fluorescence burst mechanism, and the binding process is essentially spontaneous, with the main forces manifested as hydrogen bonding, van der Waals force and electrostatic interactions. Competitive binding and molecular docking studies showed that WEL preferentially bound to HSA in substructural region IIA (site I); molecular dynamics simulations showed that HSA interacted with WEL to form a stable complex, which also induced conformational changes in HSA. The study of the interaction between WEL and HSA can provide a reference for a more in-depth study of the pharmacodynamic mechanism of WEL and its further development and utilisation.
Collapse
Affiliation(s)
- Yali Liu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Zhen Yuan
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Pan Zhao
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Changxin Li
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Lu Qin
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Tianlun Zhao
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Xiaojing Zhu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Shuai Feng
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| |
Collapse
|
21
|
Sciancalepore M, Ragnini A, Zacchi P, Borelli V, D’Andrea P, Lorenzon P, Bernareggi A. A Pharmacological Investigation of the TMEM16A Currents in Murine Skeletal Myogenic Precursor Cells. Int J Mol Sci 2024; 25:2225. [PMID: 38396901 PMCID: PMC10889721 DOI: 10.3390/ijms25042225] [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: 01/17/2024] [Revised: 02/08/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
TMEM16A is a Ca2+-activated Cl- channel expressed in various species and tissues. In mammalian skeletal muscle precursors, the activity of these channels is still poorly investigated. Here, we characterized TMEM16A channels and investigated if the pharmacological activation of Piezo1 channels could modulate the TMEM16A currents in mouse myogenic precursors. Whole-cell patch-clamp recordings combined with the pharmacological agents Ani9, T16inh-A01 and Yoda1 were used to characterize TMEM16A-mediated currents and the possible modulatory effect of Piezo1 activity on TMEM16A channels. Western blot analysis was also carried out to confirm the expression of TMEM16A and Piezo1 channel proteins. We found that TMEM16A channels were functionally expressed in fusion-competent mouse myogenic precursors. The pharmacological blockage of TMEM16A inhibited myocyte fusion into myotubes. Moreover, the specific Piezo1 agonist Yoda1 positively regulated TMEM16A currents. The findings demonstrate, for the first time, a sarcolemmal TMEM16A channel activity and its involvement at the early stage of mammalian skeletal muscle differentiation. In addition, the results suggest a possible role of mechanosensitive Piezo1 channels in the modulation of TMEM16A currents.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Annalisa Bernareggi
- Department of Life Sciences, University of Trieste, I-34127 Trieste, Italy; (M.S.); (A.R.); (P.Z.); (V.B.); (P.D.); (P.L.)
| |
Collapse
|
22
|
Li J, Sun Y, Li G, Cheng C, Sui X, Wu Q. The Extraction, Determination, and Bioactivity of Curcumenol: A Comprehensive Review. Molecules 2024; 29:656. [PMID: 38338400 PMCID: PMC10856406 DOI: 10.3390/molecules29030656] [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: 12/27/2023] [Revised: 01/18/2024] [Accepted: 01/26/2024] [Indexed: 02/12/2024] Open
Abstract
Curcuma wenyujin is a member of the Curcuma zedoaria (zedoary, Zingiberaceae) family, which has a long history in traditional Chinese medicine (TCM) due to its abundant biologically active constituents. Curcumenol, a component of Curcuma wenyujin, has several biological activities. At present, despite different pharmacological activities being reported, the clinical usage of curcumenol remains under investigation. To further determine the characteristics of curcumenol, the extraction, determination, and bioactivity of the compound are summarized in this review. Existing research has reported that curcumenol exerts different pharmacological effects in regard to a variety of diseases, including anti-inflammatory, anti-oxidant, anti-bactericidal, anti-diabetic, and anti-cancer activity, and also ameliorates osteoporosis. This review of curcumenol provides a theoretical basis for further research and clinical applications.
Collapse
Affiliation(s)
- Jie Li
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China; (J.L.)
- College of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
- School of Environmental and Chemical Engineering, Zhaoqing University, Zhaoqing 526061, China
| | - Yitian Sun
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China; (J.L.)
- College of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Guohua Li
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China; (J.L.)
- College of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Chunsong Cheng
- Lushan Botanical Garden, Chinese Academy of Sciences, Jiujiang 332900, China
| | - Xinbing Sui
- College of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Qibiao Wu
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau 999078, China; (J.L.)
- Zhuhai M.U.S.T. Science and Technology Research Institute, Zhuhai 519031, China
- Guangdong-Hong Kong-Macao Joint Laboratory for Contaminants Exposure and Health, Guangzhou 510006, China
| |
Collapse
|
23
|
Majumdar S, Rastogi H, Chowdhury PK. Bridging Soft Interaction and Excluded Volume in Crowded Milieu through Subtle Protein Dynamics. J Phys Chem B 2024; 128:716-730. [PMID: 38226816 DOI: 10.1021/acs.jpcb.3c07266] [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: 01/17/2024]
Abstract
The impact of macromolecular crowding on biological macromolecules has been elucidated through the excluded volume phenomenon and soft interactions. However, it has often been difficult to provide a clear demarcation between the two regions. Here, using temperature-dependent dynamics (local and global) of the multidomain protein human serum albumin (HSA) in the presence of commonly used synthetic crowders (Dextran 40, PEG 8, Ficoll 70, and Dextran 70), we have shown the presence of a transition that serves as a bridge between the soft and hard regimes. The bridging region is independent of the crowder identity and displays no apparent correlation with the critical overlap concentration of the polymeric crowding agents. Moreover, the dynamics of domains I and II and the protein gating motion respond differently, thereby bringing to the fore the asymmetry underlying the crowder influence on HSA. In addition, solvent-coupled and decoupled protein motions indicate the heterogeneity of the dynamic landscape in the crowded milieu. We also propose an intriguing correlation between protein stability and dynamics, with increased global stability being accompanied by eased local domain motion.
Collapse
Affiliation(s)
- Shubhangi Majumdar
- Department of Chemistry, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Harshita Rastogi
- Department of Chemistry, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Pramit K Chowdhury
- Department of Chemistry, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| |
Collapse
|
24
|
Al-Asbahy WM, Shamsi M, Senan A, Al-Areqi N. Binding mechanism, photo-induced cleavage and computational studies of interaction cefepime drug with Human serum albumin. J Biomol Struct Dyn 2024:1-11. [PMID: 38234057 DOI: 10.1080/07391102.2024.2304668] [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: 05/23/2023] [Accepted: 01/07/2024] [Indexed: 01/19/2024]
Abstract
The binding interaction of cefepime to human serum albumin (HSA) in aqueous solution was investigated by molecular spectroscopy (UV spectra, fluorescence spectra and CD spectra), photo-cleavage and modeling studies under simulative physiological conditions. Spectrophotometric results are rationalized in terms of a static quenching process and binding constant (Kb) and the number of binding sites (n ≈ 1) were calculated using fluorescence quenching approaches at three temperature settings. Thermodynamic data of ΔG, ΔH and ΔS at different temperatures were evaluated. The results showed that the electrostatic and hydrogen bonding interactions play a major role in the binding of cefepime to HSA. The value of 3.4 nm for the distance r between the donor (HSA) and acceptor (cefepime) was derived from the fluorescence resonance energy transfer (FRET). FTIR and CD measurements has been reaffirmed HSA-cefepime association and demonstrated reduction in α-helical content of HSA. Furthermore, the study of molecular modeling also indicated that cefepime could strongly bind to the site I (subdomain IIA) of HSA. Additionally, cefepime shows efficient photo- cleavage of HSA cleavage. Our results may provide valuable information to understand the pharmacological profile of cefepime drug delivery in blood stream.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Waddhaah M Al-Asbahy
- Department of Chemistry, Faculty of Applied Science, Taiz University, Taiz, Yemen
| | - Manal Shamsi
- Department of Biochemistry, Faculty of Medicine and Medical Sciences, Taiz University, Taiz, Yemen
| | - Ahmed Senan
- Department of Chemistry, Faculty of Applied Science, Taiz University, Taiz, Yemen
| | - Niyazi Al-Areqi
- Department of Chemistry, Faculty of Applied Science, Taiz University, Taiz, Yemen
| |
Collapse
|
25
|
Chakraborty P, Bhattacharyya C, Sahu R, Dua TK, Kandimalla R, Dewanjee S. Polymeric nanotherapeutics: An emerging therapeutic approach for the management of neurodegenerative disorders. J Drug Deliv Sci Technol 2024; 91:105267. [DOI: 10.1016/j.jddst.2023.105267] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
26
|
Lee C. Albumin hydrogels for repeated capture of drugs from the bloodstream and release into the tumor. J Control Release 2024; 365:384-397. [PMID: 38007193 DOI: 10.1016/j.jconrel.2023.11.027] [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: 08/03/2023] [Revised: 11/12/2023] [Accepted: 11/14/2023] [Indexed: 11/27/2023]
Abstract
Despite the efficacy of hydrogels for consistently delivering drugs to targeted areas (primarily tumors), these systems face challenges such as initial burst release, non-refillable drugs, and a lack of dosage control. To address these issues, a novel strategy has been developed to capture and release drugs from the bloodstream, thereby overcoming the limitations of traditional hydrogels. In this study, an innovative albumin hydrogel system was developed through a bioorthogonal reaction using azide-modified albumin and 4-arm PEG-DBCO. This system can repeatedly capture and release drugs over prolonged periods. Inspired by albumin-drug binding in vivo, this hydrogel can be injected intratumorally and acts as a reservoir for capturing drugs circulating in the bloodstream. Drugs captured in hydrogels are released slowly and effectively delivered to tumors through a "capture and release process." Both the in vitro and in vivo results indicated that the hydrogel effectively captured and released drugs, such as indocyanine green and doxorubicin, over repeated cycles without compromising the activity of the drugs. Moreover, implanting the hydrogel at surgical sites successfully inhibited tumor recurrence through its drug capture-release capability. These findings establish the albumin hydrogel system as a promising capture-release platform that leverages drug-binding affinity to effectively deliver drugs to tumors, offering potential advancements in cancer treatment and post-surgery recurrence prevention.
Collapse
Affiliation(s)
- Changkyu Lee
- Department of Biopharmaceutical Engineering, Division of Chemistry and Biotechnology, Dongguk University, Gyeongju 38066, Republic of Korea.
| |
Collapse
|
27
|
Hosseini L, Shahabi P, Fakhari A, Zangbar HS, Seyedaghamiri F, Sadeghzadeh J, Abolhasanpour N. Aging and age-related diseases with a focus on therapeutic potentials of young blood/plasma. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:1-13. [PMID: 37552316 DOI: 10.1007/s00210-023-02657-5] [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: 07/06/2023] [Accepted: 07/31/2023] [Indexed: 08/09/2023]
Abstract
Aging is accompanied by alterations in the body with time-related to decline of physiological integrity and functionality process, responsible for increasing diseases and vulnerability to death. Several ages associated with biomarkers were observed in red blood cells, and consequently plasma proteins have a critical rejuvenating role in the aging process and age-related disorders. Advanced age is a risk factor for a broad spectrum of diseases and disorders such as cardiovascular diseases, musculoskeletal disorders and liver, chronic kidney disease, neurodegenerative diseases, and cancer because of loss of regenerative capacity, correlated to reduced systemic factors and raise of pro-inflammatory cytokines. Most studies have shown that systemic factors in young blood/plasma can strongly protect against age-related diseases in various tissues by restoring autophagy, increasing neurogenesis, and reducing oxidative stress, inflammation, and apoptosis. Here, we focus on the current advances in using young plasma or blood to combat aging and age-related diseases and summarize the experimental and clinical evidence supporting this approach. Based on reports, young plasma or blood is new a therapeutic approach to aging and age-associated diseases.
Collapse
Affiliation(s)
- Leila Hosseini
- Research Center of Psychiatry and Behavioral Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Parviz Shahabi
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, IR, Iran
| | - Ali Fakhari
- Research Center of Psychiatry and Behavioral Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamid Soltani Zangbar
- Department of Neurosciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemehsadat Seyedaghamiri
- Department of Neurosciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jafar Sadeghzadeh
- Department of Neurosciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nasrin Abolhasanpour
- Research Center for Evidence-Based Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
28
|
Yang YD, Lu N, Tian R. Serum albumin acted as an effective carrier to improve the stability of bioactive flavonoid. Amino Acids 2023; 55:1879-1890. [PMID: 37856004 DOI: 10.1007/s00726-023-03347-5] [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: 09/22/2022] [Accepted: 10/03/2023] [Indexed: 10/20/2023]
Abstract
The health-improving functions of bioactive flavonoids in vitro and in vivo are often limited by their low stability, which could be counteracted by the application of proteins as carriers of flavonoids. Clarification of the mechanism of protein-ligand interaction is crucial for the encapsulation of bioactive components. Herein, common plasma proteins [i.e., bovine serum albumin (BSA), human serum albumin (HSA), human immunoglobulin G (IgG) and fibrinogen (FG)] were compared for their binding characteristics to quercetin, the main component of flavonoids in human diet, in the absence and presence of free Cu2+ (an accelerator for flavonoids' instability) using multi-spectroscopic and computational methods. As a flexible open structure of proteins, both BSA and HSA were found to be the most promising carriers for quercetin and Cu2+ with an affinity on the order of 104 M-1. HSA-diligand complex (i.e., HSA-quercetin-Cu2+) was successfully generated when both quercetin and Cu2+ were added to the HSA solution. The stability and free radical scavenging activity of bioactive quercetin during incubation was promoted in the HSA-diligand complex relative to quercetin-Cu2+ complex. Quercetin/Cu2+ system could induce the formation of reactive oxygen species such as hydrogen peroxide (H2O2) and hydroxide radical (·OH), which were significantly suppressed upon HSA binding. Consistently, the cytotoxicity of the quercetin/Cu2+ system to endothelial cells was reduced in the HSA-diligand complex. These results demonstrate the possibility of developing serum albumin-based carriers for the protection of bioactive flavonoids in their nutritional application.
Collapse
Affiliation(s)
- Ya-Di Yang
- Jiangxi Key Laboratory of Green Chemistry, College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, China
| | - Naihao Lu
- Jiangxi Key Laboratory of Green Chemistry, College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, China
| | - Rong Tian
- Jiangxi Key Laboratory of Green Chemistry, College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, China.
| |
Collapse
|
29
|
Rajendran D, Chandrasekaran N. Unveiling the Modification of Esterase-like Activity of Serum Albumin by Nanoplastics and Their Cocontaminants. ACS OMEGA 2023; 8:43719-43731. [PMID: 38027364 PMCID: PMC10666218 DOI: 10.1021/acsomega.3c05447] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 09/29/2023] [Indexed: 12/01/2023]
Abstract
Nanoplastics and other cocontaminants have raised concerns due to their widespread presence in the environment and their potential to enter the food chain. The harmful effects of these particles depend on various factors, such as nanoparticle size, shape, surface charge, and the nature of the cocontaminants involved. On entering the human body, human serum albumin (HSA) molecules bind and transport these particles in the blood system. The esterase-like activity of HSA, which plays a role in metabolizing drug/toxic compounds, was taken as a representative to portray the effects of these particles on HSA. Polystyrene nanoplastics (PSNPs) with different surface functionalization (plain (PS), amine (PS-NH2), and carboxy (PS-COOH)), different sizes (100 and 500 nm), and PS with cocontaminant metformin hydrochloride (Met-HCl), a widely used antidiabetic drug, were investigated in this study. Fluorescence emission spectra of HSA revealed that PS-NH2 exhibits a greater effect on protein conformation, smaller NPs have a greater influence on protein structure than larger NPs, and Met-HCl lowers PSNPs' affinity for HSA by coating the surface of the NPs, which may result in direct NP distribution to the drug's target organs and toxicity. Circular dichroism spectra also supported these results in terms of secondary structural changes. Esterase activity of HSA was inhibited by all the particles (except Met-HCl) by competitive inhibition as concluded from constant Vmax and increasing Km. Greater reduction in enzyme activity was observed for PS-NH2 among functionalizations and for 100 nm PS among sizes. Furthermore, Met-HCl lowers the inhibitory impact of PSNPs on HSA since the drug binds weakly to HSA, and so they can serve as a vector delivering PSNPs to their target organs, resulting in serious implications.
Collapse
Affiliation(s)
- Durgalakshmi Rajendran
- Centre for Nanobiotechnology, Vellore Institute of Technology (VIT University), Vellore 632014, Tamil Nadu, India
| | - Natarajan Chandrasekaran
- Centre for Nanobiotechnology, Vellore Institute of Technology (VIT University), Vellore 632014, Tamil Nadu, India
| |
Collapse
|
30
|
Garg U, Jain N, Kaul S, Nagaich U. Role of Albumin as a Targeted Drug Carrier in the Management of Rheumatoid Arthritis: A Comprehensive Review. Mol Pharm 2023; 20:5345-5358. [PMID: 37870420 DOI: 10.1021/acs.molpharmaceut.3c00581] [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: 10/24/2023]
Abstract
An endogenous transporter protein called albumin interacts with the Fc receptor to provide it with multiple substrate-binding domains, cell membrane receptor activation, and an extended circulating half-life. Albumin has the remarkable ability to bind with receptors viz. secreted protein acidic and rich in cysteine (SPARC) and scavenger protein-A (SR-A) that are overexpressed during rheumatoid arthritis (RA), enabling active targeting of the disease site instead of requiring specialized substrates to be added to the nanocarrier. RA, a chronic autoimmune illness, is characterized by the presence of a severe inflammatory response. RA patients have low serum albumin concentration, which signifies the high uptake of albumin at the inflammatory sites, giving a rationale to use albumin as a drug carrier for RA therapy. Albumin has the capacity for both passive and active targeting. It is an abundantly available protein in the bloodstream showing excellent cellular compatibility, degradability in biological tissues, nonantigenicity, and safety. There are three strategies of albumin mediated drug delivery as encapsulating therapeutics in albumin nanoparticles, chemically conjugating drugs with functional proteins, and albumin itself which is used as a targeting ligand to deliver drugs specifically to cells or tissues that express albumin-binding receptors. In the current review, an attempt has been made to highlight the significant evidence of albumin as a drug delivery carrier for the safe and effective management of RA. Evidence has been provided in the form of recent research advances, clinical trials, and patents. Additionally, this review will outline the prospective for the potential utilization of albumin as a drug vehicle for RA and suggest possible future avenues to provide the perspective for subsequent studies.
Collapse
Affiliation(s)
- Unnati Garg
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Noida, Uttar Pradesh 201303, India
| | - Neha Jain
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Noida, Uttar Pradesh 201303, India
| | - Shreya Kaul
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Noida, Uttar Pradesh 201303, India
| | - Upendra Nagaich
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Noida, Uttar Pradesh 201303, India
| |
Collapse
|
31
|
Elshami FI, Shereef HA, El-Mehasseb IM, Shaban SY, van Eldik R. Hydroxychloroquine-Loaded Chitosan Nanoparticles Induce Anticancer Activity in A549 Lung Cancer Cells: Design, BSA Binding, Molecular Docking, Mechanistic, and Biological Evaluation. Int J Mol Sci 2023; 24:14103. [PMID: 37762406 PMCID: PMC10531786 DOI: 10.3390/ijms241814103] [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: 07/30/2023] [Revised: 08/29/2023] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
The current study describes the encapsulation of hydroxychloroquine, widely used in traditional medicine due to its diverse pharmacological and medicinal uses, in chitosan nanoparticles (CNPs). This work aims to combine the HCQ drug with CS NPs to generate a novel nanocomposite with improved characteristics and bioavailability. HCQ@CS NPs are roughly shaped like roadways and have a smooth surface with an average size of 159.3 ± 7.1 nm, a PDI of 0.224 ± 0.101, and a zeta potential of +46.6 ± 0.8 mV. To aid in the development of pharmaceutical systems for use in cancer therapy, the binding mechanism and affinity of the interaction between HCQ and HCQ@CS NPs and BSA were examined using stopped-flow and other spectroscopic approaches, supplemented by molecular docking analysis. HCQ and HCQ@CS NPs binding with BSA is driven by a ground-state complex formation that may be accompanied by a non-radiative energy transfer process, and binding constants indicate that HCQ@CS NPs-BSA was more stable than HCQ-BSA. The stopped-flow analysis demonstrated that, in addition to increasing BSA affinity, the nanoformulation HCQ@CS NPS changes the binding process and may open new routes for interaction. Docking experiments verified the development of the HCQ-BSA complex, with HCQ binding to site I on the BSA structure, primarily with the amino acids, Thr 578, Gln 579, Gln 525, Tyr 400, and Asn 404. Furthermore, the nanoformulation HCQ@CS NPS not only increased cytotoxicity against the A549 lung cancer cell line (IC50 = 28.57 ± 1.72 μg/mL) compared to HCQ (102.21 ± 0.67 μg/mL), but also exhibited higher antibacterial activity against both Gram-positive and Gram-negative bacteria when compared to HCQ and chloramphenicol, which is in agreement with the binding constants. The nanoformulation developed in this study may offer a viable therapy option for A549 lung cancer.
Collapse
Affiliation(s)
- Fawzia I. Elshami
- Chemistry Department, Faculty of Science, Kafrelsheikh University, Kafrelsheikh 33516, Egypt; (F.I.E.); (I.M.E.-M.)
| | - Hadeer A. Shereef
- Clinical Pathology Department, University Hospital, Menoufia University, Shebin El-Kom 32512, Egypt;
| | - Ibrahim M. El-Mehasseb
- Chemistry Department, Faculty of Science, Kafrelsheikh University, Kafrelsheikh 33516, Egypt; (F.I.E.); (I.M.E.-M.)
| | - Shaban Y. Shaban
- Chemistry Department, Faculty of Science, Kafrelsheikh University, Kafrelsheikh 33516, Egypt; (F.I.E.); (I.M.E.-M.)
| | - Rudi van Eldik
- Department of Chemistry and Pharmacy, University of Erlangen-Nuremberg, 91058 Erlangen, Germany
- Faculty of Chemistry, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland
| |
Collapse
|
32
|
Bojilov D, Manolov S, Ahmed S, Dagnon S, Ivanov I, Marc G, Oniga S, Oniga O, Nedialkov P, Mollova S. HPLC Analysis and In Vitro and In Silico Evaluation of the Biological Activity of Polyphenolic Components Separated with Solvents of Various Polarities from Helichrysum italicum. Molecules 2023; 28:6198. [PMID: 37687028 PMCID: PMC10488648 DOI: 10.3390/molecules28176198] [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: 07/31/2023] [Revised: 08/15/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023] Open
Abstract
Helichrysum italicum has piqued the interest of many researchers in recent years, mostly for its essential oil, but increasingly for its polyphenolic content as well. In the current study, we examine the polyphenolic composition of H. italicum grown in Bulgaria. The polyphenolic complex was fractionated with solvents of various polarities, including hexane, chloroform, ethyl acetate, and butanol, in order to assess the biological impact of the components. HPLC-PDA and UHPLC-MS/MS were used to examine all fractions. The green coffee fingerprint profile was employed as a "surrogate standard" in the polyphenolic components detection approach. From the UHPLC-MS/MS analysis, we identified 60 components of the polyphenolic complex such as quercetin 3-O-glucuronide, quercetin acetyl-glycoside, isorhamnetin acetyl-glycoside, isorhamnetin caffeoyl-glycoside, quercetin caffeoyl-malonyl-glycoside, isorhamnetin coumaroyl-glycoside, coumaroyl-caffeoylquinic acid, and diCQA-acetyl-derivative were first reported in the composition of H. italicum. The biological activity of the fractions was evaluated in vitro and in silico, which included the fight against oxidative stress (hydrogen peroxide scavenging activity (HPSA), hydroxyl radical scavenging activity (HRSA), metal-chelating activity (MChA)) and nitrosative (nitric oxide scavenging activity) (NOSA)), in vitro anti-inflammatory, and anti-arthritic activity. Results are presented as IC50 ± SD μg/mL. The analysis showed that the EtOAc fraction was characterized by highest HPSA (57.12 ± 1.14 μg/mL), HRSA (92.23 ± 1.10 μg/mL), MChA (5.60 ± 0.17 μg/mL), and NOSA (89.81 ± 2.09 μg/mL), while the hexane and chloroform fractions showed significantly higher in vitro anti-inflammatory activity (30.48 ± 2.33 μg/mL, 62.50 ± 1.69 μg/mL) compared to the standard ibuprofen. All three fractions showed potential anti-arthritic activity (102.93 ± 8.62 μg/mL, 108.92 ± 4.42 μg/mL, 84.19 ± 3.89 μg/mL).
Collapse
Affiliation(s)
- Dimitar Bojilov
- Department of Organic Chemistry, Faculty of Chemistry, University of Plovdiv, 24 “Tsar Assen” Street, 4000 Plovdiv, Bulgaria; (S.A.); (S.D.); (I.I.)
| | - Stanimir Manolov
- Department of Organic Chemistry, Faculty of Chemistry, University of Plovdiv, 24 “Tsar Assen” Street, 4000 Plovdiv, Bulgaria; (S.A.); (S.D.); (I.I.)
| | - Sezan Ahmed
- Department of Organic Chemistry, Faculty of Chemistry, University of Plovdiv, 24 “Tsar Assen” Street, 4000 Plovdiv, Bulgaria; (S.A.); (S.D.); (I.I.)
| | - Soleya Dagnon
- Department of Organic Chemistry, Faculty of Chemistry, University of Plovdiv, 24 “Tsar Assen” Street, 4000 Plovdiv, Bulgaria; (S.A.); (S.D.); (I.I.)
| | - Iliyan Ivanov
- Department of Organic Chemistry, Faculty of Chemistry, University of Plovdiv, 24 “Tsar Assen” Street, 4000 Plovdiv, Bulgaria; (S.A.); (S.D.); (I.I.)
| | - Gabriel Marc
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 41 Victor Babeș Street, RO-400010 Cluj-Napoca, Romania; (G.M.); (O.O.)
| | - Smaranda Oniga
- Department of Therapeutic Chemistry, “Iuliu Hațieganu” University of Medicine and Pharmacy, 12 Ion Creangă Street, RO-400010 Cluj-Napoca, Romania;
| | - Ovidiu Oniga
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, 41 Victor Babeș Street, RO-400010 Cluj-Napoca, Romania; (G.M.); (O.O.)
| | - Paraskev Nedialkov
- Department of Pharmacognosy, Faculty of Pharmacy, Medical University of Sofia, 2 Dunav Street, 1000 Sofia, Bulgaria;
| | - Silviya Mollova
- Institute of Roses, Essential and Medical Plants, Agricultural Academy, 49 Osvobozhdenie Blvd., 6100 Kazanlak, Bulgaria;
| |
Collapse
|
33
|
Wickramasinghe NI, Corbin B, Kanakarathna DY, Pang Y, Abeywickrama CS, Wijesinghe KJ. Bright NIR-Emitting Styryl Pyridinium Dyes with Large Stokes' Shift for Sensing Applications. BIOSENSORS 2023; 13:799. [PMID: 37622885 PMCID: PMC10452306 DOI: 10.3390/bios13080799] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 08/02/2023] [Accepted: 08/07/2023] [Indexed: 08/26/2023]
Abstract
Two NIR-emitting donor-π-acceptor (D-π-A) type regioisomeric styryl pyridinium dyes (1a-1b) were synthesized and studied for their photophysical performance and environment sensitivity. The two regioisomers, 1a and 1b, exhibited interesting photophysical properties including, longer wavelength excitation (λex ≈ 530-560 nm), bright near-infrared emission (λem ≈ 690-720 nm), high-fluorescence quantum yields (ϕfl ≈ 0.24-0.72) large Stokes' shift (∆λ ≈ 150-240 nm) and high-environmental sensitivity. Probe's photophysical properties were studied in different environmental conditions such as polarity, viscosity, temperature, and concentration. Probes (1a-1b) exhibited noticeable changes in absorbance, emission and Stokes' shift while responding to the changes in physical environment. Probe 1b exhibited a significant bathochromic shift in optical spectra (∆λ ≈ 20-40 nm) compared to its isomer 1a, due to the regio-effect. Probes (1a-1b) exhibited an excellent ability to visualize bacteria (Bacillus megaterium, Escherichia coli), and yeast (Saccharomyces cerevisiae) via fluorescence microscopy.
Collapse
Affiliation(s)
| | - Brian Corbin
- Department of Chemistry, The University of Akron, Akron, OH 44325, USA
| | - Devni Y. Kanakarathna
- Department of Chemistry, Faculty of Science, University of Colombo, Colombo 00300, Sri Lanka
| | - Yi Pang
- Department of Chemistry, The University of Akron, Akron, OH 44325, USA
| | | | - Kaveesha J. Wijesinghe
- Department of Chemistry, Faculty of Science, University of Colombo, Colombo 00300, Sri Lanka
| |
Collapse
|
34
|
Alam M. Exploration of Binding Affinities of a 3β,6β-Diacetoxy-5α-cholestan-5-ol with Human Serum Albumin: Insights from Synthesis, Characterization, Crystal Structure, Antioxidant and Molecular Docking. Molecules 2023; 28:5942. [PMID: 37630192 PMCID: PMC10459092 DOI: 10.3390/molecules28165942] [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: 07/03/2023] [Revised: 08/03/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
The present study describes the synthesis, characterization, and in vitro molecular interactions of a steroid 3β,6β-diacetoxy-5α-cholestan-5-ol. Through conventional and solid-state methods, a cholestane derivative was successfully synthesized, and a variety of analytical techniques were employed to confirm its identity, including high-resolution mass spectrometry (HRMS), Fourier transforms infrared (FT-IR), nuclear magnetic resonance (NMR), elemental analysis, and X-ray single-crystal diffraction. Optimizing the geometry of the steroid was undertaken using density functional theory (DFT), and the results showed great concordance with the data from the experiments. Fluorescence spectral methods and ultraviolet-vis absorption titration were employed to study the in vitro molecular interaction of the steroid regarding human serum albumin (HSA). The Stern-Volmer, modified Stern-Volmer, and thermodynamic parameters' findings showed that steroids had a significant binding affinity to HSA and were further investigated by molecular docking studies to understand the participation of active amino acids in forming non-bonding interactions with steroids. Fluorescence studies have shown that compound 3 interacts with human serum albumin (HSA) through a static quenching mechanism. The binding affinity of compound 3 for HSA was found to be 3.18 × 104 mol-1, and the Gibbs free energy change (ΔG) for the binding reaction was -9.86 kcal mol-1 at 298 K. This indicates that the binding of compound 3 to HSA is thermodynamically favorable. The thermodynamic parameters as well as the binding score obtained from molecular docking at various Sudlow's sites was -8.2, -8.5, and -8.6 kcal/mol for Sites I, II, and III, respectively, supporting the system's spontaneity. Aside from its structural properties, the steroid demonstrated noteworthy antioxidant activity, as evidenced by its IC50 value of 58.5 μM, which is comparable to that of ascorbic acid. The findings presented here contribute to a better understanding of the pharmacodynamics of steroids.
Collapse
Affiliation(s)
- Mahboob Alam
- Department of Safety Engineering, Dongguk University Wise, 123 Dongdae-ro, Gyeongju-si 780714, Gyeongbuk, Republic of Korea
| |
Collapse
|
35
|
Sittiwanichai S, Japrung D, Mori T, Pongprayoon P. Structural and Dynamic Alteration of Glycated Human Serum Albumin in Schiff Base and Amadori Adducts: A Molecular Simulation Study. J Phys Chem B 2023. [PMID: 37267456 DOI: 10.1021/acs.jpcb.3c02048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Human serum albumin (HSA) is a protein carrier in blood transporting metabolites and drugs. Glycated HSA (GHSA) acts as a potential biomarker for diabetes. Thus, many attempts have been made to detect GHSA. Glycation was reported to damage the structure and ligand binding capability, where no molecular detail is available. Recently, the crystal structure of GHSA has been solved, where two glucose isomers (pyranose/GLC and open-chain/GLO) are located at Sudlow's site I. GLO was found to covalently bind to K195, while GLC is trapped by noncontact interactions. GHSA exists in two forms (Schiff base (SCH) and Amadori (AMA) adducts), but how both disrupt albumin activity microscopically remains unknown. To this end, molecular dynamics simulations were performed here to explore the nature of SCH and AMA. Both forms are found to alter the main protein dynamics, resulting in (i) the widening of Sudlow's site I entrance, (ii) the size reduction of nine fatty acid-binding pockets, (iii) the enlargement of Sudlow's site I and the shrinking of Sudlow's site II, (iv) the enhancement of C34 reactivity, and (v) the change in the W214 microenvironment. These unique characteristics found here can be useful for understanding the effect of glycation on the albumin function in more detail and designing specific and selective GHSA detection strategies.
Collapse
Affiliation(s)
- Sirin Sittiwanichai
- Faculty of Science, Department of Chemistry, Kasetsart University, Chatuchak, Bangkok 10900, Thailand
| | - Deanpen Japrung
- National Science and Technology Development Agency, National Nanotechnology Center, Thailand Science Park, Pathum Thani 12120, Thailand
| | - Toshifumi Mori
- Intitute for Materials Chemistry and Engineering, Kyushu University, Kasuga, Fukuoka 816-8580, Japan
- Interdisciplinary Graduate School of Engineering Science, Kyushu University, Kasuga, Fukuoka 816-8580, Japan
| | - Prapasiri Pongprayoon
- Faculty of Science, Department of Chemistry, Kasetsart University, Chatuchak, Bangkok 10900, Thailand
- Center for Advanced Studies in Nanotechnology for Chemical, Food and Agricultural Industries, KU Institute for Advanced Studies, Kasetsart University, Bangkok 10900, Thailand
| |
Collapse
|
36
|
Skuredina AA, Yakupova LR, Kopnova TY, Le-Deygen IM, Belogurova NG, Kudryashova EV. Cyclodextrins and Their Polymers Affect Human Serum Albumin's Interaction with Drugs Used in the Treatment of Pulmonary Infections. Pharmaceutics 2023; 15:1598. [PMID: 37376047 DOI: 10.3390/pharmaceutics15061598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/04/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
Respiratory infectious diseases have challenged medical communities and researchers. Ceftriaxone, meropenem and levofloxacin are widely used for bacterial infection treatment, although they possess severe side effects. To overcome this, we propose cyclodextrin (CD) and CD-based polymers as a drug delivery system for the drugs under consideration. CD polymers demonstrate higher binding affinity for levofloxacin (Ka ≈ 105 M) compared to drug-CD complexes. CDs slightly alter the drugs' affinity for human serum albumin (HSA), whereas CD polymers increase the drugs' binding affinity up to 100 times. The most significant effect was observed for more the hydrophilic drugs ceftriaxone and meropenem. The drug's encapsulation in CD carriers leads to a decrease in the degree of change in the protein's secondary structure. The drug-CD carrier-HSA complexes demonstrate satisfying antibacterial activity in vitro, and even a high binding affinity does not decrease the drug's microbiological properties after 24 h. The proposed carriers are promising for a drug form with a prolonged drug release.
Collapse
Affiliation(s)
- Anna A Skuredina
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Linara R Yakupova
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Tatiana Yu Kopnova
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Irina M Le-Deygen
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Natalya G Belogurova
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Elena V Kudryashova
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia
| |
Collapse
|
37
|
Manivel P, Marimuthu P, Ilanchelian M. Deciphering the binding site and mechanism of new methylene blue with serum albumins: A multispectroscopic and computational investigation. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2023; 300:122900. [PMID: 37244028 DOI: 10.1016/j.saa.2023.122900] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 04/28/2023] [Accepted: 05/17/2023] [Indexed: 05/29/2023]
Abstract
Herein, the interaction mechanism of new methylene blue (NMB) with human serum albumin (HSA) and bovine serum albumin (BSA) was carefully investigated both experimentally and conceptually, employing experimental and insilico analysis. The steady-state emission spectral studies showed that the emission intensity of HSA and BSA was quenched significantly by NMB. The findings of the Stern-Volmer and double logarithmic plot revealed that the observed emission quenching process was through a static quenching mechanism and the measured binding constant values (Kb) for HSA-NMB and BSA-NMB are 2.766 and 1.187 × 105 dm3 mol-1 respectively. The time-resolved fluorescence lifetime measurement and UV-vis absorption investigation further verify the complex formation between NMB and HSA/BSA. The assessment of thermodynamic parameters disclosed the binding process was spontaneous driven by hydrogen bonds (H-bond) and van der Waals interactions, which contributed a significant role in the complexation. Moreover, the secondary structural conformation and microenvironment of HSA/BSA were modified in the presence of NMB, as evidenced by circular dichroism and synchronous fluorescence data. Molecular docking study predicted a plausible binding mode of NMB inside the binding pocket of HSA and BSA. These results demonstrated that the stabilized NMB is found at the Subdomain IIA (site I) of both the proteins and the results were correlated well with the competitive binding assay. Additionally, the principal components analysis revealed less variation of docked poses for HSA, while, more dispersed docked poses were observed for the BSA model. This also highlights the effects of docking towards a modeled protein (BSA). Molecular dynamic (MD) simulation based binding free energy (ΔGmmgbsa) estimation obtained at 298, 303, 308 and 313 K, were in good agreement with our experimental (ΔGbind) values.
Collapse
Affiliation(s)
- Perumal Manivel
- Department of Chemistry, Bharathiar University, Coimbatore, Tamil Nadu, India; Bharathiar Cancer Theranostics Research Centre (RUSA-2.0), Bharathiar University, Coimbatore 641 046, Tamil Nadu, India
| | - Parthiban Marimuthu
- Structural Bioinformatics Laboratory (SBL - Biochemistry) and Pharmaceutical Science Laboratory (PSL - Pharmacy), Faculty of Science and Engineering, Åbo Akademi University, FI-20520 Turku, Finland
| | | |
Collapse
|
38
|
Tanuja Safala Bodapati A, Srinivas Reddy R, Lavanya K, Rao Madku S, Ketan Sahoo B. A comprehensive biophysical and theoretical study on the binding of dexlansoprazole with human serum albumin. J Mol Liq 2023. [DOI: 10.1016/j.molliq.2023.121777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
|
39
|
Lin HJL, Parkinson DH, Holman JC, Thompson WC, Anderson CNK, Hadfield M, Ames S, Zuniga Pina NR, Bowden JN, Quinn C, Hansen LD, Price JC. Modification of the structural stability of human serum albumin in rheumatoid arthritis. PLoS One 2023; 18:e0271008. [PMID: 36930604 PMCID: PMC10022781 DOI: 10.1371/journal.pone.0271008] [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: 06/20/2022] [Accepted: 12/29/2022] [Indexed: 03/18/2023] Open
Abstract
Differential scanning calorimetry (DSC) can indicate changes in structure and/or concentration of the most abundant proteins in a biological sample via heat denaturation curves (HDCs). In blood serum for example, HDC changes result from either concentration changes or altered thermal stabilities for 7-10 proteins and has previously been shown capable of differentiating between sick and healthy human subjects. Here, we compare HDCs and proteomic profiles of 50 patients experiencing joint-inflammatory symptoms, 27 of which were clinically diagnosed with rheumatoid arthritis (RA). The HDC of all 50 subjects appeared significantly different from expected healthy curves, but comparison of additional differences between the RA and the non-RA subjects allowed more specific understanding of RA samples. We used mass spectrometry (MS) to investigate the reasons behind the additional HDC changes observed in RA patients. The HDC differences do not appear to be directly related to differences in the concentrations of abundant serum proteins. Rather, the differences can be attributed to modified thermal stability of some fraction of the human serum albumin (HSA) proteins in the sample. By quantifying differences in the frequency of artificially induced post translational modifications (PTMs), we found that HSA in RA subjects had a much lower surface accessibility, indicating potential ligand or protein binding partners in certain regions that could explain the shift in HSA melting temperature in the RA HDCs. Several low abundance proteins were found to have significant changes in concentration in RA subjects and could be involved in or related to binding of HSA. Certain amino acid sites clusters were found to be less accessible in RA subjects, suggesting changes in HSA structure that may be related to changes in protein-protein interactions. These results all support a change in behavior of HSA which may give insight into mechanisms of RA pathology.
Collapse
Affiliation(s)
- Hsien-Jung L. Lin
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah, United States of America
| | - David H. Parkinson
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah, United States of America
| | - J. Connor Holman
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah, United States of America
| | - W. Chad Thompson
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah, United States of America
| | - Christian N. K. Anderson
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah, United States of America
| | - Marcus Hadfield
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah, United States of America
| | - Stephen Ames
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah, United States of America
| | - Nathan R. Zuniga Pina
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah, United States of America
| | - Jared N. Bowden
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah, United States of America
| | - Colette Quinn
- Applications Lab, TA Instruments, Lindon, Utah, United States of America
| | - Lee D. Hansen
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah, United States of America
| | - John C. Price
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, Utah, United States of America
| |
Collapse
|
40
|
Romani D, Marchetti F, Di Nicola C, Cuccioloni M, Gong C, Eleuteri AM, Galindo A, Fadaei-Tirani F, Nabissi M, Pettinari R. Multitarget-Directed Gallium(III) Tris(acyl-pyrazolonate) Complexes Induce Ferroptosis in Cancer Cells via Dysregulation of Cell Redox Homeostasis and Inhibition of the Mevalonate Pathway. J Med Chem 2023; 66:3212-3225. [PMID: 36802330 PMCID: PMC10009752 DOI: 10.1021/acs.jmedchem.2c01374] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
A series of Ga(Qn)3 coordination compounds have been synthesized, where HQn is 1-phenyl-3-methyl-4-RC(═O)-pyrazolo-5-one. The complexes have been characterized through analytical data, NMR and IR spectroscopy, ESI mass spectrometry, elemental analysis, X-ray crystallography, and density functional theory (DFT) studies. Cytotoxic activity against a panel of human cancer cell lines was determined by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, with interesting results in terms of both cell line selectivity and toxicity values compared with cisplatin. The mechanism of action was explored by spectrophotometric, fluorometric, chromatographic, immunometric, and cytofluorimetric assays, SPR biosensor binding studies, and cell-based experiments. Cell treatment with gallium(III) complexes promoted several cell death triggering signals (accumulation of p27, PCNA, PARP fragments, activation of the caspase cascade, and inhibition of the mevalonate pathway) and induced changes in cell redox homeostasis (decreased levels of GSH/GPX4 and NADP(H), increased reactive oxygen species (ROS) and 4-hydroxynonenal (HNE), mitochondrial damage, and increased activity of CPR and CcO), identifying ferroptosis as the mechanism responsible for cancer cell death.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Agustín Galindo
- Departamento de Química Inorgánica, Facultad de Química, Universidad de Sevilla, Aptdo 1203, 41071 Sevilla, Spain
| | - Farzaneh Fadaei-Tirani
- Institut of Chemical Sciences and Engineering, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne CH-1015, Switzerland
| | | | | |
Collapse
|
41
|
Fernández-Sainz J, Pacheco-Liñán PJ, Ripoll C, González-Fuentes J, Albaladejo J, Bravo I, Garzón-Ruiz A. Unusually High Affinity of the PLK Inhibitors RO3280 and GSK461364 to HSA and Its Possible Pharmacokinetic Implications. Mol Pharm 2023; 20:1631-1642. [PMID: 36812406 PMCID: PMC9997069 DOI: 10.1021/acs.molpharmaceut.2c00849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
The binding processes of two Polo-like kinase inhibitors, RO3280 and GSK461364, to the human serum albumin (HSA) protein as well as the protonation equilibria of both compounds have been studied combining absorbance and fluorescence spectroscopy experiments together with density functional theory calculations. We found that the charge states of RO3280 and GSK461364 are +2 and +1, respectively, at the physiological pH. Nevertheless, RO3280 binds to HSA in the charge state +1 prior to a deprotonation pre-equilibrium. Binding constants to site I of HSA of 2.23 × 106 and 8.80 × 104 M-1 were determined for RO3280 and GSK461364, respectively, at 310 K. The binding processes of RO3280 and GSK461364 to HSA are entropy- and enthalpy-driven, respectively. The positive enthalpy found for the RO3280-HSA complex formation could be related to a proton pre-equilibrium of RO3280.
Collapse
Affiliation(s)
- Jesús Fernández-Sainz
- Departamento de Química Física, Facultad de Farmacia, Universidad de Castilla-La Mancha, Av. Dr. José María Sánchez Ibáñez, s/n, 02071 Albacete, Spain
| | - Pedro J Pacheco-Liñán
- Departamento de Química Física, Facultad de Farmacia, Universidad de Castilla-La Mancha, Av. Dr. José María Sánchez Ibáñez, s/n, 02071 Albacete, Spain
| | - Consuelo Ripoll
- Departamento de Química Física, Facultad de Farmacia, Universidad de Castilla-La Mancha, Av. Dr. José María Sánchez Ibáñez, s/n, 02071 Albacete, Spain
| | - Joaquín González-Fuentes
- Centro Regional de Investigaciones Biomédicas (CRIB), Unidad Asociada de Biomedicina (UCLM-CSIC), C/ Almansa, 14, 02008 Albacete, Spain
| | - José Albaladejo
- Departamento de Química Física, Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, Avenida Camilo José Cela, 10, 13071 Ciudad Real, Spain
| | - Iván Bravo
- Departamento de Química Física, Facultad de Farmacia, Universidad de Castilla-La Mancha, Av. Dr. José María Sánchez Ibáñez, s/n, 02071 Albacete, Spain.,Centro Regional de Investigaciones Biomédicas (CRIB), Unidad Asociada de Biomedicina (UCLM-CSIC), C/ Almansa, 14, 02008 Albacete, Spain
| | - Andrés Garzón-Ruiz
- Departamento de Química Física, Facultad de Farmacia, Universidad de Castilla-La Mancha, Av. Dr. José María Sánchez Ibáñez, s/n, 02071 Albacete, Spain
| |
Collapse
|
42
|
Cooper E, Choi PJ, Hwang K, Nam KM, Kim CY, Shaban T, Schweder P, Mee E, Correia J, Turner C, Faull RLM, Denny WA, Noguchi K, Dragunow M, Jose J, Park TIH. Elucidating the cellular uptake mechanisms of heptamethine cyanine dye analogues for their use as an anticancer drug-carrier molecule for the treatment of glioblastoma. Chem Biol Drug Des 2023; 101:696-716. [PMID: 36323652 DOI: 10.1111/cbdd.14171] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 10/24/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022]
Abstract
The development of chemotherapies for glioblastoma is hindered by their limited bioavailability and toxicity on normal brain function. To overcome these limitations, we investigated the structure-dependent activity of heptamethine cyanine dyes (HMCD), a group of tumour-specific and BBB permeable near-infrared fluorescent dyes, in both commercial (U87MG) and patient-derived GBM cell lines. HMCD analogues with strongly ionisable sulphonic acid groups were not taken up by patient-derived GBM cells, but were taken up by the U87MG cell line. HMCD uptake relies on a combination of transporter uptake through organic anion-transporting polypeptides (OATPs) and endocytosis into GBM cells. The uptake of HMCDs was not affected by p-glycoprotein efflux in GBM cells. Finally, we demonstrate structure-dependent cytotoxic activity at high concentrations (EC50 : 1-100 μM), likely due to mitochondrial damage-induced apoptosis. An in vivo orthotopic glioblastoma model highlights tumour-specific accumulation of our lead HMCD, MHI-148, for up to 7 days following a single intraperitoneal injection. These studies suggest that strongly ionisable groups like sulphonic acids hamper the cellular uptake of HMCDs in patient-derived GBM cell lines, highlighting cell line-specific differences in HMCD uptake. We envisage these findings will help in the design and structural modifications of HMCDs for drug-delivery applications for glioblastoma.
Collapse
Affiliation(s)
- Elizabeth Cooper
- Department of Pharmacology, University of Auckland, Auckland, New Zealand.,Neurosurgical Research Unit, The Centre for Brain Research, University of Auckland, Auckland, New Zealand.,Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand.,The Hugh Green Biobank, The Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Peter J Choi
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Kihwan Hwang
- Department of Neurosurgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Kyung M Nam
- Department of Neurosurgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Chae-Yong Kim
- Department of Neurosurgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Tina Shaban
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Patrick Schweder
- Department of Neurosurgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea.,Department of Neurosurgery, Auckland City Hospital, Auckland, New Zealand
| | - Edward Mee
- Neurosurgical Research Unit, The Centre for Brain Research, University of Auckland, Auckland, New Zealand.,Department of Neurosurgery, Auckland City Hospital, Auckland, New Zealand
| | - Jason Correia
- Neurosurgical Research Unit, The Centre for Brain Research, University of Auckland, Auckland, New Zealand.,Department of Neurosurgery, Auckland City Hospital, Auckland, New Zealand
| | - Clinton Turner
- Neurosurgical Research Unit, The Centre for Brain Research, University of Auckland, Auckland, New Zealand.,Department of Anatomical Pathology, Pathology and Laboratory Medicine, Auckland City Hospital, Auckland, New Zealand
| | - Richard L M Faull
- Neurosurgical Research Unit, The Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - William A Denny
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Katsuya Noguchi
- Dojindo Laboratories Co., Ltd, Techno-Research Park, Kumamoto, Japan
| | - Mike Dragunow
- Department of Pharmacology, University of Auckland, Auckland, New Zealand.,Neurosurgical Research Unit, The Centre for Brain Research, University of Auckland, Auckland, New Zealand.,The Hugh Green Biobank, The Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Jiney Jose
- Auckland Cancer Society Research Centre, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| | - Thomas I-H Park
- Department of Pharmacology, University of Auckland, Auckland, New Zealand.,Neurosurgical Research Unit, The Centre for Brain Research, University of Auckland, Auckland, New Zealand.,The Hugh Green Biobank, The Centre for Brain Research, University of Auckland, Auckland, New Zealand
| |
Collapse
|
43
|
Cyclodextrin-Based Displacement Strategy of Sterigmatocystin from Serum Albumin as a Novel Approach for Acute Poisoning Detoxification. Int J Mol Sci 2023; 24:ijms24054485. [PMID: 36901918 PMCID: PMC10003537 DOI: 10.3390/ijms24054485] [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: 02/06/2023] [Revised: 02/20/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
This study demonstrates that sterigmatocystin (STC) interacts non-covalently with various cyclodextrins (CDs), showing the highest binding affinity for sugammadex (a γ-CD derivative) and γ-CD, and an almost order of magnitude lower affinity for β-CD. This difference in affinity was studied using molecular modelling and fluorescence spectroscopy, which demonstrated a better insertion of STC into larger CDs. In parallel, we showed that STC binds to human serum albumin (HSA) (a blood protein known for its role as a transporter of small molecules) with an almost two order of magnitude lower affinity compared to sugammadex and γ-CD. Competitive fluorescence experiments clearly demonstrated an efficient displacement of STC from the STC-HSA complex by cyclodextrins. These results are a proof-of-concept that CDs can be used to complex STC and related mycotoxins. Similarly, as sugammadex extracts neuromuscular relaxants (e.g., rocuronium and vecuronium) from blood and blocks their bioactivity, it could also be used as first aid upon acute intoxication to encapsulate a larger part of the STC mycotoxin from serum albumin.
Collapse
|
44
|
Zhou Y, Yu S, Chen D, Li H, Xu P, Yuan C, Jiang L, Huang M. Nafamostat Mesylate in Combination with the Mouse Amino-Terminal Fragment of Urokinase-Human Serum Albumin Improves the Treatment Outcome of Triple-Negative Breast Cancer Therapy. Mol Pharm 2023; 20:905-917. [PMID: 36463525 DOI: 10.1021/acs.molpharmaceut.2c00297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Abstract
Triple-negative breast cancer (TNBC) is highly aggressive and causes a higher proportion of metastatic cases. However, therapies directed to specific molecular targets have rarely achieved clinically meaningful improvements in the outcome of TNBC therapy. A urokinase-type plasminogen activator (uPA), one of the best-validated biomarkers of breast cancer, is an extracellular proteolytic serine protease involved in many pathological and physiological processes, including tumor cell invasion and metastasis. Nafamostat mesylate (NM) is a synthetic compound that inhibits various serine proteases and has been used as a therapeutic agent for the treatment of TNBC. Nevertheless, NM has poor specificity for serine proteases and is easy be hydrolyzed; moreover, the inhibitory mechanism of TNBC therapy is unclear. In this study, we combine NM with a macromolecular drug delivery vehicle, mouse amino-terminal fragment of urokinase-human serum albumin (mATF-HSA), to form a complex (mATF-HSA:NM) using the dilution-incubation-purification method. mATF specifically targets uPAR overexpressed on the surface of TNBC cells; moreover, HSA prevents NM from being hydrolyzed by numerous serine proteases. mATF-HSA:NM showed stronger inhibitory effects on the proliferation and metastasis of TNBC in vitro and in vivo without significant cytotoxicity on normal cells and tissues. In addition, we demonstrated that NM mediates metastasis of TNBC cells through inhibition of uPA using a stable uPA knockdown cell line (MDA-MB231 shuPA). Overall, we have developed a macromolecular complex targeted to treat high uPAR-expressing tumor types, and mATF-HSA can potentially be used to load other types of drugs with tumor-targeting specificity for mouse tumor models and is a promising tool to study tumor biology in mouse tumor models.
Collapse
Affiliation(s)
- Yang Zhou
- College of Chemistry, Fuzhou University, Fuzhou, Fujian350116, P.R. China
| | - Shujuan Yu
- College of Chemistry, Fuzhou University, Fuzhou, Fujian350116, P.R. China
| | - Dan Chen
- College of Chemistry, Fuzhou University, Fuzhou, Fujian350116, P.R. China
| | - Hanlin Li
- College of Chemistry, Fuzhou University, Fuzhou, Fujian350116, P.R. China
| | - Peng Xu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian350116, P.R. China
| | - Cai Yuan
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian350116, P.R. China
| | - Longguang Jiang
- College of Chemistry, Fuzhou University, Fuzhou, Fujian350116, P.R. China.,Fujian Key Lab Moratory of Marine Enzyme Engineering, Fuzhou University, Fuzhou, Fujian350116, P.R. China
| | - Mingdong Huang
- College of Chemistry, Fuzhou University, Fuzhou, Fujian350116, P.R. China
| |
Collapse
|
45
|
Jithinraj TK, Saheer VC, Chakkumkumarath L. Chiral 8-aminoBODIPY-based fluorescent probes with site selectivity for the quantitative detection of HSA in biological samples. Analyst 2023; 148:286-296. [PMID: 36533779 DOI: 10.1039/d2an01525k] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Human serum albumin (HSA) is one of the vital proteins in blood serum, and its optimum level is a reflection of the physiological well-being of an individual. Any abnormalities in serum HSA levels could often be a sign of disguised physiological disorders. The importance of fast and accurate determination of serum HSA levels has led to the development of various quantification methods. Among these, fluorescence-based methods employ molecular probes capable of producing selective responses on interaction with HSA. Herein, we report chiral 8-aminoBODIPY-based probes having blue emission for the quantitative detection of HSA in buffer and human blood serum. A pair of 8-aminoBODIPY enantiomers, namely R-PEB and S-PEB, were synthesized. They exhibited a fast 'turn-on' fluorescence response towards HSA, allowing its detection and quantification. In PBS buffer, R-PEB and S-PEB showed very good sensitivity with a limit of detection (LoD) of 25 nM (KD = 9.84 ± 0.14 μM) and 39 nM (KD = 18.67 ± 0.21 μM), respectively. The linear relationship observed between the fluorescence intensity of R-PEB/S-PEB and the HSA concentration in serum samples allowed us to generate a reference curve for HSA estimation for practical applications. Examination of unknown serum samples showed a good correlation with the results obtained by the benchmark BCG method. Interestingly, the difference in these probes' dissociation constants and LoD indicated their differential binding to HSA. Considering the availability of multiple ligand binding sites in HSA, their binding preferences were investigated in detail by displacement assays using site-specific drugs. These studies showed the preferential affinity of R-PEB towards site II, which was further substantiated using molecular docking studies. However, these displacement assays could not identify the preferred binding site of S-PEB. Blind docking studies indicated that S-PEB occupied a site closer to FA5. Selective binding of R-PEB to site II and its characteristic photophysical response can be utilized to quickly screen potential site II binding drugs.
Collapse
Affiliation(s)
| | | | - Lakshmi Chakkumkumarath
- Department of Chemistry, National Institute of Technology Calicut, Calicut-673601, Kerala, India.
| |
Collapse
|
46
|
Owczarzy A, Rogóż W, Kulig K, Pożycka J, Zięba A, Maciążek-Jurczyk M. Spectroscopic Studies of Quinobenzothiazine Derivative in Terms of the In Vitro Interaction with Selected Human Plasma Proteins: Part 2. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28020698. [PMID: 36677755 PMCID: PMC9865466 DOI: 10.3390/molecules28020698] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/14/2022] [Accepted: 01/05/2023] [Indexed: 01/12/2023]
Abstract
Synthesis of anticancer substances and studying their binding abilities towards human serum proteins as carriers are important parts of pharmaceutical and medical sciences development. The presented work is a continuation of studies of quinobenzothiazine derivatives binding with serum proteins. The main aim of this work was a spectroscopic analysis of second from benzothiazinium derivatives salt, 9-fluoro-5-alkyl-12(H)-quino [3,4-b][1,4]benzothiazinium chloride (Salt2), its interaction with carrier proteins, i.e., human serum albumin (HSA), α1-acid glycoprotein (AGP), human gamma globulin (HGG), and the study of protein secondary and tertiary structure changes using spectroscopic techniques (spectrofluorescence, UV-Vis and circular dichroism CD spectroscopy). In order to mimic in vivo conditions, control normal serum (CNS) was used. Using the Klotz method, both binding constants (Ka [M-1]) and the number of binding classes (n) were calculated. In addition, the percentage of displacement of binding site markers from HSA and AGP molecules has been defined. Based on the obtained data, it can be concluded that the main binding protein for Salt2 is AGP. HSA and HGG are also involved in the distribution of the studied substance in the bloodstream. Moreover, Salt2 very slightly interacts with CNS, which can cause strong therapeutic as well as toxic effects. The analysis of CD spectra confirms that there are no changes in the secondary structure of the main binding proteins in the presence of Salt2.
Collapse
Affiliation(s)
- Aleksandra Owczarzy
- Department of Physical Pharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| | - Wojciech Rogóż
- Department of Physical Pharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| | - Karolina Kulig
- Department of Physical Pharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| | - Jadwiga Pożycka
- Department of Physical Pharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| | - Andrzej Zięba
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
| | - Małgorzata Maciążek-Jurczyk
- Department of Physical Pharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland
- Correspondence: ; Tel.: +48-32-364-1582
| |
Collapse
|
47
|
Yakupova LR, Kopnova TY, Skuredina AA, Le-Deygen IM, Shustrov PN, Novoselov AM, Kudryashova EV. The Formation of β-Cyclodextrin Complexes with Levofloxacin and Ceftriaxone as an Approach to the Regulation of Drugs’ Pharmacokinetic. COLLOID JOURNAL OF THE RUSSIAN ACADEMY OF SCIENCES 2023; 85:114-127. [PMCID: PMC10112827 DOI: 10.1134/s1061933x22600488] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 08/04/2022] [Accepted: 08/25/2022] [Indexed: 06/29/2023]
Abstract
The study has been devoted to the complexation of hydroxypropyl-β-cyclodextrin (HPCD) with antibacterial drugs, namely, ceftriaxone (CT) and levofloxacin (LV), which are used to treat respiratory diseases, including bacterial infections of the respiratory tract. FTIR and NMR spectroscopic investigations have shown that the LV–HPCD complex is formed mainly due to the inclusion of the aromatic fragment of LV into the HPCD cavity; while the CT–HPCD complex is realized on the HPCD surface. Being a more hydrophobic molecule, LV forms ten times stronger complexes with HPCD than does CT: KdisLV-HPCD ~ 10–3 M, while KdisCT-HPCD ~10–2 M at pH 7.4. It has been shown that, for singly charged forms of the drugs, the complexes are two times more stable. Fluorescence spectroscopy has been employed to study the thermodynamic parameters for the interaction of dosage forms with human serum albumin. Negative values of ΔH and ΔS of the reaction have indicated both hydrogen bonding and van der Waals interactions during the complexation of both drugs with human serum albumin. It has been found that the protein is ~4 times more strongly bound to LV at 37°C as compared with CT. The data obtained will make it possible to improve the characteristics of the studied drugs and bring the methods of treating severe forms of respiratory diseases to a new level.
Collapse
Affiliation(s)
- L. R. Yakupova
- Department of Chemistry, Moscow State University, 119296 Moscow, Russia
| | - T. Yu. Kopnova
- Department of Chemistry, Moscow State University, 119296 Moscow, Russia
| | - A. A. Skuredina
- Department of Chemistry, Moscow State University, 119296 Moscow, Russia
| | - I. M. Le-Deygen
- Department of Chemistry, Moscow State University, 119296 Moscow, Russia
| | - P. N. Shustrov
- Sechenov First Moscow State Medical University, Ministry of Health of Russia (Sechenov University), 119991 Moscow, Russia
| | - A. M. Novoselov
- Department of Chemistry, Moscow State University, 119296 Moscow, Russia
| | - E. V. Kudryashova
- Department of Chemistry, Moscow State University, 119296 Moscow, Russia
| |
Collapse
|
48
|
Protein encapsulation of nanocatalysts: A feasible approach to facilitate catalytic theranostics. Adv Drug Deliv Rev 2023; 192:114648. [PMID: 36513163 DOI: 10.1016/j.addr.2022.114648] [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: 09/29/2022] [Revised: 11/14/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022]
Abstract
Enzyme-mimicking nanocatalysts, also termed nanozymes, have attracted much attention in recent years. They are considered potential alternatives to natural enzymes due to their multiple catalytic activities and high stability. However, concerns regarding the colloidal stability, catalytic specificity, efficiency and biosafety of nanomaterials in biomedical applications still need to be addressed. Proteins are biodegradable macromolecules that exhibit superior biocompatibility and inherent bioactivities; hence, the protein modification of nanocatalysts is expected to improve their bioavailability to match clinical needs. The diversity of amino acid residues in proteins provides abundant functional groups for the conjugation or encapsulation of nanocatalysts. Moreover, protein encapsulation can not only improve the overall performance of nanocatalysts in biological systems, but also bestow materials with new features, such as targeting and retention in pathological sites. This review aims to report the recent developments and perspectives of protein-encapsulated catalysts in their functional improvements, modification methods and applications in biomedicine.
Collapse
|
49
|
Ocampo-Hernández J, de Jesús Gómez-Guzmán J, Cruz-Ramírez M, Rebolledo-Chávez JPF, Mendoza A, Moreno-Esparza R, Ortiz-Frade L. Electrochemical and CD-spectroelectrochemical studies of the interaction between BSA and the complex [Cu(Bztpen)] 2+, (Bztpen = (N-benzyl-N, N', N'-tris (pyridin-2-ylmethyl) ethylenediamine). J Inorg Biochem 2022; 237:111994. [PMID: 36126431 DOI: 10.1016/j.jinorgbio.2022.111994] [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: 04/07/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 01/18/2023]
Abstract
In this work we report the electrochemical, spectroscopical and spectro-electrochemical studies of a model complex [CuΙΙ(Bztpen)]2+, (Bztpen = (N-benzyl-N,N',N'-tris(pyridin-2-ylmethyl)ethylenediamine) in order to propose a methodology to evaluate the interaction of potential metal based anticancer agents during electron transfer processes, with transport proteins such as Bovine Serum Albumin (BSA). It was possible to establish a reversible electron transfer [CuΙΙ(Bztpen)]2+ +1e → [CuΙ(Bztpen)]+ and a weak interaction energy between BSA and [CuΙΙ(Bztpen)] and [CuΙ(Bztpen)] species, with no adsorption of protein over the electrode surface. Circular Dichroism (CD) Spectroelectrochemistry, not reported before, reveals no significant changes in BSA structure during the electron transfer [CuΙΙ(Bztpen)]2+ + 1e → [CuΙ(Bztpen)]+. CD experiments at variable temperature for BSA denaturalization in the absence and in the presence of [CuΙΙ(Bztpen)]2+, shown no change in thermodynamic parameters due to low interaction between the transport protein and copper complex.
Collapse
Affiliation(s)
- Janet Ocampo-Hernández
- Departamento de Electroquímica, Centro de Investigación y Desarrollo Tecnológico en Electroquímica S.C, Parque Tecnológico Querétaro, Sanfandila, Pedro de Escobedo, C.P 76703, Querétaro, Mexico
| | - José de Jesús Gómez-Guzmán
- Departamento de Electroquímica, Centro de Investigación y Desarrollo Tecnológico en Electroquímica S.C, Parque Tecnológico Querétaro, Sanfandila, Pedro de Escobedo, C.P 76703, Querétaro, Mexico
| | - Marisela Cruz-Ramírez
- Universidad Tecnológica de San Juan del Río, División de Química y Energías Renovables, Avenida La Palma No. 125 Vista Hermosa, San Juan del Río, Querétaro, Mexico
| | - Juan Pablo F Rebolledo-Chávez
- Universidad Tecnológica de San Juan del Río, División de Química y Energías Renovables, Avenida La Palma No. 125 Vista Hermosa, San Juan del Río, Querétaro, Mexico
| | - Angel Mendoza
- Centro de Química, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Ciudad Universitaria, Col. San Manuel, 72570 Puebla, Mexico
| | - Rafael Moreno-Esparza
- Departamento de Química Inorgánica y Nuclear, Facultad de Química, Universidad Nacional Autónoma de México, Av. Universidad 3000, Ciudad Universitaria, México, D.F 04510, Mexico
| | - Luis Ortiz-Frade
- Departamento de Electroquímica, Centro de Investigación y Desarrollo Tecnológico en Electroquímica S.C, Parque Tecnológico Querétaro, Sanfandila, Pedro de Escobedo, C.P 76703, Querétaro, Mexico.
| |
Collapse
|
50
|
Sharmeen S, Kyei I, Hatch A, Hage DS. Analysis of drug interactions with serum proteins and related binding agents by affinity capillary electrophoresis: A review. Electrophoresis 2022; 43:2302-2323. [PMID: 36250426 PMCID: PMC10098505 DOI: 10.1002/elps.202200191] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 09/17/2022] [Accepted: 10/05/2022] [Indexed: 11/06/2022]
Abstract
Biomolecules such as serum proteins can interact with drugs in the body and influence their pharmaceutical effects. Specific and precise methods that analyze these interactions are critical for drug development or monitoring and for diagnostic purposes. Affinity capillary electrophoresis (ACE) is one technique that can be used to examine the binding between drugs and serum proteins, or other agents found in serum or blood. This article will review the basic principles of ACE, along with related affinity-based capillary electrophoresis (CE) methods, and examine recent developments that have occurred in this field as related to the characterization of drug-protein interactions. An overview will be given of the various formats that can be used in ACE and CE for such work, including the relative advantages or weaknesses of each approach. Various applications of ACE and affinity-based CE methods for the analysis of drug interactions with serum proteins and other binding agents will also be presented. Applications of ACE and related techniques that will be discussed include drug interaction studies with serum agents, chiral drug separations employing serum proteins, and the use of CE in hybrid methods to characterize drug binding with serum proteins.
Collapse
Affiliation(s)
- Sadia Sharmeen
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Isaac Kyei
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Arden Hatch
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - David S Hage
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| |
Collapse
|