1
|
Qu N, Song K, Ji Y, Liu M, Chen L, Lee RJ, Teng L. Albumin Nanoparticle-Based Drug Delivery Systems. Int J Nanomedicine 2024; 19:6945-6980. [PMID: 39005962 PMCID: PMC11246635 DOI: 10.2147/ijn.s467876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 05/28/2024] [Indexed: 07/16/2024] Open
Abstract
Nanoparticle-based systems are extensively investigated for drug delivery. Among others, with superior biocompatibility and enhanced targeting capacity, albumin appears to be a promising carrier for drug delivery. Albumin nanoparticles are highly favored in many disease therapies, as they have the proper chemical groups for modification, cell-binding sites for cell adhesion, and affinity to protein drugs for nanocomplex generation. Herein, this review summarizes the recent fabrication techniques, modification strategies, and application of albumin nanoparticles. We first discuss various albumin nanoparticle fabrication methods, from both pros and cons. Then, we provide a comprehensive introduction to the modification section, including organic albumin nanoparticles, metal albumin nanoparticles, inorganic albumin nanoparticles, and albumin nanoparticle-based hybrids. We finally bring further perspectives on albumin nanoparticles used for various critical diseases.
Collapse
Affiliation(s)
- Na Qu
- School of Pharmacy, Liaoning University, Shenyang, 110036, People's Republic of China
| | - Ke Song
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, 6229 ER, the Netherlands
| | - Yating Ji
- School of Pharmacy, Liaoning University, Shenyang, 110036, People's Republic of China
| | - Mingxia Liu
- School of Pharmacy, Liaoning University, Shenyang, 110036, People's Republic of China
| | - Lijiang Chen
- School of Pharmacy, Liaoning University, Shenyang, 110036, People's Republic of China
| | - Robert J Lee
- School of Life Sciences, Jilin University, Changchun, 130023, People's Republic of China
- College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Lesheng Teng
- School of Life Sciences, Jilin University, Changchun, 130023, People's Republic of China
- State Key Laboratory of Long-Acting and Targeting Drug Delivery System, Yantai, 264000, People's Republic of China
| |
Collapse
|
2
|
Gokara M, Yusuf Zamal M, Lavudiya VS, Subramanyam R. Deciphering the binding mechanism of gingerol molecules with plasma proteins: implications for drug delivery and therapeutic potential. J Biomol Struct Dyn 2024:1-18. [PMID: 38305837 DOI: 10.1080/07391102.2024.2310795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 01/22/2024] [Indexed: 02/03/2024]
Abstract
Ginger is a highly valued herb, renowned globally for its rich content of phenolic compounds. It has been traditionally used to treat various health conditions such as cardiovascular diseases, digestive issues, migraines, Alzheimer's disease, tumor reduction and chronic inflammation. Despite its potential medicinal applications, the therapeutic effectiveness of ginger is hindered by its limited availability and low plasma concentration levels. In this study, we explored the interaction of ginger's primary phenolic compounds, specifically 6-gingerol (6 G), 8-gingerol (8 G) and 10-gingerol (10 G), with plasma proteins which are human serum albumin (HSA) and α-1-acid glycoprotein (AGP). These two plasma proteins significantly influence drug distribution and disposition as they are key binding sites for most drugs. Fluorescence emission spectra indicated strong binding of 6, 8 and 10 G with HSA, with binding constants of 2.03 ± 0.01 × 104 M-1, 4.20 ± 0.01 × 104 M-1 and 6.03 ± 0.01 × 106 M-1, respectively. However, the binding of gingerols with AGP was found to be negligible. Molecular displacement by site-specific probes and molecular docking analyses revealed that gingerols bind at the IIA domain, with stability provided by hydrogen bonds, van der Waals forces, conventional hydrogen bonds, carbon-hydrogen bonds, alkyl and Pi-alkyl interactions. Further, the partial unfolding of the protein was observed upon binding the gingerol compound with HSA. In addition, molecular dynamic simulations demonstrated that gingerols remained stable in the subdomain IIA over 100 ns. This stability, coupled with Molecular Mechanics Generalized Born Surface Area indicating free energies of -43.765, -57.504 and -66.69 kcal/mol for 6, 8 and 10 G, respectively, reinforces the robust binding potential of these compounds. Circular dichroism studies suggested that the interaction of gingerols leads to the minimal transformation of HSA secondary structure, with the pattern being 10 G > 8 G > 6 G, a finding further substantiated by root mean square deviation and root mean square fluctuation fluctuations. These results propose that HSA has a stronger affinity to gingerols than AGP, which could have significant implications on the therapeutic circulating levels of gingerols.
Collapse
Affiliation(s)
- Mahesh Gokara
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Mohammad Yusuf Zamal
- Department of Plant Sciences, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Vijay Srinivas Lavudiya
- Department of Plant Sciences, School of Life Sciences, University of Hyderabad, Hyderabad, India
| | - Rajagopal Subramanyam
- Department of Plant Sciences, School of Life Sciences, University of Hyderabad, Hyderabad, India
| |
Collapse
|
3
|
Ashraf S, Qaiser H, Tariq S, Khalid A, Makeen HA, Alhazmi HA, Ul-Haq Z. Unraveling the versatility of human serum albumin - A comprehensive review of its biological significance and therapeutic potential. Curr Res Struct Biol 2023; 6:100114. [PMID: 38111902 PMCID: PMC10726258 DOI: 10.1016/j.crstbi.2023.100114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 12/20/2023] Open
Abstract
Human serum albumin (HSA) is a multi-domain macromolecule with diverse ligand binding capability because of its ability to allow allosteric modulation despite being a monomeric protein. Physiologically, HSA act as the primary carrier for various exogenous and endogenous compounds and fatty acids, and alter the pharmacokinetic properties of several drugs. It has antioxidant properties and is utilized therapeutically to improve the drug delivery of pharmacological agents for the treatment of several disorders. The flexibility of albumin in holding various types of drugs coupled with a variety of modifications makes this protein a versatile drug carrier with incalculable potential in therapeutics. This review provides a brief outline of the different structural properties of HSA, and its various binding sites, moreover, an overview of the genetic, biomedical, and allosteric modulation of drugs and drug delivery aspects of HSA is also included, which may be helpful in guiding advanced clinical applications and further research on the therapeutic potential of this extraordinary protein.
Collapse
Affiliation(s)
- Sajda Ashraf
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75210, Pakistan
| | - Hina Qaiser
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75210, Pakistan
| | - Sumayya Tariq
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75210, Pakistan
| | - Asaad Khalid
- Substance Abuse and Toxicology Research Center, Jazan University, P.O. Box: 114, Jazan, Saudi Arabia
- Medicinal and Aromatic Plants Research Institute, National Center for Research, P.O. Box: 2424, Khartoum, 11111, Sudan
| | - Hafiz A. Makeen
- Pharmacy Practice Research Unit, Clinical Pharmacy Department, Faculty of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Hassan A. Alhazmi
- Substance Abuse and Toxicology Research Center, Jazan University, P.O. Box: 114, Jazan, Saudi Arabia
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, P.O. Box 114, 45142, Jazan, Saudi Arabia
| | - Zaheer Ul-Haq
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75210, Pakistan
| |
Collapse
|
4
|
Liu Z, Trifonova D, Tulaeva I, Riabova K, Karsonova A, Kozlov E, Elisyutina O, Khaitov M, Focke-Tejkl M, Chen TH, Karaulov A, Valenta R. Albumins represent highly cross-reactive animal allergens. Front Immunol 2023; 14:1241518. [PMID: 37928538 PMCID: PMC10623431 DOI: 10.3389/fimmu.2023.1241518] [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: 06/16/2023] [Accepted: 09/20/2023] [Indexed: 11/07/2023] Open
Abstract
Albumins from animals are highly cross-reactive allergens for patients suffering from immunoglobulin E (IgE)-mediated allergy. Approximately 20-30% of cat and dog allergic patients show IgE reactivity and mount IgE-mediated allergic reactions to cat and dog albumin. It is astonishing that allergic patients can develop specific IgE responses against animal albumins because these proteins exhibit a more than 70% sequence identity to human serum albumin (HSA) which is the most abundant protein in the blood of the human body. The sequence identity of cat albumin (Fel d 2) and dog albumin (Can f 3) and HSA are 82% and 80%, respectively. Given the high degree of sequence identity between the latter two allergens and HSA one would expect that immunological tolerance would prohibit IgE sensitization to Fel d 2 and Can f 3. Here we discuss two possibilities for how IgE sensitization to Fel d 2 and Can f 3 may develop. One possibility is the failed development of immune tolerance in albumin-allergic patients whereas the other possibility is highly selective immune tolerance to HSA but not to Fel d 2 and Can f 3. If the first assumption is correct it should be possible to detect HSA-specific T cell responses and HSA-containing immune complexes in sensitized patients. In the latter scenario few differences in the sequences of Fel d 2 and Can f 3 as compared to HSA would be responsible for the development of selective T cell and B cell responses towards Fel d 2 as well as Can f 3. However, the immunological mechanisms of albumin sensitization have not yet been investigated in detail although this will be important for the development of allergen-specific prevention and allergen-specific immunotherapy (AIT) strategies for allergy to albumin.
Collapse
Affiliation(s)
- Zicheng Liu
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Daria Trifonova
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- Laboratory for Immunopathology, Department of Clinical Immunology and Allergology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Inna Tulaeva
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- Laboratory for Immunopathology, Department of Clinical Immunology and Allergology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Ksenja Riabova
- Laboratory for Immunopathology, Department of Clinical Immunology and Allergology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Antonina Karsonova
- Laboratory for Immunopathology, Department of Clinical Immunology and Allergology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Evgeny Kozlov
- Laboratory for Immunopathology, Department of Clinical Immunology and Allergology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Olga Elisyutina
- National Research Center, NRCI Institute of Immunology, Federal Medical-Biological Agency (FMBA) of Russia, Moscow, Russia
- RUDN University, Moscow, Russia
| | - Musa Khaitov
- National Research Center, NRCI Institute of Immunology, Federal Medical-Biological Agency (FMBA) of Russia, Moscow, Russia
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - Margarete Focke-Tejkl
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- Karl Landsteiner University of Healthcare, Krems, Austria
| | | | - Alexander Karaulov
- Laboratory for Immunopathology, Department of Clinical Immunology and Allergology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Rudolf Valenta
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- Laboratory for Immunopathology, Department of Clinical Immunology and Allergology, Sechenov First Moscow State Medical University, Moscow, Russia
- National Research Center, NRCI Institute of Immunology, Federal Medical-Biological Agency (FMBA) of Russia, Moscow, Russia
- Karl Landsteiner University of Healthcare, Krems, Austria
| |
Collapse
|
5
|
Ghosh R, Kishore N. Mechanistic physicochemical insights into glycation and drug binding by serum albumin: Implications in diabetic conditions. Biochimie 2021; 193:16-37. [PMID: 34688791 DOI: 10.1016/j.biochi.2021.10.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 09/16/2021] [Accepted: 10/15/2021] [Indexed: 11/02/2022]
Abstract
The drug binding ability of serum albumin might get affected as a result of its glycation under diabetic conditions. It requires not only an understanding of the effect of glycation of the protein upon association with the drug, but also calls for an assessment of structure-property-energetics relationships. A combination of ultrasensitive calorimetric, spectroscopic and chromatographic approach has been employed to correlate thermodynamic signatures with recognition, conformation and mechanistic details of the processes involved. An important observation from this work is that 3-(dansylamino) phenyl boronic acid (DnsPBA) assay cannot always determine the extent of glycation as evidenced by MALDI-TOF mass spectra of glycated HSA due to its selectivity for 1,2 or 1,3 cis-diol structures which may be absent in certain AGEs. Protein gets modified post glycation with the formation of advanced glycation end products (AGEs), which are monitored to be targeted by the guanidine group present in anti-diabetic drugs. AGEs formed in the third and fourth week of glycation are significant in the recognition of anti-diabetic drugs. The results with metformin and aminoguanidine suggest that the extent of binding depends upon the number of guanidine group(s) in the drug molecule. Open chain molecules having guanidine group(s) exhibit stronger affinity towards glycated HSA than closed ring entities like naphthalene or pyridine moiety. The observation that the drug binding ability of HSA is not adversely affected, rather strengthened upon glycation, has implications in diabetic conditions. A rigorous structure-property-energetics correlation based on thermodynamic signatures and identification of functional groups on drugs for recognition by HSA are essential in deriving guidelines for rational drug design addressing diabetes.
Collapse
Affiliation(s)
- Ritutama Ghosh
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, Mumbai, 400 076, India
| | - Nand Kishore
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, Mumbai, 400 076, India.
| |
Collapse
|
6
|
Shen Y, Shen Y, Bi X, Li J, Chen Y, Zhu Q, Wang Y, Ding F. Linoleic acid-modified liposomes for the removal of protein-bound toxins: An in vitro study. Int J Artif Organs 2020; 44:393-403. [PMID: 33135543 DOI: 10.1177/0391398820968837] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Protein-bound uremic toxins (PBUTs) and liver failure-related cholestatic solutes are associated with adverse outcomes in patients with chronic kidney disease (CKD) and liver failure, respectively, and are not easily removed by traditional dialysis therapies. We constructed linoleic acid-modified liposomes (LA-liposomes) as indirect adsorbent in the dialysate, and evaluated their effects on the clearance of the representative PBUTs and cholestatic solutes. METHODS The LA-liposomes were prepared by the thin-film hydration method. The binding rates of liposomes and protein-bound solutes were detected by the ultrafiltration column. The in vitro dialysis experiments were performed using both non-current and current devices to assay the clearing efficiency of the dialysate supported by LA-liposomes. RESULTS The LA-liposomes exhibited good binding properties to the PBUTs, bilirubin and bile acids. The LA-liposome dialysate showed higher solute reduction rates of the representative PBUTs and cholestatic solutes than the traditional dialysate or dialysate supported by the unmodified plain liposomes. Also, albumin binding of the PBUTs was significantly inhibited by the addition of linoleic acid (LA), and the removal efficiency of PBUTs was greatly enhanced by the combination of indirect adsorbent LA-liposomes and LA as the competitive displacer. CONCLUSION LA-liposomes were efficient in the clearance of the representative PBUTs and liver failure-related solutes. Moreover, the combination of indirect adsorbent LA-liposomes and competitive displacer suggested a potential application for the extremely highly-bound solutes.
Collapse
Affiliation(s)
- Yue Shen
- Division of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Yuqi Shen
- Division of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Xiao Bi
- Division of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Jiaolun Li
- Division of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Yingjie Chen
- Division of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Qiuyu Zhu
- Division of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Yifeng Wang
- Division of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Feng Ding
- Division of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| |
Collapse
|
7
|
Wenskowsky L, Wagner M, Reusch J, Schreuder H, Matter H, Opatz T, Petry SM. Resolving Binding Events on the Multifunctional Human Serum Albumin. ChemMedChem 2020; 15:738-743. [PMID: 32162429 PMCID: PMC7318646 DOI: 10.1002/cmdc.202000069] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 02/25/2020] [Indexed: 12/29/2022]
Abstract
Physiological processes rely on initial recognition events between cellular components and other molecules or modalities. Biomolecules can have multiple sites or mode of interaction with other molecular entities, so that a resolution of the individual binding events in terms of spatial localization as well as association and dissociation kinetics is required for a meaningful description. Here we describe a trichromatic fluorescent binding- and displacement assay for simultaneous monitoring of three individual binding sites in the important transporter and binding protein human serum albumin. Independent investigations of binding events by X-ray crystallography and time-resolved dynamics measurements (switchSENSE technology) confirm the validity of the assay, the localization of binding sites and furthermore reveal conformational changes associated with ligand binding. The described assay system allows for the detailed characterization of albumin-binding drugs and is therefore well-suited for prediction of drug-drug and drug-food interactions. Moreover, conformational changes, usually associated with binding events, can also be analyzed.
Collapse
Affiliation(s)
- Lea Wenskowsky
- Institute of Organic ChemistryJohannes Gutenberg-UniversityDuesbergweg 10–1455128MainzGermany
| | - Michael Wagner
- Sanofi-Aventis Deutschland GmbHIndustriepark Höchst65926Frankfurt am MainGermany
| | - Johannes Reusch
- Dynamic Biosensors GmbHLochhamer Straße 1582152Martinsried/PlaneggGermany
| | - Herman Schreuder
- Sanofi-Aventis Deutschland GmbHIndustriepark Höchst65926Frankfurt am MainGermany
| | - Hans Matter
- Sanofi-Aventis Deutschland GmbHIndustriepark Höchst65926Frankfurt am MainGermany
| | - Till Opatz
- Institute of Organic ChemistryJohannes Gutenberg-UniversityDuesbergweg 10–1455128MainzGermany
| | | |
Collapse
|
8
|
Želonková K, Havadej S, Verebová V, Holečková B, Uličný J, Staničová J. Fungicide Tebuconazole Influences the Structure of Human Serum Albumin Molecule. Molecules 2019; 24:E3190. [PMID: 31480789 PMCID: PMC6749206 DOI: 10.3390/molecules24173190] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/28/2019] [Accepted: 09/01/2019] [Indexed: 01/09/2023] Open
Abstract
Studies of interactions between pesticides and target mammalian proteins are important steps toward understanding the pesticide's toxicity. Using calorimetric and spectroscopic methods, the interaction between triazole fungicide tebuconazole and human serum albumin has been investigated. The spectroscopic techniques showed that fluorescence quenching of human serum albumin by tebuconazole was the result of the formation of tebuconazole/human serum albumin complex with the static type as the dominant mechanism. The association constant was found to be 8.51 × 103 L/mol. The thermodynamic parameters were obtained as ΔH = -56.964 kJ/mol, ΔS = -115.98 J/mol·K. The main active interactions forming the tebuconazole/human serum albumin complex were identified as the interplay between hydrogen bonds and/or van der Waals forces, based on thermodynamic experiments. These binding modes were corroborated well by the predictions of molecular modeling. Hydrogen bonding of tebuconazole with Arg222, Ala215 and Ala291 of human serum albumin played a relevant role in binding. The conformation changes in secondary structure were characterized by circular dichroism and 3D fluorescence spectra.
Collapse
Affiliation(s)
- Katarína Želonková
- Faculty of Science, Pavol Jozef Šafárik University, Jesenná 5, 041 54 Košice, Slovakia
| | - Samuel Havadej
- Faculty of Science, Pavol Jozef Šafárik University, Jesenná 5, 041 54 Košice, Slovakia.
| | - Valéria Verebová
- Department of Chemistry, Biochemistry & Biophysics, University of Veterinary Medicine & Pharmacy, Komenského 73, 041 81 Košice, Slovakia.
| | - Beáta Holečková
- Department of Biology & Genetics, University of Veterinary Medicine & Pharmacy, Komenského 73, 041 81 Košice, Slovakia.
| | - Jozef Uličný
- Faculty of Science, Pavol Jozef Šafárik University, Jesenná 5, 041 54 Košice, Slovakia.
| | - Jana Staničová
- Department of Chemistry, Biochemistry & Biophysics, University of Veterinary Medicine & Pharmacy, Komenského 73, 041 81 Košice, Slovakia.
- First Faculty of Medicine, Charles University, Kateřinská 1, 121 08 Prague, Czech Republic.
| |
Collapse
|
9
|
Börgel F, Galla F, Lehmkuhl K, Schepmann D, Ametamey SM, Wünsch B. Pharmacokinetic properties of enantiomerically pure GluN2B selective NMDA receptor antagonists with 3-benzazepine scaffold. J Pharm Biomed Anal 2019; 172:214-222. [PMID: 31060034 DOI: 10.1016/j.jpba.2019.04.032] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 04/11/2019] [Accepted: 04/15/2019] [Indexed: 01/12/2023]
Abstract
Recently, the eutomers of highly potent GluN2B selective NMDA receptor antagonists with 3-benzazepine scaffold were identified. Herein, pharmacokinetic properties regarding lipophilicity, plasma protein binding (PPB) and metabolism are analyzed. The logD7.4 values of 1.68 for phenol 1 and 2.46 for methyl ether 2 are in a very good range for CNS agents. A very similar logD7.4 value was recorded for the prototypical GluN2B antagonist ifenprodil (logD7.4 = 1.49). The herein developed high performance affinity chromatography (HPAC) method using human serum albumin as stationary phase led to PPB of 3-benzazepines (R)-1-3 and (S)-1-3 of 76-98%. Upon incubation with mouse liver microsomes, (R)-1-3 and (S)-1-3 showed moderate to high metabolic stability. The (R)-configured eutomers turned out to be metabolically more stable than their (S)-configured distomers. During phase I metabolism of 3-benzazepines 1-3 hydroxylations at both aromatic rings, the aliphatic side chain and the seven-membered ring were observed. O-demethylation of methyl ether (S)-2 was faster than O-demethylation of its enantiomer (R)-2. In phase I biotransformation the phenol eutomer (R)-1 showed comparable stability as ifenprodil. In phase II biotransformation, glucuronidation of the phenolic (only 1) and benzylic hydroxy groups was observed. Both enantiomers formed the same type of metabolites, respectively, but in different amounts. Whereas, the benzylic hydroxy group of (R)-2 was glucuronidated preferably, predominant benzylic glucuronidation of (S)-3 was detected. Mouse liver microsomes produced the glucuronide of phenol 1 (main metabolite) in larger amounts than rat liver microsomes.
Collapse
Affiliation(s)
- Frederik Börgel
- Institut für Pharmazeutische und Medizinische Chemie der Universität Münster, Corrensstraße 48, D-48149, Münster, Germany
| | - Fabian Galla
- Institut für Pharmazeutische und Medizinische Chemie der Universität Münster, Corrensstraße 48, D-48149, Münster, Germany
| | - Kirstin Lehmkuhl
- Institut für Pharmazeutische und Medizinische Chemie der Universität Münster, Corrensstraße 48, D-48149, Münster, Germany
| | - Dirk Schepmann
- Institut für Pharmazeutische und Medizinische Chemie der Universität Münster, Corrensstraße 48, D-48149, Münster, Germany
| | - Simon M Ametamey
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Bernhard Wünsch
- Institut für Pharmazeutische und Medizinische Chemie der Universität Münster, Corrensstraße 48, D-48149, Münster, Germany; Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), Westfälische Wilhelms-Universität, Münster, Germany.
| |
Collapse
|
10
|
Yamazoe H. Spectroscopic study on the conformation of serum albumin in film state. J Biosci Bioeng 2019; 127:515-519. [DOI: 10.1016/j.jbiosc.2018.09.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 08/27/2018] [Accepted: 09/23/2018] [Indexed: 11/30/2022]
|
11
|
Li C, Arakawa T. Application of native polyacrylamide gel electrophoresis for protein analysis: Bovine serum albumin as a model protein. Int J Biol Macromol 2019; 125:566-571. [DOI: 10.1016/j.ijbiomac.2018.12.090] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 11/29/2018] [Accepted: 12/09/2018] [Indexed: 11/16/2022]
|
12
|
Baghaee PT, Divsalar A, Chamani J, Donya A. Human serum albumin–malathion complex study in the presence of silver nanoparticles at different sizes by multi spectroscopic techniques. J Biomol Struct Dyn 2018; 37:2254-2264. [DOI: 10.1080/07391102.2018.1491416] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Parisa Teimoori Baghaee
- Department of Biology, Faculty of Sciences, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Adeleh Divsalar
- Department of Cell & Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Jamshikhan Chamani
- Department of Biology, Faculty of Sciences, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Atena Donya
- Department of Biology, Faculty of Sciences, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| |
Collapse
|
13
|
Asada M, Nagata M, Mizuno T, Uchida T, Kurashima N, Takahashi H, Makita K, Arai H, Echizen H, Yasuhara M. Effects of cardiopulmonary bypass on the disposition of cefazolin in patients undergoing cardiothoracic surgery. Pharmacol Res Perspect 2018; 6:e00440. [PMID: 30410768 PMCID: PMC6218359 DOI: 10.1002/prp2.440] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 09/27/2018] [Accepted: 10/02/2018] [Indexed: 11/21/2022] Open
Abstract
The aim of the study was to evaluate the disposition of plasma unbound cefazolin in patients undergoing cardiothoracic surgery with cardiopulmonary bypass (CPB). Adult patients undergoing cardiothoracic surgery with CPB were enrolled in the study. Cefazolin sodium was given intravenously before skin incision (1 g) and at the beginning of CPB (2 g). Thereafter, an additional dose (1 g) was given every 4 hours. Seven to ten blood samples were collected before and during surgery. Plasma total and unbound (ultrafiltrated) cefazolin concentrations were analyzed using an HPLC-UV method. Plasma protein binding was analyzed with the Langmuir model. Twenty-seven patients (aged 70 ± 12 years, body weight 62 ± 12 kg, mean ± SD) with GFR >30 mL min-1 completed the study. There was a significant (P < 0.001) increase in median plasma unbound fraction of cefazolin from 21% before skin incision to 45% during CPB (P < 0.001), which was accompanied by a significant (P < 0.001) reduction in median plasma albumin concentration from 36 to 27 g L-1. Plasma concentrations of unbound cefazolin exceeded the assumed target thresholds of 2 μg mL-1 in all samples and of 8 μg mL-1 in all but one of 199 samples. The increased plasma unbound fraction of cefazolin would be attributable to dilutional reduction of serum albumin at the beginning of CPB and to saturable plasma protein binding of cefazolin. These data reveal CPB may alter the plasma protein binding and possibly distribution of cefazolin. Further studies are warranted to reappraise the protocol of antimicrobial prophylaxis with cefazolin in patients undergoing surgery with CPB.
Collapse
Affiliation(s)
- Mizuho Asada
- Department of PharmacyMedical HospitalTokyo Medical and Dental University (TMDU)TokyoJapan
| | - Masashi Nagata
- Department of PharmacyMedical HospitalTokyo Medical and Dental University (TMDU)TokyoJapan
- Department of Pharmacokinetics and PharmacodynamicsGraduate School of Medical and Dental SciencesTokyo Medical and Dental University (TMDU)TokyoJapan
| | - Tomohiro Mizuno
- Department of Cardiovascular SurgeryGraduate School of Medical and Dental ScienceTokyo Medical and Dental University (TMDU)TokyoJapan
| | - Tokujiro Uchida
- Department of AnesthesiologyGraduate School of Medical and Dental SciencesTokyo Medical and Dental University (TMDU)TokyoJapan
| | - Naoki Kurashima
- Medical Engineering CenterMedical Hospital of Tokyo Medical and Dental University (TMDU)TokyoJapan
| | - Hiromitsu Takahashi
- Department of PharmacyMedical HospitalTokyo Medical and Dental University (TMDU)TokyoJapan
| | - Koshi Makita
- Department of AnesthesiologyGraduate School of Medical and Dental SciencesTokyo Medical and Dental University (TMDU)TokyoJapan
| | - Hirokuni Arai
- Department of Cardiovascular SurgeryGraduate School of Medical and Dental ScienceTokyo Medical and Dental University (TMDU)TokyoJapan
| | - Hirotoshi Echizen
- Department of PharmacotherapyMeiji Pharmaceutical UniversityTokyoJapan
| | - Masato Yasuhara
- Department of Pharmacokinetics and PharmacodynamicsGraduate School of Medical and Dental SciencesTokyo Medical and Dental University (TMDU)TokyoJapan
| |
Collapse
|
14
|
Xu L, Hu YX, Li YC, Zhang L, Ai HX, Liu HS, Liu YF, Sang YL. Study on the interaction of tussilagone with human serum albumin (HSA) by spectroscopic and molecular docking techniques. J Mol Struct 2017. [DOI: 10.1016/j.molstruc.2017.08.039] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
15
|
Maleimido-proxyl as an EPR spin label for the evaluation of conformational changes of albumin. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2017; 46:773-787. [DOI: 10.1007/s00249-017-1257-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 08/04/2017] [Accepted: 09/16/2017] [Indexed: 11/25/2022]
|
16
|
Dinis-Oliveira RJ. Metabolic profile of oxazepam and related benzodiazepines: clinical and forensic aspects. Drug Metab Rev 2017; 49:451-463. [PMID: 28903606 DOI: 10.1080/03602532.2017.1377223] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Anxiolytic drugs, namely benzodiazepines, are the most commonly used psychoactive substances since anxiety disorders are prevalent mental disorders particularly in the Western world. Oxazepam is a short-acting benzodiazepine and one of the most frequently prescribed anxiolytic drugs. It is also the active metabolite of a wide range of other benzodiazepines, such as diazepam, ketazolam, temazepam, chlordiazepoxide, demoxazepam, halazepam, medazepam, prazepam, pinazepam, and chlorazepate. Therefore, relevant clinical and forensic outocomes may arise, namely those related to interference in driving performance. It is clinically available as a racemic formulation, with S-enantiomer being more active than R-enantiomer. In humans, it is mainly polimorphically metabolized by glucuronide conjugation at the 3-carbon hydroxyl group, yielding stable diastereomeric glucuronides (R- and S-oxazepam glucuronide). Relevant metabolic and stereoselective interspecies differences have been reported. In this work, the pharmacokinetics of oxazepam with particular focus on metabolic pathways is fully reviewed. Moreover, the metabolic profile of other prescribed benzodiazepines that produce oxazepam as a metabolite is also discussed. It is aimed that knowing the metabolism of oxazepam and related benzodiazepines may lead to the development of new analytical strategies for its early detection and help in further toxicological and clinical interpretations.
Collapse
Affiliation(s)
- Ricardo Jorge Dinis-Oliveira
- a IINFACTS - Institute of Research and Advanced Training in Health Sciences and Technologies, Department of Sciences , University Institute of Health Sciences (IUCS), CESPU, CRL , Gandra , Portugal.,b UCIBIO-REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy , University of Porto , Porto , Portugal.,c Department of Public Health and Forensic Sciences, and Medical Education, Faculty of Medicine , University of Porto , Porto , Portugal
| |
Collapse
|
17
|
Thirupathi Kumara Raja S, Prakash T, Gnanamani A. Redox responsive albumin autogenic nanoparticles for the delivery of cancer drugs. Colloids Surf B Biointerfaces 2017; 152:393-405. [PMID: 28157647 DOI: 10.1016/j.colsurfb.2017.01.044] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Revised: 01/23/2017] [Accepted: 01/24/2017] [Indexed: 11/26/2022]
Abstract
The present study explores preparation and characterization of redox sensitive albumin autogenic nanoparticles (ANPs) for drug delivery applications. Human serum albumin nanoparticles are prepared by desolvation method. The particles are stabilized through self-crosslinking and no external stabilizers are involved in the preparation. ANPs are then subjected to Camptothecin (CPT) drug loading. Experiments on in vitro and in vivo release profile, cytotoxic and cytocompatability, hemocompatability, blood clearance, tracking and bio imaging are studied in detail. The redox sensitive and drug release properties of ANPs studied in the presence of glutathione. Results on the physical, chemical and instrumental characterization warrant the property of the nanoparticles. ANPs obtained in the present study is biocompatible, biodegradable, effectively entangle the chosen drug, release the drug in the controlled manner, sensitive to reducing environment, nil toxicity and appreciable uptake by cells. In the current scenario on the requirement of a drug carrier with redox sensitive property to encounter cancer cells, the results of the present study on albumin nanoparticles with redox sensitivity is smart and pave the way in the cancer therapeutics.
Collapse
Affiliation(s)
- S Thirupathi Kumara Raja
- Biological Material Laboratory, Microbiology Division, CSIR-CLRI, Adyar, Chennai 600 020, Tamil Nadu, India
| | - T Prakash
- Biological Material Laboratory, Microbiology Division, CSIR-CLRI, Adyar, Chennai 600 020, Tamil Nadu, India
| | - A Gnanamani
- Biological Material Laboratory, Microbiology Division, CSIR-CLRI, Adyar, Chennai 600 020, Tamil Nadu, India.
| |
Collapse
|
18
|
Assaran Darban R, Shareghi B, Asoodeh A, Chamani J. Multi-spectroscopic and molecular modeling studies of interaction between two different angiotensin I converting enzyme inhibitory peptides from gluten hydrolysate and human serum albumin. J Biomol Struct Dyn 2016; 35:3648-3662. [PMID: 27897084 DOI: 10.1080/07391102.2016.1264892] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The present study was carried out to characterize Angiotensin-converting enzyme (ACE) inhibitory peptides which are released from the trypsin hydrolysate of wheat gluten protein. The binding of two inhibitory peptide (P4 and P6) to human serum albumin (HSA) under physiological conditions has been investigated by multi-spectroscopic in combination with molecular modeling techniques. Time-resolved and quenching fluorescence spectroscopies results revealed that the quenching of HSA fluorescence by P4 and P6 in the binary and ternary systems caused HSA-peptides complexes formation. The results indicated that both peptides quenched the fluorescence intensity of HSA through a static mechanism. The binding affinities and number of binding sites were obtained for the HSA-peptides complexes. The circular dichroism (CD) data revealed that the presence of both peptides increased the α-helix content of HSA and induced the remarkable folding of the polypeptide of the protein. Therefore, the CD data determined that the protein structure has been stabilized in the percent of ACE inhibitory peptides in binary and ternary systems. The binding distances between HSA and both peptides were estimated by the Forster theory, and it was revealed that nonradiative energy transfer from HSA to peptides occurred with a high probability. ITC experiments reveal that, in the absence and presence of P6, the dominant forces are electrostatic in binary and ternary systems. Furthermore, molecular modeling studies confirmed the experimental results. Molecular modeling investigation suggested that P4 bound to the site IA and IIA of HSA in binary and ternary systems, respectively. This study on the interaction of peptides with HSA should prove helpful for realizing the distribution and transportation of food compliments and drugs in vivo, elucidating the action mechanism and dynamics of food compliments and drugs at the molecular level. It should moreover be of great use for understanding the pharmacokinetic and pharmacodynamic mechanism of the food compliments and drugs.
Collapse
Affiliation(s)
- Reza Assaran Darban
- a Department of Biology, Faculty of Science , University of Shahrekord , P.O. Box 115 , Shahrekord , Iran
| | - Behzad Shareghi
- a Department of Biology, Faculty of Science , University of Shahrekord , P.O. Box 115 , Shahrekord , Iran
| | - Ahmad Asoodeh
- b Department of Chemistry, Faculty of Sciences , Ferdowsi University of Mashhad , Mashhad , Iran
| | - Jamshidkhan Chamani
- c Department of Biochemistry and Biophysics, Faculty of Sciences , Mashhad Branch, Islamic Azad University , Mashhad , Iran
| |
Collapse
|
19
|
Naggar AH, El Kaoutit M, Naranjo-Rodriguez I, El-Sayed AY, Hidalgo-Hidalgo de Cisneros JL. Voltammetric and Spectroscopic Investigation of the Interaction Between 1,4-Benzodiazepines and Bovine Serum Albumin. J SOLUTION CHEM 2016. [DOI: 10.1007/s10953-016-0532-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
20
|
Yamazoe H, Ichikawa T, Hagihara Y, Iwasaki Y. Generation of a patterned co-culture system composed of adherent cells and immobilized nonadherent cells. Acta Biomater 2016; 31:231-240. [PMID: 26685756 DOI: 10.1016/j.actbio.2015.12.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 11/25/2015] [Accepted: 12/10/2015] [Indexed: 01/31/2023]
Abstract
Patterned co-culture is a promising technique used for fundamental investigation of cell-cell communication and tissue engineering approaches. However, conventional methods are inapplicable to nonadherent cells. In this study, we aimed to establish a patterned co-culture system composed of adherent and nonadherent cells. Nonadherent cells were immobilized on a substrate using a cell membrane anchoring reagent conjugated to a protein, in order to incorporate them into the co-culture system. Cross-linked albumin film, which has unique surface properties capable of regulating protein adsorption, was used to control their spatial localization. The utility of our approach was demonstrated through the fabrication of a patterned co-culture consisting of micropatterned neuroblastoma cells surrounded by immobilized myeloid cells. Furthermore, we also created a co-culture system composed of cancer cells and immobilized monocytes. We observed that monocytes enhanced the drug sensitivity of cancer cells and its influence was limited to cancer cells located near the monocytes. Therefore, the incorporation of nonadherent cells into a patterned co-culture system is useful for creating culture systems containing immune cells, as well as investigating the influence of these immune cells on cancer drug sensitivity. STATEMENT OF SIGNIFICANCE Various methods have been proposed for creating patterned co-culture systems, in which multiple cell types are attached to a substrate with a desired pattern. However, conventional methods, including our previous report published in Acta Biomaterialia (2010, 6, 526-533), are unsuitable for nonadherent cells. Here, we developed a novel method that incorporates nonadherent cells into the co-culture system, which allows us to precisely manipulate and study microenvironments containing nonadherent and adherent cells. Using this technique, we demonstrated that monocytes (nonadherent cells) could enhance the drug sensitivity of cancer cells and that their influence had a limited effective range. Thus, our technique is useful for recreating complex tissues in order to investigate cellular interactions involving nonadherent cells.
Collapse
Affiliation(s)
- Hironori Yamazoe
- National Institute of Advanced Industrial Science and Technology (AIST), 1-8-31 Midorigaoka, Ikeda, Osaka 563-8577, Japan.
| | - Takashi Ichikawa
- National Institute of Advanced Industrial Science and Technology (AIST), 1-8-31 Midorigaoka, Ikeda, Osaka 563-8577, Japan; Department of Chemistry and Materials Engineering, Faculty of Chemistry, Materials, and Bioengineering, Kansai University, 3-3-35 Yamate-cho, Suita-shi, Osaka 564-8680, Japan
| | - Yoshihisa Hagihara
- National Institute of Advanced Industrial Science and Technology (AIST), 1-8-31 Midorigaoka, Ikeda, Osaka 563-8577, Japan
| | - Yasuhiko Iwasaki
- Department of Chemistry and Materials Engineering, Faculty of Chemistry, Materials, and Bioengineering, Kansai University, 3-3-35 Yamate-cho, Suita-shi, Osaka 564-8680, Japan
| |
Collapse
|
21
|
Zsila F, Beke-Somfai T. Dimeric binding of plant alkaloid ellipticine to human serum proteins. RSC Adv 2016. [DOI: 10.1039/c6ra06078a] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Induced exciton circular dichroism signals reveal the accommodation of a pair of ellipticine molecules to the subdomain IB of human serum albumin and the β-barrel of α1-acid glycoprotein.
Collapse
Affiliation(s)
- Ferenc Zsila
- Biomolecular Self-Assembly Group
- Institute of Materials and Environmental Chemistry
- Research Centre for Natural Sciences
- Hungarian Academy of Sciences
- Budapest
| | - Tamás Beke-Somfai
- Biomolecular Self-Assembly Group
- Institute of Materials and Environmental Chemistry
- Research Centre for Natural Sciences
- Hungarian Academy of Sciences
- Budapest
| |
Collapse
|
22
|
Sekula B, Bujacz A. Structural Insights into the Competitive Binding of Diclofenac and Naproxen by Equine Serum Albumin. J Med Chem 2015; 59:82-9. [PMID: 26652101 DOI: 10.1021/acs.jmedchem.5b00909] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The binding modes to equine serum albumin (ESA) of two nonsteroidal anti-inflammatory drugs (NSAIDs), diclofenac (Dic) and naproxen (Nps), were studied by X-ray crystallography and isothermal titration calorimetry. On the basis of the crystal structure of ESA/Dic determined to a resolution of 1.92 Å and the structure of the previously described ESA/Nps complex (2.42 Å), it was found that both NSAIDs bind within drug site 2 (DS2) of ESA and both occupy secondary binding sites in separate cavities of domain II (Nps) and domain III (Dic). The two structures of the ternary complex ESA/Dic/Nps, obtained by competitive cocrystallization (2.19 Å) and through a displacement experiment (2.35 Å), were determined to investigate possible competition of these widely used pharmaceutical drugs in binding to ESA. In these complexes Nps occupies the DS2 pocket common for both drugs, whereas the other distinct binding sites of Dic and Nps remain unaffected. These results suggest that combined application of both drugs may result in increased concentration of free diclofenac in plasma.
Collapse
Affiliation(s)
- Bartosz Sekula
- Institute of Technical Biochemistry, Faculty of Biotechnology and Food Sciences, Lodz University of Technology , Stefanowskiego 4/10, 90-924 Lodz, Poland
| | - Anna Bujacz
- Institute of Technical Biochemistry, Faculty of Biotechnology and Food Sciences, Lodz University of Technology , Stefanowskiego 4/10, 90-924 Lodz, Poland
| |
Collapse
|
23
|
He LL, Wang YX, Wu XX, Liu XP, Wang X, Liu B, Wang X. Enhancement of the binding affinity of methylene blue to site I in human serum albumin by cupric and ferric ions. LUMINESCENCE 2015; 30:1380-8. [PMID: 25833007 DOI: 10.1002/bio.2910] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 03/01/2015] [Accepted: 03/02/2015] [Indexed: 12/18/2022]
Abstract
In this work, the binding characteristics of methylene blue (MB) to human serum albumin (HSA) and the influence of Cu(2+) and Fe(3+) on the binding affinity of MB to HSA were investigated using fluorescence, absorption, circular dichroism (CD) spectroscopy and molecular modelling. The results of competitive binding experiments using the site probes ketoprofen and ibuprofen as specific markers suggested that MB was located in site I within sub-domain IIA of HSA. The molecular modelling results agreed with the results of competitive site marker experiments and the results of CD spectra indicated that the interaction between MB and HSA caused the conformational changes in HSA. The binding affinity of MB to HSA was enhanced but to a different extent in the presence of Cu(2+) and Fe(3+), respectively, which indicated that the influence of different metal ions varied. Enhancement of the binding affinity of MB to HSA in the presence of Cu(2+) is due to the formation of Cu(2+)-HSA complex leading to the conformational changes in HSA, whereas in the presence of Fe(3+), enhancement of the binding affinity is due to the greater stability of the Fe(3+)-HSA-MB complex compared with the MB-HSA complex.
Collapse
Affiliation(s)
- Ling-Ling He
- College of Applied Chemistry, Shenyang University of Chemical Technology, Shenyang, 110142, China
| | - Yong-Xia Wang
- College of Applied Chemistry, Shenyang University of Chemical Technology, Shenyang, 110142, China
| | - Xiao-Xia Wu
- College of Applied Chemistry, Shenyang University of Chemical Technology, Shenyang, 110142, China
| | - Xian-Ping Liu
- College of Applied Chemistry, Shenyang University of Chemical Technology, Shenyang, 110142, China
| | - Xin Wang
- School of Pharmaceutical Sciences, Liaoning University, Shenyang, 110036, China
| | - Bin Liu
- School of Pharmaceutical Sciences, Liaoning University, Shenyang, 110036, China
| | - Xin Wang
- School of Pharmaceutical Sciences, Liaoning University, Shenyang, 110036, China
| |
Collapse
|
24
|
Abstract
In vivo modification of proteins by molecules with reactive carbonyl groups leads to intermediate and advanced glycation end products (AGE). Glucose is a significant glycation reagent due to its high physiological concentration and poorly controlled diabetics show increased albumin glycation. Increased levels of glycated and AGE-modified albumin have been linked to diabetic complications, neurodegeneration, and vascular disease. This review discusses glycated albumin formation, structural consequences of albumin glycation on drug binding, removal of circulating AGE by several scavenger receptors, as well as AGE-induced proinflammatory signaling through activation of the receptor for AGE. Analytical methods for quantitative detection of protein glycation and AGE formation are compared. Finally, the use of glycated albumin as a novel clinical marker to monitor glycemic control is discussed and compared to glycated hemoglobin (HbA1c) as long-term indicator of glycemic status.
Collapse
|
25
|
Ding Z, Zhang Z, Ma H, Chen Y. In vitro hemocompatibility and toxic mechanism of graphene oxide on human peripheral blood T lymphocytes and serum albumin. ACS APPLIED MATERIALS & INTERFACES 2014; 6:19797-19807. [PMID: 25371999 DOI: 10.1021/am505084s] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Graphene oxide (GO) has shown tremendous application potential as a biomedical material. However, its interactions with blood components are not yet well understood. In this work, we assess the toxicity of pristine GO (p-GO) and functionalized GO (GO-COOH and GO-PEI) to primary human peripheral blood T lymphocytes and human serum albumin (HSA), and also study the underlying toxic mechanism. Our results indicate that p-GO and GO-COOH have good biocompatibility to T lymphocytes at the concentration below 25 μg mL(-1), but notable cytotoxicity above 50 μg mL(-1). By contrast, GO-PEI exhibits significant toxicity even at 1.6 μg mL(-1). Further investigations show that although p-GO does not enter into the cell or damage the membrane, its presence leads to the increase in reactive oxygen species (ROS), moderate DNA damage, and T lymphocyte apoptosis. Interestingly, little effect on T lymphocyte immune response suppression is observed in this process despite p-GO inflicting cell apoptosis. The toxic mechanism is that p-GO interacts directly with the protein receptors to inhibit their ligand-binding ability, leading to ROS-dependent passive apoptosis through the B-cell lymphoma-2 (Bcl-2) pathway. Compared with p-GO, GO-COOH exhibits a similar toxic effect on T lymphocytes except keeping a normal ROS level. A proposed toxic mechanism is that GO-COOH inhibits protein receptor-ligand binding, and passes the passive apoptosis signal to nucleus DNA through a ROS-independent mechanism. On the other hand, GO-PEI shows severe hematotoxicity to T lymphocytes by inducing membrane damage. For plasma protein HSA, the binding of GO-COOH results in minimal conformational change and HSA's binding capacity to bilirubin remains unaffected, while the binding of p-GO and GO-PEI exhibits strong toxicity on HSA. These findings on the interactions of two-dimensional nanomaterials and biological systems, along with the enquiry of the mechanisms, would provide essential support for further safety evaluation of the biomedical applications of GO.
Collapse
Affiliation(s)
- Zhijia Ding
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences , Suzhou 215123, China
| | | | | | | |
Collapse
|
26
|
Identification of novel PTP1B inhibitors by pharmacophore based virtual screening, scaffold hopping and docking. Eur J Med Chem 2014; 87:578-94. [DOI: 10.1016/j.ejmech.2014.09.097] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 09/03/2014] [Accepted: 09/30/2014] [Indexed: 11/23/2022]
|
27
|
Wang YQ, Zhang HM, Cao J. Exploring the interactions of decabrominateddiphenyl ether and tetrabromobisphenol A with human serum albumin. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2014; 38:595-606. [PMID: 25194328 DOI: 10.1016/j.etap.2014.08.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Revised: 08/11/2014] [Accepted: 08/13/2014] [Indexed: 06/03/2023]
Abstract
Decabrominateddiphenyl ether (deca-BDE) and tetrabromobisphenol A (TBBPA) are known as brominated flame-retardants, which are commonly found in the environment. The binding mechanisms of deca-BDE and TBBPA with human serum albumin (HSA) are still unknown. In this report, the interactions of deca-BDE and TBBPA with HSA were investigated using different spectroscopic methods and molecular modeling. The experimental results indicated the formation of complexes between deca-BDE/TBBPA and HSA with different affinity. These interactions affected the secondary structure of HSA. Thermodynamic investigations revealed that hydrophobic forces mainly drove the binding interactions of deca-BDE/TBBPA with HSA. For TBBPA, hydrogen-bonding interactions were also involved in the binding process of TBBPA with HSA. According to the analysis of experimental and theoretical data, we concluded that the binding site of deca-BDE to HSA located in the subdomain IB, while TBBPA was near to subdomain IIA and Trp-214. The binding interactions of deca-BDE and TBBPA with the most prominent carrier protein in the human circulatory system could influence mechanisms of their biochemical processes. Thus, these binding interactions can play central roles in studying the distribution and toxicity mechanisms of brominated flame-retardants.
Collapse
Affiliation(s)
- Yan-Qing Wang
- Jiangsu Provincial Key Laboratory of Coastal Wetland Bioresources and Environmental Protection, Yancheng City, Jiangsu Province 224002, People's Republic of China; Institute of Applied Chemistry and Environmental Engineering, Yancheng Teachers University, Yancheng City, Jiangsu Province 224002, People's Republic of China.
| | - Hong-Mei Zhang
- Jiangsu Provincial Key Laboratory of Coastal Wetland Bioresources and Environmental Protection, Yancheng City, Jiangsu Province 224002, People's Republic of China; Institute of Applied Chemistry and Environmental Engineering, Yancheng Teachers University, Yancheng City, Jiangsu Province 224002, People's Republic of China
| | - Jian Cao
- Jiangsu Provincial Key Laboratory of Coastal Wetland Bioresources and Environmental Protection, Yancheng City, Jiangsu Province 224002, People's Republic of China; Institute of Applied Chemistry and Environmental Engineering, Yancheng Teachers University, Yancheng City, Jiangsu Province 224002, People's Republic of China
| |
Collapse
|
28
|
Kotani A, Kotani T, Ishii N, Hakamata H, Kusu F. The effect of hyperglycemia on the pharmacokinetics of valproic acid studied by high-performance liquid chromatography with electrochemical detection. J Pharm Biomed Anal 2014; 97:47-53. [DOI: 10.1016/j.jpba.2014.04.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 04/02/2014] [Accepted: 04/07/2014] [Indexed: 01/11/2023]
|
29
|
Mikitsh JL, Chacko AM. Pathways for small molecule delivery to the central nervous system across the blood-brain barrier. PERSPECTIVES IN MEDICINAL CHEMISTRY 2014; 6:11-24. [PMID: 24963272 PMCID: PMC4064947 DOI: 10.4137/pmc.s13384] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 04/29/2014] [Accepted: 04/29/2014] [Indexed: 01/04/2023]
Abstract
The treatment of central nervous system (CNS) disease has long been difficult due to the ineffectiveness of drug delivery across the blood-brain barrier (BBB). This review summarizes important concepts of the BBB in normal versus pathophysiology and how this physical, enzymatic, and efflux barrier provides necessary protection to the CNS during drug delivery, and consequently treatment challenging. Small molecules account for the vast majority of available CNS drugs primarily due to their ability to penetrate the phospholipid membrane of the BBB by passive or carrier-mediated mechanisms. Physiochemical and biological factors relevant for designing small molecules with optimal capabilities for BBB permeability are discussed, as well as the most promising classes of transporters suitable for small-molecule drug delivery. Clinically translatable imaging methodologies for detecting and quantifying drug uptake and targeting in the brain are discussed as a means of further understanding and refining delivery parameters for both drugs and imaging probes in preclinical and clinical domains. This information can be used as a guide to design drugs with preserved drug action and better delivery profiles for improved treatment outcomes over existing therapeutic approaches.
Collapse
Affiliation(s)
- John L Mikitsh
- Department of Radiology, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ann-Marie Chacko
- Department of Radiology, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Targeted Therapeutics and Translational Nanomedicine, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
30
|
Molecular interaction of PCB180 to human serum albumin: insights from spectroscopic and molecular modelling studies. J Mol Model 2014; 20:2098. [DOI: 10.1007/s00894-014-2098-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 11/27/2013] [Indexed: 11/27/2022]
|
31
|
Kang Y, Kim MJ, Jang HN, Bae EJ, Yun S, Cho HS, Chang SH, Park DJ. Rhabdomyolysis associated with initiation of febuxostat therapy for hyperuricaemia in a patient with chronic kidney disease. J Clin Pharm Ther 2014; 39:328-30. [PMID: 24612195 DOI: 10.1111/jcpt.12144] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 01/27/2014] [Indexed: 01/18/2023]
Abstract
WHAT IS KNOWN AND OBJECTIVE Febuxostat is now recommended as the first-line pharmacological urate-lowering therapy for gout in the American College of Rheumatology guidelines. There is no case of rhabdomyolysis associated with febuxostat among reported side effects of the drug. Our objective is to report on a case of rhabdomyolysis associated with initiation of febuxostat in a patient with chronic kidney disease (CKD). CASE SUMMARY A 73-year-old male patient visited our emergency room due to progressive weakness in both lower extremities starting 3 days earlier. Ten days before presentation, his primary physician had changed his prescription from allopurinol to febuxostat (80 mg) because of poor control of uric acid levels. There was tenderness in both thighs. Initial creatinine kinase (CK) was 7652 U/L (0-170 U/L), and a bone scan using (99m) Tc-HDP revealed strong uptake in soft tissues in both thighs and buttocks. Electromyography (EMG) and nerve conduction velocity (NCV) showed abnormal spontaneous activities (ASA), suggesting myopathy, not nerve damage. On day 7 of admission, after conservative management and febuxostat withdrawal, he could walk on the ward. He is being followed in our clinic as an outpatient with no sequelae. WHAT IS NEW AND CONCLUSION This report is first case of rhabdomyolysis associated with initiation of febuxostat. Febuxostat should be withdrawn when rhabdomyolysis is confirmed.
Collapse
Affiliation(s)
- Y Kang
- Department of Internal Medicine, Gyeongsang National University Hospital, Jinju, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Noorani L, Pourgholami MH, Liang M, Morris DL, Stenzel M. Albendazole loaded albumin nanoparticles for ovarian cancer therapy. EUROPEAN JOURNAL OF NANOMEDICINE 2014; 6. [DOI: 10.1515/ejnm-2014-0026] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2025]
Abstract
AbstractAlbendazole (ABZ), a well-established antiparasitic drug, has been shown to suppress tumor growth in a number of preclinical models of cancer. However, the low solubility of ABZ limits its use as a systemic anticancer agent. To enable systemic administration, we have formulated ABZ into albumin nanoparticles with a size range of 200–300 nm, which were cross-linked with glutaraldehyde to stabilize the nanoparticles and to introduce pH-responsive features. Drug release studies demonstrated that about 20% of ABZ was released at neutral pH within a week in comparison to 70% at slightly acidic condition (pH 5). Cellular uptake studies using ovarian cancer cells indicated that nanoparticles were internalized efficiently within 1 h of incubation. Further, cell proliferation results demonstrated that albumin nanoparticles alone showed no cytotoxicity to both normal and cancer cells. In contrast, the drug-loaded nanoparticles exhibited cellular toxicity and high killing efficacy to cancer cells compared to normal cells. Collectively, our results suggest that these albumin nanoparticles may hold great potentials as ABZ carriers for cancer therapy.
Collapse
|
33
|
Ding Z, Ma H, Chen Y. Interaction of graphene oxide with human serum albumin and its mechanism. RSC Adv 2014. [DOI: 10.1039/c4ra09613d] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
We show that GONS inhibit HSA function via two routes: blocking protein active sites, or destroying protein structure.
Collapse
Affiliation(s)
- Zhijia Ding
- Key Laboratory for Nano-Bio Interface Research
- Division of Nanobiomedicine
- Suzhou Institute of Nano-Tech and Nano-Bionics
- Chinese Academy of Sciences
- Suzhou, P. R. China
| | - Hongwei Ma
- Key Laboratory for Nano-Bio Interface Research
- Division of Nanobiomedicine
- Suzhou Institute of Nano-Tech and Nano-Bionics
- Chinese Academy of Sciences
- Suzhou, P. R. China
| | - Yanyan Chen
- Key Laboratory for Nano-Bio Interface Research
- Division of Nanobiomedicine
- Suzhou Institute of Nano-Tech and Nano-Bionics
- Chinese Academy of Sciences
- Suzhou, P. R. China
| |
Collapse
|
34
|
Zsila F. Circular Dichroism Spectroscopic Detection of Ligand Binding Induced Subdomain IB Specific Structural Adjustment of Human Serum Albumin. J Phys Chem B 2013; 117:10798-806. [DOI: 10.1021/jp4067108] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Ferenc Zsila
- Laboratory
of Chemical Pharmacology, Institute of Molecular Pharmacology, Research Centre for Natural Sciences, POB 17, H-1025, Budapest, Hungary
| |
Collapse
|
35
|
Abstract
6-Mercaptopurine (6-MP) is one of a large series of purine analogues which has been found active against human leukemias. The equilibrium dialysis, circular dichroism (CD) and molecular docking were employed to study the binding of 6-MP to human serum albumin (HSA). The binding of 6-MP to HSA in the equilibrium dialysis experiment was detected by measuring the displacement of 6-MP by specific markers for site I on HSA, warfarin (RWF), phenylbutazone (PhB) and n-butyl p-aminobenzoate (ABE). It was shown, according to CD data, that binding of 6-MP to HSA leads to alteration of HSA secondary structure. Based on the findings from displacement experiment and molecular docking simulation it was found that 6-MP was located within binding cavity of subdomain IIA and the space occupied by site markers overlapped with that of 6-MP. Displacement of 6-MP by the RWF or PhB was not up the level expected for a competitive mechanism, therefore displacement of 6-MP was rather by non-cooperative than that the direct competition. Instead, in case of the interaction between ABE and 6-MP, when the little enhancement of the binding of ABE by 6-MP was found, the interaction could be via a positively cooperative mechanism.
Collapse
|
36
|
Yamasaki K, Chuang VTG, Maruyama T, Otagiri M. Albumin-drug interaction and its clinical implication. Biochim Biophys Acta Gen Subj 2013; 1830:5435-43. [PMID: 23665585 DOI: 10.1016/j.bbagen.2013.05.005] [Citation(s) in RCA: 315] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 04/30/2013] [Accepted: 05/02/2013] [Indexed: 01/22/2023]
Abstract
BACKGROUND Human serum albumin acts as a reservoir and transport protein for endogenous (e.g. fatty acids or bilirubin) and exogenous compounds (e.g. drugs or nutrients) in the blood. The binding of a drug to albumin is a major determinant of its pharmacokinetic and pharmacodynamic profile. SCOPE OF REVIEW The present review discusses recent findings regarding the nature of drug binding sites, drug-albumin binding in certain diseased states or in the presence of coadministered drugs, and the potential of utilizing albumin-drug interactions in clinical applications. MAJOR CONCLUSIONS Drug-albumin interactions appear to predominantly occur at one or two specific binding sites. The nature of these drug binding sites has been fundamentally investigated as to location, size, charge, hydrophobicity or changes that can occur under conditions such as the content of the endogenous substances in question. Such findings can be useful tools for the analysis of drug-drug interactions or protein binding in diseased states. A change in protein binding is not always a problem in terms of drug therapy, but it can be used to enhance the efficacy of therapeutic agents or to enhance the accumulation of radiopharmaceuticals to targets for diagnostic purposes. Furthermore, several extracorporeal dialysis procedures using albumin-containing dialysates have proven to be an effective tool for removing endogenous toxins or overdosed drugs from patients. GENERAL SIGNIFICANCE Recent findings related to albumin-drug interactions as described in this review are useful for providing safer and efficient therapies and diagnoses in clinical settings. This article is part of a Special Issue entitled Serum Albumin.
Collapse
Affiliation(s)
- Keishi Yamasaki
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan; DDS Research Institute, Sojo University, 4-22-1 Ikeda, Kumamoto 860-0082, Japan
| | | | | | | |
Collapse
|
37
|
Seixas JD, Mukhopadhyay A, Santos-Silva T, Otterbein LE, Gallo DJ, Rodrigues SS, Guerreiro BH, Gonçalves AML, Penacho N, Marques AR, Coelho AC, Reis PM, Romão MJ, Romão CC. Characterization of a versatile organometallic pro-drug (CORM) for experimental CO based therapeutics. Dalton Trans 2013; 42:5985-98. [PMID: 23223860 PMCID: PMC3618497 DOI: 10.1039/c2dt32174b] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The complex fac-[Mo(CO)(3)(histidinate)]Na has been reported to be an effective CO-Releasing Molecule in vivo, eliciting therapeutic effects in several animal models of disease. The CO releasing profile of this complex in different settings both in vitro and in vivo reveals that the compound can readily liberate all of its three CO equivalents under biological conditions. The compound has low toxicity and cytotoxicity and is not hemolytic. CO release is accompanied by a decrease in arterial blood pressure following administration in vivo. We studied its behavior in solution and upon the interaction with proteins. Reactive oxygen species (ROS) generation upon exposure to air and polyoxomolybdate formation in soaks with lysozyme crystals were observed as processes ensuing from the decomposition of the complex and the release of CO.
Collapse
Affiliation(s)
- João D. Seixas
- Alfama Lda, Taguspark, núcleo central 267, 2740-122 Porto Salvo, Portugal
| | - Abhik Mukhopadhyay
- REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal
| | - Teresa Santos-Silva
- REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal
| | - Leo E Otterbein
- Harvard Medical School, Department of Surgery, Transplant Institute, Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA 02215
| | - David J. Gallo
- Harvard Medical School, Department of Surgery, Transplant Institute, Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA 02215
| | | | - Bruno H. Guerreiro
- Alfama Lda, Taguspark, núcleo central 267, 2740-122 Porto Salvo, Portugal
| | | | - Nuno Penacho
- Alfama Lda, Taguspark, núcleo central 267, 2740-122 Porto Salvo, Portugal
| | - Ana R. Marques
- Alfama Lda, Taguspark, núcleo central 267, 2740-122 Porto Salvo, Portugal
| | - Ana C. Coelho
- Instituto de Tecnologia Química e Biológica da Universidade Nova de Lisboa, Av. da República, EAN, 2780-157 Oeiras, Portugal
| | - Patrícia M. Reis
- Instituto de Tecnologia Química e Biológica da Universidade Nova de Lisboa, Av. da República, EAN, 2780-157 Oeiras, Portugal
| | - Maria J. Romão
- REQUIMTE, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal
| | - Carlos C. Romão
- Alfama Lda, Taguspark, núcleo central 267, 2740-122 Porto Salvo, Portugal
- Instituto de Tecnologia Química e Biológica da Universidade Nova de Lisboa, Av. da República, EAN, 2780-157 Oeiras, Portugal
| |
Collapse
|
38
|
Han C, Fang S, Cao H, Lu Y, Ma Y, Wei D, Xie X, Liu X, Li X, Fei D, Zhao C. Molecular interaction of PCB153 to human serum albumin: insights from spectroscopic and molecular modeling studies. JOURNAL OF HAZARDOUS MATERIALS 2013; 248-249:313-321. [PMID: 23416474 DOI: 10.1016/j.jhazmat.2012.12.056] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Revised: 12/11/2012] [Accepted: 12/30/2012] [Indexed: 06/01/2023]
Abstract
Polychlorinated biphenyls (PCBs) possessed much potential hazard to environment because of its chemical stability and biological toxicity. Here, we identified the binding mode of a representative compound, PCB153, to human serum albumin (HSA) using fluorescence and molecular dynamics simulation methods. The fluorescence study showed that the intrinsic fluorescence of HSA was quenched by addition of PCB153 through a static quenching mechanism. The thermodynamic analysis proved the binding behavior was mainly governed by hydrophobic force. Furthermore, as evidenced by site marker displacement experiments using two probe compounds, it revealed that PCB153 acted exactly on subdomain IIIA (site II) of HSA. On the other hand, the molecular dynamics studies as well as free energy calculations made another important contribution to understand the conformational changes of HSA and the stability of HSA-PCB153 system. Molecular docking revealed PCB153 can bind in a large hydrophobic activity of subdomain IIIA by the hydrophobic interaction and hydrogen bond interactions between chlorine atoms and residue ASN391. The present work provided reasonable models helping us further understand the transporting, distribution and toxicity effect of PCBs when it spread into human blood serum.
Collapse
Affiliation(s)
- Chao Han
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Seedher N, Agarwal P. Competitive binding of fluoroquinolone antibiotics and some other drugs to human serum albumin: a luminescence spectroscopic study. LUMINESCENCE 2013; 28:562-8. [PMID: 23436490 DOI: 10.1002/bio.2494] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 12/21/2012] [Accepted: 01/07/2013] [Indexed: 11/06/2022]
Abstract
Co-administration of several drugs in multidrug therapy may alter the binding of each to human serum albumin (HSA) and hence their pharmacological activity. Thirty-two frequently prescribed drug combinations, consisting of four fluoroquinolone antibiotics and eight competing drugs, have been studied using fluorescence and circular dichroism spectroscopic techniques. Competitive binding studies on the drug combinations are not available in the literature. In most cases, the presence of competing drug decreased the binding affinity of fluoroquinolone, resulting in an increase in the concentration of free pharmacologically active drug. The competitive binding mechanism involved could be interpreted in terms of the site specificity of the binding and competing drugs. For levofloxacin, the change in the binding affinity was small because in the presence of site II-specific competing drugs, levofloxacin mainly occupied site I. A competitive interference mechanism was operative for sparfloxacin, whereas competitive interference as well as site-to-site displacement of competing drugs was observed in the case of ciprofloxacin hydrochloride. For enrofloxacin, a different behavior was observed for different combinations; site-to-site displacement and conformational changes as well as independent binding has been observed for various drug combinations. Circular dichroism spectral studies showed that competitive binding did not cause any major structural changes in the HSA molecule.
Collapse
Affiliation(s)
- Neelam Seedher
- Department of Chemistry, Panjab University, Chandigarh 160014, India.
| | | |
Collapse
|
40
|
Zhang C, Jin J, Zhao J, Jiang W, Yin J. Functionalized polypropylene non-woven fabric membrane with bovine serum albumin and its hemocompatibility enhancement. Colloids Surf B Biointerfaces 2013; 102:45-52. [DOI: 10.1016/j.colsurfb.2012.08.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Revised: 08/06/2012] [Accepted: 08/06/2012] [Indexed: 10/28/2022]
|
41
|
Sharma R, Choudhary S, Kishore N. Insights into the binding of the drugs diclofenac sodium and cefotaxime sodium to serum albumin: Calorimetry and spectroscopy. Eur J Pharm Sci 2012; 46:435-45. [DOI: 10.1016/j.ejps.2012.03.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Revised: 03/15/2012] [Accepted: 03/20/2012] [Indexed: 10/28/2022]
|
42
|
Barthel BL, Rudnicki DL, Kirby TP, Colvin SM, Burkhart DJ, Koch TH. Synthesis and biological characterization of protease-activated prodrugs of doxazolidine. J Med Chem 2012; 55:6595-607. [PMID: 22742660 DOI: 10.1021/jm300714p] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Doxazolidine (doxaz) is a new anthracycline anticancer agent. While structurally similar to doxorubicin (dox), doxaz acts via a distinct mechanism to selectively enhance anticancer activity over cardiotoxicity, the most significant clinical impediment to successful anthracycline treatment. Here, we describe the synthesis and characterization of a prodrug platform designed for doxaz release mediated by secreted proteolytic activity, a common association with invasiveness and poor prognosis in cancer patients. GaFK-Doxaz is hydrolyzable by the proteases plasmin and cathepsin B, both strongly linked with cancer progression, as well as trypsin. We demonstrate that activation of GaFK-Doxaz releases highly potent doxaz that powerfully inhibits the growth of a wide variety of cancer cells (average IC(50) of 8 nM). GaFK-Doxaz is stable in human plasma and is poorly membrane permeable, thereby limiting activation to locally secreted proteolytic activity and reducing the likelihood of severe side effects.
Collapse
Affiliation(s)
- Benjamin L Barthel
- Department of Chemistry and Biochemistry, University of Colorado, Boulder, CO 80309-0215, USA
| | | | | | | | | | | |
Collapse
|
43
|
Zhang F, Xue J, Shao J, Jia L. Compilation of 222 drugs’ plasma protein binding data and guidance for study designs. Drug Discov Today 2012; 17:475-85. [DOI: 10.1016/j.drudis.2011.12.018] [Citation(s) in RCA: 142] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2011] [Revised: 10/19/2011] [Accepted: 12/15/2011] [Indexed: 01/18/2023]
|
44
|
Henoumont C, Laurent S, Muller RN, Vander Elst L. Effect of nonenzymatic glycosylation on the magnetic resonance imaging (MRI) contrast agent binding to human serum albumin. J Med Chem 2012; 55:4015-9. [PMID: 22420713 DOI: 10.1021/jm3000246] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Enhanced nonenzymatic glycosylation (NEG) of human serum albumin (HSA) is observed in diabetic patients. This modifies some of the physiological functions of HSA, as the binding of ligands. Some gadolinium complexes, commonly used as MRI contrast agents, have a high affinity for HSA, which enhances their efficacy. The aim of this study is to evaluate the possible influence of the NEG of HSA on its affinity for some gadolinium chelates.
Collapse
Affiliation(s)
- Céline Henoumont
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, 19 Avenue Maistriau, B-7000 Mons, Belgium
| | | | | | | |
Collapse
|
45
|
Xiong X, Nan Y, Zhang Q. Binding Interaction between Nicotine and Human Serum Albumin by High Performance Affinity Chromatography. Chromatographia 2011. [DOI: 10.1007/s10337-011-2043-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
46
|
Wanwimolruk S, Denton JR. Plasma Protein Binding of Quinine: Binding to Human Serum Albumin, α1-Acid Glycoprotein and Plasma from Patients with Malaria. J Pharm Pharmacol 2011; 44:806-11. [PMID: 1360505 DOI: 10.1111/j.2042-7158.1992.tb03210.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Abstract
The binding of quinine to human serum albumin (HSA), α1-acid glycoprotein (AAG) and plasma obtained from healthy subjects (10 Caucasians and 15 Thais) and from Thai patients with falciparum malaria (n = 20) has been investigated. In healthy volunteers, plasma protein binding expressed as the percentage of unbound quinine was 7·9–31·0% (69–92·1% bound). The mean percentage of unbound quinine found with essentially fatty acid-free HSA (40 g L−1) was 65·4±1 −5% (mean±s.d.) and was comparable with the value (66·3 ± 3·8%, mean ± s.d.) for Fraction V HSA (40 g L−1)- This suggests that fatty acids do not influence the plasma protein binding of quinine. Binding of quinine to 0·7 g L−1 AAG was high (mean unbound 61·0 ± 5·0%), indicating that quinine is bound primarily to AAG and albumin, although other plasma proteins such as lipoproteins may be involved. The mean percentage of unbound quinine was slightly less in Caucasians (14·8 ± 6·7% unbound), compared with healthy Thai subjects (17·0 ± 6·7% unbound). The higher binding of quinine in Caucasian subjects was associated with a higher plasma AAG concentration observed in Caucasians. Mean percentage of unbound quinine was significantly lower in Thai patients with malaria (10·9 ± 4·0%) than in the healthy Thai subjects. The increase in the extent of quinine binding corresponded with the increase in the acute-phase reactant protein, AAG in the patients with malaria. Overall, when the data were combined there was a significant correlation (r = 0·846, P < 0·005) between the binding ratio (bound/unbound) of quinine and the plasma AAG concentration. This suggests that plasma AAG concentration may serve as a useful index to predict alterations in quinine binding. Although quinine is bound to albumin, it was not bound to either site I or site II on HSA as indicated from equilibrium dialysis and fluorescent probe displacement studies. Binding displacement studies revealed that there was no marked displacement of quinine by a variety of highly bound acidic and basic drugs, including other antimalarial drugs at their therapeutic concentrations.
Collapse
Affiliation(s)
- S Wanwimolruk
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| | | |
Collapse
|
47
|
Wang NX, von Recum HA. Affinity-Based Drug Delivery. Macromol Biosci 2010; 11:321-32. [DOI: 10.1002/mabi.201000206] [Citation(s) in RCA: 139] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2010] [Revised: 09/17/2010] [Indexed: 11/06/2022]
|
48
|
Buttar D, Colclough N, Gerhardt S, MacFaul PA, Phillips SD, Plowright A, Whittamore P, Tam K, Maskos K, Steinbacher S, Steuber H. A combined spectroscopic and crystallographic approach to probing drug–human serum albumin interactions. Bioorg Med Chem 2010; 18:7486-96. [DOI: 10.1016/j.bmc.2010.08.052] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2010] [Revised: 08/26/2010] [Accepted: 08/29/2010] [Indexed: 10/19/2022]
|
49
|
Edwie F, Li Y, Chung TS. Exploration of regeneration and reusability of human serum albumin as a stereoselective ligand for chiral separation in affinity ultrafiltration. J Memb Sci 2010. [DOI: 10.1016/j.memsci.2010.07.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
50
|
Abdelmoez AA, Thurner GC, Wallnöfer EA, Klammsteiner N, Kremser C, Talasz H, Mrakovcic M, Fröhlich E, Jaschke W, Debbage P. Albumin-based nanoparticles as magnetic resonance contrast agents: II. Physicochemical characterisation of purified and standardised nanoparticles. Histochem Cell Biol 2010; 134:171-96. [PMID: 20628754 DOI: 10.1007/s00418-010-0726-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2010] [Indexed: 01/25/2023]
Abstract
We are developing a nanoparticulate histochemical reagent designed for histochemistry in living animals (molecular imaging), which should finally be useful in clinical imaging applications. The iterative development procedure employed involves conceptual design of the reagent, synthesis and testing of the reagent, then redesign based on data from the testing; each cycle of testing and development generates a new generation of nanoparticles, and this report describes the synthesis and testing of the third generation. The nanoparticles are based on human serum albumin and the imaging modality selected is magnetic resonance imaging (MRI). Testing the second particle generation with newly introduced techniques revealed the presence of impurities in the final product, therefore we replaced dialysis with diafiltration. We introduced further testing methods including thin layer chromatography, arsenazo III as chromogenic assay for gadolinium, and several versions of polyacrylamide gel electrophoresis, for physicochemical characterisation of the nanoparticles and intermediate synthesis compounds. The high grade of chemical purity achieved by combined application of these methodologies allowed standardised particle sizes to be achieved (low dispersities), and accurate measurement of critical physicochemical parameters influencing particle size and imaging properties. Regression plots confirmed the high purity and standardisation. The good degree of quantitative physicochemical characterisation aided our understanding of the nanoparticles and allowed a conceptual model of them to be prepared. Toxicological screening demonstrated the extremely low toxicity of the particles. The high magnetic resonance relaxivities and enhanced mechanical stability of the particles make them an excellent platform for the further development of MRI molecular imaging.
Collapse
Affiliation(s)
- A A Abdelmoez
- Department of Radiology, Innsbruck Medical University, 6020 Innsbruck, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|