1
|
Ali MS, Al-Lohedan HA. Spectroscopic and Molecular Docking Studies of the Interaction of Non-steroidal Anti-inflammatory Drugs with a Carrier Protein: an Interesting Case of Inner Filter Effect and Intensity Enhancement in Protein Fluorescence. J Fluoresc 2024; 34:1893-1901. [PMID: 37665513 DOI: 10.1007/s10895-023-03422-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 08/28/2023] [Indexed: 09/05/2023]
Abstract
Interaction of diclofenac and indomethacin with lysozyme was studied using several spectroscopic and molecular docking methods. Difference UV-visible spectra showed that the absorption profile of lysozyme changed when both diclofenac and indomethacin were mixed with the former. The sequential addition of both drugs to the lysozyme solution caused the decrease of the intrinsic fluorescence of the latter, however, when the data were corrected for inner filter effect, an enhancement in the fluorescence of lysozyme was detected. Accordingly, the fluorescence enhancement data were analyzed using Benesi-Hildebrand equation. Both, diclofenac and indomethacin showed good interaction with lysozyme, although, the association constants of indomethacin were nearly two-fold higher as compared to that of diclofenac. The binding was slightly more spontaneous in case of indomethacin and the major forces involved in the binding of both drugs with lysozyme were hydrogen bonding and hydrophobic interactions. Secondary structural analysis revealed that both drugs partially unfolded lysozyme. Results obtained through molecular docking were also in good agreement with the experimental outcomes. Both, diclofenac and indomethacin, are bounded at the same site inside lysozyme which is located in the big hydrophobic cavity of the protein.
Collapse
Affiliation(s)
- Mohd Sajid Ali
- Department of Chemistry, College of Science, King Saud University, P.O. Box-2455, Riyadh, 11451, Saudi Arabia.
| | - Hamad A Al-Lohedan
- Department of Chemistry, College of Science, King Saud University, P.O. Box-2455, Riyadh, 11451, Saudi Arabia
| |
Collapse
|
2
|
Pomyalov S, Minetti CA, Remeta DP, Bonala R, Johnson F, Zaitseva I, Iden C, Golebiewska U, Breslauer KJ, Shoham G, Sidorenko VS, Grollman AP. Structural and mechanistic insights into the transport of aristolochic acids and their active metabolites by human serum albumin. J Biol Chem 2024; 300:107358. [PMID: 38782206 PMCID: PMC11253539 DOI: 10.1016/j.jbc.2024.107358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 04/08/2024] [Accepted: 04/17/2024] [Indexed: 05/25/2024] Open
Abstract
Aristolochic acids I and II (AA-I/II) are carcinogenic principles of Aristolochia plants, which have been employed in traditional medicinal practices and discovered as food contaminants. While the deleterious effects of AAs are broadly acknowledged, there is a dearth of information to define the mechanisms underlying their carcinogenicity. Following bioactivation in the liver, N-hydroxyaristolactam and N-sulfonyloxyaristolactam metabolites are transported via circulation and elicit carcinogenic effects by reacting with cellular DNA. In this study, we apply DNA adduct analysis, X-ray crystallography, isothermal titration calorimetry, and fluorescence quenching to investigate the role of human serum albumin (HSA) in modulating AA carcinogenicity. We find that HSA extends the half-life and reactivity of N-sulfonyloxyaristolactam-I with DNA, thereby protecting activated AAs from heterolysis. Applying novel pooled plasma HSA crystallization methods, we report high-resolution structures of myristic acid-enriched HSA (HSAMYR) and its AA complexes (HSAMYR/AA-I and HSAMYR/AA-II) at 1.9 Å resolution. While AA-I is located within HSA subdomain IB, AA-II occupies subdomains IIA and IB. ITC binding profiles reveal two distinct AA sites in both complexes with association constants of 1.5 and 0.5 · 106 M-1 for HSA/AA-I versus 8.4 and 9.0 · 105 M-1 for HSA/AA-II. Fluorescence quenching of the HSA Trp214 suggests variable impacts of fatty acids on ligand binding affinities. Collectively, our structural and thermodynamic characterizations yield significant insights into AA binding, transport, toxicity, and potential allostery, critical determinants for elucidating the mechanistic roles of HSA in modulating AA carcinogenicity.
Collapse
Affiliation(s)
- Sergei Pomyalov
- Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Conceição A Minetti
- Department of Chemistry and Chemical Biology, Rutgers - The State University of New Jersey, Piscataway, New Jersey, USA
| | - David P Remeta
- Department of Chemistry and Chemical Biology, Rutgers - The State University of New Jersey, Piscataway, New Jersey, USA
| | - Radha Bonala
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York, USA
| | - Francis Johnson
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York, USA; Department of Chemistry, Stony Brook University, Stony Brook, New York, USA
| | - Irina Zaitseva
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York, USA
| | - Charles Iden
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York, USA
| | - Urszula Golebiewska
- Department of Physiology, Stony Brook University, Stony Brook, New York, USA; Department of Biological Sciences, Queensborough Community College, Bayside, New York, USA
| | - Kenneth J Breslauer
- Department of Chemistry and Chemical Biology, Rutgers - The State University of New Jersey, Piscataway, New Jersey, USA; Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA.
| | - Gil Shoham
- Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Viktoriya S Sidorenko
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York, USA.
| | - Arthur P Grollman
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York, USA.
| |
Collapse
|
3
|
Jevtovic V, Golubović L, Alshammari B, Alshammari MR, Rajeh SY, Alreshidi MA, Alshammari OAO, Rakić A, Dimić D. Crystal Structure, Theoretical Analysis, and Protein/DNA Binding Activity of Iron(III) Complex Containing Differently Protonated Pyridoxal- S-Methyl-Isothiosemicarbazone Ligands. Int J Mol Sci 2024; 25:7058. [PMID: 39000166 PMCID: PMC11241004 DOI: 10.3390/ijms25137058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 07/16/2024] Open
Abstract
Pyridoxal-S-methyl-isothiosemicarbazone (PLITSC) is a member of an important group of ligands characterized by different complexation modes to various transition metals. In this contribution, a new complex containing two differently protonated PLITSC ligands ([Fe(PLITSC-H)(PLITSC)]SO4)∙2.5H2O was obtained. The crystal structure was solved by the X-ray analysis and used further for the optimization at B3LYP/6-311++G(d,p)(H,C,N,O,S)/def2-TZVP(Fe) level of theory. Changes in the interaction strength and bond distance due to protonation were observed upon examination by the Quantum Theory of Atoms in Molecules. The protein binding affinity of [Fe(PLITSC-H)(PLITSC)]SO4 towards transport proteins (Bovine Serum Albumin (BSA) and Human Serum Albumin (HSA)) was investigated by the spectrofluorimetric titration and molecular docking. The interactions with the active pocket containing fluorescent amino acids were examined in detail, which explained the fluorescence quenching. The interactions between complex and DNA were followed by the ethidium-bromide displacement titration and molecular docking. The binding along the minor groove was the dominant process involving complex in the proximity of DNA.
Collapse
Affiliation(s)
- Violeta Jevtovic
- Department of Chemistry, College of Science, University Ha'il, Ha'il 81451, Saudi Arabia
| | - Luka Golubović
- Faculty of Physical Chemistry, University of Belgrade, Studentski trg 12-16, 11000 Belgrade, Serbia
| | - Badriah Alshammari
- Department of Chemistry, College of Science, University Ha'il, Ha'il 81451, Saudi Arabia
| | | | - Sahar Y Rajeh
- Department of Chemistry, College of Science, University Ha'il, Ha'il 81451, Saudi Arabia
| | - Maha Awjan Alreshidi
- Department of Chemistry, College of Science, University Ha'il, Ha'il 81451, Saudi Arabia
| | - Odeh A O Alshammari
- Department of Chemistry, College of Science, University Ha'il, Ha'il 81451, Saudi Arabia
| | - Aleksandra Rakić
- Faculty of Physical Chemistry, University of Belgrade, Studentski trg 12-16, 11000 Belgrade, Serbia
| | - Dušan Dimić
- Faculty of Physical Chemistry, University of Belgrade, Studentski trg 12-16, 11000 Belgrade, Serbia
| |
Collapse
|
4
|
Poór M, Dombi Á, Fliszár-Nyúl E, Pedroni L, Dellafiora L. Effects of Chrysin and Chrysin-7-sulfate on Ochratoxin A-Albumin Interactions and on the Plasma and Kidney Levels of the Mycotoxin in Rats. ACS OMEGA 2024; 9:17655-17666. [PMID: 38645364 PMCID: PMC11024961 DOI: 10.1021/acsomega.4c01738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/20/2024] [Accepted: 03/25/2024] [Indexed: 04/23/2024]
Abstract
The nephrotoxic mycotoxin ochratoxin A (OTA) is a common food contaminant. OTA binds to the Sudlow's Site I region of serum albumin with very high affinity, resulting in its slow elimination. The displacement of OTA from albumin may be beneficial due to the faster excretion of the mycotoxin, while it may also lead to the increased tissue uptake of OTA. Furthermore, it is challenging to displace the mycotoxin from albumin even with high-affinity Site I ligands. In this study, we tested the impacts of Site I and Heme site ligands on OTA-albumin interactions by applying fluorescence spectroscopic, ultracentrifugation, and modeling studies. Chrysin-7-sulfate (C7S) strongly displaced OTA from both human and rat albumins; therefore, the impacts of C7S (single intravenous administration) and the parent flavonoid chrysin (repeated peroral treatment) were examined on the plasma and kidney levels of OTA in rats. Chrysin barely influenced the concentrations of mycotoxin in plasma and kidneys. In the first few hours, C7S significantly decreased the plasma levels of OTA compared to the control animals; while after 24 h, only minor differences were noticed. Our study highlights the superior displacing ability of C7S vs OTA regarding human and rat albumins.
Collapse
Affiliation(s)
- Miklós Poór
- Department
of Laboratory Medicine, Medical School, University of Pécs, Ifjúság útja 13, Pécs H-7624, Hungary
- Molecular
Medicine Research Group, János Szentágothai Research
Centre, University of Pécs, Ifjúság útja
20, Pécs H-7624, Hungary
- Department
of Pharmacology, Faculty of Pharmacy, University
of Pécs, Rókus u. 2, Pécs H-7624, Hungary
| | - Ágnes Dombi
- Department
of Pharmacology, Faculty of Pharmacy, University
of Pécs, Rókus u. 2, Pécs H-7624, Hungary
| | - Eszter Fliszár-Nyúl
- Department
of Pharmacology, Faculty of Pharmacy, University
of Pécs, Rókus u. 2, Pécs H-7624, Hungary
| | - Lorenzo Pedroni
- Department
of Food and Drug, University of Parma, Via G.P. Usberti 27/A, Parma 43124, Italy
| | - Luca Dellafiora
- Department
of Food and Drug, University of Parma, Via G.P. Usberti 27/A, Parma 43124, Italy
| |
Collapse
|
5
|
Xu X, Hu J, Xue H, Hu Y, Liu YN, Lin G, Liu L, Xu RA. Applications of human and bovine serum albumins in biomedical engineering: A review. Int J Biol Macromol 2023; 253:126914. [PMID: 37716666 DOI: 10.1016/j.ijbiomac.2023.126914] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/12/2023] [Accepted: 09/13/2023] [Indexed: 09/18/2023]
Abstract
Serum albumin, commonly recognized as a predominant major plasma protein, is ubiquitously distributed among vertebrates, demonstrating versatility and widespread accessibility. Numerous studies have discussed the composition and attributes of human and bovine serum albumin; nonetheless, few systematic and comprehensive summaries on human and bovine serum albumin exist. This paper reviews the applications of human and bovine serum albumin in biomedical engineering. First, we introduce the differences in the structure of human and bovine serum albumin. Next, we describe the extraction methods for human and bovine serum albumin (fractionation process separation, magnetic adsorption, reverse micellar (RM) extraction, and genetic engineering) and the advantages and disadvantages of recently developed extraction methods. The characteristics of different processing forms of human and bovine serum albumin are also discussed, concomitantly elucidating their intrinsic properties, functions, and applications in biomedicine. Notably, their pivotal functions as carriers for drugs and tissue-engineered scaffolds, as well as their contributions to cell reproduction and bioimaging, are critically examined. Finally, to provide guidance for researchers in their future work, this review summarizes the current state of human and bovine serum albumin research and outlines potential future research topics.
Collapse
Affiliation(s)
- Xinhao Xu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China
| | - Jinyu Hu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Huaqian Xue
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China; School of Pharmacy, Ningxia Medical University, Ningxia 750004, China
| | - Yingying Hu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Ya-Nan Liu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Guanyang Lin
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Liangle Liu
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325200, China.
| | - Ren-Ai Xu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
6
|
Wong JYK, Ekanayake AI, Kharchenko S, Kirberger SE, Qiu R, Kelich P, Sarkar S, Li J, Fernandez KX, Alvizo-Paez ER, Miao J, Kalhor-Monfared S, John JD, Kang H, Choi H, Nuss JM, Vederas JC, Lin YS, Macauley MS, Vukovic L, Pomerantz WCK, Derda R. Genetically encoded discovery of perfluoroaryl macrocycles that bind to albumin and exhibit extended circulation in vivo. Nat Commun 2023; 14:5654. [PMID: 37704629 PMCID: PMC10499988 DOI: 10.1038/s41467-023-41427-y] [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: 08/25/2022] [Accepted: 08/17/2023] [Indexed: 09/15/2023] Open
Abstract
Peptide-based therapeutics have gained attention as promising therapeutic modalities, however, their prevalent drawback is poor circulation half-life in vivo. In this paper, we report the selection of albumin-binding macrocyclic peptides from genetically encoded libraries of peptides modified by perfluoroaryl-cysteine SNAr chemistry, with decafluoro-diphenylsulfone (DFS). Testing of the binding of the selected peptides to albumin identified SICRFFC as the lead sequence. We replaced DFS with isosteric pentafluorophenyl sulfide (PFS) and the PFS-SICRFFCGG exhibited KD = 4-6 µM towards human serum albumin. When injected in mice, the concentration of the PFS-SICRFFCGG in plasma was indistinguishable from the reference peptide, SA-21. More importantly, a conjugate of PFS-SICRFFCGG and peptide apelin-17 analogue (N3-PEG6-NMe17A2) showed retention in circulation similar to SA-21; in contrast, apelin-17 analogue was cleared from the circulation after 2 min. The PFS-SICRFFC is the smallest known peptide macrocycle with a significant affinity for human albumin and substantial in vivo circulation half-life. It is a productive starting point for future development of compact macrocycles with extended half-life in vivo.
Collapse
Affiliation(s)
- Jeffrey Y K Wong
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2G2, Canada
| | - Arunika I Ekanayake
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2G2, Canada
| | - Serhii Kharchenko
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2G2, Canada
| | - Steven E Kirberger
- Department of Chemistry, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Ryan Qiu
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2G2, Canada
| | - Payam Kelich
- Department of Chemistry and Biochemistry, University of Texas at El Paso, El Paso, TX, 79968, USA
| | - Susmita Sarkar
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2G2, Canada
| | - Jiaqian Li
- Department of Chemistry, University of Minnesota, Minneapolis, MN, 55455, USA
| | | | - Edgar R Alvizo-Paez
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2G2, Canada
| | - Jiayuan Miao
- Department of Chemistry, Tufts University, Medford, MA, 02155, USA
| | | | - J Dwyer John
- Ferring Research Institute, San Diego, CA, 92121, USA
| | - Hongsuk Kang
- Quantum Intelligence Corp., 31F, One IFC, 10 Gukjegeumyung-ro, Yeongdeungpo-gu-Seoul, Republic of Korea
| | - Hwanho Choi
- Quantum Intelligence Corp., 31F, One IFC, 10 Gukjegeumyung-ro, Yeongdeungpo-gu-Seoul, Republic of Korea
| | - John M Nuss
- Ferring Research Institute, San Diego, CA, 92121, USA
| | - John C Vederas
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2G2, Canada
| | - Yu-Shan Lin
- Department of Chemistry, Tufts University, Medford, MA, 02155, USA
| | - Matthew S Macauley
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2G2, Canada
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Lela Vukovic
- Department of Chemistry and Biochemistry, University of Texas at El Paso, El Paso, TX, 79968, USA
| | | | - Ratmir Derda
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2G2, Canada.
| |
Collapse
|
7
|
Structural Investigation of Diclofenac Binding to Ovine, Caprine, and Leporine Serum Albumins. Int J Mol Sci 2023; 24:ijms24021534. [PMID: 36675044 PMCID: PMC9864019 DOI: 10.3390/ijms24021534] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/05/2023] [Accepted: 01/08/2023] [Indexed: 01/15/2023] Open
Abstract
Free drug concentration in the blood sera is crucial for its appropriate activity. Serum albumin, the universal blood carrier protein, is responsible for transporting drugs and releasing them into the bloodstream. Therefore, a drug's binding to SA is especially important for its bioavailability and it is a key problem in the drug design process. In this paper, we present crystal structures of three animal serum albumin complexes: ovine, caprine, and leporine, with diclofenac, a popular non-steroidal anti-inflammatory drug that is used in therapy of chronic and acute pain. Details of diclofenac binding mode by the presented serum albumins are compared with analogous complexes of human and equine serum albumins. The analysis of the occupied binding pockets in crystal structures of the investigated serum albumins from different mammals shows that they have two common and a number of unique diclofenac binding sites. The most intriguing is the fact that the albumins from the described species are able to bind different numbers of molecules of this popular anti-inflammatory drug, but none of the binding sites overlap with ones in the human serum albumin.
Collapse
|
8
|
Effects of Albumin Supplements on In-Hospital Mortality in Patients with Sepsis or Septic Shock: A Systemic Review and Meta-Analysis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:2384730. [PMID: 36262167 PMCID: PMC9576387 DOI: 10.1155/2022/2384730] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/22/2022] [Indexed: 11/05/2022]
Abstract
Objective To explore the clinical effects of albumin supplements on the basis of crystalloid solution in patients with sepsis or septic shock. Methods The online databases including PubMed, Web of Science, Cochrane Library, and EMBASE were comprehensively searched from inception to June 28, 2021, with the keywords including “albumin,” “sepsis,” or “septic shock.” Retrospective cohort (RC) and randomized controlled trials (RCT) were included for analysis. Two authors independently searched and analyzed the literature. The in-hospital mortality at 7 days and 28 days, duration of mechanical ventilation, renal replacement therapy, length of ICU stay, and length of hospital stay were compared between patients with albumin supplements and crystalloid solution and those with crystalloid alone. Results A total of 10 studies with 6463 patients were eventually included for meta-analysis. The in-hospital mortality of patients at 7 days (OR = 1.00, 95% CI: 0.81–1.23) and 28 days (OR = 1.02, 95% CI: 0.91–1.13) did not show a significant difference between the two groups of patients. Also, the pooled results demonstrated no significant differences in duration of mechanical ventilation (OR = 0.29, 95% CI: −0.05–0.63), renal replacement therapy (WMD = 1.15, 95% CI: 0.98–1.35), length of ICU stay (WMD = −0.07, 95% CI: −0.62–0.48), and length of hospital stay (WMD = −0.09, 95% CI: −0.70–0.52) between patients receiving albumin plus crystalloid solution and those with crystalloid solution alone. Conclusion Albumin supplements on the basis of crystalloid solution did not improve the 7-day and 28-dayin-hospital mortality in patients with sepsis or septic shock compared with those with crystalloid solution alone.
Collapse
|
9
|
NMR Investigation of the Interaction of Three Non-Steroidal Anti-Inflammatory Drugs with Human Serum Albumin. Molecules 2022; 27:molecules27196647. [PMID: 36235184 PMCID: PMC9571845 DOI: 10.3390/molecules27196647] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/30/2022] [Accepted: 10/04/2022] [Indexed: 11/07/2022] Open
Abstract
The understanding of the interaction between non-steroidal anti-inflammatory drugs and human serum albumin plays a fundamental role in the development of new drugs and new therapeutic strategies. Several studies have been performed, nevertheless, the interaction phenomena are still not fully understood. In this work, high-field solution Nuclear Magnetic Resonance (NMR) spectroscopy was applied to compare the strength of the interaction of diclofenac sodium salt, ketorolac tris salt and flurbiprofen sodium salt toward albumin. To this aim, mono- and bi-selective relaxation rate measurements were performed by applying selective π-pulses at the selected frequencies and by following magnetization recovery. On the basis of the dependence of relaxation parameters on albumin concentration, normalized affinity indexes were calculated for several protons of the drugs. Affinity indexes for diclofenac were about five-fold higher in comparison with ketorolac and flurbiprofen. Aromatic moieties of the three drugs and methine protons at the chiral centers of ketorolac and flurbiprofen were more involved in the interaction with albumin. In conclusion, NMR spectroscopy allows not only for the comparison of drug-to-protein affinities but also points out the nature of the drug sites that are more extensively involved in the interaction.
Collapse
|
10
|
Assessment of the Effects of Triticonazole on Soil and Human Health. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27196554. [PMID: 36235091 PMCID: PMC9572687 DOI: 10.3390/molecules27196554] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/19/2022] [Accepted: 09/26/2022] [Indexed: 11/28/2022]
Abstract
Triticonazole is a fungicide used to control diseases in numerous plants. The commercial product is a racemate containing (R)- and (S)-triticonazole and its residues have been found in vegetables, fruits, and drinking water. This study considered the effects of triticonazole on soil microorganisms and enzymes and human health by taking into account the enantiomeric structure when applicable. An experimental method was applied for assessing the effects of triticonazole on soil microorganisms and enzymes, and the effects of the stereoisomers on soil enzymes and human health were assessed using a computational approach. There were decreases in dehydrogenase and phosphatase activities and an increase in urease activity when barley and wheat seeds treated with various doses of triticonazole were sown in chernozem soil. At least 21 days were necessary for the enzymes to recover the activities. This was consistent with the diminution of the total number of soil microorganisms in the 14 days after sowing. Both stereoisomers were able to bind to human plasma proteins and were potentially inhibitors of human cytochromes, revealing cardiotoxicity and low endocrine disruption potential. As distinct effects, (R)-TTZ caused skin sensitization, carcinogenicity, and respiratory toxicity. There were no significant differences in the interaction energies of the stereoisomers and soil enzymes, but (S)-TTZ exposed higher interaction energies with plasma proteins and human cytochromes.
Collapse
|
11
|
França VLB, Amaral JL, Martins YA, Caetano EWS, Brunaldi K, Freire VN. Characterization of the binding interaction between atrazine and human serum albumin: Fluorescence spectroscopy, molecular dynamics and quantum biochemistry. Chem Biol Interact 2022; 366:110130. [PMID: 36037875 DOI: 10.1016/j.cbi.2022.110130] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/18/2022] [Accepted: 08/20/2022] [Indexed: 11/03/2022]
Abstract
Atrazine (ATR), one of the most used herbicides worldwide, causes persistent contamination of water and soil due to its high resistance to degradation. ATR is associated with low fertility and increased risk of prostate cancer in humans, as well as birth defects, low birth weight and premature delivery. Describing ATR binding to human serum albumin (HSA) is clinically relevant to future studies about pharmacokinetics, pharmacodynamics and toxicity of ATR, as albumin is the most abundant carrier protein in plasma and binds important small biological molecules. In this work we characterize, for the first time, the binding of ATR to HSA by using fluorescence spectroscopy and performing simulations using molecular docking, classical molecular dynamics and quantum biochemistry based on density functional theory (DFT). We determine the most likely binding sites of ATR to HSA, highlighting the fatty acid binding site FA8 (located between subdomains IA-IB-IIA and IIB-IIIA-IIIB) as the most important one, and evaluate each nearby amino acid residue contribution to the binding interactions explaining the fluorescence quenching due to ATR complexation with HSA. The stabilization of the ATR/FA8 complex was also aided by the interaction between the atrazine ring and SER454 (hydrogen bond) and LEU481(alkyl interaction).
Collapse
Affiliation(s)
- Victor L B França
- Departament of Physics, Federal University of Ceará, Fortaleza, 60440-900, Brazil
| | - Jackson L Amaral
- Departament of Physics, Federal University of Ceará, Fortaleza, 60440-900, Brazil
| | - Yandara A Martins
- Departament of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, 05508-000, Brazil
| | - Ewerton W S Caetano
- Federal Institute of Education, Science and Technology of Ceará, Fortaleza, 60040-531, Brazil
| | - Kellen Brunaldi
- Departament of Physiological Sciences, State University of Maringá, Maringá, 87020-900, Brazil.
| | - Valder N Freire
- Departament of Physics, Federal University of Ceará, Fortaleza, 60440-900, Brazil.
| |
Collapse
|
12
|
Duan X, Zhang R, Zhang X, Ding X, Sun T. Identification of Prognostic Factors in Patients With Streptococcus Bloodstream Infection. Front Med (Lausanne) 2022; 9:832007. [PMID: 35559342 PMCID: PMC9087728 DOI: 10.3389/fmed.2022.832007] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/25/2022] [Indexed: 11/14/2022] Open
Abstract
Aim The purpose of this study was to explore prognostic factors of bloodstream infections (BSIs), a common severe infection and a major cause of mortality worldwide, so as to construct a prognosis model of patients with BSI. Materials and Methods Clinical and biochemical test data were obtained retrospectively from the medical records of 562 patients with BSI who had been treated at a single center; the end point was 60 days of all-cause death. The chi-square test was used to compare the mortality of patients grouped by the types of antibiotic treatment. The logistic regression analysis was adopted to identify prognostic factors; the Kaplan–Meier survival curve and log-rank test were conducted to compare the survival rate of patients with different prognostic factors; the receiver operating characteristic (ROC) curve was used to estimate the predictive value of different prognostic factors. Results Of the 562 patients, 455 survived (80.96%), and 107 died (19.04%). The mortality rate of patients treated with a combination of antibiotics (25.40%) was higher than that treated with a single antibiotic (15.82%). Univariate analysis identified 19 prognostic factors for patients with BSI, including gender, age, diabetes, malignant tumor (non-blood system), total hospitalization time, alanine aminotransferase, aspartate aminotransferase, total protein, albumin, total bilirubin, direct bilirubin, creatinine, ratio of granulocytes, fibrinogen, D-dimer, platelet, C-reactive protein, shock, and respiratory failure (P < 0.05). Multivariate analysis indicated that albumin (odds ratio [OR] = 0.94, 95% confidence interval [CI]: 0.89–0.99), fibrinogen (OR = 0.61, 95%CI: 0.46–0.82), shock (OR = 16.61, 95%CI: 7.00–39.41), and respiratory failure (OR = 47.53, 95%CI: 19.93–133.64) were independent factors. The combination of four indicators demonstrated a favorable predictive value for the 60-day outcome of patients with BSI, with an area under the ROC of 0.96 (95%CI: 0.94–0.99), sensitivity of 90.65%, specificity of 94.95%, and accuracy of 94.13%. Conclusions Shock, respiratory failure, albumin, and fibrinogen are potential independent prognostic factors for 60-day mortality.
Collapse
Affiliation(s)
- Xiaoguang Duan
- General ICU, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ruifang Zhang
- General ICU, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaojuan Zhang
- General ICU, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xianfei Ding
- General ICU, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tongwen Sun
- General ICU, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
13
|
Yang J, Duan A, Wang D, Yang X, Liu X, Yang G, Yang Q. Evaluating the effect of diclofenac on hydrogen production by anaerobic fermentation of waste activated sludge. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2022; 308:114641. [PMID: 35131705 DOI: 10.1016/j.jenvman.2022.114641] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 01/22/2022] [Accepted: 01/29/2022] [Indexed: 06/14/2023]
Abstract
Hydrogen production from waste-activated sludge (WAS) anaerobic fermentation is considered to be an effective method of resource recovery. However, the presence of a large number of complex organic compounds in sludge will affect the biological hydrogen production process. As an extensively applied prevalent anti-inflammatory drug, diclofenac (DCF) is inevitably released into the environment. However, the effect of diclofenac on hydrogen production from WAS anaerobic fermentation has not been fully investigated. This work therefore aims to comprehensively investigate the removal efficiency of DCF in mesophilic anaerobic fermentation of WAS and its effect on hydrogen yield. Experiment results showed that 32.5%-38.3% of DCF was degraded in the fermentation process when DCF concentration was ranged from 6 to 100 mg/kg TSS (total suspended solids). DCF at environmental level inhibited hydrogen production, the maximal hydrogen yield decreased from 24.2 to 15.3 mL/g VSS (volatile suspended solids) with an increase of DCF addition from 6 to 100 mg/kg TSS. This is because the presence of DCF caused inhibitions to acetogenesis and acidogenesis, the processes responsible for hydrogen production, probably due to that the polar groups of DCF (i.e., carboxyl group) could readily bind to active sites of [FeFe]- Hydrogenase. Besides, the microbial analysis revealed that DCF increased the microbial diversity but had few influences on the microbial structure.
Collapse
Affiliation(s)
- Jingnan Yang
- College of Environmental Science and Engineering, Hunan University, Changsha, 410082, PR China; Key Laboratory of Environmental Biology and Pollution Control (Hunan University), Ministry of Education, Changsha, 410082, PR China
| | - Abing Duan
- College of Environmental Science and Engineering, Hunan University, Changsha, 410082, PR China; Key Laboratory of Environmental Biology and Pollution Control (Hunan University), Ministry of Education, Changsha, 410082, PR China
| | - Dongbo Wang
- College of Environmental Science and Engineering, Hunan University, Changsha, 410082, PR China; Key Laboratory of Environmental Biology and Pollution Control (Hunan University), Ministry of Education, Changsha, 410082, PR China.
| | - Xianli Yang
- College of Environmental Science and Engineering, Hunan University, Changsha, 410082, PR China; Key Laboratory of Environmental Biology and Pollution Control (Hunan University), Ministry of Education, Changsha, 410082, PR China
| | - Xuran Liu
- College of Environmental Science and Engineering, Hunan University, Changsha, 410082, PR China; Key Laboratory of Environmental Biology and Pollution Control (Hunan University), Ministry of Education, Changsha, 410082, PR China
| | - Guojing Yang
- College of Biological and Environmental Sciences, Zhejiang Wanli University, Ningbo, 315100, PR China
| | - Qi Yang
- College of Environmental Science and Engineering, Hunan University, Changsha, 410082, PR China; Key Laboratory of Environmental Biology and Pollution Control (Hunan University), Ministry of Education, Changsha, 410082, PR China
| |
Collapse
|
14
|
Visentini FF, Perez AA, Santiago LG. Bioactive compounds: Application of albumin nanocarriers as delivery systems. Crit Rev Food Sci Nutr 2022; 63:7238-7268. [PMID: 35238254 DOI: 10.1080/10408398.2022.2045471] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Enriched products with bioactive compounds (BCs) show the capacity to produce a wide range of possible health effects. Most BCs are essentially hydrophobic and sensitive to environmental factors; so, encapsulation becomes a strategy to solve these problems. Many globular proteins have the intrinsic ability to bind, protect, encapsulate, and introduce BCs into nutraceutical or pharmaceutical matrices. Among them, albumins as human serum albumin (HSA), bovine serum albumin (BSA), ovalbumin (OVA) and α-lactalbumin (ALA) are widely abundant, available, and applied in many industrial sectors, becoming promissory materials to encapsulate BCs. Therefore, this review focuses on researches about the main groups of natural origin BCs (namely phenolic compounds, lipids, vitamins, and carotenoids), the different types of nanostructures based on albumins to encapsulate them and the main fields of application for BCs-loaded albumin systems. In this context, phenolic compounds (catechins, quercetin, and chrysin) are the most extensively BCs studied and encapsulated in albumin-based nanocarriers. Other extensively studied subgroups are stilbenes and curcuminoids. Regarding lipids and vitamins; terpenes, carotenoids (β-carotene), and xanthophylls (astaxanthin) are the most considered. The main application areas of BCs are related to their antitumor, anti-inflammatory, and antioxidant properties. Finally, BSA is the most used albumin to produced BCs-loaded nanocarriers.
Collapse
Affiliation(s)
- Flavia F Visentini
- Consejo Nacional de Investigaciones Científicas y Técnicas de la República Argentina, CONICET
- Área de Biocoloides y Nanotecnología, Instituto de Tecnología de Alimentos, Facultad de Ingeniería Química, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Adrián A Perez
- Consejo Nacional de Investigaciones Científicas y Técnicas de la República Argentina, CONICET
- Área de Biocoloides y Nanotecnología, Instituto de Tecnología de Alimentos, Facultad de Ingeniería Química, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Liliana G Santiago
- Área de Biocoloides y Nanotecnología, Instituto de Tecnología de Alimentos, Facultad de Ingeniería Química, Universidad Nacional del Litoral, Santa Fe, Argentina
| |
Collapse
|
15
|
Andrade-Villalobos F, Zúñiga-Núñez D, Fuentealba D, Fierro A. Binding of toluidine blue-myristic acid derivative to cucurbit[7]uril and human serum albumin: computational and biophysical insights towards a biosupramolecular assembly. Phys Chem Chem Phys 2022; 24:3222-3230. [PMID: 35044390 DOI: 10.1039/d1cp04307b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A new toluidine blue-myristic acid photosensitizer derivate (TBOMyr) was investigated as a design molecule to bind simultaneously to cucurbit[7]uril (CB[7]) and human serum albumin (HSA) with the aim of constructing a biosupramolecular assembly. Molecular docking and dynamics calculations revealed the main supramolecular and bio-molecular interactions of TBOMyr with the macrocycle or the protein, respectively. The addition of the negatively charged myristic acid-like tail resulted in a unique conformation of the CB[7] complex where the phenothiazine core was included in the cavity of CB[7], leaving the fatty acid portion free to interact with the protein. A favorable ternary interaction between TBOMyr, CB[7] and HSA was suggested by the calculations, and an experimental binding affinity in the order of 105 M-1 was determined for the TBOMyr@CB[7] complex with HSA. The new TBOMyr derivative could find applications in photodynamic therapy benefiting from the biosupramolecular interactions as a transport system.
Collapse
Affiliation(s)
- Felipe Andrade-Villalobos
- Departamento de Química Orgánica, Escuela de Química, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Macul, Santiago, Chile. .,Laboratorio de Química Supramolecular y Fotobiología, Departamento de Química Física, Escuela de Química, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Macul, Santiago, Chile.
| | - Daniel Zúñiga-Núñez
- Laboratorio de Química Supramolecular y Fotobiología, Departamento de Química Física, Escuela de Química, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Macul, Santiago, Chile.
| | - Denis Fuentealba
- Laboratorio de Química Supramolecular y Fotobiología, Departamento de Química Física, Escuela de Química, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Macul, Santiago, Chile.
| | - Angelica Fierro
- Departamento de Química Orgánica, Escuela de Química, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Macul, Santiago, Chile.
| |
Collapse
|
16
|
Tikhonov D, Kulikova L, Kopylov AT, Rudnev V, Stepanov A, Malsagova K, Izotov A, Kulikov D, Zulkarnaev A, Enikeev D, Potoldykova N, Kaysheva AL. Proteomic and molecular dynamic investigations of PTM-induced structural fluctuations in breast and ovarian cancer. Sci Rep 2021; 11:19318. [PMID: 34588485 PMCID: PMC8481388 DOI: 10.1038/s41598-021-98201-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 09/06/2021] [Indexed: 11/16/2022] Open
Abstract
Post-translational processing leads to conformational changes in protein structure that modulate molecular functions and change the signature of metabolic transformations and immune responses. Some post-translational modifications (PTMs), such as phosphorylation and acetylation, are strongly related to oncogenic processes and malignancy. This study investigated a PTM pattern in patients with gender-specific ovarian or breast cancer. Proteomic profiling and analysis of cancer-specific PTM patterns were performed using high-resolution UPLC-MS/MS. Structural analysis, topology, and stability of PTMs associated with sex-specific cancers were analyzed using molecular dynamics modeling. We identified highly specific PTMs, of which 12 modified peptides from eight distinct proteins derived from patients with ovarian cancer and 6 peptides of three proteins favored patients from the group with breast cancer. We found that all defined PTMs were localized in the compact and stable structural motifs exposed outside the solvent environment. PTMs increase the solvent-accessible surface area of the modified moiety and its active environment. The observed conformational fluctuations are still inadequate to activate the structural degradation and enhance protein elimination/clearance; however, it is sufficient for the significant modulation of protein activity.
Collapse
Affiliation(s)
- Dmitry Tikhonov
- Institute of Mathematical Problems of Biology RAS-the Branch of Keldysh Institute of Applied Mathematics of Russian Academy of Sciences, 142290, Pushchino, Moscow Region, Russia.,Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290, Pushchino, Moscow Region, Russia
| | - Liudmila Kulikova
- Institute of Mathematical Problems of Biology RAS-the Branch of Keldysh Institute of Applied Mathematics of Russian Academy of Sciences, 142290, Pushchino, Moscow Region, Russia.,Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290, Pushchino, Moscow Region, Russia
| | - Arthur T Kopylov
- V.N. Orekhovich Institute of Biomedical Chemistry, 119121, Moscow, Russia.
| | - Vladimir Rudnev
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290, Pushchino, Moscow Region, Russia.,V.N. Orekhovich Institute of Biomedical Chemistry, 119121, Moscow, Russia
| | - Alexander Stepanov
- V.N. Orekhovich Institute of Biomedical Chemistry, 119121, Moscow, Russia
| | - Kristina Malsagova
- V.N. Orekhovich Institute of Biomedical Chemistry, 119121, Moscow, Russia
| | - Alexander Izotov
- V.N. Orekhovich Institute of Biomedical Chemistry, 119121, Moscow, Russia
| | - Dmitry Kulikov
- Moscow Regional Research and Clinical Institute, Russian Federation, 129110, Moscow, Russia
| | - Alexey Zulkarnaev
- Moscow Regional Research and Clinical Institute, Russian Federation, 129110, Moscow, Russia
| | - Dmitry Enikeev
- Institute of Urology and Reproductive Health, Sechenov University, 119121, Moscow, Russia
| | - Natalia Potoldykova
- Institute of Urology and Reproductive Health, Sechenov University, 119121, Moscow, Russia
| | - Anna L Kaysheva
- V.N. Orekhovich Institute of Biomedical Chemistry, 119121, Moscow, Russia
| |
Collapse
|
17
|
Pannecoucke E, Van Trimpont M, Desmet J, Pieters T, Reunes L, Demoen L, Vuylsteke M, Loverix S, Vandenbroucke K, Alard P, Henderikx P, Deroo S, Baatz F, Lorent E, Thiolloy S, Somers K, McGrath Y, Van Vlierberghe P, Lasters I, Savvides SN. Cell-penetrating Alphabody protein scaffolds for intracellular drug targeting. SCIENCE ADVANCES 2021; 7:7/13/eabe1682. [PMID: 33771865 PMCID: PMC7997521 DOI: 10.1126/sciadv.abe1682] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 02/05/2021] [Indexed: 05/02/2023]
Abstract
The therapeutic scope of antibody and nonantibody protein scaffolds is still prohibitively limited against intracellular drug targets. Here, we demonstrate that the Alphabody scaffold can be engineered into a cell-penetrating protein antagonist against induced myeloid leukemia cell differentiation protein MCL-1, an intracellular target in cancer, by grafting the critical B-cell lymphoma 2 homology 3 helix of MCL-1 onto the Alphabody and tagging the scaffold's termini with designed cell-penetration polypeptides. Introduction of an albumin-binding moiety extended the serum half-life of the engineered Alphabody to therapeutically relevant levels, and administration thereof in mouse tumor xenografts based on myeloma cell lines reduced tumor burden. Crystal structures of such a designed Alphabody in complex with MCL-1 and serum albumin provided the structural blueprint of the applied design principles. Collectively, we provide proof of concept for the use of Alphabodies against intracellular disease mediators, which, to date, have remained in the realm of small-molecule therapeutics.
Collapse
Affiliation(s)
- Erwin Pannecoucke
- VIB Center for Inflammation Research, 9052 Ghent, Belgium
- Unit for Structural Biology, Department of Biochemistry and Microbiology, Ghent University, 9052 Ghent, Belgium
| | - Maaike Van Trimpont
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | | | - Tim Pieters
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Lindy Reunes
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Lisa Demoen
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | - Pieter Van Vlierberghe
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | | | - Savvas N Savvides
- VIB Center for Inflammation Research, 9052 Ghent, Belgium.
- Unit for Structural Biology, Department of Biochemistry and Microbiology, Ghent University, 9052 Ghent, Belgium
| |
Collapse
|
18
|
Computational Assessment of Chito-Oligosaccharides Interactions with Plasma Proteins. Mar Drugs 2021; 19:md19030120. [PMID: 33668290 PMCID: PMC7996291 DOI: 10.3390/md19030120] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 02/20/2021] [Accepted: 02/22/2021] [Indexed: 11/17/2022] Open
Abstract
It is widely rec ognized that chitin and chitosan are potential sources of bioactive materials and that their oligosaccharides reveal various biological activities (including antimicrobial) that are correlated with their structures and physicochemical properties. This study uses the molecular docking approach to assess the interactions of small chito-oligosaccharides (MW< 1500 Da) with plasma proteins in order to obtain information regarding their fate of distribution in the human organism. There are favorable interactions of small chito-oligomers with plasma proteins, the interactions with human serum albumin being stronger than those with α-1-acid glycoprotein. The interaction energies increase with increasing the molecular weight, decrease with increasing deacetylation degrees and are reliant on the deacetylation pattern. This study could inform the application of chito-oligosaccharides with varying molecular weights, degrees, and patterns of deacetylation in human health.
Collapse
|
19
|
Nevídalová H, Michalcová L, Glatz Z. Applicability of capillary electrophoresis-frontal analysis for displacement studies: Effect of several drugs on l-tryptophan and lidocaine binding to human serum albumin. J Sep Sci 2020; 43:4225-4233. [PMID: 32966669 DOI: 10.1002/jssc.202000594] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 08/27/2020] [Accepted: 09/17/2020] [Indexed: 01/30/2023]
Abstract
The effective concentration of a drug in the blood, i.e. the concentration of a free drug in the blood, is influenced by the strength of drug binding onto plasma proteins. Besides its efficacy, these interactions subsequently influence the liberation, absorption, distribution, metabolism, excretion, and toxicological properties of the drug. It is important to not only determine the binding strength and stoichiometry, but also the binding site of a drug on the plasma protein molecule, because the co-administration of drugs with the same binding site can affect the above-mentioned concentration and as a result the pharmacological behavior of the drugs and lead to side effects caused by the change in free drug concentration, its toxicity. In this study, the binding characteristics of six drugs with human serum albumin, the most abundant protein in human plasma, were determined by capillary electrophoresis-frontal analysis, and the obtained values of binding parameters were compared with the literature data. The effect of several drugs and site markers on the binding of l-tryptophan and lidocaine to human serum albumin was investigated in subsequent displacement studies which thus demonstrated the usability of capillary electrophoresis as an automated high-throughput screening method for drug-protein binding studies.
Collapse
Affiliation(s)
- Hana Nevídalová
- Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Lenka Michalcová
- Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Zdeněk Glatz
- Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czech Republic
| |
Collapse
|
20
|
Shabalin IG, Czub MP, Majorek KA, Brzezinski D, Grabowski M, Cooper DR, Panasiuk M, Chruszcz M, Minor W. Molecular determinants of vascular transport of dexamethasone in COVID-19 therapy. IUCRJ 2020; 7:S2052252520012944. [PMID: 33063792 PMCID: PMC7553145 DOI: 10.1107/s2052252520012944] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 09/22/2020] [Indexed: 05/06/2023]
Abstract
Dexamethasone, a widely used corticosteroid, has recently been reported as the first drug to increase the survival chances of patients with severe COVID-19. Therapeutic agents, including dexamethasone, are mostly transported through the body by binding to serum albumin. Here, the first structure of serum albumin in complex with dexamethasone is reported. Dexamethasone binds to drug site 7, which is also the binding site for commonly used nonsteroidal anti-inflammatory drugs and testosterone, suggesting potentially problematic binding competition. This study bridges structural findings with an analysis of publicly available clinical data from Wuhan and suggests that an adjustment of the dexamethasone regimen should be further investigated as a strategy for patients affected by two major COVID-19 risk factors: low albumin levels and diabetes.
Collapse
Affiliation(s)
- Ivan G. Shabalin
- Department of Molecular Physiology and Biological Physics, University of Virginia, 1340 Jefferson Park Avenue, Charlottesville, VA 22908, USA
| | - Mateusz P. Czub
- Department of Molecular Physiology and Biological Physics, University of Virginia, 1340 Jefferson Park Avenue, Charlottesville, VA 22908, USA
| | - Karolina A. Majorek
- Department of Molecular Physiology and Biological Physics, University of Virginia, 1340 Jefferson Park Avenue, Charlottesville, VA 22908, USA
| | - Dariusz Brzezinski
- Department of Molecular Physiology and Biological Physics, University of Virginia, 1340 Jefferson Park Avenue, Charlottesville, VA 22908, USA
- Center for Biocrystallographic Research, Institute of Bioorganic Chemistry, Polish Academy of Sciences, 61-704 Poznan, Poland
- Institute of Computing Science, Poznan University of Technology, 60-965 Poznan, Poland
| | - Marek Grabowski
- Department of Molecular Physiology and Biological Physics, University of Virginia, 1340 Jefferson Park Avenue, Charlottesville, VA 22908, USA
| | - David R. Cooper
- Department of Molecular Physiology and Biological Physics, University of Virginia, 1340 Jefferson Park Avenue, Charlottesville, VA 22908, USA
| | - Mateusz Panasiuk
- Department of Clinical Medicine, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Maksymilian Chruszcz
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208, USA
| | - Wladek Minor
- Department of Molecular Physiology and Biological Physics, University of Virginia, 1340 Jefferson Park Avenue, Charlottesville, VA 22908, USA
| |
Collapse
|
21
|
Leboffe L, di Masi A, Polticelli F, Trezza V, Ascenzi P. Structural Basis of Drug Recognition by Human Serum Albumin. Curr Med Chem 2020; 27:4907-4931. [DOI: 10.2174/0929867326666190320105316] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 02/12/2019] [Accepted: 03/06/2019] [Indexed: 12/18/2022]
Abstract
Background:
Human serum albumin (HSA), the most abundant protein in plasma,
is a monomeric multi-domain macromolecule with at least nine binding sites for endogenous
and exogenous ligands. HSA displays an extraordinary ligand binding capacity as a depot and
carrier for many compounds including most acidic drugs. Consequently, HSA has the potential
to influence the pharmacokinetics and pharmacodynamics of drugs.
Objective:
In this review, the structural determinants of drug binding to the multiple sites of
HSA are analyzed and discussed in detail. Moreover, insight into the allosteric and competitive
mechanisms underpinning drug recognition, delivery, and efficacy are analyzed and discussed.
Conclusion:
As several factors can modulate drug binding to HSA (e.g., concurrent administration
of drugs competing for the same binding site, ligand binding to allosteric-coupled
clefts, genetic inherited diseases, and post-translational modifications), ligand binding to HSA
is relevant not only under physiological conditions, but also in the pharmacological therapy
management.
Collapse
Affiliation(s)
- Loris Leboffe
- Department of Sciences, University Roma Tre, Viale Guglielmo Marconi 446, I-00146 Roma, Italy
| | - Alessandra di Masi
- Department of Sciences, University Roma Tre, Viale Guglielmo Marconi 446, I-00146 Roma, Italy
| | - Fabio Polticelli
- Department of Sciences, University Roma Tre, Viale Guglielmo Marconi 446, I-00146 Roma, Italy
| | - Viviana Trezza
- Department of Sciences, University Roma Tre, Viale Guglielmo Marconi 446, I-00146 Roma, Italy
| | - Paolo Ascenzi
- Interdepartmental Laboratory for Electron Microscopy, Roma Tre University, Via della Vasca Navale 79, I- 00146 Roma, Italy
| |
Collapse
|
22
|
Czub MP, Handing KB, Venkataramany BS, Cooper DR, Shabalin IG, Minor W. Albumin-Based Transport of Nonsteroidal Anti-Inflammatory Drugs in Mammalian Blood Plasma. J Med Chem 2020; 63:6847-6862. [PMID: 32469516 DOI: 10.1021/acs.jmedchem.0c00225] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Every day, hundreds of millions of people worldwide take nonsteroidal anti-inflammatory drugs (NSAIDs), often in conjunction with multiple other medications. In the bloodstream, NSAIDs are mostly bound to serum albumin (SA). We report the crystal structures of equine serum albumin complexed with four NSAIDs (ibuprofen, ketoprofen, etodolac, and nabumetone) and the active metabolite of nabumetone (6-methoxy-2-naphthylacetic acid, 6-MNA). These compounds bind to seven drug-binding sites on SA. These sites are generally well-conserved between equine and human SAs, but ibuprofen binds to both SAs in two drug-binding sites, only one of which is common. We also compare the binding of ketoprofen by equine SA to binding of it by bovine and leporine SAs. Our comparative analysis of known SA complexes with FDA-approved drugs clearly shows that multiple medications compete for the same binding sites, indicating possibilities for undesirable physiological effects caused by drug-drug displacement or competition with common metabolites. We discuss the consequences of NSAID binding to SA in a broader scientific and medical context, particularly regarding achieving desired therapeutic effects based on an individual's drug regimen.
Collapse
Affiliation(s)
- Mateusz P Czub
- Department of Molecular Physiology and Biological Physics, University of Virginia, 1340 Jefferson Park Avenue, Charlottesville, Virginia 22908, United States.,Center for Structural Genomics of Infectious Diseases (CSGID), University of Virginia, 1340 Jefferson Park Avenue, Charlottesville, Virginia 22908, United States
| | - Katarzyna B Handing
- Department of Molecular Physiology and Biological Physics, University of Virginia, 1340 Jefferson Park Avenue, Charlottesville, Virginia 22908, United States
| | - Barat S Venkataramany
- Department of Molecular Physiology and Biological Physics, University of Virginia, 1340 Jefferson Park Avenue, Charlottesville, Virginia 22908, United States
| | - David R Cooper
- Department of Molecular Physiology and Biological Physics, University of Virginia, 1340 Jefferson Park Avenue, Charlottesville, Virginia 22908, United States.,Center for Structural Genomics of Infectious Diseases (CSGID), University of Virginia, 1340 Jefferson Park Avenue, Charlottesville, Virginia 22908, United States
| | - Ivan G Shabalin
- Department of Molecular Physiology and Biological Physics, University of Virginia, 1340 Jefferson Park Avenue, Charlottesville, Virginia 22908, United States.,Center for Structural Genomics of Infectious Diseases (CSGID), University of Virginia, 1340 Jefferson Park Avenue, Charlottesville, Virginia 22908, United States
| | - Wladek Minor
- Department of Molecular Physiology and Biological Physics, University of Virginia, 1340 Jefferson Park Avenue, Charlottesville, Virginia 22908, United States.,Center for Structural Genomics of Infectious Diseases (CSGID), University of Virginia, 1340 Jefferson Park Avenue, Charlottesville, Virginia 22908, United States
| |
Collapse
|
23
|
Dargó G, Bajusz D, Simon K, Müller J, Balogh GT. Human Serum Albumin Binding in a Vial: A Novel UV-pH Titration Method To Assist Drug Design. J Med Chem 2020; 63:1763-1774. [PMID: 31995375 PMCID: PMC7307925 DOI: 10.1021/acs.jmedchem.0c00046] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
![]()
The knowledge on human serum albumin
(HSA) binding is of utmost
importance as it affects pharmacokinetic behavior and bioavailability
of drugs. In this article, we report a novel method to screen for
ionizable molecules with high HSA binding affinity based on pKa shifts using UV-pH titration. We investigated
the HSA binding of 27 drugs and compared the results to experimental
data from conventional methods. In most cases, significant shifts
(ΔpKa > 0.1) were observed for
drugs
with high HSA binding, while no change could be detected for low-affinity
binders. We showed the pivotal role of ionization centers in the formation
of strong interactions between drug and HSA using molecular docking
studies. We also verified our findings by testing five modified analogues
designed by structural considerations. Significant decreases in their
HSA binding proved that the UV-pH titration method combined with an
in silico support can be used as a medicinal chemistry tool to assist
rational molecular design.
Collapse
Affiliation(s)
- Gergő Dargó
- Department of Chemical and Environmental Process Engineering , Budapest University of Technology and Economics , Műegyetem rakpart 3 , 1111 Budapest , Hungary.,Chemistry Department , Gedeon Richter Plc. , Gyömrői út. 19-21 , 1107 Budapest , Hungary
| | - Dávid Bajusz
- Medicinal Chemistry Research Group , Research Centre for Natural Sciences , Magyar tudósok krt. 2 , 1117 Budapest , Hungary
| | - Kristóf Simon
- Department of Organic Chemistry and Technology , Budapest University of Technology and Economics , Műegyetem rakpart 3 , 1111 Budapest , Hungary
| | - Judit Müller
- Medicinal Chemistry Laboratory II , Gedeon Richter Plc. , Gyömrői út. 19-21 , 1107 Budapest , Hungary
| | - György T Balogh
- Department of Chemical and Environmental Process Engineering , Budapest University of Technology and Economics , Műegyetem rakpart 3 , 1111 Budapest , Hungary.,Chemistry Department , Gedeon Richter Plc. , Gyömrői út. 19-21 , 1107 Budapest , Hungary.,Department of Pharmacodynamics and Biopharmacy , University of Szeged , 6720 Szeged , Hungary
| |
Collapse
|
24
|
Bteich M, Poulin P, Haddad S. The potential protein-mediated hepatic uptake: discussion on the molecular interactions between albumin and the hepatocyte cell surface and their implications for the in vitro-to-in vivo extrapolations of hepatic clearance of drugs. Expert Opin Drug Metab Toxicol 2019; 15:633-658. [DOI: 10.1080/17425255.2019.1640679] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Michel Bteich
- Department of Environmental and Occupational Health, School of Public Health, Université de Montréal, Montreal, Quebec, Canada
| | - Patrick Poulin
- Department of Environmental and Occupational Health, School of Public Health, Université de Montréal, Montreal, Quebec, Canada
- Consultant Patrick Poulin Inc., Québec city, Canada
| | - Sami Haddad
- Department of Environmental and Occupational Health, School of Public Health, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
25
|
Czub MP, Venkataramany BS, Majorek KA, Handing KB, Porebski PJ, Beeram SR, Suh K, Woolfork AG, Hage DS, Shabalin IG, Minor W. Testosterone meets albumin - the molecular mechanism of sex hormone transport by serum albumins. Chem Sci 2019; 10:1607-1618. [PMID: 30842823 PMCID: PMC6371759 DOI: 10.1039/c8sc04397c] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 12/07/2018] [Indexed: 12/23/2022] Open
Abstract
Serum albumin is the most abundant protein in mammalian blood plasma and is responsible for the transport of metals, drugs, and various metabolites, including hormones. We report the first albumin structure in complex with testosterone, the primary male sex hormone. Testosterone is bound in two sites, neither of which overlaps with the previously suggested Sudlow site I. We determined the binding constant of testosterone to equine and human albumins by two different methods: tryptophan fluorescence quenching and ultrafast affinity extraction. The binding studies and similarities between residues comprising the binding sites on serum albumins suggest that testosterone binds to the same sites on both proteins. Our comparative analysis of albumin complexes with hormones, drugs, and other biologically relevant compounds strongly suggests interference between a number of compounds present in blood and testosterone transport by serum albumin. We discuss a possible link between our findings and some phenomena observed in human patients, such as low testosterone levels in diabetic patients.
Collapse
Affiliation(s)
- Mateusz P Czub
- Department of Molecular Physiology and Biological Physics , University of Virginia , 1340 Jefferson Park Avenue , Charlottesville , VA 22908 , USA . ;
- Center for Structural Genomics of Infectious Diseases (CSGID) , University of Virginia , 1340 Jefferson Park Avenue , Charlottesville , VA 22908 , USA
| | - Barat S Venkataramany
- Department of Molecular Physiology and Biological Physics , University of Virginia , 1340 Jefferson Park Avenue , Charlottesville , VA 22908 , USA . ;
| | - Karolina A Majorek
- Department of Molecular Physiology and Biological Physics , University of Virginia , 1340 Jefferson Park Avenue , Charlottesville , VA 22908 , USA . ;
| | - Katarzyna B Handing
- Department of Molecular Physiology and Biological Physics , University of Virginia , 1340 Jefferson Park Avenue , Charlottesville , VA 22908 , USA . ;
| | - Przemyslaw J Porebski
- Department of Molecular Physiology and Biological Physics , University of Virginia , 1340 Jefferson Park Avenue , Charlottesville , VA 22908 , USA . ;
- Center for Structural Genomics of Infectious Diseases (CSGID) , University of Virginia , 1340 Jefferson Park Avenue , Charlottesville , VA 22908 , USA
| | - Sandya R Beeram
- Department of Chemistry , University of Nebraska-Lincoln , Lincoln , Nebraska 68588 , USA .
| | - Kyungah Suh
- Department of Chemistry , University of Nebraska-Lincoln , Lincoln , Nebraska 68588 , USA .
| | - Ashley G Woolfork
- Department of Chemistry , University of Nebraska-Lincoln , Lincoln , Nebraska 68588 , USA .
| | - David S Hage
- Department of Chemistry , University of Nebraska-Lincoln , Lincoln , Nebraska 68588 , USA .
| | - Ivan G Shabalin
- Department of Molecular Physiology and Biological Physics , University of Virginia , 1340 Jefferson Park Avenue , Charlottesville , VA 22908 , USA . ;
- Center for Structural Genomics of Infectious Diseases (CSGID) , University of Virginia , 1340 Jefferson Park Avenue , Charlottesville , VA 22908 , USA
| | - Wladek Minor
- Department of Molecular Physiology and Biological Physics , University of Virginia , 1340 Jefferson Park Avenue , Charlottesville , VA 22908 , USA . ;
- Center for Structural Genomics of Infectious Diseases (CSGID) , University of Virginia , 1340 Jefferson Park Avenue , Charlottesville , VA 22908 , USA
| |
Collapse
|
26
|
Bertozo LDC, Tavares Neto E, Oliveira LCD, Ximenes VF. Oxidative Alteration of Trp-214 and Lys-199 in Human Serum Albumin Increases Binding Affinity with Phenylbutazone: A Combined Experimental and Computational Investigation. Int J Mol Sci 2018; 19:ijms19102868. [PMID: 30241420 PMCID: PMC6213266 DOI: 10.3390/ijms19102868] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 09/17/2018] [Accepted: 09/19/2018] [Indexed: 12/22/2022] Open
Abstract
Human serum albumin (HSA) is a target for reactive oxygen species (ROS), and alterations of its physiological functions caused by oxidation is a current issue. In this work, the amino-acid residues Trp-214 and Lys-199, which are located at site I of HSA, were experimentally and computationally oxidized, and the effect on the binding constant with phenylbutazone was measured. HSA was submitted to two mild oxidizing reagents, taurine monochloramine (Tau-NHCl) and taurine dibromamine (Tau-NBr₂). The oxidation of Trp-214 provoked spectroscopic alterations in the protein which were consistent with the formation of N'-formylkynurenine. It was found that the oxidation of HSA by Tau-NBr₂, but not by Tau-NHCl, provoked a significant increase in the association constant with phenylbutazone. The alterations of Trp-214 and Lys-199 were modeled and simulated by changing these residues using the putative oxidation products. Based on the Amber score function, the interaction energy was measured, and it showed that, while native HSA presented an interaction energy of -21.3 kJ/mol, HSA with Trp-214 altered to N'-formylkynurenine resulted in an energy of -28.4 kJ/mol, and HSA with Lys-199 altered to its carbonylated form resulted in an energy of -33.9 kJ/mol. In summary, these experimental and theoretical findings show that oxidative alterations of amino-acid residues at site I of HSA affect its binding efficacy.
Collapse
Affiliation(s)
- Luiza de Carvalho Bertozo
- Department of Chemistry, Faculty of Sciences, UNESP⁻São Paulo State University, Bauru, SP 17033-360, Brazil.
| | - Ernesto Tavares Neto
- Department of Physics⁻Institute of Biosciences, Humanities and Exact Sciences, UNESP⁻São Paulo State University, São José do Rio Preto, SP 15054-000, Brazil.
| | - Leandro Cristante de Oliveira
- Department of Physics⁻Institute of Biosciences, Humanities and Exact Sciences, UNESP⁻São Paulo State University, São José do Rio Preto, SP 15054-000, Brazil.
| | - Valdecir Farias Ximenes
- Department of Chemistry, Faculty of Sciences, UNESP⁻São Paulo State University, Bauru, SP 17033-360, Brazil.
| |
Collapse
|
27
|
Palese LL. A random version of principal component analysis in data clustering. Comput Biol Chem 2018; 73:57-64. [PMID: 29428276 DOI: 10.1016/j.compbiolchem.2018.01.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 10/05/2017] [Accepted: 01/23/2018] [Indexed: 01/01/2023]
Abstract
Principal component analysis (PCA) is a widespread technique for data analysis that relies on the covariance/correlation matrix of the analyzed data. However, to properly work with high-dimensional data sets, PCA poses severe mathematical constraints on the minimum number of different replicates, or samples, that must be included in the analysis. Generally, improper sampling is due to a small number of data respect to the number of the degrees of freedom that characterize the ensemble. In the field of life sciences it is often important to have an algorithm that can accept poorly dimensioned data sets, including degenerated ones. Here a new random projection algorithm is proposed, in which a random symmetric matrix surrogates the covariance/correlation matrix of PCA, while maintaining the data clustering capacity. We demonstrate that what is important for clustering efficiency of PCA is not the exact form of the covariance/correlation matrix, but simply its symmetry.
Collapse
Affiliation(s)
- Luigi Leonardo Palese
- University of Bari "Aldo Moro", Department of Basic Medical Sciences, Neurosciences and Sense Organs (SMBNOS), Bari 70124, Italy.
| |
Collapse
|
28
|
Gao S, Liu R. Comprehensive insights into the interaction mechanism between perfluorodecanoic acid and human serum albumin. NEW J CHEM 2018. [DOI: 10.1039/c8nj00124c] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In this investigation, we explored the toxic effects of perfluorodecanoic acid (PFDA) on human serum albumin (HSA), established the interaction mode of PFDA with HSA, and provided a new strategy for the evaluation of toxicity of PFDA on functional proteins.
Collapse
Affiliation(s)
- Sichen Gao
- School of Environmental Science and Engineering
- Shandong University
- Shandong Province
- Jinan 250100
- China
| | - Rutao Liu
- School of Environmental Science and Engineering
- Shandong University
- Shandong Province
- Jinan 250100
- China
| |
Collapse
|
29
|
Naschberger A, Orry A, Lechner S, Bowler MW, Nurizzo D, Novokmet M, Keller MA, Oemer G, Seppi D, Haslbeck M, Pansi K, Dieplinger H, Rupp B. Structural Evidence for a Role of the Multi-functional Human Glycoprotein Afamin in Wnt Transport. Structure 2017; 25:1907-1915.e5. [PMID: 29153507 DOI: 10.1016/j.str.2017.10.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 09/14/2017] [Accepted: 10/23/2017] [Indexed: 11/19/2022]
Abstract
Afamin, a human plasma glycoprotein and putative transporter of hydrophobic molecules, has been shown to act as extracellular chaperone for poorly soluble, acylated Wnt proteins, forming a stable, soluble complex with functioning Wnt proteins. The 2.1-Å crystal structure of glycosylated human afamin reveals an almost exclusively hydrophobic binding cleft capable of harboring large hydrophobic moieties. Lipid analysis confirms the presence of lipids, and density in the primary binding pocket of afamin was modeled as palmitoleic acid, presenting the native O-acylation on serine 209 in human Wnt3a. The modeled complex between the experimental afamin structure and a Wnt3a homology model based on the XWnt8-Fz8-CRD fragment complex crystal structure is compelling, with favorable interactions comparable with the crystal structure complex. Afamin readily accommodates the conserved palmitoylated serine 209 of Wnt3a, providing a structural basis how afamin solubilizes hydrophobic and poorly soluble Wnt proteins.
Collapse
Affiliation(s)
- Andreas Naschberger
- Division of Genetic Epidemiology, Medical University of Innsbruck, Schöpfstraße 41, 6020 Innsbruck, Austria; Division of Biological Chemistry, Medical University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - Andrew Orry
- MolSoft LLC, 11199 Sorrento Valley Road, San Diego, CA 92121, USA
| | - Stefan Lechner
- Division of Genetic Epidemiology, Medical University of Innsbruck, Schöpfstraße 41, 6020 Innsbruck, Austria
| | - Matthew W Bowler
- European Molecular Biology Laboratory, Grenoble Outstation, 71 Avenue des Martyrs, 38043 Grenoble, France
| | - Didier Nurizzo
- Structural Biology Group, ESRF, 71 Avenue des Martyrs, 38000 Grenoble, France
| | - Mislav Novokmet
- Genos, Glycoscience Laboratory, Hondlova 2/11, 10000 Zagreb, Croatia
| | - Markus A Keller
- Division of Human Genetics, Medical University of Innsbruck, Peter-Mayr-Straße 1, 6020 Innsbruck, Austria
| | - Gregor Oemer
- Genos, Glycoscience Laboratory, Hondlova 2/11, 10000 Zagreb, Croatia
| | - Daniele Seppi
- Division of Biological Chemistry, Medical University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - Martin Haslbeck
- Department of Chemistry, Technical University of Munich, Lichtenbergstraße 4, 85748 Garching, Germany
| | - Kathrin Pansi
- Division of Biological Chemistry, Medical University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - Hans Dieplinger
- Division of Genetic Epidemiology, Medical University of Innsbruck, Schöpfstraße 41, 6020 Innsbruck, Austria
| | - Bernhard Rupp
- Division of Genetic Epidemiology, Medical University of Innsbruck, Schöpfstraße 41, 6020 Innsbruck, Austria; k.-k. Hofkristallamt, San Diego, CA 92084, USA.
| |
Collapse
|
30
|
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: 1.1] [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]
|
31
|
Abo Dena AS, Abdel Gaber SA. In vitro drug interaction of levocetirizine and diclofenac: Theoretical and spectroscopic studies. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2017; 181:239-248. [PMID: 28371723 DOI: 10.1016/j.saa.2017.03.043] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 03/14/2017] [Accepted: 03/17/2017] [Indexed: 06/07/2023]
Abstract
Levocetirizine dihydrochloride is known to interact with some anti-inflammatory drugs. We report here a comprehensive integrated theoretical and experimental study for the in vitro drug interaction between levocetirizine dihydrochloride (LEV) and diclofenac sodium (DIC). The interaction of the two drugs was confirmed by the molecular ion peak obtained from the mass spectrum of the product. Moreover, FTIR and 1HNMR spectra of the individual drugs and their interaction product were inspected to allocate the possible sites of interaction. In addition, quantum mechanical DFT calculations were performed to search for the interaction sites and to verify the types of interactions deduced from the spectroscopic studies such as charge-transfer and non-bonding π-π interactions. It was found that the studied drugs interact with each other in aqueous solution via four types of interactions, namely, ion-pair formation, three weak hydrogen bonds, non-bonding π-π interactions and charge-transfer from DIC to LEV.
Collapse
Affiliation(s)
- Ahmed S Abo Dena
- National Organization for Drug Control and Research (NODCAR), P.O. Box 29, Giza, Egypt; Faculty of Oral and Dental Medicine, Future University in Egypt (FUE), New Cairo, Egypt.
| | - Sara A Abdel Gaber
- Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), New Cairo, Egypt
| |
Collapse
|
32
|
Sarkar U, Ravindra KC, Large E, Young CL, Rivera-Burgos D, Yu J, Cirit M, Hughes DJ, Wishnok JS, Lauffenburger DA, Griffith LG, Tannenbaum SR. Integrated Assessment of Diclofenac Biotransformation, Pharmacokinetics, and Omics-Based Toxicity in a Three-Dimensional Human Liver-Immunocompetent Coculture System. Drug Metab Dispos 2017; 45:855-866. [PMID: 28450578 PMCID: PMC5469400 DOI: 10.1124/dmd.116.074005] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 04/21/2017] [Indexed: 12/13/2022] Open
Abstract
In vitro hepatocyte culture systems have inherent limitations in capturing known human drug toxicities that arise from complex immune responses. Therefore, we established and characterized a liver immunocompetent coculture model and evaluated diclofenac (DCF) metabolic profiles, in vitro–in vivo clearance correlations, toxicological responses, and acute phase responses using liquid chromatography–tandem mass spectrometry. DCF biotransformation was assessed after 48 hours of culture, and the major phase I and II metabolites were similar to the in vivo DCF metabolism profile in humans. Further characterization of secreted bile acids in the medium revealed that a glycine-conjugated bile acid was a sensitive marker of dose-dependent toxicity in this three-dimensional liver microphysiological system. Protein markers were significantly elevated in the culture medium at high micromolar doses of DCF, which were also observed previously for acute drug-induced toxicity in humans. In this immunocompetent model, lipopolysaccharide treatment evoked an inflammatory response that resulted in a marked increase in the overall number of acute phase proteins. Kupffer cell–mediated cytokine release recapitulated an in vivo proinflammatory response exemplified by a cohort of 11 cytokines that were differentially regulated after lipopolysaccharide induction, including interleukin (IL)-1β, IL-1Ra, IL-6, IL-8, IP-10, tumor necrosis factor-α, RANTES (regulated on activation normal T cell expressed and secreted), granulocyte colony-stimulating factor, macrophage colony-stimulating factor, macrophage inflammatory protein-1β, and IL-5. In summary, our findings indicate that three-dimensional liver microphysiological systems may serve as preclinical investigational platforms from the perspective of the discovery of a set of clinically relevant biomarkers including potential reactive metabolites, endogenous bile acids, excreted proteins, and cytokines to predict early drug-induced liver toxicity in humans.
Collapse
Affiliation(s)
- Ujjal Sarkar
- Departments of Biological Engineering (U.S., K.C.R., C.L.Y., D.R.-B., J.Y., M.C., J.S.W., D.A.L., L.G.G., S.R.T.) and Chemistry (S.R.T.), Massachusetts Institute of Technology, Cambridge, Massachusetts; and CN Bio Innovations Ltd., Welwyn Garden City, Hertfordshire, United Kingdom (E.L., D.J.H.)
| | - Kodihalli C Ravindra
- Departments of Biological Engineering (U.S., K.C.R., C.L.Y., D.R.-B., J.Y., M.C., J.S.W., D.A.L., L.G.G., S.R.T.) and Chemistry (S.R.T.), Massachusetts Institute of Technology, Cambridge, Massachusetts; and CN Bio Innovations Ltd., Welwyn Garden City, Hertfordshire, United Kingdom (E.L., D.J.H.)
| | - Emma Large
- Departments of Biological Engineering (U.S., K.C.R., C.L.Y., D.R.-B., J.Y., M.C., J.S.W., D.A.L., L.G.G., S.R.T.) and Chemistry (S.R.T.), Massachusetts Institute of Technology, Cambridge, Massachusetts; and CN Bio Innovations Ltd., Welwyn Garden City, Hertfordshire, United Kingdom (E.L., D.J.H.)
| | - Carissa L Young
- Departments of Biological Engineering (U.S., K.C.R., C.L.Y., D.R.-B., J.Y., M.C., J.S.W., D.A.L., L.G.G., S.R.T.) and Chemistry (S.R.T.), Massachusetts Institute of Technology, Cambridge, Massachusetts; and CN Bio Innovations Ltd., Welwyn Garden City, Hertfordshire, United Kingdom (E.L., D.J.H.)
| | - Dinelia Rivera-Burgos
- Departments of Biological Engineering (U.S., K.C.R., C.L.Y., D.R.-B., J.Y., M.C., J.S.W., D.A.L., L.G.G., S.R.T.) and Chemistry (S.R.T.), Massachusetts Institute of Technology, Cambridge, Massachusetts; and CN Bio Innovations Ltd., Welwyn Garden City, Hertfordshire, United Kingdom (E.L., D.J.H.)
| | - Jiajie Yu
- Departments of Biological Engineering (U.S., K.C.R., C.L.Y., D.R.-B., J.Y., M.C., J.S.W., D.A.L., L.G.G., S.R.T.) and Chemistry (S.R.T.), Massachusetts Institute of Technology, Cambridge, Massachusetts; and CN Bio Innovations Ltd., Welwyn Garden City, Hertfordshire, United Kingdom (E.L., D.J.H.)
| | - Murat Cirit
- Departments of Biological Engineering (U.S., K.C.R., C.L.Y., D.R.-B., J.Y., M.C., J.S.W., D.A.L., L.G.G., S.R.T.) and Chemistry (S.R.T.), Massachusetts Institute of Technology, Cambridge, Massachusetts; and CN Bio Innovations Ltd., Welwyn Garden City, Hertfordshire, United Kingdom (E.L., D.J.H.)
| | - David J Hughes
- Departments of Biological Engineering (U.S., K.C.R., C.L.Y., D.R.-B., J.Y., M.C., J.S.W., D.A.L., L.G.G., S.R.T.) and Chemistry (S.R.T.), Massachusetts Institute of Technology, Cambridge, Massachusetts; and CN Bio Innovations Ltd., Welwyn Garden City, Hertfordshire, United Kingdom (E.L., D.J.H.)
| | - John S Wishnok
- Departments of Biological Engineering (U.S., K.C.R., C.L.Y., D.R.-B., J.Y., M.C., J.S.W., D.A.L., L.G.G., S.R.T.) and Chemistry (S.R.T.), Massachusetts Institute of Technology, Cambridge, Massachusetts; and CN Bio Innovations Ltd., Welwyn Garden City, Hertfordshire, United Kingdom (E.L., D.J.H.)
| | - Douglas A Lauffenburger
- Departments of Biological Engineering (U.S., K.C.R., C.L.Y., D.R.-B., J.Y., M.C., J.S.W., D.A.L., L.G.G., S.R.T.) and Chemistry (S.R.T.), Massachusetts Institute of Technology, Cambridge, Massachusetts; and CN Bio Innovations Ltd., Welwyn Garden City, Hertfordshire, United Kingdom (E.L., D.J.H.)
| | - Linda G Griffith
- Departments of Biological Engineering (U.S., K.C.R., C.L.Y., D.R.-B., J.Y., M.C., J.S.W., D.A.L., L.G.G., S.R.T.) and Chemistry (S.R.T.), Massachusetts Institute of Technology, Cambridge, Massachusetts; and CN Bio Innovations Ltd., Welwyn Garden City, Hertfordshire, United Kingdom (E.L., D.J.H.)
| | - Steven R Tannenbaum
- Departments of Biological Engineering (U.S., K.C.R., C.L.Y., D.R.-B., J.Y., M.C., J.S.W., D.A.L., L.G.G., S.R.T.) and Chemistry (S.R.T.), Massachusetts Institute of Technology, Cambridge, Massachusetts; and CN Bio Innovations Ltd., Welwyn Garden City, Hertfordshire, United Kingdom (E.L., D.J.H.)
| |
Collapse
|
33
|
Qi J, Zhang Y, Gou Y, Lee P, Wang J, Chen S, Zhou Z, Wu X, Yang F, Liang H. Multidrug Delivery Systems Based on Human Serum Albumin for Combination Therapy with Three Anticancer Agents. Mol Pharm 2016; 13:3098-105. [DOI: 10.1021/acs.molpharmaceut.6b00277] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Jinxu Qi
- State
Key Laboratory for the Chemistry and Molecular Engineering of Medicinal
Resources, Ministry of Science and Technology of China, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Yao Zhang
- State
Key Laboratory for the Chemistry and Molecular Engineering of Medicinal
Resources, Ministry of Science and Technology of China, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Yi Gou
- State
Key Laboratory for the Chemistry and Molecular Engineering of Medicinal
Resources, Ministry of Science and Technology of China, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Philbert Lee
- Ben
May Department for Cancer Research, University of Chicago, Chicago, Illinois 60637, United States
| | - Jun Wang
- State
Key Laboratory for the Chemistry and Molecular Engineering of Medicinal
Resources, Ministry of Science and Technology of China, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Shifang Chen
- State
Key Laboratory for the Chemistry and Molecular Engineering of Medicinal
Resources, Ministry of Science and Technology of China, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Zuping Zhou
- Guangxi
Universities Key Laboratory of Stem Cell and Pharmaceutical Biotechnology, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Xiaoyang Wu
- Ben
May Department for Cancer Research, University of Chicago, Chicago, Illinois 60637, United States
| | - Feng Yang
- State
Key Laboratory for the Chemistry and Molecular Engineering of Medicinal
Resources, Ministry of Science and Technology of China, Guangxi Normal University, Guilin, Guangxi 541004, China
- Guangxi
Universities Key Laboratory of Stem Cell and Pharmaceutical Biotechnology, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Hong Liang
- State
Key Laboratory for the Chemistry and Molecular Engineering of Medicinal
Resources, Ministry of Science and Technology of China, Guangxi Normal University, Guilin, Guangxi 541004, China
| |
Collapse
|
34
|
Structural evidence of the species-dependent albumin binding of the modified cyclic phosphatidic acid with cytotoxic properties. Biosci Rep 2016; 36:BSR20160089. [PMID: 27129297 PMCID: PMC5293571 DOI: 10.1042/bsr20160089] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 04/15/2016] [Indexed: 12/12/2022] Open
Abstract
Cytotoxic properties of a new phosphorodithioate myristoyl derivative of cyclic phosphatidic acid as well as detailed binding mode of this ligand by human and equine serum albumins based on two crystal structures are presented. Cyclic phosphatidic acids (cPAs) are naturally occurring, very active signalling molecules, which are involved in several pathological states, such as cancer, diabetes or obesity. As molecules of highly lipidic character found in the circulatory system, cPAs are bound and transported by the main extracellular lipid binding protein–serum albumin. Here, we present the detailed interactions between human serum albumin (HSA) and equine serum albumin (ESA) with a derivative of cPA, 1-O-myristoyl-sn-glycerol-2,3-cyclic phosphorodithioate (Myr-2S-cPA). Initial selection of the ligand used for the structural study was made by the analysis of the therapeutically promising properties of the sulfur containing analogues of cPA in respect to the unmodified lysophospholipids (LPLs). Substitution of one or two non-bridging oxygen atoms in the phosphate group with one or two sulfur atoms increases the cytotoxic effect of cPAs up to 60% on the human prostate cancer (PC) cells. Myr-2S-cPA reduces cancer cell viability in a dose-dependent manner, with IC50 value of 29.0 μM after 24 h incubation, which is almost 30% lower than IC50 of single substituted phosphorothioate cPA. Although, the structural homology between HSA and ESA is big, their crystal complexes with Myr-2S-cPA demonstrate significantly different mode of binding of this LPL analogue. HSA binds three molecules of Myr-2S-cPA, whereas ESA only one. Moreover, none of the identified Myr-2S-cPA binding sites overlap in both albumins.
Collapse
|