1
|
Mehmood H, Haroon M, Akhtar T, Woodward S, Haq S, M Alshehri S. Synthesis, anti-diabetic profiling and molecular docking studies of 2-(2-arylidenehydrazinyl)thiazol-4(5 H)-ones. Future Med Chem 2024; 16:1255-1266. [PMID: 38989987 PMCID: PMC11244696 DOI: 10.1080/17568919.2024.2342700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 04/08/2024] [Indexed: 07/12/2024] Open
Abstract
Aim: To synthesize novel more potent anti-diabetic agents. Methodology: A simple cost effective Hantzsch's synthetic strategy was used to synthesize 2-(2-arylidenehydrazinyl)thiazol-4(5H)-ones. Results: Fifteen new 2-(2-arylidenehydrazinyl)thiazol-4(5H)-ones were established to check their anti-diabetic potential. From alpha(α)-amylase inhibition, anti-glycation and anti-oxidant activities it is revealed that most of the compounds possess good anti-diabetic potential. All tested compounds were found to be more potent anti-diabetic agents via anti-glycation mode. The results of α-amylase and anti-oxidant inhibition revealed that compounds are less active against α-amylase and anti-oxidant assays. Conclusion: This study concludes that introduction of various electron withdrawing groups at the aryl ring and substitution of different functionalities around thiazolone nucleus could help to find out better anti-diabetic drug.
Collapse
Affiliation(s)
- Hasnain Mehmood
- Department of Chemistry, Mirpur University of Science & Technology (MUST), 10250, Mirpur (AJK), Pakistan
| | - Muhammad Haroon
- Department of Chemistry & Biochemistry, Miami University 651, E. High Street, Oxford, 45056, Ohio, USA
| | - Tashfeen Akhtar
- Department of Chemistry, Mirpur University of Science & Technology (MUST), 10250, Mirpur (AJK), Pakistan
| | - Simon Woodward
- GSK, Carbon Neutral Laboratories for Sustainable Chemistry, University Park Nottingham, NG7 2RD, United Kingdom
| | - Saadia Haq
- Institute of Chemistry, Khwaja Fareed University of Engineering & Information Technology, Rahim Yar Khan, 64200, Pakistan
- Centre for Theoretical & Computational Research, Khwaja Fareed University of Engineering & Information Technology, Rahim Yar Khan, 64200, Pakistan
| | - Saad M Alshehri
- Department of Chemistry, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| |
Collapse
|
2
|
Marson NA, Gallio AE, Mandal SK, Laskowski RA, Raven EL. In silico prediction of heme binding in proteins. J Biol Chem 2024; 300:107250. [PMID: 38569935 PMCID: PMC11101860 DOI: 10.1016/j.jbc.2024.107250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/11/2024] [Accepted: 03/26/2024] [Indexed: 04/05/2024] Open
Abstract
The process of heme binding to a protein is prevalent in almost all forms of life to control many important biological properties, such as O2-binding, electron transfer, gas sensing or to build catalytic power. In these cases, heme typically binds tightly (irreversibly) to a protein in a discrete heme binding pocket, with one or two heme ligands provided most commonly to the heme iron by His, Cys or Tyr residues. Heme binding can also be used as a regulatory mechanism, for example in transcriptional regulation or ion channel control. When used as a regulator, heme binds more weakly, with different heme ligations and without the need for a discrete heme pocket. This makes the characterization of heme regulatory proteins difficult, and new approaches are needed to predict and understand the heme-protein interactions. We apply a modified version of the ProFunc bioinformatics tool to identify heme-binding sites in a test set of heme-dependent regulatory proteins taken from the Protein Data Bank and AlphaFold models. The potential heme binding sites identified can be easily visualized in PyMol and, if necessary, optimized with RosettaDOCK. We demonstrate that the methodology can be used to identify heme-binding sites in proteins, including in cases where there is no crystal structure available, but the methodology is more accurate when the quality of the structural information is high. The ProFunc tool, with the modification used in this work, is publicly available at https://www.ebi.ac.uk/thornton-srv/databases/profunc and can be readily adopted for the examination of new heme binding targets.
Collapse
Affiliation(s)
- Noa A Marson
- School of Chemistry, University of Bristol, Bristol, UK
| | | | | | - Roman A Laskowski
- European Bioinformatics Institute (EMBL-EBI), European Molecular Biology Laboratory, Wellcome Trust Genome Campus, Cambridge, UK
| | - Emma L Raven
- School of Chemistry, University of Bristol, Bristol, UK.
| |
Collapse
|
3
|
Enomoto K, Torisu T, Mizuguchi J, Tanoue R, Uchiyama S. Structure of Human Serum Albumin at a Foam Surface. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:8774-8783. [PMID: 38587054 DOI: 10.1021/acs.jafc.3c09357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Proteins can be adsorbed on the air-water interface (AWI), and the structural changes in proteins at the AWI are closely related to the foaming properties of foods and beverages. However, how these structural changes in proteins at the AWI occur is not well understood. We developed a method for the structural assessment of proteins in the foam state using hydrogen/deuterium exchange mass spectrometry. Adsorption sites and structural changes in human serum albumin (HSA) were identified in situ at the peptide-level resolution. The N-terminus and the loop (E492-T506), which contains hydrophobic amino acids, were identified as adsorption sites. Both the structural flexibility and hydrophobicity were considered to be critical factors for the adsorption of HSA at the AWI. Structural changes in HSA were observed after more than one minute of foaming and were spread widely throughout the structure. These structural changes at the foam AWI were reversible.
Collapse
Affiliation(s)
- Kanta Enomoto
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Tetsuo Torisu
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Junya Mizuguchi
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Ryosuke Tanoue
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Susumu Uchiyama
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Exploratory Research Center on Life andLiving Systems, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan
| |
Collapse
|
4
|
Wang L, Wang T, Wu G, Tian D. An HDBB-based fluorescent probe for the sensitive detection of human serum albumin. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:427-433. [PMID: 38165671 DOI: 10.1039/d3ay01733h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2024]
Abstract
The detection of human serum albumin (HSA) in bodily fluids is of great significance in the biomedical area because HSA in bodily fluids is commonly used as a biomarker for the early diagnosis of diseases. To detect HSA, we employed HDBB, 4,4'-(hydrazine-1,2-diylidene bis(methanylylidene)) bis(3-hydroxybenzoic acid), as a fluorescent probe with a large Stokes shift. HDBB had obvious excited state intramolecular proton transfer (ESIPT) and aggregation-induced emission (AIE) features. We elucidated the ESIPT characteristics of HDBB through the DFT approach. We also performed a molecular docking simulation between HDBB and HSA, showing that HDBB primarily bonded to HSA via hydrophobic force and hydrogen bonds. The FL intensities of HDBB with HSA concentrations had a linear range of 0.01-0.2 mg mL-1 (R2 = 0.9995), and the LOD was 1.104 μg mL-1. We also used the probe to detect HSA in urine, with spiked recoveries of 98.10-105.33%. Given its high selectivity and feasible synthesis, HDBB has potential applications in detecting HSA in real biological systems.
Collapse
Affiliation(s)
- Liwen Wang
- Hubei Key Laboratory of Biological Resources Protection and Utilization, School of Chemical and Environmental Engineering, Hubei Minzu University, Enshi 445000, PR China.
| | - Tengfei Wang
- Hubei Key Laboratory of Biological Resources Protection and Utilization, School of Chemical and Environmental Engineering, Hubei Minzu University, Enshi 445000, PR China.
| | - Guang Wu
- Hubei Key Laboratory of Biological Resources Protection and Utilization, School of Chemical and Environmental Engineering, Hubei Minzu University, Enshi 445000, PR China.
| | - Dating Tian
- Hubei Key Laboratory of Biological Resources Protection and Utilization, School of Chemical and Environmental Engineering, Hubei Minzu University, Enshi 445000, PR China.
| |
Collapse
|
5
|
Cvjetan N, Schuler LD, Ishikawa T, Walde P. Optimization and Enhancement of the Peroxidase-like Activity of Hemin in Aqueous Solutions of Sodium Dodecylsulfate. ACS OMEGA 2023; 8:42878-42899. [PMID: 38024761 PMCID: PMC10652838 DOI: 10.1021/acsomega.3c05915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 10/10/2023] [Accepted: 10/13/2023] [Indexed: 12/01/2023]
Abstract
Iron porphyrins play several important roles in present-day living systems and probably already existed in very early life forms. Hemin (= ferric protoporphyrin IX = ferric heme b), for example, is the prosthetic group at the active site of heme peroxidases, catalyzing the oxidation of a number of different types of reducing substrates after hemin is first oxidized by hydrogen peroxide as the oxidizing substrate of the enzyme. The active site of heme peroxidases consists of a hydrophobic pocket in which hemin is embedded noncovalently and kept in place through coordination of the iron atom to a proximal histidine side chain of the protein. It is this partially hydrophobic local environment of the enzyme which determines the efficiency with which the sequential reactions of the oxidizing and reducing substrates proceed at the active site. Free hemin, which has been separated from the protein moiety of heme peroxidases, is known to aggregate in an aqueous solution and exhibits low catalytic activity. Based on previous reports on the use of surfactant micelles to solubilize free hemin in a nonaggregated state, the peroxidase-like activity of hemin in the presence of sodium dodecyl sulfate (SDS) at concentrations below and above the critical concentration for SDS micelle formation (critical micellization concentration (cmc)) was systematically investigated. In most experiments, 3,3',5,5'-tetramethylbenzidine (TMB) was applied as a reducing substrate at pH = 7.2. The presence of SDS clearly had a positive effect on the reaction in terms of initial reaction rate and reaction yield, even at concentrations below the cmc. The highest activity correlated with the cmc value, as demonstrated for reactions at three different HEPES concentrations. The 4-(2-hydroxyethyl)-1-piperazineethanesulfonate salt (HEPES) served as a pH buffer substance and also had an accelerating effect on the reaction. At the cmc, the addition of l-histidine (l-His) resulted in a further concentration-dependent increase in the peroxidase-like activity of hemin until a maximal effect was reached at an optimal l-His concentration, probably corresponding to an ideal mono-l-His ligation to hemin. Some of the results obtained can be understood on the basis of molecular dynamics simulations, which indicated the existence of intermolecular interactions between hemin and HEPES and between hemin and SDS. Preliminary experiments with SDS/dodecanol vesicles at pH = 7.2 showed that in the presence of the vesicles, hemin exhibited similar peroxidase-like activity as in the case of SDS micelles. This supports the hypothesis that micelle- or vesicle-associated ferric or ferrous iron porphyrins may have played a role as primitive catalysts in membranous prebiotic compartment systems before cellular life emerged.
Collapse
Affiliation(s)
- Nemanja Cvjetan
- Department
of Materials, ETH-Zürich, Leopold-Ruzicka-Weg 4, 8093 Zürich, Switzerland
| | | | - Takashi Ishikawa
- Department
of Biology and Chemistry, Paul Scherrer Institute and Department of
Biology, ETH-Zürich, Forschungsstrasse 111, 5232 Villigen PSI, Switzerland
| | - Peter Walde
- Department
of Materials, ETH-Zürich, Leopold-Ruzicka-Weg 4, 8093 Zürich, Switzerland
| |
Collapse
|
6
|
Dent MR, DeMartino AW. Nitric oxide and thiols: Chemical biology, signalling paradigms and vascular therapeutic potential. Br J Pharmacol 2023:10.1111/bph.16274. [PMID: 37908126 PMCID: PMC11058123 DOI: 10.1111/bph.16274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 09/18/2023] [Accepted: 10/09/2023] [Indexed: 11/02/2023] Open
Abstract
Nitric oxide (• NO) interactions with biological thiols play crucial, but incompletely determined, roles in vascular signalling and other biological processes. Here, we highlight two recently proposed signalling paradigms: (1) the formation of a vasodilating labile nitrosyl ferrous haem (NO-ferrohaem) facilitated by thiols via thiyl radical generation and (2) polysulfides/persulfides and their interaction with • NO. We also describe the specific (bio)chemical routes in which • NO and thiols react to form S-nitrosothiols, a broad class of small molecules, and protein post-translational modifications that can influence protein function through catalytic site or allosteric structural changes. S-Nitrosothiol formation depends upon cellular conditions, but critically, an appropriate oxidant for either the thiol (yielding a thiyl radical) or • NO (yielding a nitrosonium [NO+ ]-donating species) is required. We examine the roles of these collective • NO/thiol species in vascular signalling and their cardiovascular therapeutic potential.
Collapse
Affiliation(s)
- Matthew R. Dent
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Anthony W. DeMartino
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
7
|
DeMartino AW, Poudel L, Dent MR, Chen X, Xu Q, Gladwin BS, Tejero J, Basu S, Alipour E, Jiang Y, Rose JJ, Gladwin MT, Kim-Shapiro DB. Thiol-catalyzed formation of NO-ferroheme regulates intravascular NO signaling. Nat Chem Biol 2023; 19:1256-1266. [PMID: 37710075 PMCID: PMC10897909 DOI: 10.1038/s41589-023-01413-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 07/27/2023] [Indexed: 09/16/2023]
Abstract
Nitric oxide (NO) is an endogenously produced signaling molecule that regulates blood flow and platelet activation. However, intracellular and intravascular diffusion of NO are limited by scavenging reactions with several hemoproteins, raising questions as to how free NO can signal in hemoprotein-rich environments. We explore the hypothesis that NO can be stabilized as a labile ferrous heme-nitrosyl complex (Fe2+-NO, NO-ferroheme). We observe a reaction between NO, labile ferric heme (Fe3+) and reduced thiols to yield NO-ferroheme and a thiyl radical. This thiol-catalyzed reductive nitrosylation occurs when heme is solubilized in lipophilic environments such as red blood cell membranes or bound to serum albumin. The resulting NO-ferroheme resists oxidative inactivation, is soluble in cell membranes and is transported intravascularly by albumin to promote potent vasodilation. We therefore provide an alternative route for NO delivery from erythrocytes and blood via transfer of NO-ferroheme and activation of apo-soluble guanylyl cyclase.
Collapse
Affiliation(s)
- Anthony W DeMartino
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Laxman Poudel
- Department of Physics, Wake Forest University, Winston-Salem, NC, USA
| | - Matthew R Dent
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xiukai Chen
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Qinzi Xu
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Brendan S Gladwin
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jesús Tejero
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Swati Basu
- Department of Physics, Wake Forest University, Winston-Salem, NC, USA
- Translational Science Center, Wake Forest University, Winston-Salem, NC, USA
| | - Elmira Alipour
- Department of Physics, Wake Forest University, Winston-Salem, NC, USA
| | - Yiyang Jiang
- Department of Physics, Wake Forest University, Winston-Salem, NC, USA
| | - Jason J Rose
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mark T Gladwin
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Daniel B Kim-Shapiro
- Department of Physics, Wake Forest University, Winston-Salem, NC, USA.
- Translational Science Center, Wake Forest University, Winston-Salem, NC, USA.
| |
Collapse
|
8
|
Vaishnav MS, Kumari N, Srikanta S, Krishnaswamy PR, Balaram P, Bhat N. Differential Spectrum of Albumin Glycation, Oxidation, and Truncation in Type 2 and Type 1 Diabetes: Clinical and Biological Implications. Metab Syndr Relat Disord 2023; 21:397-409. [PMID: 37471231 DOI: 10.1089/met.2023.0079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/22/2023] Open
Abstract
Background: Albumin, the most abundant and physiologically vital serum protein, accumulates a range of chemical modifications, as consequence of encounters with large number of reactive molecules whose concentrations increase in serum under pathological conditions. Methods: In a "proof of concept" study, mass spectrometric analysis was utilized to quantitate albumin post-translational modifications (glycation, oxidation, and truncation; individual isoforms and total) in four informative subject groups [type 1 diabetes (T1DM), type 2 diabetes (T2DM), prediabetes-obesity and healthy; all with estimated glomerular filtration rate ≥60 mL/(min·m2)]. Besides glycated albumin (GA/mass spectrometry), glycated serum protein (GSP/nitro blue tetrazolium colorimetry), and glycated hemoglobin (HbA1c/high-performance liquid chromatography) were also measured. Results: A wide spectrum of albumin molecular modifications was identified in diabetes, with significant differences between T2DM and T1DM. Albumin glycation: GA correlated more strongly with HbA1c in T1DM, compared to T2DM. Higher albumin glycation isoforms (human serum albumin +3G/2G) were more stable and discriminative markers of mean glycemia. Albumin oxidation: T2DM, in comparison with T1DM, showed enhanced oxidative and dual (glycation plus oxidation) modifications, representing extreme molecular pathology. Albumin truncation: There was dramatic reduction ("deficiency") of truncated albumin isoforms in T2DM, and significant reduction in T1DM. Albumin truncation negatively correlated with severity of albumin glycation (mean glycemia) and albumin oxidation (cysteinylation). Possible mechanisms of insulin resistance, with associated increased free fatty acids binding to albumin, in stimulating albumin oxidation and inhibiting albumin glycation ("metabolic cross talks") are reviewed. Conclusions: Albumin molecular modifications in diabetes, together with significant differences between T2DM and T1DM, suggest possible role for insulin resistance in their genesis and consequent cell, tissue, and vascular dysfunction/damage. Albumin molecular fingerprinting can provide valuable insights into pathogenesis, diagnosis, monitoring, and future therapies for diabetes. Identification of biomarker battery ("albuminomics," "diabetomics") driven diverse "healthy," prediabetes, obesity, and T2DM phenotypes represents additional novel step toward precision medicine in diabetes and related disorders.
Collapse
Affiliation(s)
- Madhumati S Vaishnav
- Centre for Nano Science and Engineering, Indian Institute of Science, Bengaluru, India
- Samatvam Endocrinology Diabetes Center, Jnana Sanjeevini Diabetes Hospital and Medical Center, Bengaluru, India
| | - Namita Kumari
- Centre for Nano Science and Engineering, Indian Institute of Science, Bengaluru, India
| | - Sathyanarayana Srikanta
- Samatvam Endocrinology Diabetes Center, Jnana Sanjeevini Diabetes Hospital and Medical Center, Bengaluru, India
| | - Patnam R Krishnaswamy
- Centre for Nano Science and Engineering, Indian Institute of Science, Bengaluru, India
| | - Padmanabhan Balaram
- Molecular Biophysics Unit, Indian Institute of Science, Bengaluru, India
- National Centre for Biological Sciences, Bengaluru, India
| | - Navakanta Bhat
- Centre for Nano Science and Engineering, Indian Institute of Science, Bengaluru, India
| |
Collapse
|
9
|
Pires IS, Berthiaume F, Palmer AF. Engineering Therapeutics to Detoxify Hemoglobin, Heme, and Iron. Annu Rev Biomed Eng 2023; 25:1-21. [PMID: 37289555 DOI: 10.1146/annurev-bioeng-081622-031203] [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] [Indexed: 06/10/2023]
Abstract
Hemolysis (i.e., red blood cell lysis) can increase circulatory levels of cell-free hemoglobin (Hb) and its degradation by-products, namely heme (h) and iron (Fe). Under homeostasis, minor increases in these three hemolytic by-products (Hb/h/Fe) are rapidly scavenged and cleared by natural plasma proteins. Under certain pathophysiological conditions, scavenging systems become overwhelmed, leading to the accumulation of Hb/h/Fe in the circulation. Unfortunately, these species cause various side effects such as vasoconstriction, hypertension, and oxidative organ damage. Therefore, various therapeutics strategies are in development, ranging from supplementation with depleted plasma scavenger proteins to engineered biomimetic protein constructs capable of scavenging multiple hemolytic species. In this review, we briefly describe hemolysis and the characteristics of the major plasma-derived protein scavengers of Hb/h/Fe. Finally, we present novel engineering approaches designed to address the toxicity of these hemolytic by-products.
Collapse
Affiliation(s)
- Ivan S Pires
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio, USA;
| | - François Berthiaume
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey, USA
| | - Andre F Palmer
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio, USA;
| |
Collapse
|
10
|
Turilli-Ghisolfi ES, Lualdi M, Fasano M. Ligand-Based Regulation of Dynamics and Reactivity of Hemoproteins. Biomolecules 2023; 13:683. [PMID: 37189430 PMCID: PMC10135655 DOI: 10.3390/biom13040683] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/14/2023] [Accepted: 04/15/2023] [Indexed: 05/17/2023] Open
Abstract
Hemoproteins include several heme-binding proteins with distinct structure and function. The presence of the heme group confers specific reactivity and spectroscopic properties to hemoproteins. In this review, we provide an overview of five families of hemoproteins in terms of dynamics and reactivity. First, we describe how ligands modulate cooperativity and reactivity in globins, such as myoglobin and hemoglobin. Second, we move on to another family of hemoproteins devoted to electron transport, such as cytochromes. Later, we consider heme-based reactivity in hemopexin, the main heme-scavenging protein. Then, we focus on heme-albumin, a chronosteric hemoprotein with peculiar spectroscopic and enzymatic properties. Eventually, we analyze the reactivity and dynamics of the most recently discovered family of hemoproteins, i.e., nitrobindins.
Collapse
Affiliation(s)
| | | | - Mauro Fasano
- Department of Science and High Technology, University of Insubria, 22100 Como, Italy
| |
Collapse
|
11
|
Mehmood H, Akhtar T, Haroon M, Khalid M, Woodward S, Asghar MA, Baby R, Orfali R, Perveen S. Synthesis of Fluorinated Hydrazinylthiazole Derivatives: A Virtual and Experimental Approach to Diabetes Management. ACS OMEGA 2023; 8:11433-11446. [PMID: 37008089 PMCID: PMC10061536 DOI: 10.1021/acsomega.3c00265] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 03/07/2023] [Indexed: 06/19/2023]
Abstract
A novel series of fluorophenyl-based thiazoles was synthesized following the Hanztsch method. All of the compounds were initially verified with physical parameters (color, melting point, retardation factor (R f)), which were further confirmed by several spectroscopic methods, including ultraviolet-visible (UV-visible), Fourier-transform infrared (FTIR), 1H, 13C, 19F NMR, and high-resolution mass spectrometry (HRMS). The binding interactions of all compounds were studied using a molecular docking simulation approach. Furthermore, each compound was evaluated for its alpha(α)-amylase, antiglycation, and antioxidant potentials. The biocompatibility of all compounds was checked with an in vitro hemolytic assay. All synthesized scaffolds were found biocompatible with minimal lysis of human erythrocytes as compared to the standard Triton X-100. Among the tested compounds, the analogue 3h (IC50 = 5.14 ± 0.03 μM) was found to be a highly potent candidate against α-amylase as compared to the standard (acarbose, IC50 = 5.55 ± 0.06 μM). The compounds 3d, 3f, 3i, and 3k exhibited excellent antiglycation inhibition potential with their IC50 values far less than the standard amino guanidine (IC50 = 0.403 ± 0.001 mg/mL). The antidiabetic potential was further supported by docking studies. Docking studies revealed that all synthesized compounds exhibited various interactions along enzyme active sites (pi-pi, H-bonding, van der Waals) with varied binding energies.
Collapse
Affiliation(s)
- Hasnain Mehmood
- Department
of Chemistry, Mirpur University of Science
and Technology (MUST), Mirpur 10250, Azad Jammu and Kashmir, Pakistan
| | - Tashfeen Akhtar
- Department
of Chemistry, Mirpur University of Science
and Technology (MUST), Mirpur 10250, Azad Jammu and Kashmir, Pakistan
| | - Muhammad Haroon
- Department
of Chemistry, Mirpur University of Science
and Technology (MUST), Mirpur 10250, Azad Jammu and Kashmir, Pakistan
- Department
of Chemistry, Government Major Muhammad Afzal Khan (Shaheed), Boys
Degree College Afzalpur, Mirpur (Affiliated
with Mirpur University of Science and Technology (MUST)), Mirpur 10250, Azad Jammu and Kashmir, Pakistan
| | - Muhammad Khalid
- Institute
of Chemistry, Khwaja Fareed University of
Engineering & Information Technology, Rahim Yar Khan 64200, Pakistan
- Center
for Theoretical and Computational Research, Khwaja Fareed University of Engineering & Information Technology, Rahim Yar Khan 64200, Pakistan
| | - Simon Woodward
- GSK,
Carbon Neutral Laboratories for Sustainable Chemistry, University Park Nottingham, Nottingham NG7 2RD, U.K.
| | - Muhammad Adnan Asghar
- Department
of Chemistry, Division of Science and Technology, University of Education, Lahore 54770, Pakistan
| | - Rabia Baby
- Department
of Education, Sukkur IBA University, Sukkur 65200, Sindh, Pakistan
| | - Raha Orfali
- Department
of Pharmacognosy, Collage of Pharmacy, King
Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Shagufta Perveen
- Department
of Chemistry, School of Computer, Mathematical and Natural Sciences, Morgan State University, Baltimore, Maryland 21251, United States
| |
Collapse
|
12
|
De Simone G, Varricchio R, Ruberto TF, di Masi A, Ascenzi P. Heme Scavenging and Delivery: The Role of Human Serum Albumin. Biomolecules 2023; 13:biom13030575. [PMID: 36979511 PMCID: PMC10046553 DOI: 10.3390/biom13030575] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/10/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Heme is the reactive center of several metal-based proteins that are involved in multiple biological processes. However, free heme, defined as the labile heme pool, has toxic properties that are derived from its hydrophobic nature and the Fe-atom. Therefore, the heme concentration must be tightly controlled to maintain cellular homeostasis and to avoid pathological conditions. Therefore, different systems have been developed to scavenge either Hb (i.e., haptoglobin (Hp)) or the free heme (i.e., high-density lipoproteins (HDL), low-density lipoproteins (LDL), hemopexin (Hx), and human serum albumin (HSA)). In the first seconds after heme appearance in the plasma, more than 80% of the heme binds to HDL and LDL, and only the remaining 20% binds to Hx and HSA. Then, HSA slowly removes most of the heme from HDL and LDL, and finally, heme transits to Hx, which releases it into hepatic parenchymal cells. The Hx:heme or HSA:heme complexes are internalized via endocytosis mediated by the CD91 and CD71 receptors, respectively. As heme constitutes a major iron source for pathogens, bacteria have evolved hemophores that can extract and uptake heme from host proteins, including HSA:heme. Here, the molecular mechanisms underlying heme scavenging and delivery from HSA are reviewed. Moreover, the relevance of HSA in disease states associated with increased heme plasma concentrations are discussed.
Collapse
Affiliation(s)
- Giovanna De Simone
- Department of Sciences, Section of Biomedical Sciences and Technologies, Roma Tre University, 00146 Roma, Italy
| | - Romualdo Varricchio
- Department of Sciences, Section of Biomedical Sciences and Technologies, Roma Tre University, 00146 Roma, Italy
| | - Tommaso Francesco Ruberto
- Department of Sciences, Section of Biomedical Sciences and Technologies, Roma Tre University, 00146 Roma, Italy
| | - Alessandra di Masi
- Department of Sciences, Section of Biomedical Sciences and Technologies, Roma Tre University, 00146 Roma, Italy
- Centro Linceo Interdisciplinare Beniamino Segre, Accademia Nazionale dei Lincei, 00165 Roma, Italy
| | - Paolo Ascenzi
- Department of Sciences, Section of Biomedical Sciences and Technologies, Roma Tre University, 00146 Roma, Italy
- Accademia Nazionale dei Lincei, 00165 Roma, Italy
| |
Collapse
|
13
|
Mariño-Ocampo N, Rodríguez DF, Guerra Díaz D, Zúñiga-Núñez D, Duarte Y, Fuentealba D, Zacconi FC. Direct Oral FXa Inhibitors Binding to Human Serum Albumin: Spectroscopic, Calorimetric, and Computational Studies. Int J Mol Sci 2023; 24:ijms24054900. [PMID: 36902328 PMCID: PMC10002493 DOI: 10.3390/ijms24054900] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/26/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
Direct FXa inhibitors are an important class of bioactive molecules (rivaroxaban, apixaban, edoxaban, and betrixaban) applied for thromboprophylaxis in diverse cardiovascular pathologies. The interaction of active compounds with human serum albumin (HSA), the most abundant protein in blood plasma, is a key research area and provides crucial information about drugs' pharmacokinetics and pharmacodynamic properties. This research focuses on the study of the interactions between HSA and four commercially available direct oral FXa inhibitors, applying methodologies including steady-state and time-resolved fluorescence, isothermal titration calorimetry (ITC), and molecular dynamics. The HSA complexation of FXa inhibitors was found to occur via static quenching, and the complex formation in the ground states affects the fluorescence of HSA, with a moderate binding constant of 104 M-1. However, the ITC studies reported significantly different binding constants (103 M-1) compared with the results obtained through spectrophotometric methods. The suspected binding mode is supported by molecular dynamics simulations, where the predominant interactions were hydrogen bonds and hydrophobic interactions (mainly π-π stacking interactions between the phenyl ring of FXa inhibitors and the indole moiety of Trp214). Finally, the possible implications of the obtained results regarding pathologies such as hypoalbuminemia are briefly discussed.
Collapse
Affiliation(s)
- Nory Mariño-Ocampo
- Escuela de Química, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| | - Diego F. Rodríguez
- Escuela de Química, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| | - Daniel Guerra Díaz
- Escuela de Química, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| | - Daniel Zúñiga-Núñez
- Escuela de Química, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| | - Yorley Duarte
- Center for Bioinformatics and Integrative Biology, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago 8370035, Chile
| | - Denis Fuentealba
- Escuela de Química, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
- Correspondence: (D.F.); (F.C.Z.)
| | - Flavia C. Zacconi
- Escuela de Química, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
- Centro de Investigaciones en Nanotecnología y Materiales Avanzados, CIEN-UC, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
- Center for Nanomedicine, Diagnostic & Drug Development (ND3), Universidad de Talca, Talca 3460000, Chile
- Correspondence: (D.F.); (F.C.Z.)
| |
Collapse
|
14
|
DeMartino AW, Poudel L, Dent MR, Chen X, Xu Q, Gladwin BS, Tejero J, Basu S, Alipour E, Jiang Y, Rose JJ, Gladwin MT, Kim-Shapiro DB. Thiol catalyzed formation of NO-ferroheme regulates canonical intravascular NO signaling. RESEARCH SQUARE 2023:rs.3.rs-2402224. [PMID: 36711928 PMCID: PMC9882697 DOI: 10.21203/rs.3.rs-2402224/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Nitric oxide (NO) is an endogenously produced physiological signaling molecule that regulates blood flow and platelet activation. However, both the intracellular and intravascular diffusion of NO is severely limited by scavenging reactions with hemoglobin, myoglobin, and other hemoproteins, raising unanswered questions as to how free NO can signal in hemoprotein-rich environments, like blood and cardiomyocytes. We explored the hypothesis that NO could be stabilized as a ferrous heme-nitrosyl complex (Fe 2+ -NO, NO-ferroheme) either in solution within membranes or bound to albumin. Unexpectedly, we observed a rapid reaction of NO with free ferric heme (Fe 3+ ) and a reduced thiol under physiological conditions to yield NO-ferroheme and a thiyl radical. This thiol-catalyzed reductive nitrosylation reaction occurs readily when the hemin is solubilized in lipophilic environments, such as red blood cell membranes, or bound to serum albumin. NO-ferroheme albumin is stable, even in the presence of excess oxyhemoglobin, and potently inhibits platelet activation. NO-ferroheme-albumin administered intravenously to mice dose-dependently vasodilates at low- to mid-nanomolar concentrations. In conclusion, we report the fastest rate of reductive nitrosylation observed to date to generate a NO-ferroheme molecule that resists oxidative inactivation, is soluble in cell membranes, and is transported intravascularly by albumin to promote potent vasodilation.
Collapse
Affiliation(s)
- Anthony W. DeMartino
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Laxman Poudel
- Department of Physics, Wake Forest University, Winston-Salem, NC 27109, USA
| | - Matthew R. Dent
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Xiukai Chen
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Qinzi Xu
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Brendan S. Gladwin
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Jesús Tejero
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | - Swati Basu
- Department of Physics, Wake Forest University, Winston-Salem, NC 27109, USA
- Translational Science Center, Wake Forest University, Winston-Salem, NC 27109, USA
| | - Elmira Alipour
- Department of Physics, Wake Forest University, Winston-Salem, NC 27109, USA
| | - Yiyang Jiang
- Department of Physics, Wake Forest University, Winston-Salem, NC 27109, USA
| | - Jason J. Rose
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Mark T. Gladwin
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Daniel B. Kim-Shapiro
- Department of Physics, Wake Forest University, Winston-Salem, NC 27109, USA
- Translational Science Center, Wake Forest University, Winston-Salem, NC 27109, USA
| |
Collapse
|
15
|
Abstract
Ferric heme b (= ferric protoporphyrin IX = hemin) is an important prosthetic group of different types of enzymes, including the intensively investigated and widely applied horseradish peroxidase (HRP). In HRP, hemin is present in monomeric form in a hydrophobic pocket containing among other amino acid side chains the two imidazoyl groups of His170 and His42. Both amino acids are important for the peroxidase activity of HRP as an axial ligand of hemin (proximal His170) and as an acid/base catalyst (distal His42). A key feature of the peroxidase mechanism of HRP is the initial formation of compound I under heterolytic cleavage of added hydrogen peroxide as a terminal oxidant. Investigations of free hemin dispersed in aqueous solution showed that different types of hemin dimers can form, depending on the experimental conditions, possibly resulting in hemin crystallization. Although it has been recognized already in the 1970s that hemin aggregation can be prevented in aqueous solution by using micelle-forming amphiphiles, it remains a challenge to prepare hemin-containing micellar and vesicular systems with peroxidase-like activities. Such systems are of interest as cheap HRP-mimicking catalysts for analytical and synthetic applications. Some of the key concepts on which research in this fascinating and interdisciplinary field is based are summarized, along with major accomplishments and possible directions for further improvement. A systematic analysis of the physico-chemical properties of hemin in aqueous micellar solutions and vesicular dispersions must be combined with a reliable evaluation of its catalytic activity. Future studies should show how well the molecular complexity around hemin in HRP can be mimicked by using micelles or vesicles. Because of the importance of heme b in virtually all biological systems and the fact that porphyrins and hemes can be obtained under potentially prebiotic conditions, ideas exist about the possible role of heme-containing micellar and vesicular systems in prebiotic times.
Collapse
|
16
|
Cvjetan N, Kissner R, Bajuk-Bogdanović D, Ćirić-Marjanović G, Walde P. Hemin-catalyzed oxidative oligomerization of p-aminodiphenylamine (PADPA) in the presence of aqueous sodium dodecylbenzenesulfonate (SDBS) micelles. RSC Adv 2022; 12:13154-13167. [PMID: 35520130 PMCID: PMC9063397 DOI: 10.1039/d2ra02198f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 04/18/2022] [Indexed: 11/23/2022] Open
Abstract
In a previous report on the enzymatic synthesis of the conductive emeraldine salt form of polyaniline (PANI-ES) in aqueous solution using PADPA (p-aminodiphenylamine) as monomer, horseradish peroxidase isoenzyme C (HRPC) was applied as a catalyst at pH = 4.3 with H2O2 as a terminal oxidant. In that work, anionic vesicles were added to the reaction mixture for (i) guiding the reaction to obtain poly(PADPA) products that resemble PANI-ES, and for (ii) preventing product precipitation (known as the “template effect”). In the work now presented, instead of native HRPC, only its prosthetic group ferric heme b (= hemin) was utilized as a catalyst, and micelles formed from SDBS (sodium dodecylbenzenesulfonate) served as templates. For the elaborated optimal reaction conditions, complementary UV/vis/NIR, EPR, and Raman spectroscopy measurements clearly showed that the reaction mixture obtained after completion of the reaction contained PANI-ES-like products as dominating species, very similar to the products formed with HRPC as catalyst. HEPES (4-(2-hydroxyethyl)-1-piperazineethanesulfonate) was found to have a positive effect on the reaction rate as compared to dihydrogenphosphate. This work is the first on the template-assisted formation of PANI-ES type products under mild, environmentally friendly conditions using hemin as a cost-effective catalyst. Polyaniline emeraldine salt-type products were synthesized under mild, environmentally friendly conditions using hemin as a cost-effective catalyst, p-aminodiphenylamine (PADPA) as a monomer, and micelles formed from SDBS as templates.![]()
Collapse
Affiliation(s)
- Nemanja Cvjetan
- Department of Materials, Laboratory for Multifunctional Materials, ETH Zürich Vladimir-Prelog-Weg 5 8093 Zürich Switzerland
| | - Reinhard Kissner
- Department of Chemistry and Applied Biosciences, Laboratory of Inorganic Chemistry Vladimir-Prelog-Weg 2 8093 Zürich Switzerland
| | - Danica Bajuk-Bogdanović
- Faculty of Physical Chemistry, University of Belgrade Studentski trg 12-16 11158 Belgrade Serbia
| | - Gordana Ćirić-Marjanović
- Faculty of Physical Chemistry, University of Belgrade Studentski trg 12-16 11158 Belgrade Serbia
| | - Peter Walde
- Department of Materials, Laboratory for Multifunctional Materials, ETH Zürich Vladimir-Prelog-Weg 5 8093 Zürich Switzerland
| |
Collapse
|
17
|
Progress of albumin-polymer conjugates as efficient drug carriers. PURE APPL CHEM 2022. [DOI: 10.1515/pac-2021-2006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
Albumin is a protein that has garnered wide attention in nanoparticle-based drug delivery of cancer therapeutics due to its natural abundance and unique cancer-targeting ability. The propensity of albumin to naturally accumulate in tumours, further augmented by the incorporation of targeting ligands, has made the field of albumin-polymer conjugate development a much pursued one. Polymerization techniques such as RAFT and ATRP have paved the path to incorporate various polymers in the design of albumin-polymer hybrids, indicating the advancement of the field since the first instance of PEGylated albumin in 1977. The synergistic combination of albumin and polymer endows manifold features to these macromolecular hybrids to evolve as next generation therapeutics. The current review is successive to our previously published review on drug delivery vehicles based on albumin-polymer conjugates and aims to provide an update on the progress of albumin-polymer conjugates. This review also highlights the alternative of exploring albumin-polymer conjugates formed via supramolecular, non-covalent interactions. Albumin-based supramolecular polymer systems provide a versatile platform for functionalization, thereby, holding great potential in enhancing cytotoxicity and controlled delivery of therapeutic agents.
Collapse
|
18
|
Variations in the Human Serum Albumin Gene: Molecular and Functional Aspects. Int J Mol Sci 2022; 23:ijms23031159. [PMID: 35163085 PMCID: PMC8835714 DOI: 10.3390/ijms23031159] [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: 12/24/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 01/14/2023] Open
Abstract
The human albumin gene, the most abundant serum protein, is located in the long arm of chromosome 4, near the centromere, position 4q11–3. It is divided by 14 intervening introns into 15 exons, the last of which is untranslated. To date, 74 nucleotide substitutions (mainly missense) have been reported, determining the circulating variants of albumin or pre-albumin. In a heterozygous state, this condition is known as alloalbuminaemia or bisalbuminaemia (OMIM # 103600). The genetic variants are not associated with disease, neither in the heterozygous nor in the homozygous form. Only the variants resulting in familial dysalbuminaemic hyperthyroxinaemia and hypertriiodothyroninaemia are of clinical relevance because affected individuals are at risk of inappropriate treatment or may have adverse drug effects. In 28 other cases, the pathogenic variants (mainly affecting splicing, nonsense, and deletions), mostly in the homozygous form, cause a premature stop in the synthesis of the protein and lead to the condition known as congenital analbuminaemia. In this review, we will summarize the current knowledge of genetic and molecular aspects, functional consequences and potential therapeutic uses of the variants. We will also discuss the molecular defects resulting in congenital analbuminaemia, as well as the biochemical and clinical features of this rare condition
Collapse
|
19
|
Divergent roles of haptoglobin and hemopexin deficiency for disease progression of Shiga-toxin-induced hemolytic-uremic syndrome in mice. Kidney Int 2022; 101:1171-1185. [PMID: 35031328 DOI: 10.1016/j.kint.2021.12.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 12/05/2021] [Accepted: 12/16/2021] [Indexed: 02/06/2023]
Abstract
Thrombotic microangiopathy, hemolysis and acute kidney injury are typical clinical characteristics of hemolytic-uremic syndrome (HUS), which is predominantly caused by Shiga-toxin-producing Escherichia coli. Free heme aggravates organ damage in life-threatening infections, even with a low degree of systemic hemolysis. Therefore, we hypothesized that the presence of the hemoglobin- and the heme-scavenging proteins, haptoglobin and hemopexin, respectively impacts outcome and kidney pathology in HUS. Here, we investigated the effect of haptoglobin and hemopexin deficiency (haptoglobin-/-, hemopexin-/-) and haptoglobin treatment in a murine model of HUS-like disease. Seven-day survival was decreased in haptoglobin-/- (25%) compared to wild type mice (71.4%), whereas all hemopexin-/- mice survived. Shiga-toxin-challenged hemopexin-/- mice showed decreased kidney inflammation and attenuated thrombotic microangiopathy, indicated by reduced neutrophil recruitment and platelet deposition. These observations were associated with supranormal haptoglobin plasma levels in hemopexin-/- mice. Low dose haptoglobin administration to Shiga-toxin-challenged wild type mice attenuated kidney platelet deposition and neutrophil recruitment, suggesting that haptoglobin at least partially contributes to the beneficial effects. Surrogate parameters of hemolysis were elevated in Shiga-toxin-challenged wild type and haptoglobin-/- mice, while signs for hepatic hemoglobin degradation like heme oxygenase-1, ferritin and CD163 expression were only increased in Shiga-toxin-challenged wild type mice. In line with this observation, haptoglobin-/- mice displayed tubular iron deposition as an indicator for kidney hemoglobin degradation. Thus, haptoglobin and hemopexin deficiency play divergent roles in Shiga-toxin-mediated HUS, suggesting haptoglobin is involved, and hemopexin is redundant for the resolution of HUS pathology.
Collapse
|
20
|
Szkudlarek A, Pożycka J, Kulig K, Owczarzy A, Rogóż W, Maciążek-Jurczyk M. Changes in Glycated Human Serum Albumin Binding Affinity for Losartan in the Presence of Fatty Acids In Vitro Spectroscopic Analysis. Molecules 2022; 27:401. [PMID: 35056715 PMCID: PMC8778988 DOI: 10.3390/molecules27020401] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/04/2022] [Accepted: 01/06/2022] [Indexed: 01/11/2023] Open
Abstract
Conformational changes in human serum albumin due to numerous modifications that affect its stability and biological activity should be constantly monitored, especially in elderly patients and those suffering from chronic diseases (which include diabetes, obesity, and hypertension). The main goal of this study was to evaluate the effect of a mixture of fatty acids (FA) on the affinity of losartan (LOS, an angiotensin II receptor (AT1) blocker used in hypertension, a first-line treatment with coexisting diabetes) for glycated albumin-simulating the state of diabetes in the body. Individual fatty acid mixtures corresponded to the FA content in the physiological state and in various clinical states proceeding with increased concentrations of saturated (FAS) and unsaturated (FAUS) acids. Based on fluorescence studies, we conclude that LOS interacts with glycated human serum albumin (af)gHSA in the absence and in the presence of fatty acids ((af)gHSAphys, (af)gHSA4S, (af)gHSA8S, (af)gHSA4US, and (af)gHSA8US) and quenches the albumin fluorescence intensity via a static quenching mechanism. LOS not only binds to its specific binding sites in albumins but also non-specifically interacts with the hydrophobic fragments of its surface. Incorrect contents of fatty acids in the body affect the drug pharmacokinetics. A higher concentration of both FAS and FAUS acids in glycated albumin reduces the stability of the complex formed with losartan. The systematic study of FA and albumin interactions using an experimental model mimicking pathological conditions in the body may result in new tools for personalized pharmacotherapy.
Collapse
Affiliation(s)
- Agnieszka Szkudlarek
- Department of Physical Pharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 40-055 Katowice, Poland; (J.P.); (K.K.); (A.O.); (W.R.); (M.M.-J.)
| | | | | | | | | | | |
Collapse
|
21
|
Badawy MA, Yasseen BA, El-Messiery RM, Abdel-Rahman EA, Elkhodiry AA, Kamel AG, El-Sayed H, Shedra AM, Hamdy R, Zidan M, Al-Raawi D, Hammad M, Elsharkawy N, El Ansary M, Al-Halfawy A, Elhadad A, Hatem A, Abouelnaga S, Dugan LL, Ali SS. Neutrophil-mediated oxidative stress and albumin structural damage predict COVID-19-associated mortality. eLife 2021; 10:69417. [PMID: 34821549 PMCID: PMC8641949 DOI: 10.7554/elife.69417] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 11/24/2021] [Indexed: 12/15/2022] Open
Abstract
Human serum albumin (HSA) is the frontline antioxidant protein in blood with established anti-inflammatory and anticoagulation functions. Here, we report that COVID-19-induced oxidative stress inflicts structural damages to HSA and is linked with mortality outcome in critically ill patients. We recruited 39 patients who were followed up for a median of 12.5 days (1–35 days), among them 23 had died. Analyzing blood samples from patients and healthy individuals (n=11), we provide evidence that neutrophils are major sources of oxidative stress in blood and that hydrogen peroxide is highly accumulated in plasmas of non-survivors. We then analyzed electron paramagnetic resonance spectra of spin-labeled fatty acids (SLFAs) bound with HSA in whole blood of control, survivor, and non-survivor subjects (n=10–11). Non-survivors’ HSA showed dramatically reduced protein packing order parameter, faster SLFA correlational rotational time, and smaller S/W ratio (strong-binding/weak-binding sites within HSA), all reflecting remarkably fluid protein microenvironments. Following loading/unloading of 16-DSA, we show that the transport function of HSA may be impaired in severe patients. Stratified at the means, Kaplan–Meier survival analysis indicated that lower values of S/W ratio and accumulated H2O2 in plasma significantly predicted in-hospital mortality (S/W≤0.15, 81.8% (18/22) vs. S/W>0.15, 18.2% (4/22), p=0.023; plasma [H2O2]>8.6 μM, 65.2% (15/23) vs. 34.8% (8/23), p=0.043). When we combined these two parameters as the ratio ((S/W)/[H2O2]) to derive a risk score, the resultant risk score lower than the mean (<0.019) predicted mortality with high fidelity (95.5% (21/22) vs. 4.5% (1/22), log-rank χ2=12.1, p=4.9×10−4). The derived parameters may provide a surrogate marker to assess new candidates for COVID-19 treatments targeting HSA replacements and/or oxidative stress.
Collapse
Affiliation(s)
| | - Basma A Yasseen
- Research Department, Children's Cancer Hospital, Cairo, Egypt
| | - Riem M El-Messiery
- Infectious Disease Unit, Internal Medicine Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Engy A Abdel-Rahman
- Research Department, Children's Cancer Hospital, Cairo, Egypt.,Pharmacology Department, Faculty of Medicine, Assuit University, Assuit, Egypt
| | - Aya A Elkhodiry
- Research Department, Children's Cancer Hospital, Cairo, Egypt
| | - Azza G Kamel
- Research Department, Children's Cancer Hospital, Cairo, Egypt
| | - Hajar El-Sayed
- Research Department, Children's Cancer Hospital, Cairo, Egypt
| | - Asmaa M Shedra
- Research Department, Children's Cancer Hospital, Cairo, Egypt
| | - Rehab Hamdy
- Research Department, Children's Cancer Hospital, Cairo, Egypt
| | - Mona Zidan
- Research Department, Children's Cancer Hospital, Cairo, Egypt
| | - Diaa Al-Raawi
- Research Department, Children's Cancer Hospital, Cairo, Egypt
| | - Mahmoud Hammad
- Pediatric Oncology Department, National Cancer Institute, Cairo University and Children's Cancer Hospital, Cairo, Egypt
| | - Nahla Elsharkawy
- Clinical pathology department, National Cancer Institute, Cairo University and Children's Cancer Hospital, Cairo, Egypt
| | - Mohamed El Ansary
- Department of Intensive Care, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Ahmed Al-Halfawy
- Department of Pulmonary Medicine, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Alaa Elhadad
- Pediatric Oncology Department, National Cancer Institute, Cairo University and Children's Cancer Hospital, Cairo, Egypt
| | - Ashraf Hatem
- Department of Chest Diseases, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Sherif Abouelnaga
- Pediatric Oncology Department, National Cancer Institute, Cairo University and Children's Cancer Hospital, Cairo, Egypt
| | - Laura L Dugan
- Division of Geriatric Medicine, Department of Medicine, Vanderbilt University Medical Center; and VATennessee Valley Geriatric Research, Education and Clinical Center (GRECC), Nashville, United States
| | - Sameh Saad Ali
- Research Department, Children's Cancer Hospital, Cairo, Egypt
| |
Collapse
|
22
|
Buehler PW, Swindle D, Pak DI, Ferguson SK, Majka SM, Karoor V, Moldovan R, Sintas C, Black J, Gentinetta T, Buzzi RM, Vallelian F, Wassmer A, Edler M, Bain J, Schu D, Hassell K, Nuss R, Schaer DJ, Irwin DC. Hemopexin dosing improves cardiopulmonary dysfunction in murine sickle cell disease. Free Radic Biol Med 2021; 175:95-107. [PMID: 34478834 PMCID: PMC9231663 DOI: 10.1016/j.freeradbiomed.2021.08.238] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/26/2021] [Accepted: 08/29/2021] [Indexed: 01/13/2023]
Abstract
Hemopexin (Hpx) is a crucial defense protein against heme liberated from degraded hemoglobin during hemolysis. High heme stress creates an imbalance in Hpx bioavailability, favoring heme accumulation and downstream pathophysiological responses leading to cardiopulmonary disease progression in sickle cell disease (SCD) patients. Here, we evaluated a model of murine SCD, which was designed to accelerate red blood cell sickling, pulmonary hypertension, right ventricular dysfunction, and exercise intolerance by exposure of the mice to moderate hypobaric hypoxia. The sequence of pathophysiology in this model tracks with circulatory heme accumulation, lipid oxidation, extensive remodeling of the pulmonary vasculature, and fibrosis. We hypothesized that Hpx replacement for an extended period would improve exercise tolerance measured by critical speed as a clinically meaningful therapeutic endpoint. Further, we sought to define the effects of Hpx on upstream cardiopulmonary function, histopathology, and tissue oxidation. Our data shows that tri-weekly administrations of Hpx for three months dose-dependently reduced heme exposure and pulmonary hypertension while improving cardiac pressure-volume relationships and exercise tolerance. Furthermore, Hpx administration dose-dependently attenuated pulmonary fibrosis and oxidative modifications in the lung and myocardium of the right ventricle. Observations in our SCD murine model are consistent with pulmonary vascular and right ventricular pathology at autopsy in SCD patients having suffered from severe pulmonary hypertension, right ventricular dysfunction, and sudden cardiac death. This study provides a translational evaluation supported by a rigorous outcome analysis demonstrating therapeutic proof-of-concept for Hpx replacement in SCD.
Collapse
Affiliation(s)
- Paul W Buehler
- University of Maryland, Department of Pathology and the Center for Blood Oxygen Transport, Department of Pediatrics, School of Medicine, Baltimore, MD, USA.
| | - Delaney Swindle
- Cardiovascular and Pulmonary Research Laboratory, Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - David I Pak
- Cardiovascular and Pulmonary Research Laboratory, Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Scott K Ferguson
- Cardiovascular and Pulmonary Research Laboratory, Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA; Department of Kinesiology and Exercise Sciences, College of Natural and Health Sciences, University of Hawaii at Hilo, Hilo, HI, USA
| | - Susan M Majka
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Hospital, Denver, CO, USA
| | - Vijaya Karoor
- Cardiovascular and Pulmonary Research Laboratory, Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Radu Moldovan
- Advanced Light Microscopy Core, CU Anschutz Medical Campus, Aurora,, CO, USA
| | - Chantal Sintas
- Department of Pathology and Laboratory Medicine at Children's Hospital Colorado, USA
| | - Jennifer Black
- Department of Pathology, Pediatrics, University of Colorado School of Medicine, USA
| | | | - Raphael M Buzzi
- Division of Internal Medicine, University and University Hospital of Zurich, Zurich, Switzerland
| | - Florence Vallelian
- Division of Internal Medicine, University and University Hospital of Zurich, Zurich, Switzerland
| | | | - Monika Edler
- CSL Behring AG, Research and Development, Bern, Switzerland
| | - Joseph Bain
- CSL Behring AG, Innovations GmbH, Marburg, Germany
| | - Daniel Schu
- CSL Behring AG, Innovations GmbH, Marburg, Germany
| | - Kathryn Hassell
- Division of Hematology Colorado Sickle Cell Treatment and Research Center, School of Medicine, Anschutz Medical Campus, University of Colorado-Denver School of Medicine, Aurora,, CO, USA
| | - Rachelle Nuss
- Division of Hematology Colorado Sickle Cell Treatment and Research Center, School of Medicine, Anschutz Medical Campus, University of Colorado-Denver School of Medicine, Aurora,, CO, USA
| | - Dominik J Schaer
- Division of Internal Medicine, University and University Hospital of Zurich, Zurich, Switzerland
| | - David C Irwin
- Cardiovascular and Pulmonary Research Laboratory, Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
23
|
Lysine reactivity profiling reveals molecular insights into human serum albumin-small-molecule drug interactions. Anal Bioanal Chem 2021; 413:7431-7440. [PMID: 34676431 DOI: 10.1007/s00216-021-03700-1] [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: 07/04/2021] [Revised: 08/31/2021] [Accepted: 09/27/2021] [Indexed: 10/20/2022]
Abstract
Human serum albumin (HSA) is one of the most important serum carrier proteins that deliver small-molecule drugs to their specific targets. Clarifying the molecular mechanism of the interaction between natural HSA and drugs in an aqueous solution has been a hot topic in pharmaceutical chemistry, clinical medicine, and biochemistry in recent years, but it is still challenging. In this paper, the details of molecular interactions of HSA with a variety of therapeutic drugs including ibuprofen, indomethacin, phenylbutazone, and warfarin are systematically investigated using a mass spectrometry (MS)-based lysine reactivity profiling (LRP) strategy. The results reaffirm that the major ligand binding sites (including Sites I and II) of HSA are located in subdomains IIA and IIIA, while several potential drug-binding areas at subdomain IIIB and α helix IIB-IIIA are newly characterized. The MS-LRP strategy may have important application prospects in pharmacodynamics, pharmacokinetics, and safety evaluation of small-molecule drugs.
Collapse
|
24
|
Serum Albumin: A Multifaced Enzyme. Int J Mol Sci 2021; 22:ijms221810086. [PMID: 34576249 PMCID: PMC8466385 DOI: 10.3390/ijms221810086] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 02/06/2023] Open
Abstract
Human serum albumin (HSA) is the most abundant protein in plasma, contributing actively to oncotic pressure maintenance and fluid distribution between body compartments. HSA acts as the main carrier of fatty acids, recognizes metal ions, affects pharmacokinetics of many drugs, provides the metabolic modification of some ligands, renders potential toxins harmless, accounts for most of the anti-oxidant capacity of human plasma, and displays esterase, enolase, glucuronidase, and peroxidase (pseudo)-enzymatic activities. HSA-based catalysis is physiologically relevant, affecting the metabolism of endogenous and exogenous compounds including proteins, lipids, cholesterol, reactive oxygen species (ROS), and drugs. Catalytic properties of HSA are modulated by allosteric effectors, competitive inhibitors, chemical modifications, pathological conditions, and aging. HSA displays anti-oxidant properties and is critical for plasma detoxification from toxic agents and for pro-drugs activation. The enzymatic properties of HSA can be also exploited by chemical industries as a scaffold to produce libraries of catalysts with improved proficiency and stereoselectivity for water decontamination from poisonous agents and environmental contaminants, in the so called “green chemistry” field. Here, an overview of the intrinsic and metal dependent (pseudo-)enzymatic properties of HSA is reported to highlight the roles played by this multifaced protein.
Collapse
|
25
|
Moreno S, Boye S, Ajeilat HGA, Michen S, Tietze S, Voit B, Lederer A, Temme A, Appelhans D. Multivalent Protein-Loaded pH-Stable Polymersomes: First Step toward Protein Targeted Therapeutics. Macromol Biosci 2021; 21:e2100102. [PMID: 34355506 DOI: 10.1002/mabi.202100102] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 07/22/2021] [Indexed: 12/19/2022]
Abstract
Synthetic platforms for mimicking artificial organelles or for designing multivalent protein therapeutics for targeting cell surface, extracellular matrix, and tissues are in the focus of this study. Furthermore, the availability of a multi-functionalized and stimuli-responsive carrier system is required that can be used for sequential in situ and/or post loading of different proteins combined with post-functionalization steps. Until now, polymersomes exhibit excellent key characteristics to fulfill those requirements, which allow specific transport of proteins and the integration of proteins in different locations of polymeric vesicles. Herein, different approaches to fabricate multivalent protein-loaded, pH-responsive, and pH-stable polymersomes are shown, where a combination of therapeutic action and targeting can be achieved, by first choosing two model proteins such as human serum albumin and avidin. Validation of the molecular parameters of the multivalent biohybrids is performed by dynamic light scattering, cryo-TEM, fluorescence spectroscopy, and asymmetrical flow-field flow fractionation combined with light scattering techniques. To demonstrate targeting functions of protein-loaded polymersomes, avidin post-functionalized polymersomes are used for the molecular recognition of biotinylated cell surface receptors. These versatile protein-loaded polymersomes present new opportunities for designing sophisticated biomolecular nanoobjects in the field of (extracellular matrix) protein therapeutics.
Collapse
Affiliation(s)
- Silvia Moreno
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Straße 6, Dresden, 01069, Germany
| | - Susanne Boye
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Straße 6, Dresden, 01069, Germany
| | | | - Susanne Michen
- Department of Neurosurgery, Section Experimental Neurosurgery/Tumor Immunology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, 01307, Germany
| | - Stefanie Tietze
- Department of Neurosurgery, Section Experimental Neurosurgery/Tumor Immunology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, 01307, Germany
| | - Brigitte Voit
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Straße 6, Dresden, 01069, Germany.,Faculty of Chemistry, Technische Universität Dresden, Dresden, 01062, Germany
| | - Albena Lederer
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Straße 6, Dresden, 01069, Germany.,Department of Chemistry and Polymer Science, Stellenbosch University, Private Bag X1, Matieland, 7602, South Africa
| | - Achim Temme
- Department of Neurosurgery, Section Experimental Neurosurgery/Tumor Immunology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, 01307, Germany.,German Cancer Consortium (DKTK), partner site Dresden, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany, National Center for Tumor Diseases (NCT), Fetscherstraße 74, Dresden, 01307, Germany
| | - Dietmar Appelhans
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Straße 6, Dresden, 01069, Germany
| |
Collapse
|
26
|
Structural and Biochemical Features of Human Serum Albumin Essential for Eukaryotic Cell Culture. Int J Mol Sci 2021; 22:ijms22168411. [PMID: 34445120 PMCID: PMC8395139 DOI: 10.3390/ijms22168411] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/30/2021] [Accepted: 08/04/2021] [Indexed: 12/16/2022] Open
Abstract
Serum albumin physically interacts with fatty acids, small molecules, metal ions, and several other proteins. Binding with a plethora of bioactive substances makes it a critical transport molecule. Albumin also scavenges the reactive oxygen species that are harmful to cell survival. These properties make albumin an excellent choice to promote cell growth and maintain a variety of eukaryotic cells under in vitro culture environment. Furthermore, purified recombinant human serum albumin is mostly free from impurities and modifications, providing a perfect choice as an additive in cell and tissue culture media while avoiding any regulatory constraints. This review discusses key features of human serum albumin implicated in cell growth and survival under in vitro conditions.
Collapse
|
27
|
Johnson AS, Winlow W. COVID-19 vulnerabilities are intensified by declining human serum albumin levels. Exp Physiol 2021; 107:674-682. [PMID: 34275164 PMCID: PMC8447469 DOI: 10.1113/ep089703] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 07/12/2021] [Indexed: 12/16/2022]
Abstract
What is the topic of this review? Human serum albumin (HSA) a common factor in COVID‐19 vulnerabilities. What advances does it highlight? Understanding of HSA capacity, and systemic vulnerabilities to COVID‐19. Raising HSA in COVID‐19 patients may alleviate systemic injury caused by diminished native HSA binding. A change in fluid therapy administration into the portal system of the liver is proposed to safely raise HSA levels.
Collapse
Affiliation(s)
- Andrew S Johnson
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - William Winlow
- Department of Biology, University of Naples Federico II, Naples, Italy.,Institute of Ageing and Chronic Diseases, University of Liverpool, Liverpool, UK
| |
Collapse
|
28
|
Kang N, Pei S, Zhang C, Zhang G, Zhou Y, Fan L, Yao Q, Wang W, Shuang S, Dong C. A red emitting fluorescent probe based on TICT for selective detection and imaging of HSA. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2021; 250:119409. [PMID: 33422865 DOI: 10.1016/j.saa.2020.119409] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/21/2020] [Accepted: 12/29/2020] [Indexed: 06/12/2023]
Abstract
A red emitting fluorescence probe, TPA-CPO, based on twisted intra-molecular charge transfer (TICT) was designed and synthesized. The spectra results displayed that TPA-CPO could sense HSA with excellent properties including significant fluorescence enhancement, long emission wavelength, large stokes shift, and wide linear range. The recognition mechanism was proved that TPA-CPO could bind to domain IB of HSA and its TICT process was suppressed by utilizing hydrophobic cavity and low polarity of HSA. TPA-CPO bind to domain IB instead of common drug sites of HSA could effectively avoid interference from most drugs. The selective response of TPA-CPO allowed quantitative detection of HSA with sensitivity limit of 13.65 µg/mL. What's more, it successfully achieved HSA imaging in HeLa cells.
Collapse
Affiliation(s)
- Na Kang
- School of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, China
| | - Shizeng Pei
- School of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, China
| | - Caihong Zhang
- School of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, China.
| | - Guomei Zhang
- School of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, China
| | - Ying Zhou
- School of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, China
| | - Li Fan
- Institute of Environmental Science, Shanxi University, Taiyuan 030006, China
| | - QingJia Yao
- School of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, China
| | - Wen Wang
- School of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, China; Institute of Acoustics, Chinese Academy of Sciences, Beijing 100190, China
| | - Shaomin Shuang
- School of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, China
| | - Chuan Dong
- Institute of Environmental Science, Shanxi University, Taiyuan 030006, China.
| |
Collapse
|
29
|
Hopp MT, Imhof D. Linking Labile Heme with Thrombosis. J Clin Med 2021; 10:427. [PMID: 33499296 PMCID: PMC7865584 DOI: 10.3390/jcm10030427] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/14/2021] [Accepted: 01/19/2021] [Indexed: 12/14/2022] Open
Abstract
Thrombosis is one of the leading causes of death worldwide. As such, it also occurs as one of the major complications in hemolytic diseases, like hemolytic uremic syndrome, hemorrhage and sickle cell disease. Under these conditions, red blood cell lysis finally leads to the release of large amounts of labile heme into the vascular compartment. This, in turn, can trigger oxidative stress and proinflammatory reactions. Moreover, the heme-induced activation of the blood coagulation system was suggested as a mechanism for the initiation of thrombotic events under hemolytic conditions. Studies of heme infusion and subsequent thrombotic reactions support this assumption. Furthermore, several direct effects of heme on different cellular and protein components of the blood coagulation system were reported. However, these effects are controversially discussed or not yet fully understood. This review summarizes the existing reports on heme and its interference in coagulation processes, emphasizing the relevance of considering heme in the context of the treatment of thrombosis in patients with hemolytic disorders.
Collapse
Affiliation(s)
| | - Diana Imhof
- Pharmaceutical Biochemistry and Bioanalytics, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany;
| |
Collapse
|
30
|
Zhou L, Luo M, Tian R, Zeng XP, Peng YY, Lu N. Generation of a Bovine Serum Albumin-Diligand Complex for the Protection of Bioactive Quercetin and Suppression of Heme Toxicity. Chem Res Toxicol 2021; 34:920-928. [PMID: 33464047 DOI: 10.1021/acs.chemrestox.0c00537] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
As an abundant protein in milk and blood serum, bovine serum albumin (BSA) contains various sites to bind a lot of bioactive components, generating BSA-monoligand complex. Demonstration of the interaction between BSA and bioactive components (such as heme, flavonoids) is important to develop effective carrier for the protection of bioactive ligands and to reduce cytotoxicity of heme. Herein, the bindings of BSA to quercetin and/or heme were investigated by multispectroscopic and molecular docking methods. The fluorescence of protein was significantly quenched by both quercetin and heme in a static mode (i.e., generation of BSA-ligand complex). Although quercetin had lower affinity to protein than heme, the interactions of both compounds with protein did locate in site I (i.e., subdomain IIA). BSA-diligand complex was successfully generated after the coaddition of quercetin and heme. The cytotoxicity of free heme to endothelial cells was reduced in the BSA-diligand complex relative to that of heme or BSA-monoligand complex, while the stability of bioactive quercetin was promoted in the complex relative to free flavonoid. The complex provided a better inhibition on the cytotoxicity of heme than BSA-monoligand complex, in which the copresence of quercetin played a vital role.
Collapse
Affiliation(s)
- Lan Zhou
- Key Laboratory of Functional Small Organic Molecule, Ministry of Education; College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, Jiangxi China
| | - Mengjuan Luo
- Key Laboratory of Functional Small Organic Molecule, Ministry of Education; College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, Jiangxi China
| | - Rong Tian
- Key Laboratory of Functional Small Organic Molecule, Ministry of Education; College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, Jiangxi China
| | - Xing-Ping Zeng
- Key Laboratory of Functional Small Organic Molecule, Ministry of Education; College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, Jiangxi China
| | - Yi-Yuan Peng
- Key Laboratory of Functional Small Organic Molecule, Ministry of Education; College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, Jiangxi China
| | - Naihao Lu
- Key Laboratory of Functional Small Organic Molecule, Ministry of Education; College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, Jiangxi China
| |
Collapse
|
31
|
Gáll T, Pethő D, Nagy A, Balla G, Balla J. Therapeutic Potential of Carbon Monoxide (CO) and Hydrogen Sulfide (H 2S) in Hemolytic and Hemorrhagic Vascular Disorders-Interaction between the Heme Oxygenase and H 2S-Producing Systems. Int J Mol Sci 2020; 22:ijms22010047. [PMID: 33374506 PMCID: PMC7793096 DOI: 10.3390/ijms22010047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/18/2020] [Accepted: 12/19/2020] [Indexed: 02/07/2023] Open
Abstract
Over the past decades, substantial work has established that hemoglobin oxidation and heme release play a pivotal role in hemolytic/hemorrhagic disorders. Recent reports have shown that oxidized hemoglobins, globin-derived peptides, and heme trigger diverse biological responses, such as toll-like receptor 4 activation with inflammatory response, reprogramming of cellular metabolism, differentiation, stress, and even death. Here, we discuss these cellular responses with particular focus on their mechanisms that are linked to the pathological consequences of hemorrhage and hemolysis. In recent years, endogenous gasotransmitters, such as carbon monoxide (CO) and hydrogen sulfide (H2S), have gained a lot of interest in connection with various human pathologies. Thus, many CO and H2S-releasing molecules have been developed and applied in various human disorders, including hemolytic and hemorrhagic diseases. Here, we discuss our current understanding of oxidized hemoglobin and heme-induced cell and tissue damage with particular focus on inflammation, cellular metabolism and differentiation, and endoplasmic reticulum stress in hemolytic/hemorrhagic human diseases, and the potential beneficial role of CO and H2S in these pathologies. More detailed mechanistic insights into the complex pathology of hemolytic/hemorrhagic diseases through heme oxygenase-1/CO as well as H2S pathways would reveal new therapeutic approaches that can be exploited for clinical benefit.
Collapse
Affiliation(s)
- Tamás Gáll
- Division of Nephrology, Department of Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (T.G.); (D.P.); (A.N.)
- HAS-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, University of Debrecen, 4032 Debrecen, Hungary;
| | - Dávid Pethő
- Division of Nephrology, Department of Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (T.G.); (D.P.); (A.N.)
- Faculty of Medicine, University of Debrecen, Kálmán Laki Doctoral School, 4032 Debrecen, Hungary
| | - Annamária Nagy
- Division of Nephrology, Department of Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (T.G.); (D.P.); (A.N.)
- Faculty of Medicine, University of Debrecen, Kálmán Laki Doctoral School, 4032 Debrecen, Hungary
| | - György Balla
- HAS-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, University of Debrecen, 4032 Debrecen, Hungary;
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - József Balla
- Division of Nephrology, Department of Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (T.G.); (D.P.); (A.N.)
- Correspondence: ; Tel.: +36-52-255-500/55004
| |
Collapse
|
32
|
Shityakov S, Fischer A, Su KP, Hussein AA, Dandekar T, Broscheit J. Novel Approach for Characterizing Propofol Binding Affinities to Serum Albumins from Different Species. ACS OMEGA 2020; 5:25543-25551. [PMID: 33073080 PMCID: PMC7557242 DOI: 10.1021/acsomega.0c01295] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 05/25/2020] [Indexed: 05/09/2023]
Abstract
The interaction between the main carrier (serum albumin, SA) of endogenous and exogenous compounds in the bloodstream of different species (human, bovine, canine, rat, rabbit, and sheep) and a general anesthetic agent (propofol, PR) was investigated using an experimental technique (high-performance liquid chromatography) and computational methods (molecular docking, molecular dynamics, sequence, and phylogenetic analyses). The obtained results revealed the differences in the PR binding affinity to various homologous forms of this protein with reliable statistics (R 2 = 0.9 and p-value < 0.005), correlating with the evolutionary relationships among SAs from different species. Additionally, the protein conformational changes (root-mean-square deviation ≈ 1.0 Å) and amino acid conservation of binding sites in protein domains were detected, contributing to the SA-PR binding modes. Overall, the outcomes from this study might provide a novel methodology to assess protein-ligand interactions and to gain some interesting insights into drug pharmacokinetics and pharmacodynamics to explain its variations among different species.
Collapse
Affiliation(s)
- Sergey Shityakov
- Department
of Psychiatry and Mind-Body Interface Laboratory (MBI-Lab), China Medical University Hospital, Taichung 40402, Taiwan
- Department
of Bioinformatics, Würzburg University, Würzburg 97074, Germany
- College
of Medicine, China Medical University, Taichung 404, Taiwan
- . Phone: +49-931-318-4550. Fax: +49-931-318-4552
| | - Anneli Fischer
- Department
of Anesthesia and Critical Care, Würzburg
University Hospital, Würzburg 97080, Germany
| | - Kuan-Pin Su
- Department
of Psychiatry and Mind-Body Interface Laboratory (MBI-Lab), China Medical University Hospital, Taichung 40402, Taiwan
- College
of Medicine, China Medical University, Taichung 404, Taiwan
| | - Aqeel A. Hussein
- Faculty
of Dentistry, University of Al-Ameed, 56001 Karbala, Iraq
- Department
of Chemistry, University of Southampton, Southampton SO17 1BJ, U.K.
| | - Thomas Dandekar
- Department
of Bioinformatics, Würzburg University, Würzburg 97074, Germany
- Phone: +49 (0)931 31-84551. Fax: +49-931-318-4552
| | - Jens Broscheit
- Department
of Anesthesia and Critical Care, Würzburg
University Hospital, Würzburg 97080, Germany
| |
Collapse
|
33
|
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
|
34
|
Amoorahim M, Ashrafi-Kooshk MR, Esmaeili S, Shahlaei M, Moradi S, Khodarahmi R. Physiological changes in the albumin-bound non-esterified free fatty acids critically influence heme/bilirubin binding properties of the protein: A comparative, in vitro, spectroscopic study using the endogenous biomolecules. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2020; 235:118298. [PMID: 32294588 DOI: 10.1016/j.saa.2020.118298] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/21/2020] [Accepted: 03/23/2020] [Indexed: 06/11/2023]
Abstract
Heme and bilirubin (BR), as by-products of red blood cells (and hemoglobin) degradation, show increased plasma concentrations in some diseases. These two toxic hydrophobic molecules are mainly transported in the blood-stream by human serum albumin (HSA) that carries a wide variety of ligands. Under normal physiological conditions, ~3 fatty acid (FA) molecules are bound to each HSA; and its possible effect on BR/heme binding remains to be more clarified. In the present study, to provide deeper insight on this issue, we purified albumin from healthy individuals (as purified non-defatted albumin or PA) with normal plasma levels of FA, then defatted some of the purified protein (as defatted-HSA; or DA). In the next step, using various spectroscopic methods, their interactions with heme and BR were investigated. By 1: 1 binding of the ligands, quenching and thermodynamic analysis of parameters indicated that binding constants (Kb) values of bilirubin and heme for PA and DA are different. It could be perceived that the presence of FAs in high-affinity FA binding sites (FABSs) exerted considerable conformational changes in the structure followed by an improved BR binding while hindered heme interaction. The data was confirmed by determining surface hydrophobicity of the purified albumin (PA) and DA, and then supported by bioinformatics analyses. The physiological and clinical relevance of the observed dynamic interactions is also discussed. This study, also, re-confirmed that the primary BR binding site is subdomain IIA not subdomain IB.
Collapse
Affiliation(s)
- Mahtab Amoorahim
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Reza Ashrafi-Kooshk
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sajjad Esmaeili
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohsen Shahlaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sajad Moradi
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Reza Khodarahmi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Pharmacognosy and Biotechnology, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
35
|
Caviglia H, Daffunchio C, Galatro G, Cambiaggi G, Oneto P, Douglas Price AL, Landro ME, Etulain J. Inhibition of Fenton reaction is a novel mechanism to explain the therapeutic effect of intra-articular injection of PRP in patients with chronic haemophilic synovitis. Haemophilia 2020; 26:e187-e193. [PMID: 32530133 DOI: 10.1111/hae.14075] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/13/2020] [Accepted: 05/20/2020] [Indexed: 12/13/2022]
Abstract
INTRODUCTION AND AIM Haemarthroses cause major morbidity in haemophilia resulting in chronic haemophilic synovitis (CHS) and arthropathy. Oxidation of haemoglobin-coupled iron released in synovium after haemolysis induces chondrocytes death and cartilage damage, allowing postulate using iron-chelating drugs as potential therapeutic tool for haemophilic joint damage. Considering that albumin, the most abundant plasma protein, is a physiologic iron chelator, we aim to demonstrate that impediment of haemoglobin oxidation is exerted by plasma as a mechanism involved in the therapeutic effect of intra-articular injection of platelet-rich plasma in CHS. METHODS Oxidation of haemoglobin (Hb) to methaemoglobin (MeHb) through Fenton reaction was induced in vitro by addition of potassium ferricyanide in the presence or absence of peripheral blood-derived platelets-rich or platelets-poor plasma (PRP/PPP) or albumin. The relevance of in vitro findings was analysed in synovial fluid (SF) samples from one patient with CHS obtained before and after 6 months of PRP intra-articular injection. RESULTS MeHb formation was completely impaired either by of PPP, PRP or albumin indicating that PRP exerts an anti-oxidative effect, probably due by plasma albumin. Analysis of SF samples revealed the presence of MeHb levels and haemosiderin-laden macrophages in SF obtained before PRP treatment. Reduction of synovial MeHb, normalization of cellular composition and improvement of health joint haemophilic score, pain and bleeding episodes were registered after 6 months of PRP intra-articular injection. CONCLUSION Inhibition of Fenton reaction and the consequent normalization of joint cellular composition is a noncanonical mechanism underlying the therapeutic effect of PRP intra-articular injection in CHS.
Collapse
Affiliation(s)
| | | | | | | | - Paula Oneto
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET, National Academy of Medicine, CABA, Argentina
| | | | | | - Julia Etulain
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET, National Academy of Medicine, CABA, Argentina
| |
Collapse
|
36
|
Munoz CJ, Pires IS, Baek JH, Buehler PW, Palmer AF, Cabrales P. Apohemoglobin-haptoglobin complex attenuates the pathobiology of circulating acellular hemoglobin and heme. Am J Physiol Heart Circ Physiol 2020; 318:H1296-H1307. [PMID: 32302494 DOI: 10.1152/ajpheart.00136.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Haptoglobin (Hp) is the plasma protein that binds and clears cell-free hemoglobin (Hb), whereas apohemoglobin (apoHb, i.e., Hb devoid of heme) can bind heme. Therefore, the apoHb-Hp protein complex should facilitate holoHb-apoHb αβ-dimer exchange and apoHb-heme intercalation. Thus, we hypothesized that apoHb-Hp could facilitate both Hb and heme clearance, which, if not alleviated, could have severe microcirculatory consequences. In this study, we characterized apoHb-Hp and Hb/heme ligand interactions and assessed their in vivo consequences. Hb exchange and heme binding with the apoHb-Hp complex was studied with transfer assays using size-exclusion high-performance liquid chromatography coupled with UV-visible spectrophotometry. Exchange/transfer experiments were conducted in guinea pigs dosed with Hb or heme-albumin followed by a challenge with equimolar amounts of apoHb-Hp. Finally, systemic and microcirculatory parameters were studied in hamsters instrumented with a dorsal window chamber via intravital microscopy. In vitro and in vivo Hb exchange and heme transfer experiments demonstrated proof-of-concept Hb/heme ligand transfer to apoHb-Hp. Dosing with the apoHb-Hp complex reversed Hb- and heme-induced systemic hypertension and microvascular vasoconstriction, reduced microvascular blood flow, and diminished functional capillary density. Therefore, this study highlights the apoHb-Hp complex as a novel therapeutic strategy to attenuate the adverse systemic and microvascular responses to intravascular Hb and heme exposure.NEW & NOTEWORTHY This study highlights the apoHb-Hp complex as a novel therapeutic strategy to attenuate the adverse systemic and microvascular responses to intravascular Hb and heme exposure. In vitro and in vivo Hb exchange and heme transfer experiments demonstrated proof-of-concept Hb/heme ligand transfer to apoHb-Hp. The apoHb-Hp complex reverses Hb- and heme-induced systemic hypertension and microvascular vasoconstriction, preserves microvascular blood flow, and functional capillary density. In summary, the unique properties of the apoHb-Hp complex prevent adverse systemic and microvascular responses to Hb and heme-albumin exposure and introduce a novel therapeutic approach to facilitate simultaneous removal of extracellular Hb and heme.
Collapse
Affiliation(s)
- Carlos J Munoz
- Department of Bioengineering, University of California San Diego, La Jolla, California
| | - Ivan S Pires
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio
| | - Jin Hyen Baek
- Division of Blood Components and Devices, Office of Blood Research and Review, Laboratory of Biochemistry and Vascular Biology, Food and Drug Administration, Silver Spring, Maryland
| | - Paul W Buehler
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland.,The Center for Blood Oxygen Transport and Hemostasis, Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland
| | - Andre F Palmer
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio
| | - Pedro Cabrales
- Department of Bioengineering, University of California San Diego, La Jolla, California
| |
Collapse
|
37
|
Pires IS, Savla C, Palmer AF. Poly(ethylene glycol) Surface-Conjugated Apohemoglobin as a Synthetic Heme Scavenger. Biomacromolecules 2020; 21:2155-2164. [DOI: 10.1021/acs.biomac.0c00141] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Ivan S. Pires
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Chintan Savla
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Andre F. Palmer
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
38
|
Synthesis and HSA-interaction of a new mixed ligand Cu-isothiosemicarbazonato complex with adenine nucleobase. Polyhedron 2020. [DOI: 10.1016/j.poly.2020.114357] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
39
|
Remy KE, Cortés-Puch I, Sun J, Feng J, Lertora JJ, Risoleo T, Katz J, Basu S, Liu X, Perlegas A, Kim-Shapiro DB, Klein HG, Natanson C, Solomon SB. Haptoglobin therapy has differential effects depending on severity of canine septic shock and cell-free hemoglobin level. Transfusion 2019; 59:3628-3638. [PMID: 31639229 PMCID: PMC8216248 DOI: 10.1111/trf.15567] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 09/12/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND During sepsis, higher plasma cell-free hemoglobin (CFH) levels portend worse outcomes. In sepsis models, plasma proteins that bind CFH improve survival. In our canine antibiotic-treated Staphylococcus aureus pneumonia model, with and without red blood cell (RBC) exchange transfusion, commercial human haptoglobin (Hp) concentrates bound and compartmentalized CFH intravascularly, increased CFH clearance, and lowered iron levels, improving shock, lung injury, and survival. We now investigate in our model how very high CFH levels and treatment time affect Hp's beneficial effects. MATERIALS AND METHODS Two separate canine pneumonia sepsis Hp studies were undertaken: one with exchange transfusion of RBCs after prolonged storage to raise CFH to very high levels and another with rapidly lethal sepsis alone to shorten time to treat. All animals received continuous standard intensive care unit supportive care for 96 hours. RESULTS Older RBCs markedly elevated plasma CFH levels and, when combined with Hp therapy, created supraphysiologic CFH-Hp complexes that did not increase CFH or iron clearance or improve lung injury and survival. In a rapidly lethal bacterial challenge model without RBC transfusion, Hp binding did not increase clearance of complexes or iron or show benefits seen previously in the less lethal model. DISCUSSION High-level CFH-Hp complexes may impair clearance mechanisms and eliminate Hp's beneficial effect during sepsis. Rapidly lethal sepsis narrows the therapeutic window for CFH and iron clearance, also decreasing Hp's beneficial effects. In designing clinical trials, dosing and kinetics may be critical factors if Hp infusion is used to treat sepsis.
Collapse
Affiliation(s)
- Kenneth E. Remy
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland;,Department of Pediatrics, Division of Critical Care, Washington University in St. Louis, St. Louis, Missouri
| | - Irene Cortés-Puch
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland;,Division of Pulmonary, Critical Care and Sleep Medicine, University of California Davis, Sacramento, California
| | - Junfeng Sun
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Jing Feng
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Juan J. Lertora
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana
| | - Thomas Risoleo
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Julia Katz
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Swati Basu
- Department of Biochemistry, Wake Forest University Health Sciences, Winston-Salem, North Carolina
| | - Xiaohua Liu
- Department of Physics, Wake Forest University, Winston-Salem, North Carolina
| | - Andreas Perlegas
- Department of Physics, Wake Forest University, Winston-Salem, North Carolina
| | | | - Harvey G. Klein
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Charles Natanson
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Steven B. Solomon
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
40
|
Chaves OA, Menezes LB, Iglesias BA. Multiple spectroscopic and theoretical investigation of meso-tetra-(4-pyridyl)porphyrin‑ruthenium(II) complexes in HSA-binding studies. Effect of Zn(II) in protein binding. J Mol Liq 2019. [DOI: 10.1016/j.molliq.2019.111581] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
41
|
Yasrebi SA, Takjoo R, Riazi GH. HSA-interaction studies of uranyl complexes of alkyl substituted isothiosemicarbazone. J Mol Struct 2019. [DOI: 10.1016/j.molstruc.2019.04.126] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
42
|
Pratesi A, Cirri D, Fregona D, Ferraro G, Giorgio A, Merlino A, Messori L. Structural Characterization of a Gold/Serum Albumin Complex. Inorg Chem 2019; 58:10616-10619. [DOI: 10.1021/acs.inorgchem.9b01900] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Alessandro Pratesi
- Laboratory of Metals in Medicine (MetMed), Department of Chemistry “U. Schiff”, University of Florence, via della Lastruccia 3, 50019 Sesto Fiorentino, Italy
| | - Damiano Cirri
- Laboratory of Metals in Medicine (MetMed), Department of Chemistry “U. Schiff”, University of Florence, via della Lastruccia 3, 50019 Sesto Fiorentino, Italy
| | - Dolores Fregona
- Department of Chemical Sciences, University of Padova, via Marzolo 1, 35131 Padova, Italy
| | - Giarita Ferraro
- Department of Chemical Sciences, University of Naples Federico II, via Cinthia, 80126 Napoli, Italy
| | - Anna Giorgio
- Department of Chemical Sciences, University of Naples Federico II, via Cinthia, 80126 Napoli, Italy
| | - Antonello Merlino
- Department of Chemical Sciences, University of Naples Federico II, via Cinthia, 80126 Napoli, Italy
| | - Luigi Messori
- Laboratory of Metals in Medicine (MetMed), Department of Chemistry “U. Schiff”, University of Florence, via della Lastruccia 3, 50019 Sesto Fiorentino, Italy
| |
Collapse
|
43
|
Al-Harthi S, Lachowicz JI, Nowakowski ME, Jaremko M, Jaremko Ł. Towards the functional high-resolution coordination chemistry of blood plasma human serum albumin. J Inorg Biochem 2019; 198:110716. [PMID: 31153112 DOI: 10.1016/j.jinorgbio.2019.110716] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 05/07/2019] [Accepted: 05/13/2019] [Indexed: 12/11/2022]
Abstract
Human serum albumin (HSA) is a monomeric, globular, multi-carrier and the most abundant protein in the blood. HSA displays multiple ligand binding sites with extraordinary binding capacity for a wide range of ions and molecules. For decades, HSA's ability to bind to various ligands has led many scientists to study its physiological properties and protein structure; indeed, a better understanding of HSA-ligand interactions in human blood, at the atomic level, will likely foster the development of more potent, and overall more performant, diagnostic and therapeutic tools against serious human disorders such as diabetes, cardiovascular disorders, and cancer. Here, we present a concise overview of the current knowledge of HSA's structural characteristics, and its coordination chemistry with transition metal ions, within the scope and limitations of current techniques and biophysical methods to reach atomic resolution in solution and in blood serum. We also highlight the overwhelming need of a detailed atomistic understanding of HSA dynamic structures and interactions that are transient, weak, multi-site and multi-step, and allosterically affected by each other. Considering the fact that HSA is a current clinical tool for drug delivery systems and a potential contender as molecular cargo and nano-vehicle used in biophysical, clinical and industrial fields, we underline the emerging need for novel approaches to target the dynamic functional coordination chemistry of the human blood serum albumin in solution, at the atomic level.
Collapse
Affiliation(s)
- Samah Al-Harthi
- King Abdullah University of Science and Technology (KAUST), Biological and Environmental Science and Engineering Division (BESE), 23955-6900 Thuwal, Saudi Arabia
| | - Joanna Izabela Lachowicz
- Dipartimento di Scienze Chimiche e Geologiche, Università di Cagliari, Cittadella Universitaria, I-09042 Monserrato, Cagliari, Italy
| | - Michal Eligiusz Nowakowski
- King Abdullah University of Science and Technology (KAUST), Biological and Environmental Science and Engineering Division (BESE), 23955-6900 Thuwal, Saudi Arabia; Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Żwirki i Wigury 101, 02-089 Warszawa, Poland
| | - Mariusz Jaremko
- King Abdullah University of Science and Technology (KAUST), Biological and Environmental Science and Engineering Division (BESE), 23955-6900 Thuwal, Saudi Arabia
| | - Łukasz Jaremko
- King Abdullah University of Science and Technology (KAUST), Biological and Environmental Science and Engineering Division (BESE), 23955-6900 Thuwal, Saudi Arabia.
| |
Collapse
|
44
|
Novel combinations of experimental and computational analysis tested on the binding of metalloprotoporphyrins to albumin. Int J Biol Macromol 2019; 134:445-457. [PMID: 31078597 DOI: 10.1016/j.ijbiomac.2019.05.060] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 05/08/2019] [Accepted: 05/08/2019] [Indexed: 02/02/2023]
Abstract
The evidence that Human Serum Albumin (HSA) binds metal ions and organometallic compounds has generated interest in its physiological role as a metalloprotein and as a vehicle for synthetic biology applications (e.g., synthetic blood and solar energy conversion). HSA has been shown to bind metallo-porphyrins, however, the structural details of such interactions are available only for the HSA:heme complex. A typical challenge for studying the interaction of proteins with metalloporphyrins is the poor solubility of the ligands that affect the characterization the complexes. The manuscript shows that a combination of dialysis and centrifugation yields aqueous solutions that contain >90% HSA:porphyrin complexes and virtually eliminate aggregated ligands. The removal of aggregates increases the quality of the optical spectroscopy data which, in turn, yield more accurate binding constants (~0.1 and 2.1 × 106 M-1) and reveal FRET between Trp214 and the porphyrins. The Trp-porphyrin distance was estimated to be within the 28-34 Å range and was used to guide the search of binding sites through a novel feedback approach with docking simulations. Results suggest while some protoporphyrins (metal-free, Fe(III)PPIX and Mg(II)PPIX) bind HSA at the heme site, others (Zn(II)PPIX, Mn(III)PPIX and Sn(IV)PPIX) are more likely to bind the Cys34.
Collapse
|
45
|
Gan N, Sun Q, Tang P, Wu D, Xie T, Zhang Y, Li H. Determination of interactions between human serum albumin and niraparib through multi-spectroscopic and computational methods. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2019; 206:126-134. [PMID: 30096696 DOI: 10.1016/j.saa.2018.07.100] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 07/25/2018] [Accepted: 07/31/2018] [Indexed: 06/08/2023]
Abstract
The interactions between 2-{4-[(3S)-piperidin-3-yl] phenyl}-2H-indazole-7-carboxamide (niraparib) and human serum albumin (HSA) were investigated through fluorescence and computational studies. Fluorescence experiments showed that the static quenching mechanism and the binding constant of the HSA-niraparib system at a single binding site was approximately 4 × 104 L mol-1. Thermodynamic constants indicated that the binding of niraparib to HSA was mainly driven by electrostatic interactions. Competition experiments and molecular docking simulations revealed that niraparib bound to site III of HSA. Synchronous fluorescence and Fourier transform infrared spectroscopy (FT-IR) results suggested that interactions between niraparib and HSA could affect the conformation and microenvironment of HSA. Circular dichroism (CD) measurements revealed that the α-helix contents of HSA negligibly increased after binding with niraparib. Molecular dynamics simulations demonstrated the stability of the binary HSA-niraparib system and confirmed that electrostatic forces accounted for the dominant contribution to system energy between HSA and niraparib.
Collapse
Affiliation(s)
- Na Gan
- School of Chemical Engineering, Sichuan University, Chengdu 610065, Sichuan, China
| | - Qiaomei Sun
- School of Chemical Engineering, Sichuan University, Chengdu 610065, Sichuan, China
| | - Peixiao Tang
- School of Chemical Engineering, Sichuan University, Chengdu 610065, Sichuan, China.
| | - Di Wu
- Key Laboratory of Meat Processing of Sichuan, College of Pharmacy and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Tonghui Xie
- School of Chemical Engineering, Sichuan University, Chengdu 610065, Sichuan, China.
| | - Yongkui Zhang
- School of Chemical Engineering, Sichuan University, Chengdu 610065, Sichuan, China
| | - Hui Li
- School of Chemical Engineering, Sichuan University, Chengdu 610065, Sichuan, China
| |
Collapse
|
46
|
Ascenzi P, Coletta M. Peroxynitrite Detoxification by Human Haptoglobin:Hemoglobin Complexes: A Comparative Study. J Phys Chem B 2018; 122:11100-11107. [DOI: 10.1021/acs.jpcb.8b05340] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Paolo Ascenzi
- Interdepartmental Laboratory for Electron Microscopy, Roma Tre University, Via della Vasca Navale 79, I-00146 Roma, Italy
| | - Massimo Coletta
- Department of Clinical Sciences and Translational Medicine, University of Roma “Tor Vergata”, Via Montpellier 1, I-00133 Roma, Italy
- Interuniversity Consortium for the Research on the Chemistry of Metals in Biological Systems, Via Celso Ulpiani 27, I-70126 Bari, Italy
| |
Collapse
|
47
|
Johnson CN, Gorbet GE, Ramsower H, Urquidi J, Brancaleon L, Demeler B. Multi-wavelength analytical ultracentrifugation of human serum albumin complexed with porphyrin. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2018; 47:789-797. [PMID: 29675648 DOI: 10.1007/s00249-018-1301-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 03/26/2018] [Accepted: 04/10/2018] [Indexed: 11/30/2022]
Abstract
The new Beckman Coulter Optima AUC instrument, which features multi-wavelength detection that couples the hydrodynamic separation of colloidal mixtures to spectral deconvolution of interacting and non-interacting solutes present in a mixture, was used to analyze the composition of human serum albumin (HSA) bound to metallo-protoporphyrin. We present new methods implemented in UltraScan that permit Optima AUC-derived multi-wavelength data to be spectrally decomposed in the same fashion as has been made possible for the Cölfen detector earlier. We demonstrate this approach by spectrally separating sedimentation velocity experimental data from mixtures of apo-HSA and HSA complexed to different metallo-protoporphyrins. We further demonstrate how multi-wavelength AUC can accurately recover percentages of metallo-protoporphyrin-bound HSA and apo-HSA from mixtures and how multi-wavelength AUC permits the calculation of molar extinction coefficients for porphyrins bound to HSA. The presented method has broad applicability to other complex systems where mixtures of molecules with different spectral properties need to be characterized.
Collapse
Affiliation(s)
- Courtney N Johnson
- Department of Biochemistry and Structural Biology, UTHSCSA, San Antonio, TX, USA
| | - Gary E Gorbet
- Department of Biochemistry and Structural Biology, UTHSCSA, San Antonio, TX, USA
| | - Heidi Ramsower
- Department of Physics and Astronomy, UTSA, San Antonio, TX, USA
| | - Julio Urquidi
- Department of Physics and Astronomy, UTSA, San Antonio, TX, USA
| | | | - Borries Demeler
- Department of Biochemistry and Structural Biology, UTHSCSA, San Antonio, TX, USA.
| |
Collapse
|
48
|
Iavarone F, Desiderio C, Vitali A, Messana I, Martelli C, Castagnola M, Cabras T. Cryptides: latent peptides everywhere. Crit Rev Biochem Mol Biol 2018; 53:246-263. [PMID: 29564928 DOI: 10.1080/10409238.2018.1447543] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Proteomic surveys with top-down platforms are today revealing thousands of naturally occurring fragments of bigger proteins. Some of them have not functional meaning because they derive from pathways responsible for protein degradation, but many have specific functions, often completely different from that one of the parent proteins. These peptides encrypted in the protein sequence are nowadays called cryptides. They are frequent in the animal and plant kingdoms and represent a new interesting -omic field of investigation. To point out how much widespread is their presence, we describe here the most studied cryptides from very common sources such as serum albumin, immunoglobulins, hemoglobin, and from saliva and milk proteins. Given its vastness, it is unfeasible to cover the topic exhaustively, therefore only several selected examples of cryptides from other sources are thereafter reported. Demanding is the development of new -omic platforms for the functional screening of new cryptides, which could provide suggestion for peptides and peptido-mimetics with variegate fields of application.
Collapse
Affiliation(s)
- Federica Iavarone
- a Istituto di Biochimica e Biochimica Clinica, Università Cattolica , Roma , Italy
| | - Claudia Desiderio
- b Istituto di Chimica del Riconoscimento Molecolare, CNR , Roma , Italy
| | - Alberto Vitali
- b Istituto di Chimica del Riconoscimento Molecolare, CNR , Roma , Italy
| | - Irene Messana
- b Istituto di Chimica del Riconoscimento Molecolare, CNR , Roma , Italy
| | - Claudia Martelli
- a Istituto di Biochimica e Biochimica Clinica, Università Cattolica , Roma , Italy
| | - Massimo Castagnola
- a Istituto di Biochimica e Biochimica Clinica, Università Cattolica , Roma , Italy.,b Istituto di Chimica del Riconoscimento Molecolare, CNR , Roma , Italy
| | - Tiziana Cabras
- c Dipartimento di Scienze della Vita e dell'Ambiente , Università di Cagliari , Cagliari , Italy
| |
Collapse
|
49
|
Exploring the interaction between “site-markers, aspirin and esterase-like activity” ternary systems on the human serum albumin: direct evidence for modulation of catalytic activity of the protein in different inhibition modes. JOURNAL OF THE IRANIAN CHEMICAL SOCIETY 2017. [DOI: 10.1007/s13738-017-1256-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
50
|
Ascenzi P, Bocedi A, Gioia M, Fanali G, Fasano M, Coletta M. Warfarin inhibits allosterically the reductive nitrosylation of ferric human serum heme-albumin. J Inorg Biochem 2017; 177:63-75. [PMID: 28926756 DOI: 10.1016/j.jinorgbio.2017.08.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 08/30/2017] [Indexed: 12/31/2022]
Abstract
Human serum heme-albumin (HSA-heme-Fe) displays heme-based ligand binding and (pseudo-)enzymatic properties. Here, the effect of the prototypical drug warfarin on kinetics and thermodynamics of NO binding to ferric and ferrous HSA-heme-Fe (HSA-heme-Fe(III) and HSA-heme-Fe(II), respectively) and on the NO-mediated reductive nitrosylation of the heme-Fe atom is reported; data were obtained between pH5.5 and 9.5 at 20.0°C. Since warfarin is a common drug, its effect on the reactivity of HSA-heme-Fe represents a relevant issue in the pharmacological therapy management. The inhibition of NO binding to HSA-heme-Fe(III) and HSA-heme-Fe(II) as well as of the NO-mediated reductive nitrosylation of the heme-Fe(III) atom by warfarin has been ascribed to drug binding to the fatty acid binding site 2 (FA2), shifting allosterically the penta-to-six coordination equilibrium of the heme-Fe atom toward the low reactive species showing the six-coordinated metal center by His146 and Tyr161 residues. These data: (i) support the role of HSA-heme-Fe in trapping NO, (ii) highlight the modulation of the heme-Fe-based reactivity by drugs, and (iii) could be relevant for the modulation of HSA functions by drugs in vivo.
Collapse
Affiliation(s)
- Paolo Ascenzi
- Interdepartmental Laboratory for Electron Microscopy, Roma Tre University, I-00146 Roma, Italy.
| | - Alessio Bocedi
- Department of Chemical Sciences and Technology, University of Roma "Tor Vergata", I-00133 Roma, Italy
| | - Magda Gioia
- Department of Clinical Sciences and Translational Medicine, University of Roma "Tor Vergata", I-00133 Roma, Italy; Interuniversity Consortium for the Research on the Chemistry of Metals in Biological Systems, I-70126 Bari, Italy
| | | | - Mauro Fasano
- Department of Science and High Technology, University of Insubria, I-21052 Busto Arsizio, VA, Italy; Neuroscience Research Center, University of Insubria, I-21052 Busto Arsizio, VA, Italy
| | - Massimo Coletta
- Department of Clinical Sciences and Translational Medicine, University of Roma "Tor Vergata", I-00133 Roma, Italy; Interuniversity Consortium for the Research on the Chemistry of Metals in Biological Systems, I-70126 Bari, Italy
| |
Collapse
|