Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (27)

Search Parameters:
Keywords = heme-albumin

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
14 pages, 6857 KB  
Article
Structural Alterations of Human Erythrocytes Induced by Minocycline
by Elena Baeva, Marina Holyavka, Valery Artyukhov and Maxim Kondratyev
Cells 2025, 14(22), 1787; https://doi.org/10.3390/cells14221787 - 14 Nov 2025
Viewed by 458
Abstract
The non-antibacterial effects of the tetracycline antibiotic minocycline on human erythrocytes are currently under investigation. Our data indicate alterations in the surface structure of erythrocytes; the antibiotic promotes the redistribution of cellular transformational forms during preliminary in vitro incubation (1 h and 24 [...] Read more.
The non-antibacterial effects of the tetracycline antibiotic minocycline on human erythrocytes are currently under investigation. Our data indicate alterations in the surface structure of erythrocytes; the antibiotic promotes the redistribution of cellular transformational forms during preliminary in vitro incubation (1 h and 24 h) with the modifier. The degree of surface relief changes increases over time, leading to the formation of erythrocytes displaying outgrowths and ridges, spherulation, and “deflated ball”-shaped cells (after 1 day). These alterations are largely reversible, as washing the erythrocyte suspensions with a 1% bovine serum albumin solution reduces the number of echinocytes and irreversibly transformed spherocytes with spikes. Spectrophotometric analysis has shown that minocycline stabilizes the spatial organization of hemoprotein molecules, as it does not lead to increased methemoglobin formation in the samples over time. The antibiotic appears to bind primarily to amino acid residues within heme pockets, as confirmed by molecular docking. Our findings suggest a potential risk of reduced oxygen transport function in red blood cells when taking this antibiotic, highlighting the need to consider potential erythrocyte-related side effects during long-term minocycline therapy. Full article
(This article belongs to the Special Issue Highlights in Red Blood Cell Research)
Show Figures

Graphical abstract

12 pages, 221 KB  
Review
Comparative Analysis of 5-ALA and Fluorescent Techniques in High-Grade Glioma Treatment
by José E. Valerio, Guillermo de Jesús Aguirre Vera, Jorge Zumaeta, Noe Santiago Rea, Maria P. Fernandez Gomez, Penelope Mantilla-Farfan, Laurel Valente and Andrés M. Alvarez-Pinzon
Biomedicines 2025, 13(5), 1161; https://doi.org/10.3390/biomedicines13051161 - 10 May 2025
Cited by 6 | Viewed by 3938
Abstract
Background: 5-Aminolevulinic acid (5-ALA) serves as a precursor in the heme biosynthesis pathway, resulting in the selective accumulation of protoporphyrin IX (PpIX) within glioma cells. This property facilitates fluorescence-guided resection (FGR) in high-grade gliomas (HGGs), enhancing surgical precision and oncological results. Nonetheless, its [...] Read more.
Background: 5-Aminolevulinic acid (5-ALA) serves as a precursor in the heme biosynthesis pathway, resulting in the selective accumulation of protoporphyrin IX (PpIX) within glioma cells. This property facilitates fluorescence-guided resection (FGR) in high-grade gliomas (HGGs), enhancing surgical precision and oncological results. Nonetheless, its clinical implementation is restricted by factors such as accessibility, cost, and technical limitations. Methods: A systematic review of PubMed literature (2019–2024) was conducted to assess the efficacy of 5-ALA in HGG surgery compared to conventional white light microscopy. Studies focusing on non-neurosurgical applications, pediatric populations, and non-HGG indications were excluded. Results: Nineteen articles met the criteria. Recent studies indicate that 5-ALA-guided resection significantly enhances gross total resection (GTR) rates compared to white light surgery (75.4% vs. 54.3%, p < 0.001). Patients receiving 5-ALA-assisted resection exhibit enhanced progression-free survival (PFS) at 6 months (median 8.1 months compared to 5.4 months, p = 0.002) and overall survival (OS) (median 15.2 months versus 12.3 months, p = 0.008). The necessity for specialized neurosurgical microscopes equipped with blue light filters restricts accessibility, especially in low-resource environments. Recent advancements in fluorescence-enhancing technologies, particularly loupe-based systems, have demonstrated increases in fluorescence intensity by up to tenfold through direct emission. Sodium fluorescein, originally designed for ophthalmological use, has been adapted for enhancing contrast in intracranial tumors; however, its non-specific binding to serum albumin restricts its accuracy in glioma resection. Conclusions: Recent publications demonstrate that 5-ALA fluorescence-guided surgery significantly improves gross total resection rates and survival outcomes in patients with high-grade gliomas. Although it offers clinical advantages, cost and equipment constraints continue to pose substantial obstacles to broad implementation. Additional research is required to enhance fluorescence-guided techniques and increase accessibility in resource-constrained environments. Full article
(This article belongs to the Special Issue Advanced Cancer Diagnosis and Treatment: Second Edition)
21 pages, 4291 KB  
Article
Analysis of Ferric Protoporphyrin IX Effects on Human Platelets: Hematin Is a More Potent Agonist than Hemin
by Diana M. Mikhailova, Julia Sudnitsyna, Polina Kovgan, Lidia Naida, Alexandra Kharazova, Igor Mindukshev and Stepan Gambaryan
Cells 2025, 14(4), 255; https://doi.org/10.3390/cells14040255 - 11 Feb 2025
Cited by 3 | Viewed by 1440
Abstract
Hemolysis during severe diseases (malaria, hemorrhagic stroke, sickle cell disease, etc.) and blood transfusion induces the release of free hemoglobin, which degrades to highly reactive and toxic compounds—hemin and hematin. Oxidized heme derivatives induce platelet activation, aggregation, and degranulation, leading to prothrombotic and [...] Read more.
Hemolysis during severe diseases (malaria, hemorrhagic stroke, sickle cell disease, etc.) and blood transfusion induces the release of free hemoglobin, which degrades to highly reactive and toxic compounds—hemin and hematin. Oxidized heme derivatives induce platelet activation, aggregation, and degranulation, leading to prothrombotic and inflammatory events. In the present study, we showed that hematin is a more potent agonist of platelet activation than hemin, and using several methods, including the original laser diffraction method, flow cytometry, and confocal microscopy, we demonstrated that hematin at low doses induces platelet activation and aggregation without reducing cell viability and affecting calcium efflux. On the contrary, hematin at high concentrations triggered phosphatidylserine exposure, severe loss of platelet viability, and calcium dysregulation, which was not inhibited by cGMP/PKG and cAMP/PKA pathways. Additionally, we showed that albumin could initiate disaggregation processes in hematin-activated platelets. Full article
(This article belongs to the Special Issue Heme Biology in Health and Disease)
Show Figures

Graphical abstract

12 pages, 2675 KB  
Article
Urobilin Derived from Bilirubin Bioconversion Binds Albumin and May Interfere with Bilirubin Interacting with Albumin: Implications for Disease Pathology
by Kevin I. Williams, Priyanka Suryadevara, Chang-Guo Zhan, Terry D. Hinds and Zachary A. Kipp
Biomedicines 2025, 13(2), 302; https://doi.org/10.3390/biomedicines13020302 - 26 Jan 2025
Cited by 5 | Viewed by 3491
Abstract
Background/Objectives: Bilirubin is a hydrophobic molecule that binds the carrier protein albumin for transport through systemic circulation. Bilirubin is cleared from the body through the liver and excreted into the intestines, where the microbiota modifies the chemical structure, forming urobilin, which can be [...] Read more.
Background/Objectives: Bilirubin is a hydrophobic molecule that binds the carrier protein albumin for transport through systemic circulation. Bilirubin is cleared from the body through the liver and excreted into the intestines, where the microbiota modifies the chemical structure, forming urobilin, which can be reabsorbed into circulation by the hepatic portal vein. Urobilin has no known function. It is also unknown whether urobilin binds albumin for transport in circulation. We hypothesized that because of the likeness of their chemical structures, urobilin would also bind albumin like bilirubin does. Methods: First, we used in silico docking to predict if urobilin would bind to albumin and compared it to the bilirubin binding sites. To test this binding in vitro, we applied bilirubin’s fluorescent property, which occurs when it is bound to a protein, including albumin, and exposed to light. We also used this method to determine if urobilin could exhibit autofluorescence when protein bound. Results: We found that bilirubin was predicted to bind albumin at amino acids E208, K212, D237, and K240 through hydrogen bonds. However, urobilin was predicted to bind albumin using different hydrogen bonds at amino acids H67, K240, and E252. We found that urobilin has a fluorescent property that can be quantified when bound to albumin. We performed a concentration response for urobilin–albumin fluorescent binding and observed a direct relationship between the urobilin level and the fluorescence intensity. Conclusions: The in silico docking analysis and autofluorescence results demonstrate that urobilin binds to albumin and might compete with bilirubin. This is the first study to identify a urobilin-binding protein and the important aspects of its physiological function and transport in circulation. Full article
(This article belongs to the Section Endocrinology and Metabolism Research)
Show Figures

Figure 1

22 pages, 5018 KB  
Article
Hematin- and Hemin-Induced Spherization and Hemolysis of Human Erythrocytes Are Independent of Extracellular Calcium Concentration
by Diana M. Mikhailova, Elisaveta Skverchinskaya, Julia Sudnitsyna, Kirill R. Butov, Ekaterina M. Koltsova, Igor V. Mindukshev and Stepan Gambaryan
Cells 2024, 13(6), 554; https://doi.org/10.3390/cells13060554 - 21 Mar 2024
Cited by 3 | Viewed by 3993
Abstract
Pathologies such as malaria, hemorrhagic stroke, sickle cell disease, and thalassemia are characterized by the release of hemoglobin degradation products from damaged RBCs. Hematin (liganded with OH) and hemin (liganded with Cl)—are the oxidized forms of heme with toxic [...] Read more.
Pathologies such as malaria, hemorrhagic stroke, sickle cell disease, and thalassemia are characterized by the release of hemoglobin degradation products from damaged RBCs. Hematin (liganded with OH) and hemin (liganded with Cl)—are the oxidized forms of heme with toxic properties due to their hydrophobicity and the presence of redox-active Fe3. In the present study, using the original LaSca-TM laser particle analyzer, flow cytometry, and confocal microscopy, we showed that both hematin and hemin induce dose-dependent RBC spherization and hemolysis with ghost formation. Hematin and hemin at nanomolar concentrations increased [Ca2+]i in RBC; however, spherization and hemolysis occurred in the presence and absence of calcium, indicating that both processes are independent of [Ca2+]i. Both compounds triggered acute phosphatidylserine exposure on the membrane surface, reversible after 60 min of incubation. A comparison of hematin and hemin effects on RBCs revealed that hematin is a more reactive toxic metabolite than hemin towards human RBCs. The toxic effects of heme derivatives were reduced and even reversed in the presence of albumin, indicating the presence in RBCs of the own recovery system against the toxic effects of heme derivatives. Full article
(This article belongs to the Special Issue Highlights in Red Blood Cell Research)
Show Figures

Figure 1

18 pages, 4888 KB  
Article
Histidine-Bound Dinitrosyl Iron Complexes: Antioxidant and Antiradical Properties
by Konstantin B. Shumaev, Olga V. Kosmachevskaya, Elvira I. Nasybullina, Enno K. Ruuge, Elena I. Kalenikova and Alexey F. Topunov
Int. J. Mol. Sci. 2023, 24(24), 17236; https://doi.org/10.3390/ijms242417236 - 7 Dec 2023
Cited by 9 | Viewed by 2308
Abstract
Dinitrosyl iron complexes (DNICs) are important physiological derivatives of nitric oxide. These complexes have a wide range of biological activities, with antioxidant and antiradical ones being of particular interest and importance. We studied the interaction between DNICs associated with the dipeptide L-carnosine or [...] Read more.
Dinitrosyl iron complexes (DNICs) are important physiological derivatives of nitric oxide. These complexes have a wide range of biological activities, with antioxidant and antiradical ones being of particular interest and importance. We studied the interaction between DNICs associated with the dipeptide L-carnosine or serum albumin and prooxidants under conditions mimicking oxidative stress. The ligands of these DNICs were histidine residues of carnosine or His39 and Cys34 in bovine serum albumin. Carnosine-bound DNICs reduced the level of piperazine free radicals in the reaction system containing tert-butyl hydroperoxide (t-BOOH), bivalent iron ions, a nitroxyl anion donor (Angeli’s salt), and HEPES buffer. The ability of carnosine DNICs to intercept organic free radicals produced from t-BOOH decay could lead to this effect. In addition, carnosine DNICs reacted with the superoxide anion radical (O2•−) formed in the xanthine/xanthine oxidase enzymatic system. They also reduced the oxoferryl form of the heme group formed in the reaction of myoglobin with t-BOOH. DNICs associated with serum albumin were found to be rapidly destroyed in a model system containing metmyoglobin and t-BOOH. At the same time, these protein DNICs inhibited the t-BOOH-induced oxidative degradation of coenzymes Q9 and Q10 in rat myocardial homogenate. The possible mechanisms of the antioxidant and antiradical action of the DNICs studied and their role in the metabolism of reactive oxygen and nitrogen species are discussed. Full article
(This article belongs to the Collection Feature Papers Collection in Biochemistry)
Show Figures

Figure 1

15 pages, 3226 KB  
Article
Metalloporphyrins Reduce Proteinuria in Podocyte Immune Injury: The Role of Metal and Porphyrin Moieties
by Elias A. Lianos, Gia Nghi Phung, Michelle Foster, Jianping Zhou and Mukut Sharma
Int. J. Mol. Sci. 2023, 24(16), 12777; https://doi.org/10.3390/ijms241612777 - 14 Aug 2023
Cited by 5 | Viewed by 1904
Abstract
Depending on their central metal atom, metalloporphyrins (MPs) can attenuate or exacerbate the severity of immune-mediated kidney injury, and this has been attributed to the induction or inhibition of heme oxygenase (HO) activity, particularly the inducible isoform (HO-1) of this enzyme. The role [...] Read more.
Depending on their central metal atom, metalloporphyrins (MPs) can attenuate or exacerbate the severity of immune-mediated kidney injury, and this has been attributed to the induction or inhibition of heme oxygenase (HO) activity, particularly the inducible isoform (HO-1) of this enzyme. The role of central metal or porphyrin moieties in determining the efficacy of MPs to attenuate injury, as well as mechanisms underlying this effect, have not been assessed. Using an antibody-mediated complement-dependent model of injury directed against rat visceral glomerular epithelial cells (podocytes) and two MPs (FePPIX, CoPPIX) that induce both HO-1 expression and HO enzymatic activity in vivo but differ in their chelated metal, we assessed their efficacy in reducing albuminuria. Podocyte injury was induced using rabbit immune serum raised against the rat podocyte antigen, Fx1A, and containing an anti-Fx1A antibody that activates complement at sites of binding. FePPIX or CoPPIX were injected intraperitoneally (5 mg/kg) 24 h before administration of the anti-Fx1A serum and on days 1, 3, 6, and 10 thereafter. Upon completion of urine collection on day 14, the kidney cortex was obtained for histopathology and isolation of glomeruli, from which total protein extracts were obtained. Target proteins were analyzed by capillary-based separation and immunodetection (Western blot analysis). Both MPs had comparable efficacy in reducing albuminuria in males, but the efficacy of CoPPIX was superior in female rats. The metal-free protoporphyrin, PPIX, had minimal or no effect on urine albumin excretion. CoPPIX was also the most potent MP in inducing glomerular HO-1, reducing complement deposition, and preserving the expression of the complement regulatory protein (CRP) CD55 but not that of CD59, the expression of which was reduced by both MPs. These observations demonstrate that the metal moiety of HO-1-inducing MPs plays an important role in reducing proteinuria via mechanisms involving reduced complement deposition and independently of an effect on CRPs. Full article
(This article belongs to the Special Issue Together and Apart: Acute Kidney Injury and Chronic Kidney Disease)
Show Figures

Figure 1

17 pages, 2794 KB  
Review
Heme Interactions as Regulators of the Alternative Pathway Complement Responses and Implications for Heme-Associated Pathologies
by Stefanos A. Tsiftsoglou
Curr. Issues Mol. Biol. 2023, 45(6), 5198-5214; https://doi.org/10.3390/cimb45060330 - 16 Jun 2023
Cited by 6 | Viewed by 3435
Abstract
Heme (Fe2+-protoporphyrin IX) is a pigment of life, and as a prosthetic group in several hemoproteins, it contributes to diverse critical cellular processes. While its intracellular levels are tightly regulated by networks of heme-binding proteins (HeBPs), labile heme can be hazardous [...] Read more.
Heme (Fe2+-protoporphyrin IX) is a pigment of life, and as a prosthetic group in several hemoproteins, it contributes to diverse critical cellular processes. While its intracellular levels are tightly regulated by networks of heme-binding proteins (HeBPs), labile heme can be hazardous through oxidative processes. In blood plasma, heme is scavenged by hemopexin (HPX), albumin and several other proteins, while it also interacts directly with complement components C1q, C3 and factor I. These direct interactions block the classical pathway (CP) and distort the alternative pathway (AP). Errors or flaws in heme metabolism, causing uncontrolled intracellular oxidative stress, can lead to several severe hematological disorders. Direct interactions of extracellular heme with alternative pathway complement components (APCCs) may be implicated molecularly in diverse conditions at sites of abnormal cell damage and vascular injury. In such disorders, a deregulated AP could be associated with the heme-mediated disruption of the physiological heparan sulphate–CFH coat of stressed cells and the induction of local hemostatic responses. Within this conceptual frame, a computational evaluation of HBMs (heme-binding motifs) aimed to determine how heme interacts with APCCs and whether these interactions are affected by genetic variation within putative HBMs. Combined computational analysis and database mining identified putative HBMs in all of the 16 APCCs examined, with 10 exhibiting disease-associated genetic (SNPs) and/or epigenetic variation (PTMs). Overall, this article indicates that among the pleiotropic roles of heme reviewed, the interactions of heme with APCCs could induce differential AP-mediated hemostasis-driven pathologies in certain individuals. Full article
Show Figures

Figure 1

21 pages, 2841 KB  
Review
Ligand-Based Regulation of Dynamics and Reactivity of Hemoproteins
by Emily Samuela Turilli-Ghisolfi, Marta Lualdi and Mauro Fasano
Biomolecules 2023, 13(4), 683; https://doi.org/10.3390/biom13040683 - 17 Apr 2023
Cited by 9 | Viewed by 3581
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, [...] Read more.
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. Full article
Show Figures

Figure 1

15 pages, 1699 KB  
Review
Heme Scavenging and Delivery: The Role of Human Serum Albumin
by Giovanna De Simone, Romualdo Varricchio, Tommaso Francesco Ruberto, Alessandra di Masi and Paolo Ascenzi
Biomolecules 2023, 13(3), 575; https://doi.org/10.3390/biom13030575 - 22 Mar 2023
Cited by 29 | Viewed by 8700
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 [...] Read more.
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. Full article
Show Figures

Figure 1

42 pages, 6472 KB  
Article
Measurement Precision and Thermal and Absorption Properties of Nanostructures in Aqueous Solutions by Transient and Steady-State Thermal-Lens Spectrometry
by Vladislav R. Khabibullin, Liliya O. Usoltseva, Polina A. Galkina, Viktoriya R. Galimova, Dmitry S. Volkov, Ivan V. Mikheev and Mikhail A. Proskurnin
Physchem 2023, 3(1), 156-197; https://doi.org/10.3390/physchem3010012 - 9 Mar 2023
Cited by 15 | Viewed by 4976
Abstract
A simultaneous steady-state and transient photothermal-lens modality was used for both the thermal and optical parameters of aqueous dispersed systems (carbon and silica nanoparticles, metal iodides, surfactants, heme proteins, albumin, and their complexes). Heat-transfer parameters (thermal diffusivity and thermal effusivity), the temperature gradient [...] Read more.
A simultaneous steady-state and transient photothermal-lens modality was used for both the thermal and optical parameters of aqueous dispersed systems (carbon and silica nanoparticles, metal iodides, surfactants, heme proteins, albumin, and their complexes). Heat-transfer parameters (thermal diffusivity and thermal effusivity), the temperature gradient of the refractive index, light absorption, and concentration parameters were assessed. To simultaneously measure thermal and optical parameters, the time scale of thermal lensing (characteristic time, tc) should correspond to an excitation beam size of 60–300 µm, and the relative time intervals 0.5÷5tc and (5÷20)tc should be selected for transient and steady-state measurements, respectively. Dual-beam thermal-lens spectrometers in a mode-mismatched optical schematic at various excitation wavelengths were built. The spectrometers implement back-synchronized detection, providing different measurement conditions for the heating and cooling parts of the thermal-lens cycle. By varying the measurement parameters depending on the dispersed system, the conditions providing the suitable precision (replicability, repeatability, and reproducibility) of thermal-lens measurements were found; setups with a broad excitation beam (waist size, 150 and 300 μm) provide longer times to attain a thermal equilibrium and, thus, the better precision of measurements of thermal diffusivity. Full article
(This article belongs to the Section Application of Lasers to Physical Chemistry)
Show Figures

Figure 1

15 pages, 4801 KB  
Article
Dissecting the Interactions between Chlorin e6 and Human Serum Albumin
by Alessia Marconi, Edoardo Jun Mattioli, Filippo Ingargiola, Giulia Giugliano, Tainah Dorina Marforio, Luca Prodi, Matteo Di Giosia and Matteo Calvaresi
Molecules 2023, 28(5), 2348; https://doi.org/10.3390/molecules28052348 - 3 Mar 2023
Cited by 15 | Viewed by 3437
Abstract
Chlorin e6 (Ce6) is among the most used sensitizers in photodynamic (PDT) and sonodynamic (SDT) therapy; its low solubility in water, however, hampers its clinical exploitation. Ce6 has a strong tendency to aggregate in physiological environments, reducing its performance as a photo/sono-sensitizer, as [...] Read more.
Chlorin e6 (Ce6) is among the most used sensitizers in photodynamic (PDT) and sonodynamic (SDT) therapy; its low solubility in water, however, hampers its clinical exploitation. Ce6 has a strong tendency to aggregate in physiological environments, reducing its performance as a photo/sono-sensitizer, as well as yielding poor pharmacokinetic and pharmacodynamic properties. The interaction of Ce6 with human serum albumin (HSA) (i) governs its biodistribution and (ii) can be used to improve its water solubility by encapsulation. Here, using ensemble docking and microsecond molecular dynamics simulations, we identified the two Ce6 binding pockets in HSA, i.e., the Sudlow I site and the heme binding pocket, providing an atomistic description of the binding. Comparing the photophysical and photosensitizing properties of Ce6@HSA with respect to the same properties regarding the free Ce6, it was observed that (i) a red-shift occurred in both the absorption and emission spectra, (ii) a maintaining of the fluorescence quantum yield and an increase of the excited state lifetime was detected, and (iii) a switch from the type II to the type I mechanism in a reactive oxygen species (ROS) production, upon irradiation, took place. Full article
(This article belongs to the Special Issue Novel Agents in Photodynamic Therapy)
Show Figures

Figure 1

18 pages, 1931 KB  
Article
Effects of Heme Site (FA1) Ligands Bilirubin, Biliverdin, Hemin, and Methyl Orange on the Albumin Binding of Site I Marker Warfarin: Complex Allosteric Interactions
by Beáta Lemli, Zuzana Lomozová, Tamás Huber, András Lukács and Miklós Poór
Int. J. Mol. Sci. 2022, 23(22), 14007; https://doi.org/10.3390/ijms232214007 - 13 Nov 2022
Cited by 16 | Viewed by 3582
Abstract
Human serum albumin (HSA) is the most abundant plasma protein in circulation. The three most important drug-binding sites on HSA are Sudlow’s Site I (subdomain IIA), Sudlow’s Site II (subdomain IIIA), and Heme site (subdomain IB). Heme site and Site I are allosterically [...] Read more.
Human serum albumin (HSA) is the most abundant plasma protein in circulation. The three most important drug-binding sites on HSA are Sudlow’s Site I (subdomain IIA), Sudlow’s Site II (subdomain IIIA), and Heme site (subdomain IB). Heme site and Site I are allosterically coupled; therefore, their ligands may be able to allosterically modulate the binding affinity of each other. In this study, the effects of four Heme site ligands (bilirubin, biliverdin, hemin, and methyl orange) on the interaction of the Site I ligand warfarin with HSA were tested, employing fluorescence spectroscopic, ultrafiltration, and ultracentrifugation studies. Our major results/conclusions are the following. (1) Quenching studies indicated no relevant interaction, while the other fluorescent model used suggested that each Heme site ligand strongly decreases the albumin binding of warfarin. (2) Ultrafiltration and ultracentrifugation studies demonstrated the complex modulation of warfarin–HSA interaction by the different Heme site markers; for example, bilirubin strongly decreased while methyl orange considerably increased the bound fraction of warfarin. (3) Fluorescence spectroscopic studies showed misleading results in these diligand–albumin interactions. (4) Different Heme site ligands can increase or decrease the albumin binding of warfarin and the outcome can even be concentration dependent (e.g., biliverdin and hemin). Full article
(This article belongs to the Special Issue Proteins and Protein-Ligand Interactions)
Show Figures

Figure 1

11 pages, 2243 KB  
Article
Interaction of the Emerging Mycotoxins Beauvericin, Cyclopiazonic Acid, and Sterigmatocystin with Human Serum Albumin
by Eszter Fliszár-Nyúl, Zelma Faisal, Renáta Skaper, Beáta Lemli, Bayarsaikhan Bayartsetseg, Csaba Hetényi, Patrik Gömbös, András Szabó and Miklós Poór
Biomolecules 2022, 12(8), 1106; https://doi.org/10.3390/biom12081106 - 11 Aug 2022
Cited by 26 | Viewed by 2941
Abstract
Beauvericin (BEA), cyclopiazonic acid (CPA), and sterigmatocystin (STC) are emerging mycotoxins. They appear as contaminants in food and animal feed, leading to economic losses and health risks. Human serum albumin (HSA) forms stable complexes with certain mycotoxins, including ochratoxins, alternariol, citrinin, and zearalenone. [...] Read more.
Beauvericin (BEA), cyclopiazonic acid (CPA), and sterigmatocystin (STC) are emerging mycotoxins. They appear as contaminants in food and animal feed, leading to economic losses and health risks. Human serum albumin (HSA) forms stable complexes with certain mycotoxins, including ochratoxins, alternariol, citrinin, and zearalenone. HSA binding can influence the toxicokinetics of xenobiotics, and albumin can also be considered and applied as a relatively cheap affinity protein. Therefore, we examined the potential interactions of BEA, CPA, and STC with HSA employing fluorescence spectroscopy, ultracentrifugation, ultrafiltration, and molecular modeling. Spectroscopic and ultracentrifugation studies demonstrated the formation of low-affinity BEA–HSA (Ka ≈ 103 L/mol) and moderately strong CPA–HSA and STC–HSA complexes (Ka ≈ 104 L/mol). In ultrafiltration experiments, CPA slightly displaced each site marker (warfarin, naproxen, and camptothecin) tested, while BEA and STC did not affect significantly the albumin binding of these drugs. Modeling studies suggest that CPA occupies Sudlow’s site I, while STC binds to the Heme site (FA1) on HSA. Considering the interactions of CPA with the site markers, the CPA–HSA interaction may have toxicological importance. Full article
Show Figures

Figure 1

19 pages, 7930 KB  
Article
Starch-Based Hydrogel Nanoparticles Loaded with Polyphenolic Compounds of Moringa Oleifera Leaf Extract Have Hepatoprotective Activity in Bisphenol A-Induced Animal Models
by Hend Mohamed Hasanin Abou El-Naga, Samah A. El-Hashash, Ensaf Mokhtar Yasen, Stefano Leporatti and Nemany A. N. Hanafy
Polymers 2022, 14(14), 2846; https://doi.org/10.3390/polym14142846 - 13 Jul 2022
Cited by 18 | Viewed by 4376
Abstract
Bisphenol A (BPA) is an xenoestrogenic chemical used extensively in the fabrication of baby bottles, reusable plastic water bottles and polycarbonate plastic containers. The current study aims to investigate the hepatoprotective activity of Moringa oleifera Lam leaf extract (MOLE) and hydrogel NPs made [...] Read more.
Bisphenol A (BPA) is an xenoestrogenic chemical used extensively in the fabrication of baby bottles, reusable plastic water bottles and polycarbonate plastic containers. The current study aims to investigate the hepatoprotective activity of Moringa oleifera Lam leaf extract (MOLE) and hydrogel NPs made of starch-MOLE-Bovine Serum Albumin (BSA) against Bisphenol A-induced liver toxicity in male rats. Fabrication and characterization of hydrogel NPs formed of starch-MOLE-BSA were investigated using FTIR, TEM, zeta potential, UV-visible spectroscopy and fluorescence spectrophotometer. The potential efficacy of hydrogel NPs was studied. Compared to the results of control, the level of liver function, oxidative stress markers and lipid profile status were remodulated in the groups treated with MOLE and hydrogel NPs (Encap. MOLE). Meanwhile, the administration of MOLE and Encap MOLE significantly increased antioxidant activity and decreased the level of apoptotic pathways. Heme oxygenase (HO)-1 and growth arrest -DNA damage-inducible gene 45b (Gadd45b) were also regulated in the groups treated with MOLE and Encap. MOLE compared to the group which received BPA alone. In the present study, MOLE and hydrogel NPs led to remarkable alterations in histological changes during BPA administration. Overall, MOLE has a potential antioxidant activity which can be used in the treatment of liver disorders. Full article
Show Figures

Figure 1

Back to TopTop