Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.256
Filtrer
1.
Chirality ; 36(7): e23696, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38965734

RÉSUMÉ

Through molecular recognition, drugs can interact and complex with macromolecules circulating in the body. The serum albumin transport protein, found in several mammals, has several interaction sites where these molecules can be located. The drug sulfasalazine (SSZ) is known in the literature to complex at drug site 1 (DS1) in human serum (HSA) and bovine serum (BSA) proteins. This complexation can be studied using various spectroscopic techniques. With the techniques used in this work, absorption in the ultraviolet and visible regions (UV-Vis) and electronic circular dichroism (ECD), a significant difference was observed in the results involving HSA and BSA. The application of theoretical methodologies, such as TD-DFT and molecular docking, suggests that the conformation that SSZ assumes in DS1 of the two proteins is different, which exposes it to different amino acid residues and different hydrophobicities. This difference in conformation may be related to the location of DS1 where the drug interacts or to the possibility of SSZ moving in the BSA site, due to its larger size, and moving less freely in HSA.


Sujet(s)
Simulation de docking moléculaire , Sérumalbumine bovine , Sulfasalazine , Sulfasalazine/composition chimique , Sérumalbumine bovine/composition chimique , Humains , Bovins , Animaux , Stéréoisomérie , Dichroïsme circulaire , Sérum-albumine humaine/composition chimique , Théorie de la fonctionnelle de la densité
2.
ACS Appl Mater Interfaces ; 16(28): 36983-37006, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-38953207

RÉSUMÉ

Repairing multiphasic defects is cumbersome. This study presents new soft and hard scaffold designs aimed at facilitating the regeneration of multiphasic defects by enhancing angiogenesis and improving cell attachment. Here, the nonimmunogenic, nontoxic, and cost-effective human serum albumin (HSA) fibril (HSA-F) was used to fabricate thermostable (up to 90 °C) and hard printable polymers. Additionally, using a 10.0 mg/mL HSA-F, an innovative hydrogel was synthesized in a mixture with 2.0% chitosan-conjugated arginine, which can gel in a cell-friendly and pH physiological environment (pH 7.4). The presence of HSA-F in both hard and soft scaffolds led to an increase in significant attachment of the scaffolds to the human periodontal ligament fibroblast (PDLF), human umbilical vein endothelial cell (HUVEC), and human osteoblast. Further studies showed that migration (up to 157%), proliferation (up to 400%), and metabolism (up to 210%) of these cells have also improved in the direction of tissue repair. By examining different in vitro and ex ovo experiments, we observed that the final multiphasic scaffold can increase blood vessel density in the process of per-vascularization as well as angiogenesis. By providing a coculture environment including PDLF and HUVEC, important cross-talk between these two cells prevails in the presence of roxadustat drug, a proangiogenic in this study. In vitro and ex ovo results demonstrated significant enhancements in the angiogenic response and cell attachment, indicating the effectiveness of the proposed design. This approach holds promise for the regeneration of complex tissue defects by providing a conducive environment for vascularization and cellular integration, thus promoting tissue healing.


Sujet(s)
Cellules endothéliales de la veine ombilicale humaine , Néovascularisation physiologique , Structures d'échafaudage tissulaires , Humains , Structures d'échafaudage tissulaires/composition chimique , Néovascularisation physiologique/effets des médicaments et des substances chimiques , Sérum-albumine humaine/composition chimique , Glycine/composition chimique , Glycine/pharmacologie , Glycine/analogues et dérivés , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/cytologie , Fibroblastes/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Amyloïde/composition chimique , Amyloïde/métabolisme , Ostéoblastes/effets des médicaments et des substances chimiques , Ostéoblastes/cytologie , Ostéoblastes/métabolisme , Desmodonte/cytologie , Desmodonte/effets des médicaments et des substances chimiques , Ingénierie tissulaire , Hydrogels/composition chimique , Hydrogels/pharmacologie , Température , Isoquinoléines
3.
Molecules ; 29(13)2024 Jun 24.
Article de Anglais | MEDLINE | ID: mdl-38998953

RÉSUMÉ

Ketoprofen (KTF) and ketorolac (KTL) are among the most primarily used non-steroidal anti-inflammatory drugs (NSAIDs) in humans to alleviate moderate pain and to treat inflammation. Their binding affinity with albumin (the main globular protein responsible for the biodistribution of drugs in the bloodstream) was previously determined by spectroscopy without considering some conventional pitfalls. Thus, the present work updates the biophysical characterization of the interactions of HSA:KTF and HSA:KTL by 1H saturation-transfer difference nuclear magnetic resonance (1H STD-NMR), ultraviolet (UV) absorption, circular dichroism (CD), steady-state, and time-resolved fluorescence spectroscopies combined with in silico calculations. The binding of HSA:NSAIDs is spontaneous, endothermic, and entropically driven, leading to a conformational rearrangement of HSA with a slight decrease in the α-helix content (7.1% to 7.6%). The predominance of the static quenching mechanism (ground-state association) was identified. Thus, both Stern-Volmer quenching constant (KSV) and binding constant (Kb) values enabled the determination of the binding affinity. In this sense, the KSV and Kb values were found in the order of 104 M-1 at human body temperature, indicating moderate binding affinity with differences in the range of 0.7- and 3.4-fold between KTF and KTL, which agree with the previously reported experimental pharmacokinetic profile. According to 1H STD-NMR data combined with in silico calculations, the aromatic groups in relation to the aliphatic moiety of the drugs interact preferentially with HSA into subdomain IIIA (site II) and are stabilized by interactions via hydrogen bonding and hydrophobic forces. In general, the data obtained in this study have been revised and updated in comparison to those previously reported by other authors who did not account for inner filter corrections, spectral backgrounds, or the identification of the primary mathematical approach for determining the binding affinity of HSA:KTF and HSA:KTL.


Sujet(s)
Anti-inflammatoires non stéroïdiens , Kétoprofène , Kétorolac , Liaison aux protéines , Sérum-albumine humaine , Humains , Kétoprofène/composition chimique , Kétoprofène/métabolisme , Kétoprofène/pharmacocinétique , Anti-inflammatoires non stéroïdiens/composition chimique , Anti-inflammatoires non stéroïdiens/métabolisme , Anti-inflammatoires non stéroïdiens/pharmacocinétique , Kétorolac/composition chimique , Kétorolac/métabolisme , Kétorolac/pharmacocinétique , Sérum-albumine humaine/composition chimique , Sérum-albumine humaine/métabolisme , Dichroïsme circulaire , Thermodynamique , Spectrométrie de fluorescence , Sites de fixation
4.
Inorg Chem ; 63(28): 12992-13004, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38949627

RÉSUMÉ

In recent years, the coordination chemistry of high-spin Fe(III) complexes has increasingly attracted interest due to their potential as effective alternatives to Gd(III)-based MRI contrast agents. This paper discusses the results from our study on Fe(III) complexes with two EDTA derivatives, each modified with either one (EDTA-BOM) or two (EDTA-BOM2) benzyloxymethylene (BOM) groups on the acetic arm(s). These pendant hydrophobic groups enable the complexes to form noncovalent adducts with human serum albumin (HSA), leading to an observed increase in relaxivity due to the reduction in molecular tumbling. Our research involved detailed relaxometric measurements and analyses of both 1H and 17O NMR data at varying temperatures and magnetic field strengths, which is conducted with and without the presence of a protein. A significant finding of this study is the effect of electronic relaxation time on the effectiveness of [Fe(EDTA-BOM)(H2O)]- and [Fe(EDTA-BOM2)(H2O)]- as diagnostic MRI probes. By integrating these relaxometric results with comprehensive thermodynamic, kinetic, and electrochemical data, we have thoroughly characterized how structural modifications to the EDTA base ligand influence the properties of the complexes.


Sujet(s)
Acide édétique , Sérum-albumine humaine , Humains , Sérum-albumine humaine/composition chimique , Acide édétique/composition chimique , Acide édétique/analogues et dérivés , Complexes de coordination/composition chimique , Complexes de coordination/synthèse chimique , Composés du fer III/composition chimique , Structure moléculaire , Thermodynamique , Imagerie par résonance magnétique
5.
Drug Deliv ; 31(1): 2380538, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39044468

RÉSUMÉ

Rheumatoid arthritis (RA) is a chronic inflammatory joint disease accompanied by energy depletion and accumulation of reactive oxygen species (ROS). Inorganic nanoparticles (NPs) offer great promise for the treatment of RA because they mostly have functions beyond being drug carriers. However, conventional nanomaterials become coated with a protein corona (PC) or lose their cargo prematurely in vivo, reducing their therapeutic efficacy. To avoid these problems, we loaded methotrexate (MTX) into hollow structured manganese dioxide nanoparticles (H-MnO2 NPs), then coated them with a 'pseudo-corona' of human serum albumin (HSA) at physiological concentrations to obtain HSA-MnO2@MTX NPs. Efficacy of MTX, MnO2@MTX, and HSA-MnO2@MTX NPs was compared in vitro and in vivo. Compared to MnO2@MTX, HSA-coated NPs were taken up better by lipopolysaccharide-activated RAW264.7 and were more effective at lowering levels of pro-inflammatory cytokines and preventing ROS accumulation. HSA-MnO2@MTX NPs were also more efficient at blocking the proliferation and migration of fibroblast-like synoviocytes from rats with collagen-induced arthritis. In this rat model, HSA-MnO2@MTX NPs showed better biodistribution than other treatments, specifically targeting the ankle joint. Furthermore, HSA-MnO2@MTX NPs reduced swelling in the paw, regulated pro-inflammatory cytokine production, and limited cartilage degradation and signs of inflammation. These results establish the therapeutic potential of HSA-MnO2@MTX NPs against RA.


Sujet(s)
Polyarthrite rhumatoïde , Vecteurs de médicaments , Composés du manganèse , Méthotrexate , Nanoparticules , Oxydes , Espèces réactives de l'oxygène , Sérum-albumine humaine , Animaux , Composés du manganèse/composition chimique , Oxydes/composition chimique , Polyarthrite rhumatoïde/traitement médicamenteux , Souris , Rats , Nanoparticules/composition chimique , Sérum-albumine humaine/composition chimique , Humains , Méthotrexate/pharmacologie , Méthotrexate/administration et posologie , Méthotrexate/pharmacocinétique , Méthotrexate/composition chimique , Espèces réactives de l'oxygène/métabolisme , Cellules RAW 264.7 , Vecteurs de médicaments/composition chimique , Mâle , Arthrite expérimentale/traitement médicamenteux , Distribution tissulaire , Antirhumatismaux/pharmacologie , Antirhumatismaux/administration et posologie , Antirhumatismaux/pharmacocinétique , Rat Sprague-Dawley , Cytokines/métabolisme
6.
Article de Anglais | MEDLINE | ID: mdl-38959705

RÉSUMÉ

This study established a method to prepare and detect OPs adducts on butyrylcholinesterase (BChE) and human serum albumin (HSA). OPs (methyl paraoxon, ethyl paraoxon, methyl parathion, parathion) were incubated with BChE or HSA in vitro, and the adducts of OPs-BChE or OPs-HSA were prepared and qualitatively analyzed by ultra-performance liquid chromatography data-dependent high-resolution tandem mass spectrometry (UPLC-ddHRMS/MS). The amounts of BChE and HSA in the incubating systems were varied and the resulting amounts of the adducts were determined using linear regression. OPs-BChE in the blood were isolated by immunomagnetic separation (IMS), and then digested into the OPs-nonapeptide adduct by pepsin. The proteins in the remaining blood plasma were precipitated and digested by pronase to OPs-tyrosines(OPs-Tyr), which were quantified by UPLC-ddHRMS/MS. 4 OPs-nonapeptides and 4 OPs-Tyr adducts were obtained through the process above. The relative mass deviation of incubated adducts between the actual and theoretical exact masses was less than 10 ppm, and further confirmed by fragmentation mass spectra analysis. Calibration curves were linear for all adducts with a coefficient of determination value (R2) ≥0.995. The limits of detection (LOD) and limits of quantification (LOQ) for adducts detected by MS ranged from 0.05 to 1.0 ng/mL, and from 0.1 to 2.0 ng/mL, respectively. The recovery percentages for adducts ranged from 76.1 % to 107.1 %, matrix effects ranged from 83.4 % to 102.1 %. The inter-day and intra-day precision were 6.1-10.1 % and 6.9-12.9 % for adducts. This study provides a new reference method for the detection of organophosphorus pesticide poisoning. In addition, two blood samples with organophosphorus poisoning were tested by the designed method, and the corresponding adducts were detected in both samples.


Sujet(s)
Butyrylcholine esterase , Composés organiques du phosphore , Spectrométrie de masse en tandem , Humains , Butyrylcholine esterase/sang , Butyrylcholine esterase/composition chimique , Butyrylcholine esterase/métabolisme , Composés organiques du phosphore/composition chimique , Composés organiques du phosphore/sang , Composés organiques du phosphore/analyse , Spectrométrie de masse en tandem/méthodes , Modèles linéaires , Chromatographie en phase liquide à haute performance/méthodes , Pesticides/sang , Pesticides/analyse , Pesticides/composition chimique , Limite de détection , Sérum-albumine humaine/composition chimique , Sérum-albumine humaine/analyse , Reproductibilité des résultats
7.
Int J Mol Sci ; 25(13)2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-39000166

RÉSUMÉ

Pyridoxal-S-methyl-isothiosemicarbazone (PLITSC) is a member of an important group of ligands characterized by different complexation modes to various transition metals. In this contribution, a new complex containing two differently protonated PLITSC ligands ([Fe(PLITSC-H)(PLITSC)]SO4)∙2.5H2O was obtained. The crystal structure was solved by the X-ray analysis and used further for the optimization at B3LYP/6-311++G(d,p)(H,C,N,O,S)/def2-TZVP(Fe) level of theory. Changes in the interaction strength and bond distance due to protonation were observed upon examination by the Quantum Theory of Atoms in Molecules. The protein binding affinity of [Fe(PLITSC-H)(PLITSC)]SO4 towards transport proteins (Bovine Serum Albumin (BSA) and Human Serum Albumin (HSA)) was investigated by the spectrofluorimetric titration and molecular docking. The interactions with the active pocket containing fluorescent amino acids were examined in detail, which explained the fluorescence quenching. The interactions between complex and DNA were followed by the ethidium-bromide displacement titration and molecular docking. The binding along the minor groove was the dominant process involving complex in the proximity of DNA.


Sujet(s)
ADN , Simulation de docking moléculaire , Liaison aux protéines , Sérumalbumine bovine , Ligands , ADN/composition chimique , ADN/métabolisme , Humains , Sérumalbumine bovine/composition chimique , Sérumalbumine bovine/métabolisme , Cristallographie aux rayons X , Thiosemicarbazones/composition chimique , Thiosemicarbazones/métabolisme , Bovins , Complexes de coordination/composition chimique , Complexes de coordination/métabolisme , Animaux , Protons , Composés du fer III/composition chimique , Sérum-albumine humaine/composition chimique , Sérum-albumine humaine/métabolisme , Sites de fixation , Fer/composition chimique , Fer/métabolisme
8.
ACS Nano ; 18(29): 19332-19344, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-38990329

RÉSUMÉ

The therapeutic response of microsatellite instability-high (MSI-H) colorectal cancer (CRC) to immune checkpoint inhibitors (ICI) is indeed surprising; however, the emergence of acquired resistance poses an even greater threat to the survival of these patients. Herein, bioinformatics analysis of MSI-H CRC samples revealed that Wnt signaling pathway represents a promising target for acquired immune reactivation, while subsequent analysis and biochemical testing substantiated the inclination of Wnt-hyperactive CRC cells to engage in macropinocytosis with human serum albumin (HSA). These findings have inspired us to develop an engineered HSA that not only possesses the ability to specifically target cancer cells but also effectively suppresses the Wnt/ß-catenin cascade within these malignant cells. In pursuit of this objective, a comprehensive screening of reported Wnt small-molecule inhibitors was conducted to evaluate their affinity with HSA, and it was discovered that Carnosic acid (CA) exhibited the highest affinity while simultaneously revealing multiple binding sites. Further investigation revealed that CA HSA the capability to engineer HSA into spherical and size-tunable nanostructures known as eHSA (Engineering HSA particle), which demonstrated optimized macropinocytosis-dependent cellular internalization. As anticipated, eHSA effectively suppressed the Wnt signaling pathway and reactivated the acquired immune response in vivo. Furthermore, eHSA successfully restored sensitivity to Anti-PD1's anticancer effects in both subcutaneous and orthotopic mouse homograft models of MSI-H CRC, as well as a humanized hu-PBMC patient-derived orthotopic xenograft (PDOX) mouse model of MSI-H CRC, all while maintaining a favorable safety profile. The collective implementation of this clinically viable immune reactivation strategy not only enables the delivery of Wnt inhibitors for CRC therapy, but also serves as an exemplary demonstration of precision-medicine-guided nanopharmaceutical development that effectively harnesses specific cellular indications in pathological states.


Sujet(s)
Tumeurs colorectales , Immunothérapie , Instabilité des microsatellites , Sérum-albumine humaine , Humains , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/génétique , Tumeurs colorectales/immunologie , Tumeurs colorectales/métabolisme , Animaux , Instabilité des microsatellites/effets des médicaments et des substances chimiques , Souris , Sérum-albumine humaine/composition chimique , Voie de signalisation Wnt/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique , Souris nude , Prolifération cellulaire/effets des médicaments et des substances chimiques
9.
PLoS One ; 19(7): e0303706, 2024.
Article de Anglais | MEDLINE | ID: mdl-39042609

RÉSUMÉ

Serine proteases (SPs) are distributed among all living cells accounting for almost one-third of all proteases. Dysregulation of SPs during inflammation and/or infection can result in devastating consequences, such as skin and lung inflammation, neuroinflammation, arthritis, as well as metastasis of cancerous cells. Such activities are tightly regulated by various inhibitors known as serine protease inhibitors (SERPIN). The thermodynamic investigations previously revealed that L-ascorbic acid binds to trypsin more firmly than pepsin and the binding force of L-ascorbic acid is driven by hydrogen bonds and van der Waals forces. However, the physiochemical effects of such interaction on trypsin and/or pepsin have not yet been reported. Ascorbic acid, also known as vitamin C, is one of the essential nutrients and most common food supplements, fortificants, and preservatives. The aim of this study was to explore the inhibitory effects of ascorbic acid on serine proteases at various concentrations on the in-vitro digestion and/or hydrolysis of intercellular matrix of cell monolayer and human serum albumin (HSA). The inhibitory effects of ascorbic on trypsin are investigated by qualitative and quantitative analysis using SDS-PAGE imaging and NIH densitometric software. Upon the addition of ascorbic acid in both indicator systems, the detachment and/or dissociation of cell monolayer and the digestion of HSA were inhibited in the presence of EDTA-Trypsin. The inhibitory effect of ascorbic acid on the digestion of intercellular matrix and/or hydrolysis of HSA showed a dose-dependent trend until it reached the maximum extent of inhibition. At an equal concentration (2.5mg/mL) ascorbic acid and EDTA-Trypsin exhibited the most potent inhibitory effect on the in vitro digestion of protein either in the form of intercellular matrix in cell monolayer and/or HSA respectively. Overall, our results based on two indicator systems strongly indicate that ascorbic acid may function as a serine protease inhibitor (SERPIN) beyond other important functions.


Sujet(s)
Acide ascorbique , Inhibiteurs de la sérine protéinase , Humains , Acide ascorbique/pharmacologie , Acide ascorbique/composition chimique , Inhibiteurs de la sérine protéinase/pharmacologie , Inhibiteurs de la sérine protéinase/composition chimique , Inhibiteurs de la sérine protéinase/métabolisme , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Sérum-albumine humaine/métabolisme , Sérum-albumine humaine/composition chimique , Trypsine/métabolisme , Trypsine/composition chimique , Lignée cellulaire tumorale , Cellules A549
10.
Talanta ; 277: 126337, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-38823331

RÉSUMÉ

Depletion and separation of histidine-rich proteins from complicated biosamples are crucial for various downstream applications in proteome research and clinical diagnosis. Herein, porous polymer microspheres coated with polyacrylic acid (SPSDVB-PAA) were fabricated through double emulsion interfacial polymerization technique and followed by immobilization of Cu2+ ions on the surface of SPSDVB-PAA. The as-prepared SPSDVB-PAA-Cu with uniform size and nanoscale pore structure enabled coordination interaction of Cu2+ with histidine residues in his-rich proteins, resulting in high-performance adsorption. As metal affinity adsorbent, the SPSDVB-PAA-Cu exhibited favorable selectivity for adsorbing hemoglobin (Hb) and human serum albumin (HSA) with the maximum adsorption capacities of 152.2 and 100.7 mg g-1. Furthermore, the polymer microspheres were used to isolate histidine-rich proteins from human whole blood and plasma, underscoring their effectiveness. The liquid chromatography tandem mass spectrometry (LC-MS/MS) results indicated that the content of 14 most abundant proteins in human plasma was depleted from 81.6 % to 30.7 % and low-abundance proteins were enriched from 18.4 % to 69.3 % after treatment with SPSDVB-PAA-Cu, illustrating potential application of SPSDVB-PAA-Cu in proteomic research.


Sujet(s)
Cuivre , Microsphères , Protéines , Cuivre/composition chimique , Humains , Porosité , Protéines/composition chimique , Protéines/isolement et purification , Protéines/analyse , Adsorption , Chélateurs/composition chimique , Résines acryliques/composition chimique , Histidine/composition chimique , Polymères/composition chimique , Sérum-albumine humaine/composition chimique , Sérum-albumine humaine/analyse
11.
Int J Mol Sci ; 25(12)2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38928485

RÉSUMÉ

Gyrophoric acid (GA), a lichen secondary metabolite, has attracted more attention during the last years because of its potential biological effects. Until now, its effect in vivo has not yet been demonstrated. The aim of our study was to evaluate the basic physicochemical and pharmacokinetic properties of GA, which are directly associated with its biological activities. The stability of the GA in various pH was assessed by conducting repeated UV-VIS spectral measurements. Microsomal stability in rat liver microsomes was performed using Ultra-Performance LC/MS. Binding to human serum albumin (HSA) was assessed using synchronous fluorescence spectra, and molecular docking analysis was used to reveal the binding site of GA to HSA. In the in vivo experiment, 24 Sprague-Dawley rats (Velaz, Únetice, Czech Republic) were used. The animals were divided as follows. The first group (n = 6) included healthy males as control intact rats (♂INT), and the second group (n = 6) included healthy females as controls (♀INT). Groups three and four (♂GA/n = 6 and ♀GA/n = 6) consisted of animals with daily administered GA (10 mg/kg body weight) in an ethanol-water solution per os for a one-month period. We found that GA remained stable under various pH and temperature conditions. It bonded to human serum albumin with the binding constant 1.788 × 106 dm3mol-1 to reach the target tissue via this mechanism. In vivo, GA did not influence body mass gain, food, or fluid intake during the experiment. No liver toxicity was observed. However, GA increased the rearing frequency in behavioral tests (p < 0.01) and center crossings in the elevated plus-maze (p < 0.01 and p < 0.001, respectively). In addition, the time spent in the open arm was prolonged (p < 0.01 and p < 0.001, respectively). Notably, GA was able to pass through the blood-brain barrier, indicating its ability to permeate into the brain and to stimulate neurogenesis in the hilus and subgranular zone of the hippocampus. These observations highlight the potential role of GA in influencing brain function and neurogenesis.


Sujet(s)
Simulation de docking moléculaire , Rat Sprague-Dawley , Animaux , Rats , Mâle , Femelle , Humains , Microsomes du foie/métabolisme , Concentration en ions d'hydrogène , Sérum-albumine humaine/métabolisme , Sérum-albumine humaine/composition chimique , Liaison aux protéines
12.
J Colloid Interface Sci ; 672: 53-62, 2024 Oct 15.
Article de Anglais | MEDLINE | ID: mdl-38830318

RÉSUMÉ

Increasing evidence suggests that the accumulations of reactive oxygen species (ROS), ß-amyloid (Aß), and neuroinflammation are crucial pathological hallmarks for the onset of Alzheimer's disease (AD), yet there are few effective treatment strategies. Therefore, design of nanomaterials capable of simultaneously elimination of ROS and inhibition of Aß aggregation and neuroinflammation is urgently needed for AD treatment. Herein, we designed human serum albumin (HSA)-embedded ultrasmall copper nanoclusters (CuNCs@HSA) via an HSA-mediated fabrication strategy. The as-prepared CuNCs@HSA exhibited outstanding multiple enzyme-like properties, including superoxide dismutase (>5000 U/mg), catalase, and glutathione peroxidase activities as well as hydroxyl radicals scavenging ability. Besides, CuNCs@HSA prominently inhibited Aß fibrillization, and its inhibitory potency was 2.5-fold higher than native HSA. Moreover, CuNCs@HSA could significantly increase the viability of Aß-treated cells from 60 % to over 96 % at 40 µg/mL and mitigate Aß-induced oxidative stresses. The secretion of neuroinflammatory cytokines by lipopolysaccharide-induced BV-2 cells, including tumor necrosis factor-α and interleukin-6, was alleviated by CuNCs@HSA. In vivo studies manifested that CuNCs@HSA effectively suppressed the formation of plaques in transgenic C. elegans, reduced ROS levels, and extended C. elegans lifespan by 5 d. This work, using HSA as a template to mediate the fabrication of copper nanoclusters with robust ROS scavenging capability, exhibited promising potentials in inhibiting Aß aggregation and neuroinflammation for AD treatment.


Sujet(s)
Maladie d'Alzheimer , Peptides bêta-amyloïdes , Cuivre , Nanoparticules métalliques , Cuivre/composition chimique , Cuivre/pharmacologie , Peptides bêta-amyloïdes/métabolisme , Peptides bêta-amyloïdes/antagonistes et inhibiteurs , Peptides bêta-amyloïdes/composition chimique , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Humains , Animaux , Nanoparticules métalliques/composition chimique , Souris , Espèces réactives de l'oxygène/métabolisme , Maladies neuro-inflammatoires/traitement médicamenteux , Sérum-albumine humaine/composition chimique , Caenorhabditis elegans/métabolisme , Taille de particule , Stress oxydatif/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire , Sérumalbumine/composition chimique , Sérumalbumine/pharmacologie , Propriétés de surface
13.
Spectrochim Acta A Mol Biomol Spectrosc ; 320: 124549, 2024 Nov 05.
Article de Anglais | MEDLINE | ID: mdl-38870694

RÉSUMÉ

Ferulic acid ethyl ester (FAEE) is an essential raw material for the formulation of drugs for cardiovascular and cerebrovascular diseases and leukopenia. It is also used as a fixed aroma agent for food production due to its high pharmacological activity. In this study, the interaction of FAEE with Human serum albumin (HSA) and Lysozyme (LZM) was characterized by multi-spectrum and molecular dynamics simulations at four different temperatures. Additionally, the quenching mechanism of FAEE-HSA and FAEE-LZM were explored. Meanwhile, the binding constants, binding sites, thermodynamic parameters, molecular dynamics, molecular docking binding energy, and the influence of metal ions in the system were evaluated. The results of Synchronous fluorescence spectroscopy, UV-vis spectroscopy, CD, three-dimensional fluorescence spectrum, and resonance light scattering showed that the microenvironment of HSA and LZM and the protein conformation changed in the presence of FAEE. Furthermore, the effects of some common metal ions on the binding constants of FAEE-HSA and FAEE-LZM were investigated. Overall, the experimental results provide a theoretical basis for promoting the application of FAEE in the cosmetics, food, and pharmaceutical industries and significant guidance for food safety, drug design, and development.


Sujet(s)
Acides coumariques , Simulation de docking moléculaire , Lysozyme , Liaison aux protéines , Sérum-albumine humaine , Spectrométrie de fluorescence , Humains , Lysozyme/composition chimique , Lysozyme/métabolisme , Acides coumariques/composition chimique , Acides coumariques/métabolisme , Sérum-albumine humaine/métabolisme , Sérum-albumine humaine/composition chimique , Simulation de dynamique moléculaire , Thermodynamique , Sites de fixation , Dichroïsme circulaire , Spectrophotométrie UV , Acides caféiques
14.
Int J Biol Macromol ; 273(Pt 2): 133082, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38878923

RÉSUMÉ

The Shark-derived immunoglobulin new antigen receptors (IgNARs) have gained increasing attention for their high solubility, exceptional thermal stability, and intricate sequence variation. In this study, we immunized whitespotted bamboo shark (Chiloscyllium plagiosum) to create phage display library of variable domains of IgNAR (VNARs) for screening against Human Serum Albumin (HSA), a versatile vehicle in circulation due to its long in vivo half-life. We identified two HSA-binding VNAR clones, 2G5 and 2G6, and enhanced their expression in E. coli with the FKPA chaperone. 2G6 exhibited a strong binding affinity of 13 nM with HSA and an EC50 of 1 nM. In vivo study with a murine model further provided initial validation of 2G6's ability to prolong circulation time by binding to HSA. Additionally, we employed computational molecular docking to predict the binding affinities of both 2G6 and its humanized derivative, H2G6, to HSA. Our analysis unveiled that the complementarity-determining regions (CDR1 and CDR3) are pivotal in the antigen recognition process. Therefore, our study has advanced the understanding of the potential applications of VNARs in biomedical research aimed at extending drug half-life, holding promise for future therapeutic and diagnostic progressions.


Sujet(s)
Simulation de docking moléculaire , Sérum-albumine humaine , Requins , Animaux , Humains , Sérum-albumine humaine/composition chimique , Sérum-albumine humaine/métabolisme , Souris , Récepteurs aux antigènes/composition chimique , Récepteurs aux antigènes/génétique , Récepteurs aux antigènes/métabolisme , Liaison aux protéines , Banque de peptides , Séquence d'acides aminés
15.
J Phys Chem B ; 128(27): 6622-6637, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38937939

RÉSUMÉ

Extensive research has been carried out to investigate the stability and function of human serum albumin (HSA) when exposed to surface-active ionic liquids (SAILs) with different head groups (imidazolium, morpholinium, and pyridinium) and alkyl chain lengths (ranging from decyl to tetradecyl). Analysis of the protein fluorescence spectra indicates noticeable changes in the secondary structure of HSA with varying concentrations of all SAILs tested. Helicity calculations based on the Fourier transform infrared (FTIR) data show that HSA becomes more organized at the micellar concentration of SAILs, leading to an increased protein activity at this level. Small-angle neutron scattering (SANS) data confirm the formation of a bead-necklace structure between the SAILs and HSA. Atomistic molecular dynamics (MD) simulation results identify several hotspots on the protein surface for interaction with SAIL, which results in the modulation of protein conformational fluctuation and stability. Furthermore, fluorescence resonance energy transfer (FRET) experiments with the intramolecular charge transfer (ICT) probe trans-ethyl p-(dimethylamino) cinnamate (EDAC) demonstrate that higher alkyl chain lengths and SAIL concentrations result in a significantly increased energy transfer efficiency. The findings of this study provide a detailed molecular-level understanding of how the protein structure and function are affected by the presence of SAILs, with potential implications for a wide range of applications involving protein-SAIL composite systems.


Sujet(s)
Liquides ioniques , Simulation de dynamique moléculaire , Sérum-albumine humaine , Liquides ioniques/composition chimique , Humains , Sérum-albumine humaine/composition chimique , Sérum-albumine humaine/métabolisme , Transfert d'énergie par résonance de fluorescence , Liaison aux protéines , Conformation des protéines , Diffusion aux petits angles , Tensioactifs/composition chimique
16.
J Biotechnol ; 391: 11-19, 2024 Aug 10.
Article de Anglais | MEDLINE | ID: mdl-38844246

RÉSUMÉ

Ecallantide comprises Kunitz Domain 1 of Tissue Factor Pathway Inhibitor, mutated at seven amino acid positions to inhibit plasma kallikrein (PK). It is used to treat acute hereditary angioedema (HAE). We appended hexahistidine tags to the N- or C-terminus of recombinant Ecallantide (rEcall) and expressed and purified the resulting proteins, with or without fusion to human serum albumin (HSA), using Pichia pastoris. The inhibitory constant (Ki) of rEcall-H6 or H6-rEcall for PK was not increased by albumin fusion. When 125I-labelled rEcall proteins were injected intravenously into mice, the area under the clearance curve (AUC) was significantly increased, 3.4- and 3.6-fold, for fusion proteins H6-rEcall-HSA and HSA-rEcall-H6 versus their unfused counterparts but remained 2- to 3-fold less than that of HSA-H6. The terminal half-life of H6-rEcall-HSA and HSA-H6 did not differ, although that of HSA-rEcall-H6 was significantly shorter than either other protein. Receptor Associated Protein (RAP), a Low-density lipoprotein Receptor-related Protein (LRP1) antagonist, competed H6-rEcall-HSA clearance more effectively than intravenous immunoglobulin (IVIg), a neonatal Fc receptor (FcRn) antagonist. HSA fusion decreases rEcall clearance in vivo, but LRP1-mediated clearance remains more important than FcRn-mediated recycling for rEcall fusion proteins. The properties of H6-rEcall-HSA warrant investigation in a murine model of HAE.


Sujet(s)
Protéines de fusion recombinantes , Animaux , Protéines de fusion recombinantes/pharmacocinétique , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/pharmacologie , Protéines de fusion recombinantes/composition chimique , Souris , Humains , Période , Kallicréine plasmatique/métabolisme , Kallicréine plasmatique/génétique , Sérum-albumine humaine/composition chimique , Sérum-albumine humaine/génétique , Sérum-albumine humaine/métabolisme , Saccharomycetales/génétique , Saccharomycetales/métabolisme , Récepteur Fc , Antigènes d'histocompatibilité de classe I
17.
Org Biomol Chem ; 22(25): 5087-5092, 2024 06 26.
Article de Anglais | MEDLINE | ID: mdl-38835316

RÉSUMÉ

We have devised a supported-amine-catalyzed efficient synthesis of spiro-thiazolone-tetrahydrothiophenes via a sulfa-Michael/aldol cascade approach. The catalyst demonstrated sustained efficacy over 21 cycles. These derivatives were found to exhibit excellent binding abilities with purified human serum albumin as indicated by both in silico and in vitro-based experiments.


Sujet(s)
Amines , Thiophènes , Humains , Catalyse , Thiophènes/composition chimique , Thiophènes/synthèse chimique , Amines/composition chimique , Amines/synthèse chimique , Sérum-albumine humaine/composition chimique , Sérum-albumine humaine/métabolisme , Liaison aux protéines , Spiranes/composition chimique , Spiranes/synthèse chimique , Structure moléculaire , Thiazoles/composition chimique , Thiazoles/synthèse chimique , Simulation de docking moléculaire
18.
Biochemistry ; 63(13): 1697-1707, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38889356

RÉSUMÉ

Human serum albumin (HSA) is a protein carrier that transports a wide range of drugs and nutrients. The amount of glycated HSA (GHSA) is used as a diabetes biomarker. To quantify the GHSA amount, the fluorescent graphene-based aptasensor has been a successful method. In aptasensors, the key mechanism is the adsorption/desorption of albumin from the aptamer-graphene complex. Recently, the graphene quantum dot (GQD) has been reported to be an aptamer sorbent. Due to its comparable size to aptamers, it is attractive enough to explore the possibility of GQD as a part of an albumin aptasensor. Therefore, molecular dynamics (MD) simulations were performed here to reveal the binding mechanism of albumin to an aptamer-GQD complex in molecular detail. GQD saturated by albumin-selective aptamers (GQDA) is studied, and GHSA and HSA are studied in comparison to understand the effect of glycation. Fast and spontaneous albumin-GQDA binding was observed. While no specific GQDA-binding site on both albumins was found, the residues used for binding were confined to domains I and III for HSA and domains II and III for GHSA. Albumins were found to bind preferably to aptamers rather than to GQD. Lysines and arginines were the main contributors to binding. We also found the dissociation of GLC from all GHSA trajectories, which highlights the role of GQDA in interfering with the ligand binding affinity in Sudlow site I. The binding of GQDA appears to impair albumin structure and function. The insights obtained here will be useful for the future design of diabetes aptasensors.


Sujet(s)
Aptamères nucléotidiques , Albumine sérique glycosylée , Graphite , Simulation de dynamique moléculaire , Boîtes quantiques , Sérum-albumine humaine , Graphite/composition chimique , Humains , Aptamères nucléotidiques/composition chimique , Aptamères nucléotidiques/métabolisme , Boîtes quantiques/composition chimique , Sérum-albumine humaine/composition chimique , Sérum-albumine humaine/métabolisme , Sérumalbumine/composition chimique , Sérumalbumine/métabolisme , Liaison aux protéines , Sites de fixation , Agrégats de protéines
19.
Spectrochim Acta A Mol Biomol Spectrosc ; 320: 124598, 2024 Nov 05.
Article de Anglais | MEDLINE | ID: mdl-38850819

RÉSUMÉ

The interactions between gold nanoclusters (AuNCs) and proteins have been extensively investigated. Nevertheless, the structure-activity relationship between gold nanoclusters and proteins in terms of ligand isomerization remained unclear. Here, interactions between Au25NCs modified with para-, inter- and ortho-mercaptobenzoic acid (p/m/o-MBA-Au25NCs) and human serum albumin (HSA) were analyzed. The results of the multispectral approach showed that all three gold nanoclusters bound to the site I in dynamic modes to increase the stability of HSA. There were significant differences in the binding intensity, thermodynamic parameters, main driving forces, and binding ratios between these three gold nanoclusters and HSA, which might be related to the existence forms of the three ligands on the surface of AuNCs. Due to the different polarities of AuNCs themselves, the impact of three AuNCs on the microenvironment of amino acid residues in HSA was also different. It could be seen that ligand isomerization significantly affected the interactions between gold nanoclusters and proteins. This work will provide theoretical guidance for ligand selection and biological applications of metal nanoclusters.


Sujet(s)
Or , Nanoparticules métalliques , Sérum-albumine humaine , Thermodynamique , Or/composition chimique , Humains , Nanoparticules métalliques/composition chimique , Ligands , Sérum-albumine humaine/composition chimique , Sérum-albumine humaine/métabolisme , Relation structure-activité , Isomérie , Liaison aux protéines
20.
Biotechnol J ; 19(5): e2400154, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38719568

RÉSUMÉ

Maximizing product yield in biopharmaceutical manufacturing processes is a critical factor in determining the overall cost of goods, especially given the high value of these biological products. However, there has been relatively limited research on the quantitative analysis of protein losses due to adsorption and fouling during the different membrane filtration processes employed in typical downstream operations. This study aims to provide a comprehensive analysis of protein loss in the range of membrane systems used in downstream processing including clarification, virus removal filtration, ultrafiltration/diafiltration for formulation, and final sterile filtration, all using commercially available membranes with three model proteins (bovine serum albumin, human serum albumin, and immunoglobulin G). The correlation between protein loss and various parameters (i.e., protein type, protein concentration, throughput, membrane morphology, and protein removal mechanism) was also investigated. This study provides important insights into the nature of protein loss during membrane processes as well as a methodology for quantifying protein yield loss in bioprocesses.


Sujet(s)
Membrane artificielle , Ultrafiltration , Humains , Ultrafiltration/méthodes , Filtration/méthodes , Animaux , Produits biologiques/composition chimique , Sérumalbumine bovine/composition chimique , Immunoglobuline G/composition chimique , Adsorption , Bovins , Sérum-albumine humaine/composition chimique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE