Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
J Phys Chem B ; 128(5): 1121-1138, 2024 Feb 08.
Article in English | MEDLINE | ID: mdl-38266243

ABSTRACT

Human serum albumin (HSA) acts as a repository and transporter of substances in the blood. An abnormal concentration may indicate the occurrence of liver- and kidney-related diseases, which has attracted people to investigate the precise quantification of HSA in body fluids. Fluorescent probes can combine with HSA covalently or noncovalently to quantify HSA in urine and plasma. Moreover, probes combined with HSA can improve its photophysical properties; probe-HSA has been applied in real-time monitoring and photothermal and photodynamic therapy in vivo. This Review will introduce fluorescent probes for quantitative HSA according to the three reaction mechanisms of spatial structure, enzymatic reaction, and self-assembly and systematically introduce the application of probes combined with HSA in disease imaging and phototherapy. It will help develop multifunctional applications for HSA probes and provide assistance in the early diagnosis and treatment of diseases.


Subject(s)
Photochemotherapy , Serum Albumin, Human , Humans , Serum Albumin, Human/chemistry , Fluorescent Dyes/therapeutic use , Fluorescent Dyes/chemistry , Phototherapy/methods
2.
Environ Sci Technol ; 57(48): 20118-20126, 2023 Dec 05.
Article in English | MEDLINE | ID: mdl-37916746

ABSTRACT

Layered black phosphorus (LBP) is drawing increasing attention because of its excellent potential in biomedical applications. Properties and bioeffects of LBP depend on its layer number (LN). However, the variation of LN during applications, especially in organisms, is largely unknown. Herein, LBP is found to be exfoliated by human serum albumin (HSA) after the formation of protein coronas. The sorption of HSA on LBP exhibits multiple intermediate equilibrium and size-dependent capacity and is distinguished from traditional multilayer sorption. The loss of LN for LBP increases with the increase of HSA concentrations, e.g., 2, 4, and 6 layers of LBP are exfoliated at 35, 135, and 550 mg/L HSA, respectively. The energy distribution shows that at low HSA concentrations, exfoliation is mainly driven by electrostatic and hydrogen bond interactions. With middle or high HSA concentrations, exfoliation is mainly driven by p-π or hydrophobic interactions, respectively. Layer exfoliation causes the continuous emergence of an unsaturated LBP surface available for adsorbing further HSA, breaking previous sorption saturations. The complete exfoliation of LBP weakens cytotoxicity and promotes internalization to the A-549 cell line compared with pristine or less exfoliated LBP. This finding unveils the exfoliation mechanism of proteins toward LBP and is of benefit to evaluating application performance and biosafety of LBP.


Subject(s)
Phosphorus , Serum Albumin, Human , Humans , Serum Albumin, Human/chemistry
3.
Nat Commun ; 14(1): 6567, 2023 10 17.
Article in English | MEDLINE | ID: mdl-37848496

ABSTRACT

Human serum albumin (HSA) based drug delivery platforms that feature desirable biocompatibility and pharmacokinetic property are rapidly developed for tumor-targeted drug delivery. Even though various HSA-based platforms have been established, it is still of great significance to develop more efficient preparation technology to broaden the therapeutic applications of HSA-based nano-carriers. Here we report a bridging strategy that unfastens HSA to polypeptide chains and subsequently crosslinks these chains by a bridge-like molecule (BPY-Mal2) to afford the HSA reassemblies formulation (BPY@HSA) with enhanced loading capacity, endowing the BPY@HSA with uniformed size, high photothermal efficacy, and favorable therapeutic features. Both in vitro and in vivo studies demonstrate that the BPY@HSA presents higher delivery efficacy and more prominent photothermal therapeutic performance than that of the conventionally prepared formulation. The feasibility in preparation, stability, high photothermal conversion efficacy, and biocompatibility of BPY@HSA may facilitate it as an efficient photothermal agents (PTAs) for tumor photothermal therapy (PTT). This work provides a facile strategy to enhance the loading capacity of HSA-based crosslinking platforms in order to improve delivery efficacy and therapeutic effect.


Subject(s)
Nanoparticles , Neoplasms , Humans , Serum Albumin, Human/chemistry , Photothermal Therapy , Cell Line, Tumor , Neoplasms/therapy , Drug Delivery Systems , Nanoparticles/chemistry , Phototherapy
4.
PLoS One ; 17(5): e0264460, 2022.
Article in English | MEDLINE | ID: mdl-35617167

ABSTRACT

Interaction of thymol, carvacrol and linalool with fungal lipase and Human Serum Albumin (HSA) have been investigated employing UV-Vis spectroscopy Fluorescence and Circular dichroism spectroscopy (CD) along with docking studies. Thymol, carvacrol and linalool displayed approximately 50% inhibition at 1.5 mmol/litre concentrations using para-nitrophenyl palmitate (pNPP). UV-Vis spectroscopy give evidence of the formation of lipase-linalool, lipase-carvacrol and lipase-thymol complex at the ground state. Three molecules also showed complex formation with HSA at the ground state. Fluorescence spectroscopy shows strong binding of lipase to thymol (Ka of 2.6 x 109 M-1) as compared to carvacrol (4.66 x 107 M-1) and linalool (5.3 x 103 M-1). Number of binding sites showing stoichiometry of association process on lipase is found to be 2.52 (thymol) compared to 2.04 (carvacrol) and 1.12 (linalool). Secondary structure analysis by CD spectroscopy results, following 24 hours incubation at 25°C, with thymol, carvacrol and linalool revealed decrease in negative ellipticity for lipase indicating loss in helical structure as compared with the native protein. The lowering in negative ellipticity was in the order of thymol > carvacrol > linalool. Fluorescence spectra following binding of all three molecules with HSA caused blue shift which suggests the compaction of the HSA structure. Association constant of thymol and HSA is 9.6 x 108 M-1 which along with 'n' value of 2.41 suggests strong association and stable complex formation, association constant for carvacrol and linalool was in range of 107 and 103 respectively. Docking results give further insight into strong binding of thymol, carvacrol and linalool with lipase having free energy of binding as -7.1 kcal/mol, -5.0 kcal/mol and -5.2 kcal/mol respectively. To conclude, fungal lipases can be attractive target for controlling their growth and pathogenicity. Employing UV-Vis, Fluorescence and Circular dichroism spectroscopy we have shown that thymol, carvacrol and linalool strongly bind and disrupt structure of fungal lipase, these three phytochemicals also bind well with HSA. Based on disruption of lipase structure and its binding nature with HSA, we concluded thymol as a best anti-lipase molecule among three molecules tested. Results of Fluorescence and CD spectroscopy taken together suggests that thymol and carvacrol are profound disrupter of lipase structure.


Subject(s)
Lipase , Thymol , Binding Sites , Circular Dichroism , Humans , Molecular Docking Simulation , Protein Binding , Serum Albumin, Human/chemistry , Spectrometry, Fluorescence , Thermodynamics , Thymol/pharmacology
5.
J Phys Chem Lett ; 13(7): 1825-1832, 2022 Feb 24.
Article in English | MEDLINE | ID: mdl-35171617

ABSTRACT

Cyanine molecules are important phototheranostic compounds given their high fluorescence yield in the near-infrared region of the spectrum. We report on the frequency and time-resolved spectroscopy of the S2 state of IR806, which demonstrates enhanced emission upon binding to the hydrophobic pocket of human serum albumin (HSA). From excitation-emission matrix spectra and electronic structure calculations, we identify the emission as one associated with a state having the polymethine chain twisted out of plane by 103°. In addition, we find that this configuration is significantly stabilized as the concentration of HSA increases. Spectroscopic changes associated with the S1 and S2 states of IR806 as a function of HSA concentration, as well as anisotropy measurements, confirm the formation of HSA dimers at concentrations greater than 10 µM. These findings imply that the longer-lived S2 state configuration can lead to more efficient phototherapy agents, and cyanine S2 spectroscopy may be a useful tool to determine the oligomerization state of HSA.


Subject(s)
Carbocyanines/chemistry , Serum Albumin, Human/chemistry , Binding Sites , Carbocyanines/metabolism , Density Functional Theory , Dimerization , Humans , Molecular Docking Simulation , Protein Binding , Serum Albumin, Human/metabolism , Spectrometry, Fluorescence , Thermodynamics
6.
Int J Biol Macromol ; 199: 181-188, 2022 Feb 28.
Article in English | MEDLINE | ID: mdl-34973990

ABSTRACT

Intensive research in the field of protein aggregation confirmed that the deposition of amyloid fibrils of proteins are the major cause for the development of various neurotoxic and neurodegenerative diseases, which could be controlled by ensuring the efficient inhibition of aggregation using anti aggregation strategies. Herein, we elaborated the anti amyloidogenic potential of Sunset Yellow (SY) dye against Human Serum Albumin (HSA) fibrillogenesis utilising different biophysical, computational and microscopic techniques. The inhibitory effect of sunset yellow was confirmed by Rayleigh Light Scattering (RLS) measurements along with different dye binding assays (ANS, ThT and CR) by showing concentration dependent reduction in scattering intensity and fluorescence intensity respectively. Further, destabilization and anti fibrillation activity of HSA aggregates were characterized through spectroscopic techniques like Circular Dichroism (CD) and other microscopic techniques like Transmission Electron Microscopy (TEM) for elucidating the structural properties. The SDS-PAGE was also carried out that render the disaggregation effect of the dye on the protein. Moreover, Molecular Docking studies revealed the binding parameters justifying the stable protein-dye complex. Simulation studies were also performed accordingly. Thus, this dye which is used as food additive can serve as a potential aggregation inhibiting agent that can aid in the prevention of amyloidogenic diseases.


Subject(s)
Naturopathy , Serum Albumin, Human , Amyloid/chemistry , Azo Compounds , Circular Dichroism , Humans , Molecular Docking Simulation , Protein Aggregates , Serum Albumin, Human/chemistry
7.
Molecules ; 26(21)2021 Oct 29.
Article in English | MEDLINE | ID: mdl-34770947

ABSTRACT

Boron neutron capture therapy is a unique form of adjuvant cancer therapy for various malignancies including malignant gliomas. The conjugation of boron compounds and human serum albumin (HSA)-a carrier protein with a long plasma half-life-is expected to extend systemic circulation of the boron compounds and increase their accumulation in human glioma cells. We report on the synthesis of fluorophore-labeled homocystamide conjugates of human serum albumin and their use in thiol-'click' chemistry to prepare novel multimodal boronated albumin-based theranostic agents, which could be accumulated in tumor cells. The novelty of this work involves the development of the synthesis methodology of albumin conjugates for the imaging-guided boron neutron capture therapy combination. Herein, we suggest using thenoyltrifluoroacetone as a part of an anticancer theranostic construct: approximately 5.4 molecules of thenoyltrifluoroacetone were bound to each albumin. Along with its beneficial properties as a chemotherapeutic agent, thenoyltrifluoroacetone is a promising magnetic resonance imaging agent. The conjugation of bimodal HSA with undecahydro-closo-dodecaborate only slightly reduced human glioma cell line viability in the absence of irradiation (~30 µM of boronated albumin) but allowed for neutron capture and decreased tumor cell survival under epithermal neutron flux. The simultaneous presence of undecahydro-closo-dodecaborate and labeled amino acid residues (fluorophore dye and fluorine atoms) in the obtained HSA conjugate makes it a promising candidate for the combination imaging-guided boron neutron capture therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Boron Compounds/therapeutic use , Boron Neutron Capture Therapy , Drug Delivery Systems , Homocysteine/chemistry , Serum Albumin, Human/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Boron Compounds/chemical synthesis , Boron Compounds/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Homocysteine/analogs & derivatives , Humans , Molecular Structure
8.
PLoS One ; 16(8): e0256186, 2021.
Article in English | MEDLINE | ID: mdl-34411162

ABSTRACT

Three new uranyl complexes [(UO2)(OAc)2(CMZ)], [(UO2)(OAc)2(MP)] and [(UO2)(OAc)2(SCZ)] were synthesized and characterized by elemental analysis, FT-IR, UV-Vis spectroscopy, powder XRD analysis, and molar conductivity. The IR analysis confirmed binding to the metal ion by the sulfur and ethoxy oxygen atoms in the carbimazole (CMZ) ligand, while in the 6-mercaptopurine (MP) ligand, the sulfur and the N7 nitrogen atom of a purine coordinated binding to the metal ion. The third ligand showed a 1:1 molar ratio and bound via sulfonamide oxygen and the nitrogen of the pyrimidine ring. Analysis of the synthesized complexes also showed that acetate groups had monodentate binding to the (UO22+). Density Functional Theory (DFT) calculations at the B3LYP level showed similar structures to the experimental results. Theoretical quantum parameters predicted the reactivity of the complexes in the order, [(UO2)(OAc)2(SCZ)] > [(UO2)(OAc)2(MP)]> [(UO2)(OAc)2(CMZ)]. DNA binding studies revealed that [(UO2)(OAc)2(SCZ)] and [(UO2)(OAc)2(CMZ)] have the highest binding constant (Kb) among the uranyl complexes. Additionally, strong binding of the MP and CMZ metal complexes to human serum albumin (HSA) were observed by both absorbance and fluorescence approaches. The antibacterial activity of the complexes was also evaluated against four bacterial strains: two gram-negative; Escherichia coli and Klebsiella pneumonia, and two gram-positive; Staphylococcus aureus and Streptococcus mutans. [(UO2)(OAc)2(MP)] had the greatest antibacterial activity against Klebsiella pneumonia, the gram-positive bacteria, with even higher activity than the standard antibiotic. In vitro cytotoxicity tests were also performed against three human cancer lines, and revealed the most cytotoxic complexes to be [(UO2)(OAc)2(SCZ)], which showed moderate activity against a colon cancer cell line. Thus, uranyl addition enhances the antibacterial and anticancer properties of the free ligands.


Subject(s)
Carbimazole/pharmacology , Coordination Complexes/chemistry , Mercaptopurine/chemistry , Uranium/chemistry , Coordination Complexes/chemical synthesis , Coordination Complexes/pharmacology , Escherichia coli/drug effects , Humans , Ligands , Molecular Structure , Nitrogen , Oxygen/chemistry , Serum Albumin, Human/chemistry , Spectroscopy, Fourier Transform Infrared , Staphylococcus aureus/drug effects , Sulfonamides/chemistry
9.
J Inorg Biochem ; 223: 111534, 2021 10.
Article in English | MEDLINE | ID: mdl-34273715

ABSTRACT

The reaction of the dioxouranium(VI) ion with a series of non-steroidal anti-inflammatory drugs (NSAIDs), namely mefenamic acid, indomethacin, diclofenac, diflunisal and tolfenamic acid, as ligands in the absence or presence of diverse N,N'-donors (1,10-phenanthroline,2,2'-bipyridine or 2,2'-bipyridylamine) as co-ligands led to the formation of ten complexes bearing the formulas [UO2(NSAID-O,O')2(O-donor)2] or [UO2(NSAID-O,O')2(N,N'-donor)], respectively. The complexes were characterized with diverse spectroscopic techniques and the crystal structures of three complexes were determined by single-crystal X-ray crystallography. The biological profile of the resultant complexes was assessed in vitro and in silico. The in vitro studies include their antioxidant properties (ability to scavenge free radicals 1,1-diphenyl-picrylhydrazyl and 2,2'-azinobis(3-ethylbenzothiazoline-6-sulfonic acid) and to reduce H2O2), their interaction with DNA (linear calf-thymus DNA or supercoiled circular pBR322 plasmid DNA) and their affinity for serum albumins (bovine and human serum albumin). In silico molecular docking calculations were performed regarding the behavior of the complexes towards DNA and their binding to both albumins.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemistry , Coordination Complexes/chemistry , Free Radical Scavengers/chemistry , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Cattle , Coordination Complexes/chemical synthesis , Coordination Complexes/metabolism , DNA/chemistry , DNA/metabolism , Free Radical Scavengers/chemical synthesis , Free Radical Scavengers/metabolism , Humans , Ligands , Molecular Docking Simulation , Molecular Structure , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , Serum Albumin, Bovine/chemistry , Serum Albumin, Bovine/metabolism , Serum Albumin, Human/chemistry , Serum Albumin, Human/metabolism , Uranium/chemistry
10.
ACS Appl Mater Interfaces ; 13(26): 30373-30382, 2021 Jul 07.
Article in English | MEDLINE | ID: mdl-34180234

ABSTRACT

Phytotherapeutic approaches are of immense value in the treatment of advanced Alzheimer's disease (AD) because of their diverse biological components and potential multitarget mechanisms. In this study, quercetin, a natural neuroprotective flavonoid, was encapsulated in human serum albumin to obtain HSA@QC nanoparticles (HQ NPs) as a natural phyto-antioxidant albumin nanoagent for the treatment of advanced AD. HQ NPs showed excellent antioxidant effects and protected PC12 cells from H2O2-induced oxidative damage. The intranasal administration of HQ NPs in 11-month-old APP/PS1 mice, which represented advanced AD, effectively prevented the loss of body weight, increased survival rates, and significantly reduced oxidative stress, Aß aggregation, neuronal apoptosis, and synaptic damage in the brain. It also ultimately reversed severely impaired cognitive function. In addition to their favorable anti-AD effects, HQ NPs exhibited excellent biosafety and biocompatibility owing to their natural composition and are expected to become an ideal choice for future drug development and clinical applications.


Subject(s)
Alzheimer Disease/drug therapy , Drug Carriers/chemistry , Free Radical Scavengers/therapeutic use , Nanoparticles/chemistry , Quercetin/therapeutic use , Serum Albumin, Human/chemistry , Alzheimer Disease/complications , Alzheimer Disease/pathology , Animals , Apoptosis/drug effects , Body Weight/drug effects , Brain/pathology , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Cognitive Dysfunction/pathology , Drug Carriers/chemical synthesis , Drug Carriers/toxicity , Female , Free Radical Scavengers/toxicity , Humans , Mice, Inbred C57BL , Morris Water Maze Test/drug effects , Nanoparticles/toxicity , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/toxicity , Oxidative Stress/drug effects , PC12 Cells , Quercetin/toxicity , Rats , Serum Albumin, Human/toxicity
11.
Ultrason Sonochem ; 76: 105617, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34126523

ABSTRACT

The focus of this study is the preparation of proteinaceous human serum albumin (HSA) nanocapsules with biocompatible plant oil cores avoiding toxic cross-linker and noxious non-aqueous liquids. The sonochemical preparation of HSA capsules with different plant oils yields particles with narrow size distribution forming suspensions stable for at least 14 days and enabling long-term storage by freezing. Furthermore, wheat germ agglutinin (WGA) as a targeting molecule was successfully embedded into the proteinaceous particle shell at a molar ratio of 7:1 (HSA/WGA). As urothelial cell binding studies revealed up to 55% higher cell binding potential of WGA-grafted particles than those without a targeter, targeted protein nanocapsules represent the first step towards new and innovative formulations.


Subject(s)
Drug Carriers/chemistry , Nanocapsules/chemistry , Plant Oils/chemistry , Serum Albumin, Human/chemistry , Ultrasonic Waves , Humans
12.
ACS Appl Mater Interfaces ; 13(22): 25701-25714, 2021 Jun 09.
Article in English | MEDLINE | ID: mdl-34041901

ABSTRACT

It has been a great challenge to simultaneously inhibit the outgrowth of both the primary tumor and metastasis in metastatic cancer treatment. Substantial studies have evidenced that the interaction of platelets and cancer cells supports tumor metastasis, and platelets are considered to have metastasis-targeting property. Inspired by injury-targeting and metastasis-targeting properties of platelets, we constructed a photothermal therapy strategy with activated platelet-targeting albumin-based nanoparticles, PSN-HSA-PTX-IR780, to amplify drug delivery in the primary tumor at mild temperatures and simultaneously inhibit metastasis via a "platelet bridge". Human serum albumin (HSA) was premodified with a P-selectin-targeting peptide (PSN peptide) or IR780 serving as a photosensitizer. Hybrid albumin nanoparticles were assembled via the disulfide reprogramming method and encapsulated paclitaxel (PTX) to formulate PSN-HSA-PTX-IR780. The PSN-modified albumin nanoparticles could bind with upregulated P-selectin on activated platelets and subsequently target cancer cells by using platelets as a "bridge". In addition, nanoparticle-generated hyperthermia induced tissue injury and increased tumor-infiltrating platelets, thereby recruiting more nanoparticles into the tumor in a self-promoted way. In vivo studies showed that the drug accumulation of PSN-HSA-PTX-IR780 was 2.86-fold higher than that of HSA-PTX-IR780 at the optimal temperature (45 °C), which consequently improved the therapeutic outcome. Moreover, PSN-HSA-PTX-IR780 also effectively targets and inhibits lung metastasis by binding with metastasis-infiltrating platelets. Altogether, the self-promoted nanoplatform provides a unique and promising strategy for metastatic cancer treatment with enhanced drug delivery efficacy.


Subject(s)
Blood Platelets/chemistry , Breast Neoplasms/therapy , Hyperthermia, Induced/methods , Lung Neoplasms/therapy , Nanoparticles/administration & dosage , Paclitaxel/pharmacology , Serum Albumin, Human/metabolism , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Movement , Cell Proliferation , Combined Modality Therapy , Drug Delivery Systems , Female , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Serum Albumin, Human/chemistry , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
Nat Nanotechnol ; 16(7): 830-839, 2021 07.
Article in English | MEDLINE | ID: mdl-33958764

ABSTRACT

Nanoparticulate albumin bound paclitaxel (nab-paclitaxel, nab-PTX) is among the most widely prescribed nanomedicines in clinical use, yet it remains unclear how nanoformulation affects nab-PTX behaviour in the tumour microenvironment. Here, we quantified the biodistribution of the albumin carrier and its chemotherapeutic payload in optically cleared tumours of genetically engineered mouse models, and compared the behaviour of nab-PTX with other clinically relevant nanoparticles. We found that nab-PTX uptake is profoundly and distinctly affected by cancer-cell autonomous RAS signalling, and RAS/RAF/MEK/ERK inhibition blocked its selective delivery and efficacy. In contrast, a targeted screen revealed that IGF1R kinase inhibitors enhance uptake and efficacy of nab-PTX by mimicking glucose deprivation and promoting macropinocytosis via AMPK, a nutrient sensor in cells. This study thus shows how nanoparticulate albumin bound drug efficacy can be therapeutically improved by reprogramming nutrient signalling and enhancing macropinocytosis in cancer cells.


Subject(s)
MAP Kinase Signaling System/drug effects , Mutation , Nanoparticles , Neoplasms, Experimental/drug therapy , Paclitaxel , Proto-Oncogene Proteins p21(ras)/genetics , Serum Albumin, Human , Animals , Cell Line, Tumor , Glucose/deficiency , Glucose/metabolism , Humans , Mice , Mice, Transgenic , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Paclitaxel/pharmacokinetics , Paclitaxel/pharmacology , Pinocytosis , Proto-Oncogene Proteins p21(ras)/metabolism , RAW 264.7 Cells , Serum Albumin, Human/chemistry , Serum Albumin, Human/pharmacology , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics
14.
Nanotechnology ; 32(31)2021 May 10.
Article in English | MEDLINE | ID: mdl-33794506

ABSTRACT

To overcome multi-drug resistance in microbes, highly efficient antimicrobial substances are required that have a controllable antibacterial effect and are biocompatible. In the present study, an efficient phototherapeutic antibacterial agent, human serum albumin (HSA)/reduced graphene oxide (rGO)/Cladophora glomeratabionanocomposite was synthesized by the incorporation of rGO nanoparticles with HSA, forming protein-rGO, and decorated with a natural freshwater seaweedCladophora glomerata. The prepared HSA/rGO/Cladophora glomeratabionanocomposite was characterized by spectroscopic (UV-vis, FTIR, XRD and Raman) and microscopic (TEM and SEM) techniques. The as-synthesized bionanocomposite showed that sunlight/NIR irradiation stimulated ROS-generating dual-phototherapic effects against antibiotic-resistant bacteria. The bionanocomposite exerted strong antibacterial effects (above 96 %) against amoxicillin-resistantP. aeruginosaandS. aureus, in contrast to single-model-phototherapy. The bionanocomposite not only generated abundant ROS for killing bacteria, but also expressed a fluorescence image for bacterial tracking under sunlight/NIR irradiation. Additionally, the bionanocomposite displayed pronounced antioxidant activity.


Subject(s)
Chlorophyta/physiology , Graphite/chemistry , Oxidative Stress/drug effects , Pseudomonas aeruginosa/growth & development , Serum Albumin, Human/chemistry , Staphylococcus aureus/growth & development , Bacterial Load/drug effects , Drug Resistance, Multiple, Bacterial/drug effects , HeLa Cells , Humans , Microbial Viability/drug effects , Nanocomposites , Particle Size , Photochemotherapy , Photothermal Therapy , Pseudomonas aeruginosa/drug effects , Reactive Oxygen Species/metabolism , Staphylococcus aureus/drug effects
15.
Int J Nanomedicine ; 16: 1473-1485, 2021.
Article in English | MEDLINE | ID: mdl-33654397

ABSTRACT

PURPOSE: The near-infrared fluorescent dye indocyanine green (ICG) has shown great potential in the photodynamic therapy (PDT) and photothermal therapy (PTT) of cancer. However, its disadvantages of instability in aqueous solution, short half-life, and non-targeting accumulation limit the effectiveness of ICG PDT/PTT. To overcome the disadvantages of ICG in tumor treatment, we designed PEGylated-human serum albumin (PHSA)-ICG-TAT. In this nanoparticle, PEG4000, the HSA package, and nuclear targeting peptide TAT (human immunodeficiency virus 1 [HIV-1]-transactivator protein) were used to improve the water solubility of ICG, prolong the life span of ICG in vivo, and target the nuclei of tumor cells, respectively. METHODS: The PHSA-ICG-TAT was characterized in terms of morphology and size, ultraviolet spectrum, dispersion stability, singlet oxygen and cellular uptake, and colocalization using transmission electron microscopy and dynamic light scattering, and fluorescence assay, respectively. Subsequently, the anti-tumor effect of PHSA-ICG-TAT was investigated via in vitro and in vivo experiments, including cell viability, apoptosis, comet assays, histopathology, and inhibition curves. RESULTS: The designed ICG-loaded nanoparticle had a higher cell uptake rate and stronger PDT/PTT effect than free ICG. The metabolism of PHSA-ICG-TAT in normal mice revealed that there was no perceptible toxicity. In vivo imaging of mice showed that PHSA-ICG-TAT had a good targeting effect on tumors. PHSA-ICG-TAT was used for the phototherapy of tumors, and significantly suppressed the tumor growth. The tumor tissue sections showed that the cell gap and morphology of the tumor tissue had been obviously altered after treatment with PHSA-ICG-TAT. CONCLUSION: These results indicate that the PHSA-ICG-TAT had a significant therapeutic effect against tumors.


Subject(s)
Breast Neoplasms/drug therapy , Cell Nucleus/metabolism , Nanoparticles/chemistry , Photochemotherapy , Photosensitizing Agents/therapeutic use , Photothermal Therapy , Animals , Breast Neoplasms/pathology , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Dynamic Light Scattering , Female , Fluorescence , Fluorescent Dyes/chemistry , Humans , Indocyanine Green/chemistry , Mice, Inbred BALB C , Nanoparticles/ultrastructure , Photosensitizing Agents/pharmacology , Polyethylene Glycols/chemistry , Serum Albumin, Human/chemistry
16.
Chem Res Toxicol ; 34(1): 144-153, 2021 01 18.
Article in English | MEDLINE | ID: mdl-33410325

ABSTRACT

Emerging evidence suggests that chronic exposure to aristolochic acids (AAs) is one of the etiological pathways leading to chronic kidney disease (CKD). Due to the traditional practice of herbal medicine and AA-containing plants being used extensively as medicinal herbs, over 100 million East Asians are estimated to be at risk of AA poisoning. Given that the chronic nephrotoxicity of AAs only manifests itself after decades of exposure, early diagnosis of AA exposure could allow for timely intervention and disease risk reduction. However, an early detection method is not yet available, and diagnosis can only be established at the end stage of CKD. The goal of this study was to develop a highly sensitive and selective method to quantitate protein adducts of aristolochic acid I (AAI) as a biomarker of AA exposure. The method entails the release of protein-bound aristolactam I (ALI) by heat-assisted alkaline hydrolysis, extraction of ALI, addition of internal standard, and quantitation by liquid chromatography-tandem mass spectrometric analysis. Accuracy and precision of the method were critically evaluated using a synthetic ALI-containing glutathione adduct. The validated method was subsequently used to detect dose-dependent formation of ALI-protein adducts in human serum albumin exposed to AAI and in proteins isolated from the tissues and sera of AAI-exposed rats. Our time-dependent study showed that ALI-protein adducts remained detectable in rats even at 28 days postdosing. It is anticipated that the developed method will fill the technical gap in diagnosing AA intoxication and facilitate the biomonitoring of human exposures to AAs.


Subject(s)
Aristolochic Acids/analysis , Biological Monitoring/methods , Chromatography, Liquid/methods , Glutathione/analysis , Serum Albumin, Human/chemistry , Tandem Mass Spectrometry/methods , Administration, Oral , Animals , Aristolochic Acids/administration & dosage , Biomarkers/analysis , Humans , Male , Molecular Structure , Rats , Rats, Sprague-Dawley
17.
Mol Biol (Mosk) ; 54(4): 653-666, 2020.
Article in Russian | MEDLINE | ID: mdl-32799228

ABSTRACT

Artemisinins are secondary metabolites of the medicinal plant Artemisia annua, have anti-inflammatory, anticarcinogenic, immunomodulating, antimicrobial and other properties. However, the pharmacokinetics, pharmacodynamics, exact molecular targets of artemisinin are not well known. The interaction of artemisinin with human serum albumin was studied both in vitro and in silico, and compared with dexamethasone. The quenching of the fluorescence emission of human serum albumin with artemisinin at different temperatures proceeded according to a single mechanism and indicated the static nature, which is similar to the effect of dexamethasone. Artemisinin and dexamethasone interact with Drug site I on human serum albumin. We have shown for the first time the formation of hydrogen bond with Arg218, which plays a crucial role in the binding of drugs at site I. Dexamethasone forms hydrogen bonds with the side chain of Arg218 and Arg222 and the main chain of Val343. The amino acids of subdomains IIA and IIIA of human serum albumin coincide for both compounds. Studies of the electrophoretic mobility of DNA of sarcoma S-180 cells show that artemisinin does not interact directly with DNA. Therefore, we assume that one of the main transporters of artemisinin is human serum albumin. Moreover, the interaction parameters of artemisinin with human serum albumin coincide with those of dexamethasone.


Subject(s)
Artemisinins/chemistry , Serum Albumin, Human/chemistry , Artemisia annua , Cell Line, Tumor , Computer Simulation , Dexamethasone/chemistry , Humans
18.
Food Chem ; 330: 127241, 2020 Nov 15.
Article in English | MEDLINE | ID: mdl-32540526

ABSTRACT

Curcumin (CUR) is a promising edible phytochemical compound with ideal ulcerative colitis (UC) treatment activity; however, it is characteristically instable in the digestive tract and has a short retention time in colon. Therefore, we designed and fabricated an oral food-grade nanocarrier composed of tannic acid (TA)-coated, Genipin (Gnp)-crosslinked human serum albumin (HSA) to encapsulate CUR (TA/CUR-NPs). The resulting CUR nanoparticles (NPs) were about 220 nm and -28.8 mV. With the assistance of TA layer and Gnp-crosslinking, the entire nano-scaled system effectively delayed CUR release in simulated gastric fluid, prolonged its colon adhesion and increased its uptake in Caco-2 cells. As expected, TA/CUR-NPs oral administration significantly alleviated colitis symptoms in DSS-treated mice when compared with controls by inhibiting the TLR4-linked NF-κB signaling pathway. Collectively, this study indicates that we have developed a convenient, eco-friendly, nano-scaled vehicle for oral delivery of CUR with anti-UC benefit.


Subject(s)
Colitis, Ulcerative/drug therapy , Curcumin/chemistry , Iridoids/chemistry , Serum Albumin, Human/chemistry , Tannins/chemistry , Administration, Oral , Animals , Caco-2 Cells , Curcumin/administration & dosage , Drug Delivery Systems , Humans , Male , Mice , Mice, Inbred BALB C , Nanoparticles/chemistry
19.
Molecules ; 25(10)2020 May 17.
Article in English | MEDLINE | ID: mdl-32429512

ABSTRACT

The interaction of drugs with human serum albumin (HSA) is an important element of therapy. Albumin affects the distribution of the drug substance in the body, as well as its pharmacokinetic and pharmacodynamic properties. On the one hand, inflammation and protein glycation, directly associated with many pathological conditions and old age, can cause structural and functional modification of HSA, causing binding disorders. On the other hand, the widespread availability of various dietary supplements that affect the content of fatty acids in the body means that knowledge of the binding activity of transporting proteins, especially in people with chronic diseases, e.g., diabetes, will achieve satisfactory results of the selected therapy. Therefore, the aim of the present study was to evaluate the effect of a mixture of fatty acids (FA) with different saturated and unsaturated acids on the affinity of acetohexamide (AH), a drug with hypoglycaemic activity for glycated albumin, simulating the state of diabetes in the body. Based on fluorescence studies, we can conclude that the presence of both saturated and unsaturated FA disturbs the binding of AH to glycated albumin. Acetohexamide binds more strongly to defatted albumin than to albumin in the presence of fatty acids. The competitive binding of AH and FA to albumin may influence the concentration of free drug fraction and thus its therapeutic effect.


Subject(s)
Acetohexamide/chemistry , Fatty Acids/chemistry , Hypoglycemic Agents/chemistry , Serum Albumin, Human/chemistry , Serum Albumin/chemistry , Binding, Competitive , Glucose/chemistry , Glycation End Products, Advanced , Humans , Linoleic Acid/chemistry , Molecular Dynamics Simulation , Myristic Acid/chemistry , Oleic Acid/chemistry , Palmitic Acid/chemistry , Protein Binding , Protein Conformation , Solutions , Glycated Serum Albumin
20.
Daru ; 28(1): 209-219, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32270402

ABSTRACT

BACKGROUND: Many solutions have been evaluated to deal with "chemotherapy and radiation-resistant cancer cells' as well as "severe complications of chemotherapy drugs". One of these solutions is the use of herbal compounds with antioxidant properties. Among these antioxidant compounds, curcumin is identified as the strongest one to inhibit cancerous cells proliferation. However, its clinical trials have encountered many constraints, because curcumin is insoluble in water and unstable in physiological conditions. To overcome these limitations, in this study, curcumin was conjugated with human serum albumin (HSA) and its effects on breast cancer cell lines were also measured. METHODS: After making of HSA-curcumin nanoparticles (NPs) by the desolvation technique, they were characterized by the FTIR, DLS, TEM, and SEM method. At the end, its anticancer effects have been examined using MTT test and apoptosis assay. RESULTS: The FTIR graph confirmed that curcumin and HSA have been conjugated along with each other. Particles size was reported to be 220 nm and 180 nm by DLS and SEM, respectively. The zeta potential of HSA-curcumin NPs was -7 mV, while it was -37 mV for curcumin. The MTT and apoptosis assay results indicated that the toxicity of HSA-curcumin NPs on the normal cell are less than curcumin; however, its anti-cancer effects on the cancer cells are much greater, compared to curcumin. CONCLUSION: HSA-curcumin NPs increase curcumin solubility in water as well as its stability in physiological and acidic conditions. These factors have the ability of overwhelming the limitations on using curcumin alone, and they could result in a significant increase in the toxicity of curcumin on the cancer cells without increasing its toxicity on the normal cells. Grapical abstract.


Subject(s)
Anticarcinogenic Agents/administration & dosage , Antineoplastic Agents/administration & dosage , Curcumin/administration & dosage , Drug Carriers/administration & dosage , Nanoparticles/administration & dosage , Serum Albumin, Human/administration & dosage , Anticarcinogenic Agents/chemistry , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Curcumin/chemistry , Drug Carriers/chemistry , Drug Liberation , Humans , Nanoparticles/chemistry , Neoplasms/prevention & control , Serum Albumin, Human/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL