Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 286
Filter
1.
Food Res Int ; 192: 114830, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39147519

ABSTRACT

The natural dual nanofibril system consisting of the rigid semicrystalline nanofibrils disintegrated from citrus fiber (CF) and soft semiflexible nanofibrils self-assembled from glycyrrhizic acid (GA) has been recently shown to be effective structural building blocks for fabrication of emulsion gels. In this work, the effect of the CF nanofibrils prepared by different mechanical disintegration approaches (i.e., high-pressure microfluidization and hydrodynamic cavitation) on the interfibrillar CF-GA interactions and the subsequent formation and properties of emulsion gels were investigated, with the aim of evaluating the potential of the dual nanofibril-stabilized emulsion gels as templates for synthesizing all-natural edible oleogels. The obtained results demonstrate that compared to the cavitation, the high-pressure microfluidization is more capable of generating CF nanofibrils with a higher degree of nanofibrillation and individualization, thus forming a denser CF-GA gel network with higher viscoelasticity and structural stability due to the stronger multiple intrafibrillar and interfibrillar interactions. The emulsion gels stabilized by the dual nanofibril system are demonstrated to be an efficient template to fabricate solid-like oleogels, and the structural properties of the oleogels can be well tuned by the mechanical disintegration of CF and the GA nanofibril concentration. The prepared oleogels possess high oil loading capacity, dense network microstructure, superior rheological and large deformation compression performances, and satisfactory thermal stability, which is attributed to the compact and ordered CF-GA dual nanofibrillar network via multiple hydrogen-bonding interactions in the continuous phase as well as at the droplet surface. This study highlights the unique use of all-natural dual nanofibrils to develop oil structured soft materials for sustainable applications.


Subject(s)
Citrus , Emulsions , Gels , Glycyrrhizic Acid , Nanofibers , Organic Chemicals , Emulsions/chemistry , Glycyrrhizic Acid/chemistry , Citrus/chemistry , Nanofibers/chemistry , Organic Chemicals/chemistry , Gels/chemistry , Rheology , Viscosity
2.
Colloids Surf B Biointerfaces ; 241: 114068, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38954940

ABSTRACT

Inspired by glycyrrhizin's strong pharmacological activities and the directed self-assembly into hydrogels, we created a novel carrier-free, injectable hydrogel (CAR@glycygel) by combining glycyrrhizin with carvacrol (CAR), without any other chemical crosslinkers, to promote wound healing on bacteria-infected skin. CAR appeared to readily dissolve and load into CAR@glycygel. CAR@glycygel had a dense, porous, sponge structure and strong antioxidant characteristics. In vitro, it showed better antibacterial ability than free CAR. For methicillin-resistant Staphylococcus aureus (MRSA), Staphylococcus aureus, and Escherichia coli, the diameter of inhibition zone values of CAR@glycygel were 3.80 ± 0.04, 3.31 ± 0.20 and 3.12 ± 0.24 times greater, respectively, than those of free CAR. The MICs for CAR@glycygel was 156.25 µg/mL while it was 1250.00 µg/mL for free CAR to these three bacteria. Its antibacterial mechanism appeared to involve destruction of the integrity of the bacterial cell wall and biomembrane, leading to a leakage of AKP and inhibition of biofilm formation. In vivo, CAR@glycygel effectively stopped bleeding. When applied to skin wounds on rats infected with MRSA, CAR@glycygel had strong bactericidal activity and improved wound healing. The wound healing rates for CAR@glycygel were 49.59 ± 15.78 %, 93.02 ± 3.09 % and 99.02 ± 0.55 % on day 3, day 7, and day 11, respectively, which were much better than blank control and positive control groups. Mechanisms of CAR@glycygel accelerating wound healing involved facilitating epidermis remolding, promoting the growth of hair follicles, stimulating collagen deposition, mitigating inflammation, and promoting angiogenesis. Overall, CAR@glycygel showed great potential as wound dressing for infected skin wounds.


Subject(s)
Anti-Bacterial Agents , Cymenes , Glycyrrhizic Acid , Hydrogels , Methicillin-Resistant Staphylococcus aureus , Microbial Sensitivity Tests , Wound Healing , Wound Healing/drug effects , Methicillin-Resistant Staphylococcus aureus/drug effects , Hydrogels/chemistry , Hydrogels/pharmacology , Animals , Glycyrrhizic Acid/pharmacology , Glycyrrhizic Acid/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Cymenes/pharmacology , Cymenes/chemistry , Rats , Rats, Sprague-Dawley , Male , Escherichia coli/drug effects , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology
3.
Inorg Chem ; 63(29): 13653-13663, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-38967129

ABSTRACT

The development and exploration of uranium decorporation agents with straightforward synthesis, high removal ability, and low toxicity are crucial guarantees for the safety of workers in the nuclear industry and the public. Herein, we report the use of traditional Chinese medicine licorice for uranium decorporation. Licorice has good adsorption performance and excellent selectivity for uranium in the simulated human environment. Glycyrrhizic acid (GL) has a high affinity for uranium (p(UO2) = 13.67) and will complex with uranium at the carbonyl site. Both licorice and GL exhibit lower cytotoxicity compared to the commercial clinical decorporation agent diethylenetriamine pentaacetate sodium salts (CaNa3-DTPA). Notably, at the cellular level, the uranium removal efficiency of GL is eight times higher than that of CaNa3-DTPA. Administration of GL by prophylactic intraperitoneal injection demonstrates that its uranium removal efficiency from kidneys and bones is 55.2 and 23.9%, while CaNa3-DTPA shows an insignificant effect. The density functional theory calculation of the bonding energy between GL and uranium demonstrates that GL exhibits a higher binding affinity (-2.01 vs -1.15 eV) to uranium compared to DTPA. These findings support the potential of licorice and its active ingredient, GL, as promising candidates for uranium decorporation agents.


Subject(s)
Biological Products , Glycyrrhiza , Glycyrrhizic Acid , Uranium , Glycyrrhiza/chemistry , Uranium/chemistry , Uranium/isolation & purification , Humans , Glycyrrhizic Acid/chemistry , Glycyrrhizic Acid/pharmacology , Glycyrrhizic Acid/isolation & purification , Biological Products/chemistry , Biological Products/pharmacology , Biological Products/isolation & purification , Biological Products/chemical synthesis , Animals , Density Functional Theory , Mice , Cell Survival/drug effects
4.
Langmuir ; 40(29): 14941-14952, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38980061

ABSTRACT

The objective of the current study is to prepare amorphous solid dispersions (ASDs) containing piperine (PIP) by utilizing organic acid glycyrrhizic acid (GA) and inorganic disordered mesoporous silica 244FP (MSN/244FP) as carriers and to investigate their dissolution mechanism. The physicochemical properties of ASDs were characterized with scanning electron microscopy (SEM), powder X-ray diffraction (PXRD), and differential scanning calorimetry (DSC). Fourier transform infrared spectroscopy (FTIR) and one-dimensional proton nuclear magnetic resonance (1H NMR) studies collectively proved that strong hydrogen-bonding interactions formed between PIP and the carriers in ASDs. Additionally, molecular dynamic (MD) simulation was conducted to simulate and predict the physical stability and dissolution mechanisms of the ASDs. Interestingly, it revealed a significant increase in the dissolution of amorphous PIP in ASDs in in vitro dissolution studies. Rapid dissolution of GA in pH 6.8 medium resulted in the immediate release of PIP drugs into a supersaturated state, acting as a dissolution-control mechanism. This exhibited a high degree of fitting with the pseudo-second-order dynamic model, with an R2 value of 0.9996. Conversely, the silanol groups on the outer surface of the MSN and its porous nanostructures enabled PIP to display a unique two-step drug release curve, indicating a diffusion-controlled mechanism. This curve conformed to the Ritger-Peppas model, with an R2 > 0.9. The results obtained provide a clear evidence of the proposed transition of dissolution mechanism within the same ASD system, induced by changes in the properties of carriers in a solution medium of varying pH levels.


Subject(s)
Alkaloids , Benzodioxoles , Piperidines , Polyunsaturated Alkamides , Silicon Dioxide , Piperidines/chemistry , Benzodioxoles/chemistry , Polyunsaturated Alkamides/chemistry , Alkaloids/chemistry , Porosity , Silicon Dioxide/chemistry , Glycyrrhizic Acid/chemistry , Solubility , Molecular Dynamics Simulation , Drug Carriers/chemistry , Particle Size
5.
Arch Biochem Biophys ; 758: 110080, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38960345

ABSTRACT

Glycyrrhizinic acid (GA) is one of the active substances in licorice root. It exhibits antiviral activity against various enveloped viruses, for example, SARS-CoV-2. GA derivatives are promising biologically active compounds from perspective of developing broad-spectrum antiviral agents. Given that GA nicotinate derivatives (Glycyvir) demonstrate activity against various DNA- and RNA-viruses, a search for a possible mechanism of action of these compounds is required. In the present paper, the interaction of Glycyvir with the transmembrane domain of the SARS-CoV-2 E-protein (ETM) in a model lipid membrane was investigated by NMR spectroscopy and molecular dynamics simulation. The lipid-mediated influence on localization of the SARS-CoV-2 E-protein by Glycyvir was observed. The presence of Glycyvir leads to deeper immersion of the ETM in lipid bilayer. Taking into account that E-protein plays a significant role in virus production and takes part in virion assembly and budding, the data on the effect of potential antiviral agents on ETM localization and structure in the lipid environment may provide a basis for further studies of potential coronavirus E-protein inhibitors.


Subject(s)
Antiviral Agents , Glycyrrhizic Acid , Lipid Bilayers , Molecular Dynamics Simulation , SARS-CoV-2 , Lipid Bilayers/metabolism , Lipid Bilayers/chemistry , Glycyrrhizic Acid/pharmacology , Glycyrrhizic Acid/chemistry , SARS-CoV-2/drug effects , SARS-CoV-2/metabolism , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Coronavirus Envelope Proteins/metabolism , Coronavirus Envelope Proteins/chemistry , Humans , Protein Domains , COVID-19 Drug Treatment
6.
Article in English | MEDLINE | ID: mdl-39068869

ABSTRACT

In this study, high-performance liquid chromatography was used to determine four components of Shaoyao Gancao Decoction (SGD), and the effect of purification was evaluated using fingerprints, similarity analysis and cell experiments. An effective method for isolation and purification of SGD was established. The adsorption/desorption properties of SGD were evaluated using resin screening, isothermal analysis, adsorption kinetics, and dynamic adsorption-desorption experiments. It was shown that the Langmuir equation fitted the isotherm data well and that a pseudo-second-order model accurately described kinetic adsorption on AB-8 resin. Analysis of thermodynamic parameters showed that the adsorption process was exothermic. Under the optimal process conditions, the concentrations of albiflorin, paeoniflorin, liquiritin and ammonium glycyrrhizinate in the product were 73.05, 134.04, 45.04 and 75.00 mg/g, respectively. The yields of the four components were 71.89 %-86.19 %. Cell experiments showed that the purified SGD retained anti-inflammatory activity. This research lays the foundation for the separation and purification of SGD and subsequent preparation research.


Subject(s)
Drugs, Chinese Herbal , Glucosides , Monoterpenes , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/isolation & purification , Chromatography, High Pressure Liquid/methods , Glucosides/isolation & purification , Glucosides/chemistry , Monoterpenes/isolation & purification , Monoterpenes/chemistry , Adsorption , Flavanones/isolation & purification , Flavanones/chemistry , Flavanones/analysis , Animals , Glycyrrhizic Acid/isolation & purification , Glycyrrhizic Acid/chemistry , Glycyrrhizic Acid/analysis , Bridged-Ring Compounds/chemistry , Bridged-Ring Compounds/isolation & purification , Mice , Resins, Synthetic/chemistry , Anti-Inflammatory Agents/isolation & purification , Anti-Inflammatory Agents/chemistry , Humans , RAW 264.7 Cells
7.
Int J Pharm ; 660: 124311, 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-38848798

ABSTRACT

The challenges in treating oral cancer include the limited effectiveness and systemic side effects of conventional chemotherapy and radiation therapy. Hyaluronic acid (HA) based Glycyrrhizin (GL) and Methotrexate (MT) loaded localized delivery systems, specifically nanofiber (NF) based platforms, were developed to address these challenges. The electrospinning method was used for the successful fabrication of a homogenous NF membrane and characterized for morphology, drug entrapment efficiency, tensile strength, and ex-vivo mucoadhesive study. Also, it was evaluated for in-vitro drug release profile, ex-vivo drug permeability, in-vitro anti-inflammatory, apoptosis assay by MTT and flow, and against specific cell lines in order to determine their potential for therapeutic use. Superior tensile breaking force (50 g), mucoadhesive strength of 153 gm/cm2, drug permeability, and releasing properties of designed NF, making them perfect requirements for oral cavity delivery. The anticancer potential of MT in the MTT assay and flow cytometry analysis was significantly increased in oral epidermal carcinoma cell (KB cell) for drug-loaded NF with 63.97 ± 1.99 % apoptosis, at 24 h. With these incorporated, GL with MT in NF had an anti-inflammatory potential, also demonstrated in-vitro and in-vivo. In the Ehrlich Ascites Carcinoma (EAC) induced mice model, the optimal formulation's shows better potential for tumor regression when comparing the developed NF formulation to the drugs. Experimental results show that by lowering mucositis-related inflammation and enhancing the effectiveness of oral cancer treatment, a developed nanofiber-based local drug delivery system offers a feasible strategy for managing oral cancer.


Subject(s)
Apoptosis , Drug Liberation , Glycyrrhizic Acid , Hyaluronic Acid , Methotrexate , Mouth Neoplasms , Nanofibers , Hyaluronic Acid/chemistry , Nanofibers/chemistry , Animals , Methotrexate/administration & dosage , Methotrexate/chemistry , Methotrexate/pharmacology , Mouth Neoplasms/drug therapy , Humans , Glycyrrhizic Acid/chemistry , Glycyrrhizic Acid/administration & dosage , Cell Line, Tumor , Apoptosis/drug effects , Drug Delivery Systems/methods , Mice , Male , Drug Carriers/chemistry , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology
8.
Molecules ; 29(9)2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38731484

ABSTRACT

In this study, we developed a green and multifunctional bioactive nanoemulsion (BBG-NEs) of Blumea balsamifera oil using Bletilla striata polysaccharide (BSP) and glycyrrhizic acid (GA) as natural emulsifiers. The process parameters were optimized using particle size, PDI, and zeta potential as evaluation parameters. The physicochemical properties, stability, transdermal properties, and bioactivities of the BBG-NEs under optimal operating conditions were investigated. Finally, network pharmacology and molecular docking were used to elucidate the potential molecular mechanism underlying its wound-healing properties. After parameter optimization, BBG-NEs exhibited excellent stability and demonstrated favorable in vitro transdermal properties. Furthermore, it displayed enhanced antioxidant and wound-healing effects. SD rats wound-healing experiments demonstrated improved scab formation and accelerated healing in the BBG-NE treatment relative to BBO and emulsifier groups. Pharmacological network analyses showed that AKT1, CXCL8, and EGFR may be key targets of BBG-NEs in wound repair. The results of a scratch assay and Western blotting assay also demonstrated that BBG-NEs could effectively promote cell migration and inhibit inflammatory responses. These results indicate the potential of the developed BBG-NEs for antioxidant and skin wound applications, expanding the utility of natural emulsifiers. Meanwhile, this study provided a preliminary explanation of the potential mechanism of BBG-NEs to promote wound healing through network pharmacology and molecular docking, which provided a basis for the mechanistic study of green multifunctional nanoemulsions.


Subject(s)
Antioxidants , Emulsifying Agents , Emulsions , Glycyrrhizic Acid , Molecular Docking Simulation , Wound Healing , Wound Healing/drug effects , Animals , Emulsions/chemistry , Emulsifying Agents/chemistry , Emulsifying Agents/pharmacology , Rats , Antioxidants/pharmacology , Antioxidants/chemistry , Antioxidants/chemical synthesis , Glycyrrhizic Acid/pharmacology , Glycyrrhizic Acid/chemistry , Polysaccharides/chemistry , Polysaccharides/pharmacology , Green Chemistry Technology , Humans , Rats, Sprague-Dawley , Nanoparticles/chemistry , Plant Oils/chemistry , Plant Oils/pharmacology , Fabaceae/chemistry , Male , Particle Size , Cell Movement/drug effects
9.
Chem Biol Drug Des ; 103(5): e14536, 2024 May.
Article in English | MEDLINE | ID: mdl-38725079

ABSTRACT

This research was designed to prospect the mechanism and impact of glycyrrhizic acid (GA) on DNA damage repair and cisplatin (CP)-induced apoptosis of melanoma cells. First, human melanoma cell SK-MEL-28 was stimulated using GA for 24, 48, and 72 h. Then, the optimal treatment time and dosage were selected. After that, cell counting kit-8 (CCK-8) was employed for testing the cell viability, flow cytometry for the apoptosis, comet assay for the DNA damage of cells, and western blot for the cleaved-Caspase3, Caspase3, Bcl-2, and γH2AX protein expression levels. The experimental outcomes exhibited that as the GA concentration climbed up, the SK-MEL-28 cell viability dropped largely, while the apoptosis level raised significantly, especially at the concentration of 100 µm. In addition, compared with GA or CPtreatment only, CP combined with GA notably suppressed the viability of melanoma cells and promoted cell apoptosis at the cytological level. At the protein level, the combined treatment notably downregulated the Bcl-2 and Caspase3 expression levels, while significantly upregulated the cleaved-Caspase3 and γH2AX expression levels. Besides, CP + GA treatment promoted DNA damage at the DNA molecular level. Collectively, both GA and CP can inhibit DNA damage repair and enhance the apoptosis of SK-MEL-28 cells, and the synergistic treatment of both exhibits better efficacy.


Subject(s)
Apoptosis , Cisplatin , DNA Damage , DNA Repair , Glycyrrhizic Acid , Melanoma , Cisplatin/pharmacology , Humans , Glycyrrhizic Acid/pharmacology , Glycyrrhizic Acid/chemistry , Apoptosis/drug effects , Melanoma/drug therapy , Melanoma/metabolism , Melanoma/pathology , Cell Line, Tumor , DNA Damage/drug effects , DNA Repair/drug effects , Cell Survival/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Caspase 3/metabolism , Drug Synergism , Proto-Oncogene Proteins c-bcl-2/metabolism
10.
Int J Pharm ; 658: 124213, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38729382

ABSTRACT

Safe and effective Cu2+ supplementation in local lesion is crucial for minimizing toxicity of DSF-based chemotherapy. Targeted delivery of Cu2+ appears more promising. Intraperitoneal chemotherapy for peritoneal carcinoma (PC) establishes "face-to-face" contact between targeted nanocarriers and tumor tissue. Herein, this study developed a biodegradable, injectable thermosensitive hydrogel that coencapsulating DSF submicroemulsion (DSF-SE) and folate-modified liposome loading glycyrrhizic acid-Cu (FCDL). FCDL acted as 'beneficial horse' to target the tumor-localized folate receptor, thus liberating Cu2+ in tumor nidus. The prepared FCDL and DSF-SE were found with uniform sizes (160.2 nm, 175.4 nm), low surface charge (-25.77 mV, -16.40 mV) and high encapsulation efficiency (97.93 %, 90.08 %). In vitro drug release profile of FCDL, DSF-SE and FCDL&DSF-SE@G followed a sustained release pattern. And the release behavior of Cu2+ from FCDL was pH-related, i.e., Cu2+ was released faster under acidic condition. When FCDL and DSF-SE were loaded into an PLGA-PEG-PLGA-based hydrogel system, FCDL&DSF-SE@G was formed to ensure separated delivery of Cu2+ and DSF in space but synchronized release over time. The rheology experiment showed a satisfactory gelling temperature of 32.7 °C. In vitro cytotoxicity study demonstrated that FCDL&DSF-SE@G significantly lowered the IC50 of free Cu2+/DSF, Cu2+/DSF hydrogel and non-targeted analogue by almost 70 %, 65 % and 32 %, respectively. Accordingly, in tumor-bearing mice, FCDL&DSF-SE@G augmented the tumor inhibition rates for the same formulations by 352 %, 145 % and 44 %, respectively. The main mechanism was attributed to higher uptake of FCDL and DSF-SE, resulting in increased Cu(DDTC)2 formation, ROS production and cell apoptosis. In conclusion, this targeted nanotherapy approach with dual-nanocarriers loaded hydrogel system, with its focus on face-to-face contact between nanocarriers and tumor tissues in the peritoneal cavity, holds significant promise for intraperitoneal chemotherapy in PC.


Subject(s)
Copper , Delayed-Action Preparations , Drug Liberation , Folic Acid , Liposomes , Folic Acid/chemistry , Folic Acid/administration & dosage , Animals , Copper/chemistry , Copper/administration & dosage , Cell Line, Tumor , Humans , Glycyrrhizic Acid/chemistry , Glycyrrhizic Acid/administration & dosage , Hydrogels/chemistry , Nanoparticles/chemistry , Mice, Inbred BALB C , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Mice , Temperature , Cell Survival/drug effects , Female , Mice, Nude , Drug Carriers/chemistry , Polyethylene Glycols/chemistry
11.
Pharm Dev Technol ; 29(6): 551-558, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38808380

ABSTRACT

The purpose of this study was to investigate the impact of different functional stabilizers on in vitro/in vivo drug performances after oral administration of drug nanocrystals. Quercetin nanocrystals (QT-NCs) respectively stabilized by five types of functional stabilizers, including hydroxypropyl methyl cellulose E15 (HPMC E15), poloxamer 407 (P407), poloxamer 188 (P188), D-α-tocopherol polyethylene glycol succinate (TPGS), and glycyrrhizin acid (GL), were fabricated by wet media milling technique. The particle size, morphology, physical state, drug solubility, drug dissolution in vitro, and orally pharmacokinetic behaviors of all QT-NCs were investigated. All QT-NCs with similar particle size about 200 nm were obtained by controlling milling speed and milling time. No significant differences in particles shape and crystalline nature were found for QT-NCs stabilized by different functional stabilizers. But the solubility and dissolution of QT-NCs were significantly influenced by the different functional stabilizers. The AUC0∼t of all QT-NCs after oral administration was in the following order: QT-NCs/P188 ≈ QT-NCs/HPMC E15 > QT-NCs/GL > QT-NCs/P407 ≈ QT-NCs/TPGS, and the Cmax showed an order of QT-NCs/P407 > QT-NCs/P188 ≈ QT-NCs/GL > QT-NCs/HPMC E15 > QT-NCs/TPGS. Both of QT-NCs/P407 and QT-NCs/TPGS exhibited faster oral absorption with Tmax at 0.5 h and 0.83 h, respectively, while the other three QT-NCs (QT-NCs/P188, QT-NCs/GL and QT-NCs/HPMC E15) showed a relatively slow absorption with same Tmax at 5.33 h. The longest MRT0∼t (11.72 h) and t1/2z (32.22 h) were observed for QT-NCs/HPMC E15. These results suggested that the different functional stabilizers could significantly influence on drug solubility, drug dissolution in vitro and orally pharmacokinetic behavior of QT-NCs, and it is possible to alter the drug dissolution in vitro, oral absorption and drug retention in vivo by changing the type of functional stabilizers in NCs preparation.


Subject(s)
Biological Availability , Nanoparticles , Quercetin , Solubility , Quercetin/pharmacokinetics , Quercetin/administration & dosage , Quercetin/chemistry , Quercetin/pharmacology , Nanoparticles/chemistry , Animals , Administration, Oral , Male , Particle Size , Rats, Sprague-Dawley , Drug Liberation , Rats , Excipients/chemistry , Poloxamer/chemistry , Glycyrrhizic Acid/chemistry , Glycyrrhizic Acid/pharmacokinetics , Glycyrrhizic Acid/administration & dosage , Vitamin E/chemistry , Vitamin E/pharmacokinetics
12.
Antiviral Res ; 227: 105920, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38821317

ABSTRACT

COVID-19 pandemic is predominantly caused by SARS-CoV-2, with its main protease, Mpro, playing a pivotal role in viral replication and serving as a potential target for inhibiting different variants. In this study, potent Mpro inhibitors were identified from glycyrrhizic acid (GL) derivatives with amino acid methyl/ethyl esters. Out of the 17 derivatives semisynthesized, Compounds 2, 6, 9, and 15, with methionine methyl esters, D-tyrosine methyl esters, glutamic acid methyl esters, and methionines in the carbohydrate moiety, respectively, significantly inhibited wild-type SARS-CoV-2 Mpro-mediated proteolysis, with IC50 values ranging from 0.06 µM to 0.84 µM. They also demonstrated efficacy in inhibiting trans-cleavage by mutant Mpro variants (Mpro_P132H, Mpro_E166V, Mpro_P168A, Mpro_Q189I), with IC50 values ranging from 0.05 to 0.92 µM, surpassing nirmatrelvir (IC50: 1.17-152.9 µM). Molecular modeling revealed stronger interactions with Valine166 in the structural complex of Mpro_E166V with the compounds compared to nirmatrelvir. Moreover, these compounds efficiently inhibited the post-entry viral processes of wild-type SARS-CoV-2 single-round infectious particles (SRIPs), mitigating viral cytopathic effects and reducing replicon-driven GFP reporter signals, as well as in vitro infectivity of wild-type, Mpro_E166V, and Mpro_Q189I SRIPs, with EC50 values ranging from 0.02 to 0.53 µM. However, nirmatrelvir showed a significant decrease in inhibiting the replication of mutant SARS-CoV-2 SRIPs carrying Mpro_E166V (EC50: >20 µM) and Mpro_Q189I (EC50: 13.2 µM) compared to wild-type SRIPs (EC50: 0.06 µM). Overall, this study identifies four GL derivatives as promising lead compounds for developing treatments against various SARS-CoV-2 strains, including Omicron, and nirmatrelvir-resistant variants.


Subject(s)
Antiviral Agents , Coronavirus 3C Proteases , Drug Resistance, Viral , Glycyrrhizic Acid , SARS-CoV-2 , SARS-CoV-2/drug effects , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Glycyrrhizic Acid/pharmacology , Glycyrrhizic Acid/chemistry , Humans , Coronavirus 3C Proteases/antagonists & inhibitors , Coronavirus 3C Proteases/metabolism , Esters/pharmacology , Esters/chemistry , Chlorocebus aethiops , COVID-19 Drug Treatment , Animals , Vero Cells , Molecular Docking Simulation , Virus Replication/drug effects , Protease Inhibitors/pharmacology , Protease Inhibitors/chemistry , COVID-19/virology , Amino Acids/pharmacology , Indoles/pharmacology , Indoles/chemistry , Mutation , Lactams , Leucine , Nitriles , Proline
13.
Int J Biol Macromol ; 266(Pt 2): 131368, 2024 May.
Article in English | MEDLINE | ID: mdl-38580025

ABSTRACT

This study presents an innovative approach for targeted drug delivery through the development of Glycyrrhizic acid-loaded zein nanoparticles (GA-LNPs) as a proficient carrier system. The juxtaposition of zein, a hydrophobic biological macromolecule as a protein carrier, and Glycyrrhizic acid (GA), a hydrophilic therapeutic compound, exemplifies the adaptability of hydrocolloids within cutting-edge drug delivery systems. The characterization and functional traits of research encompass multifaceted analyses of natural macromolecules, which elucidate the homogeneous and spherical morphology of GA-LNPs with an average size of 170.49 nm. The controlled drug release profile of GA, orchestrated under simulated gastrointestinal conditions, adheres to diffusion-based Higuchi kinetics, reflecting the controlled release of the natural macromolecules. The intermolecular interactions among Zein, GA, and cross-linker EDC, facilitated through molecular dynamics simulations, fortify the structural integrity of the encapsulation matrix. In Vitro studies revealed enhanced cellular uptake of GA-LNPs in MCF-7 breast cancer cells. This cellular internalization was further confirmed through cytotoxicity assessments using MTT and apoptosis assays (fluorescence microscopy), which demonstrated the prominent anticancer effects of GA-LNPs on MCF-7 in time/dose-dependent manner. The successful formulation of GA-LNPs, coupled with their sustained release and potent anticancer properties, makes them a potential platform for advanced targeted therapeutic strategies in biomedical applications.


Subject(s)
Breast Neoplasms , Drug Carriers , Glycyrrhizic Acid , Nanoparticles , Zein , Glycyrrhizic Acid/chemistry , Glycyrrhizic Acid/pharmacology , Zein/chemistry , Humans , Nanoparticles/chemistry , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , MCF-7 Cells , Drug Carriers/chemistry , Drug Delivery Systems , Drug Liberation , Apoptosis/drug effects , Molecular Dynamics Simulation , Female , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Computer Simulation , Cell Survival/drug effects
14.
Int J Pharm ; 656: 124111, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38609057

ABSTRACT

Methotrexate (MTX) is recognized as the golden standard for rheumatoid arthritis (RA) treatment. However, it can cause liver damage in long-term application. Although nanomedicines can target to inflamed sites, most of them tend to accumulate in liver. Glycyrrhizinic acid (GA) holds potential to reverse MTX-associated hepatotoxicity. The combination of GA and MTX might achieve a synergistic anti-inflammatory efficacy and reduced hepatotoxicity. As MTX and GA have totally different in vivo performance, it is necessary to co-encapsulate them in one carrier to coordinate their in vivo fates. Here, we co-delivered MTX and GA to arthritic joints using a human serum albumin-based nanoparticle (HSN). We found the dual drug-loaded albumin nanoparticles (HSN/MTX/GA) could preferentially distribute in inflamed joints, where GA can extend MTX retention by inhibiting the expression of efflux pumps for MTX, thereby exerting synergistic therapeutic effect. In liver tissues, GA was able to reverse the MTX-induced liver damage by activating anti-oxidant defense Nrf2/HO-1 and anti-apoptosis Bcl-2/Bax signaling. We offer a combinational strategy to effectively overcome the MTX-induced hepatotoxicity and enhance the anti-rheumatic efficacy simultaneously. Furthermore, we verified the underlying mechanism about how GA cooperated with MTX in vivo for the first time. Our findings can provide valuable insights for long-term treatment of RA.


Subject(s)
Antirheumatic Agents , Chemical and Drug Induced Liver Injury , Glycyrrhizic Acid , Methotrexate , Nanoparticles , Methotrexate/administration & dosage , Animals , Antirheumatic Agents/administration & dosage , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/etiology , Glycyrrhizic Acid/administration & dosage , Glycyrrhizic Acid/chemistry , Glycyrrhizic Acid/pharmacology , Arthritis, Rheumatoid/drug therapy , Liver/drug effects , Liver/metabolism , Liver/pathology , Mice , Serum Albumin, Human/chemistry , Serum Albumin, Human/administration & dosage , Male , Drug Synergism , Humans , Drug Carriers/chemistry , Arthritis, Experimental/drug therapy
15.
Int J Biol Macromol ; 267(Pt 2): 131423, 2024 May.
Article in English | MEDLINE | ID: mdl-38583832

ABSTRACT

This article reveals the binding mechanism between glycyrrhizic acid (GA) and α-synuclein to may provide further information for the modulation of synucleinopathies using bioactive compounds. Therefore, the inhibitory activities of GA against α-synuclein aggregation and induced neurotoxicity were evaluated using different assays. Results showed that α-synuclein-GA binding was mediated by intermolecular hydrogen bonds leading to the formation of a slightly folded complex. Theoretical studies revealed that GA binds to the N-terminal domain of α-synuclein and triggers a compact structure around a major part of the N-terminal and the NAC regions along with fluctuations in the C-terminal domain, which are prerequisites for the inhibition of α-synuclein aggregation. Then, the cellular assays showed that GA as a potential small molecule can inhibit the oligomerization of α-synuclein and relevant neurotoxicity through modulation of neural viability, membrane leakage, and ROS formation in a concentration-dependent manner. As a result, the primary mechanism of GA's anti-aggregation and neuroprotective activities is the reorganized α-synuclein structure and fluctuating C-terminal domain, which promotes long-range transient intramolecular contacts between the N-terminal and the C-terminal domain.


Subject(s)
Glycyrrhizic Acid , Protein Aggregates , Synucleinopathies , alpha-Synuclein , Humans , alpha-Synuclein/metabolism , alpha-Synuclein/chemistry , Cell Survival/drug effects , Glycyrrhizic Acid/pharmacology , Glycyrrhizic Acid/chemistry , Hydrogen Bonding , Molecular Docking Simulation , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemistry , Protein Aggregates/drug effects , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/drug therapy , Protein Binding , Reactive Oxygen Species/metabolism , Synucleinopathies/metabolism , Synucleinopathies/pathology
16.
Int J Biol Macromol ; 269(Pt 2): 131896, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38677681

ABSTRACT

The recovery of infectious wound tissues presents a significant global health challenge due to the impediments posed by the harsh healing microenvironment, which includes ongoing bacterial invasion, high oxidative stress, inflammatory response, and impaired angiogenesis. To overcome the above issues, we propose a composite hydrogel based on the multiple-crosslinked mechanism involving the covalent network of CC bonds within catechol and maleic-modified HA (CMHA), the self-assembly network of glycyrrhizic acid (GA), and the metal-polyphenol coordination induced by ZHMCe for accelerating infectious wound healing. The resulting CMHA/GA/ZHMCe hydrogels demonstrate enhanced mechanical, adhesive, antioxidative, and antibacterial properties. Importantly, the hydrogel system possesses wound environment-responsive properties that allow it to adapt to the specific therapeutic requirements of different stages by regulating various enzyme activities in the healing of infected wounds. Furthermore, the biocompatible CMHA/GA/ZHMCe shows the ability to promote cell migration and angiogenesis in vitro while reprogramming macrophages toward an anti-inflammatory phenotype due to the effective release of active ingredients. In vivo experiments confirm that the CMHA/GA/ZHMCe hydrogel significantly enhances infectious wound healing by accelerating re-epithelialization, promoting collagen deposition, regulating inflammation, and contributing to vascularization. These findings underscore the therapeutic potential of our hydrogel dressings for the treatment of bacterially infected cutaneous wound healing.


Subject(s)
Hyaluronic Acid , Hydrogels , Wound Healing , Hydrogels/chemistry , Hydrogels/pharmacology , Wound Healing/drug effects , Animals , Mice , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Glycyrrhizic Acid/pharmacology , Glycyrrhizic Acid/chemistry , Male , RAW 264.7 Cells , Humans , Wound Infection/drug therapy , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Antioxidants/pharmacology , Antioxidants/chemistry
17.
Adv Healthc Mater ; 13(18): e2304300, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38589053

ABSTRACT

Spinal cord injury (SCI) often leads to a severe permanent disability. A poor inflammatory microenvironment and nerve electric signal conduction block are the main reasons for difficulty in spinal cord nerve regeneration. In this study, black phosphorus (BP) and glycyrrhizic acid (GA) are integrated into methacrylate-modified silk fibroin (SF) to construct a bifunctional injectable hydrogel (SF/BP/GA) with appropriate conductivity and the ability to inhibit inflammation to promote neuronal regeneration after SCI. This work discovers that the SF/BP/GA hydrogel can reduce the oxidative damage mediated by oxygen free radicals, promote the polarization of macrophages toward the anti-inflammatory M2 phenotype, reduce the expression of inflammatory factors, and improve the inflammatory microenvironment. Moreover, it induces neural stem cell (NSC) differentiation and neurosphere formation, restores signal conduction at the SCI site in vivo, and ameliorates motor function in mice with spinal cord hemisection, revealing a significant neural repair effect. An injectable, electroconductive, free-radical-scavenging hydrogel is a promising therapeutic strategy for SCI repair.


Subject(s)
Fibroins , Glycyrrhizic Acid , Hydrogels , Nanocomposites , Phosphorus , Spinal Cord Injuries , Animals , Fibroins/chemistry , Fibroins/pharmacology , Hydrogels/chemistry , Hydrogels/pharmacology , Spinal Cord Injuries/therapy , Spinal Cord Injuries/drug therapy , Mice , Phosphorus/chemistry , Glycyrrhizic Acid/chemistry , Glycyrrhizic Acid/pharmacology , Nanocomposites/chemistry , Free Radical Scavengers/pharmacology , Free Radical Scavengers/chemistry , Neural Stem Cells/drug effects , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Electric Conductivity , Spinal Cord Regeneration/drug effects , Cell Differentiation/drug effects
18.
Adv Healthc Mater ; 13(20): e2400580, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38574340

ABSTRACT

High glucose blood and bacterial infection remain major issues for the slow healing of diabetic wounds, so developing functional biosensing composite with excellent antibacterial and remarkable glucose response sensitivity is necessary and prospective. Herein, by in situ synthesis AgNPs on the surface of self-prepared PTIGA elastomers, PTIGA-AgNPs conductive composites are obtained with efficient synergistic antibacterial effect, excellent mechanical and self-healing properties. The strain of the composites can reach 1800%, and its self-healing efficiency exceeds 90% at 60 °C within 8 h. Both elastomers and composites represent excellent biocompatibility and the antibacterial rate against E. coli and S. aureus exceeded 90%. Moreover, the biosensor assembled from the conductive composites exhibits excellent glucose response sensitivity and stability, with a sensitivity coefficient of 0.518 mA mm-1 in the range of 0.2-3.6 × 10-3 m glucose concentration, as well as a low detection limit of 0.08 × 10-3 m. Furthermore, based on the remarkable antibacterial performance and bioactivity derived from GA, the composites reduce the expression of pro-inflammatory factors and promote the production of anti-inflammatory factors, and effectively promote the regeneration of skin and granulation tissue of wounds in a diabetic full-thickness skin defect model, demonstrating the enormous therapeutic potential in diabetic wound healing.


Subject(s)
Anti-Bacterial Agents , Biosensing Techniques , Escherichia coli , Glycyrrhizic Acid , Silver , Staphylococcus aureus , Wound Healing , Wound Healing/drug effects , Biosensing Techniques/methods , Animals , Escherichia coli/drug effects , Glycyrrhizic Acid/chemistry , Glycyrrhizic Acid/pharmacology , Staphylococcus aureus/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Silver/chemistry , Silver/pharmacology , Mice , Metal Nanoparticles/chemistry , Blood Glucose/analysis , Male
19.
Recent Pat Anticancer Drug Discov ; 19(2): 154-164, 2024.
Article in English | MEDLINE | ID: mdl-38214355

ABSTRACT

BACKGROUND: Multidrug resistance (MDR) of cancer cells is a major obstacle to efficient cancer chemotherapy. Combination therapy is expected to enhance the anticancer effect and reverse MDR. Numerous patents involve different kinds of nanoparticles for the co-delivery of multiple chemotherapeutics, but the FDA has approved none. OBJECTIVE: In this study, oxymatrine (OMT) and glycyrrhizin (GL) were co-loaded into phytosomes as the core of nanocarriers, and the shell was cross-linked with chitosan (CS) and hyaluronic acid (HA) with the capability for the controlled, sequential release and the targeted drug uptake. METHODS: Phospholipid complexes of OMT and GL (OGPs) were prepared by a solvent evaporation technique and could self-assemble in an aqueous solution to form phytosomes. CS and HA were sequentially coated on the surface of OGPs via electrostatic interactions to obtain CS coated OGPs (CS-OGPs) and HA modified CS-OGPs (HA-CS-OGPs), respectively. The particle size and zeta potential were measured to optimize the formulations. In vitro cytotoxicity and cellular uptake experiments on HepG2 cells were performed to evaluate the anticancer activity. RESULTS: OGPs were obtained with nano-size around 100 nm, and CS and HA coating on phytosomes could change the particle size and surface potential. The drug loading of OMT and GL showed that the nanocarriers could maintain a fixed ratio of 1:1. The in vitro release experiments indicated the release of OMT and GL was pH-dependent and sequential: the release of OMT from CS-OGPs and HA-CS-OGPs was significantly increased at pH 5.0 compared to the release at pH 7.4, while GL exhibited sustained released from CS-OGPs and HA-CS-OGPs at pH 5.0. Furthermore, in vitro cytotoxicity and cellular uptake experiments on HepG2 cells demonstrated that the co-delivery system based on phytosomes had significant synergistic anti-tumor activities, and the effects were enhanced by CS and HA modification. CONCLUSION: The delivery of OMT and GL via HA-CS-OGPs might be a promising treatment to reverse MDR in cancer therapy.


Subject(s)
Chitosan , Matrines , Nanoparticles , Humans , Chitosan/chemistry , Phytosomes , Hyaluronic Acid/chemistry , Glycyrrhizic Acid/pharmacology , Glycyrrhizic Acid/chemistry , Patents as Topic , Drug Delivery Systems/methods , Drug Carriers/chemistry
20.
BMC Complement Med Ther ; 24(1): 58, 2024 Jan 27.
Article in English | MEDLINE | ID: mdl-38280993

ABSTRACT

Acute pancreatitis (AP) is a severe gastrointestinal inflammatory disease with increasing mortality and morbidity. Glycyrrhiza glabra, commonly known as Liquorice, is a widely used plant containing bioactive compounds like Glycyrrhizin, which possesses diverse medicinal properties such as anti-inflammatory, antioxidant, antiviral, and anticancer activities. The objective of this study is to investigate the active components, relevant targets, and underlying mechanisms of the traditional Chinese medicine Glycyrrhiza glabra in the treatment of AP. Utilizing various computational biology methods, we explored the potential targets and molecular mechanisms through Glycyrrhizin supplementation. Computational results indicated that Glycyrrhizin shows promising pharmacological potential, particularly with mitogen-activated protein kinase 3 (MAPK3) protein (degree: 70), forming stable complexes with Glycyrrhizin through ionic and hydrogen bonding interactions, with a binding free energy (ΔGbind) of -33.01 ± 0.08 kcal/mol. Through in vitro experiments, we validated that Glycyrrhizin improves primary pancreatic acinar cell injury by inhibiting the MAPK/STAT3/AKT signaling pathway. Overall, MAPK3 emerges as a reliable target for Glycyrrhizin's therapeutic effects in AP treatment. This study provides novel insights into the active components and potential targets and molecular mechanisms of natural products.


Subject(s)
Glycyrrhiza , Pancreatitis , Glycyrrhizic Acid/pharmacology , Glycyrrhizic Acid/chemistry , Glycyrrhizic Acid/metabolism , Network Pharmacology , Acute Disease , Pancreatitis/drug therapy , Signal Transduction , Glycyrrhiza/chemistry , Glycyrrhiza/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL