Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.822
Filter
1.
Int J Mol Sci ; 25(14)2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39063101

ABSTRACT

Astaxanthin (ATX) is a carotenoid nutraceutical with poor bioavailability due to its high lipophilicity. We tested a new tailored nanodroplet capable of solubilizing ATX in an oil-in-water micro-environment (LDS-ATX) for its capacity to improve the ATX pharmacokinetic profile and therapeutic efficacy. We used liquid chromatography tandem mass spectrometry (LC-MS/MS) to profile the pharmacokinetics of ATX and LDS-ATX, superoxide mutase (SOD) activity to determine their antioxidant capacity, protein carbonylation and lipid peroxidation to compare their basal and lipopolysaccharide (LPS)-induced oxidative damage, and ELISA-based detection of IL-2 and IFN-γ to determine their anti-inflammatory capacity. ATX and LDS-ATX corrected only LPS-induced SOD inhibition and oxidative damage. SOD activity was restored only by LDS-ATX in the liver and brain and by both ATX and LDS-ATX in muscle. While in the liver and muscle, LDS-ATX attenuated oxidative damage to proteins and lipids better than ATX; only oxidative damage to lipids was preferably corrected by LDS-ATX in the brain. IL-2 and IFN-γ pro-inflammatory response was corrected by LDS-ATX and not ATX in the liver and brain, but in muscle, the IL-2 response was not corrected and the IFN-γ response was mitigated by both. These results strongly suggest an organ-dependent improvement of ATX bioavailability and efficacy by the LDS-ATX nanoformulation.


Subject(s)
Anti-Inflammatory Agents , Antioxidants , Xanthophylls , Xanthophylls/pharmacokinetics , Xanthophylls/pharmacology , Xanthophylls/chemistry , Xanthophylls/administration & dosage , Antioxidants/pharmacokinetics , Antioxidants/pharmacology , Antioxidants/chemistry , Anti-Inflammatory Agents/pharmacokinetics , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Animals , Mice , Oxidative Stress/drug effects , Male , Nanoparticles/chemistry , Tandem Mass Spectrometry/methods , Liver/metabolism , Liver/drug effects , Drug Carriers/chemistry , Lipid Peroxidation/drug effects , Superoxide Dismutase/metabolism , Lipopolysaccharides , Interferon-gamma/metabolism
2.
Pharm Res ; 41(6): 1183-1199, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38849712

ABSTRACT

AIM: This study aimed to fabricate dexamethasone sodium phosphate loaded microneedle arrays (MNA) and investigate their efficiency in combination with iontophoresis for the treatment of hind paw oedema in rats. METHODS: Drug loaded polyvinyl alcohol, polyvinyl pyrrolidone and D-sorbitol-based MNA11 were fabricated by vacuum micromolding. Physicochemical, morphological, thermal, in-silico, in-vitro insertion ability (on parafilm) and drug release studies were performed. Ex-vivo permeation, in-vivo insertion and anti-inflammatory studies were performed in combination with iontophoresis. RESULTS: MNA11 displayed sharp-tipped projections and acceptable physicochemical features. Differential scanning calorimetry results indicated that drug loaded MNA11 were amorphous solids. Drug interacted with PVP and PVA predominately via hydrogen bonding. Parafilm displayed conspicuously engraved complementary structure of MNA11. Within 60 min, 91.50 ± 3.1% drug released from MNA11. A significantly higher i.e., 95.06 ± 2.5% permeation of drug was observed rapidly (within 60 min) from MNA11-iontophoresis combination than MNA11 i.e., 84.07 ± 3.5% within 240 min. Rat skin treated using MNA11 and MNA11-iontophoresis showed disruptions / microchannels in the epidermis without any damage to underlying anatomical structures. MNA11-iontophoresis combination led to significant reduction (83.02 ± 3.9%) in paw oedema as compared to MNA11 alone (72.55 ± 4.1%). CONCLUSION: MNA11-iontophoresis combination can act as a promising candidate to deliver drugs transcutaneously for treating inflammatory diseases.


Subject(s)
Administration, Cutaneous , Anti-Inflammatory Agents , Dexamethasone , Drug Delivery Systems , Edema , Iontophoresis , Needles , Skin Absorption , Skin , Animals , Iontophoresis/methods , Dexamethasone/administration & dosage , Dexamethasone/pharmacokinetics , Dexamethasone/analogs & derivatives , Rats , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacokinetics , Edema/drug therapy , Drug Delivery Systems/methods , Skin/metabolism , Skin/drug effects , Male , Drug Liberation , Inflammation/drug therapy , Rats, Sprague-Dawley
3.
Pharm Res ; 41(7): 1493-1505, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38918308

ABSTRACT

PURPOSE: Joint destruction is a major burden and an unsolved problem in rheumatoid arthritis (RA) patients. We designed an intra-articular mesoporous silica nanosystem (MSN-TP@PDA-GlcN) with anti-inflammatory and joint protection effects. The nanosystem was synthesized by encapsulating triptolide (TP) in mesoporous silica nanoparticles and coating it with pH-sensitive polydopamine (PDA) and glucosamine (GlcN) grafting on the PDA. The nano-drug delivery system with anti-inflammatory and joint protection effects should have good potency against RA. METHODS: A template method was used to synthesize mesoporous silica (MSN). MSN-TP@PDA-GlcN was synthesized via MSN loading with TP, coating with PDA and grafting of GlcN on PDA. The drug release behavior was tested. A cellular inflammatory model and a rat RA model were used to evaluate the effects on RA. In vivo imaging and microdialysis (MD) system were used to analyze the sustained release and pharmacokinetics in RA rats. RESULTS: TMSN-TP@PDA-GlcN was stable, had good biocompatibility, and exhibited sustained release of drugs in acidic environments. It had excellent anti-inflammatory effects in vitro and in vivo. It also effectively repaired joint destruction in vivo without causing any tissue toxicity. In vivo imaging and pharmacokinetics experiments showed that the nanosystem prolonged the residence time, lowered the Cmax value and enhanced the relative bioavailability of TP. CONCLUSIONS: These results demonstrated that MSN-TP@PDA-GlcN sustained the release of drugs in inflammatory joints and produced effective anti-inflammatory and joint protection effects on RA. This study provides a new strategy for the treatment of RA.


Subject(s)
Anti-Inflammatory Agents , Arthritis, Rheumatoid , Diterpenes , Drug Liberation , Indoles , Nanoparticles , Phenanthrenes , Polymers , Silicon Dioxide , Animals , Silicon Dioxide/chemistry , Arthritis, Rheumatoid/drug therapy , Nanoparticles/chemistry , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacokinetics , Phenanthrenes/chemistry , Phenanthrenes/administration & dosage , Phenanthrenes/pharmacokinetics , Phenanthrenes/pharmacology , Rats , Diterpenes/administration & dosage , Diterpenes/chemistry , Diterpenes/pharmacokinetics , Diterpenes/pharmacology , Indoles/administration & dosage , Indoles/chemistry , Indoles/pharmacokinetics , Indoles/pharmacology , Polymers/chemistry , Porosity , Male , Epoxy Compounds/chemistry , Epoxy Compounds/administration & dosage , Glucosamine/chemistry , Glucosamine/administration & dosage , Rats, Sprague-Dawley , Drug Carriers/chemistry , Humans , Mice , Delayed-Action Preparations , Inflammation/drug therapy , Inflammation/prevention & control
4.
J Med Chem ; 67(13): 10946-10966, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38913497

ABSTRACT

Thrombo-inflammation is closely associated with a few severe cardiovascular and infectious diseases. Factor XIIa (FXIIa) in the intrinsic coagulation pathway plays a pivotal role in the development of thrombo-inflammation and its inhibition has emerged as a potential therapeutic approach for thrombo-inflammatory disorders. Nonetheless, as of now, few small-molecule FXIIa inhibitors have demonstrated notable effectiveness against thrombo-inflammation, with none progressing into clinical stages. Herein, we present potent, covalent, reversible, and selective small-molecule FXIIa inhibitors such as 4a and 4j obtained through structure-based drug design. Compounds 4a and 4j showed significant anticoagulation and substantial anti-inflammatory effects in vitro, coupled with exceptional plasma stability. Furthermore, in carrageenan-induced thrombosis models, 4a and 4j demonstrated remarkable dual antithrombotic and anti-inflammatory activity when administered orally. Compound 4j exhibited a favorable safety profile without obvious tissue toxicity in mice, suggesting its potential as an oral therapeutic option for thrombo-inflammation.


Subject(s)
Factor XIIa , Thrombosis , Animals , Thrombosis/drug therapy , Mice , Humans , Factor XIIa/antagonists & inhibitors , Factor XIIa/metabolism , Administration, Oral , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacokinetics , Structure-Activity Relationship , Carrageenan , Drug Discovery , Inflammation/drug therapy , Male , Anticoagulants/pharmacology , Anticoagulants/therapeutic use , Anticoagulants/chemistry , Fibrinolytic Agents/pharmacology , Fibrinolytic Agents/therapeutic use , Fibrinolytic Agents/chemistry , Biological Availability
5.
AAPS PharmSciTech ; 25(6): 145, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38918292

ABSTRACT

The objective of the current research was to develop abietic acid (AA)-loaded hybrid polymeric nanoparticles (HNPs) for anti-inflammatory and antioxidant activity after oral administration. AAHNPs were developed by microinjection technique and optimized by 3-factor 3-level Box-Behnken design. The AAHNPs were evaluated for morphology, FTIR, X-ray diffraction, in-vitro release, ex-vivo permeation, in-vitro antioxidant, and in-vivo anti-inflammatory activity. The optimized AAHNPs (AAHNPsopt) displayed 384.5 ± 6.36nm of PS, 0.376 of PDI, 23.0 mV of ZP, and 80.01 ± 1.89% of EE. FTIR and X-ray diffraction study results revealed that AA was encapsulated into a HNPs matrix. The AAHNPsopt showed significant (P < 0.05) high and sustained release of AA (86.72 ± 4.92%) than pure AA (29.87 ± 3.11%) in 24h. AAHNPsopt showed an initial fast release of AA (20.12 ± 3.07% in 2h), which succeeded in reaching the therapeutic concentration. The AAHNPsopt showed 2.49-fold higher ex-vivo gut permeation flux than pure AA due to the presence of lipid and surfactant. The AAHNPsopt exhibited significantly (P < 0.05, P < 0.01, P < 0.001) higher antioxidant activity as compared to pure AA at each concentration. AAHNPsopt formulation displayed a significantly (P < 0.05) higher anti-inflammatory effect (21.51 ± 2.23% swelling) as compared to pure AA (46.51 ± 1.74% swelling). From the in-vitro and in-vivo finding, it was concluded that HNPs might be a suitable carrier for the improvement of the therapeutic efficacy of the drug.


Subject(s)
Abietanes , Anti-Inflammatory Agents , Antioxidants , Drug Carriers , Lipids , Nanoparticles , Polymers , Nanoparticles/chemistry , Animals , Antioxidants/pharmacology , Antioxidants/chemistry , Antioxidants/administration & dosage , Antioxidants/pharmacokinetics , Rats , Polymers/chemistry , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacokinetics , Lipids/chemistry , Drug Carriers/chemistry , Abietanes/pharmacology , Abietanes/administration & dosage , Abietanes/chemistry , X-Ray Diffraction/methods , Drug Liberation , Administration, Oral , Male , Particle Size , Rats, Wistar , Chemistry, Pharmaceutical/methods
6.
Int J Pharm ; 658: 124222, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38735632

ABSTRACT

Dry eye disease (DED) is a chronic multifactorial disorder of the ocular surface caused by tear film dysfunction and constitutes one of the most common ocular conditions worldwide. However, its treatment remains unsatisfactory. While artificial tears are commonly used to moisturize the ocular surface, they do not address the underlying causes of DED. Apigenin (APG) is a natural product with anti-inflammatory properties, but its low solubility and bioavailability limit its efficacy. Therefore, a novel formulation of APG loaded into biodegradable and biocompatible nanoparticles (APG-NLC) was developed to overcome the restricted APG stability, improve its therapeutic efficacy, and prolong its retention time on the ocular surface by extending its release. APG-NLC optimization, characterization, biopharmaceutical properties and therapeutic efficacy were evaluated. The optimized APG-NLC exhibited an average particle size below 200 nm, a positive surface charge, and an encapsulation efficiency over 99 %. APG-NLC exhibited sustained release of APG, and stability studies demonstrated that the formulation retained its integrity for over 25 months. In vitro and in vivo ocular tolerance studies indicated that APG-NLC did not cause any irritation, rendering them suitable for ocular topical administration. Furthermore, APG-NLC showed non-toxicity in an epithelial corneal cell line and exhibited fast cell internalization. Therapeutic benefits were demonstrated using an in vivo model of DED, where APG-NLC effectively reversed DED by reducing ocular surface cellular damage and increasing tear volume. Anti-inflammatory assays in vivo also showcased its potential to treat and prevent ocular inflammation, particularly relevant in DED patients. Hence, APG-NLC represent a promising system for the treatment and prevention of DED and its associated inflammation.


Subject(s)
Apigenin , Drug Carriers , Dry Eye Syndromes , Lipids , Nanoparticles , Animals , Apigenin/administration & dosage , Apigenin/chemistry , Apigenin/pharmacology , Apigenin/pharmacokinetics , Drug Carriers/chemistry , Dry Eye Syndromes/drug therapy , Humans , Rabbits , Lipids/chemistry , Lipids/administration & dosage , Cell Line , Nanoparticles/chemistry , Administration, Ophthalmic , Drug Liberation , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/pharmacokinetics , Particle Size , Nanostructures/administration & dosage , Nanostructures/chemistry , Male
7.
Int J Pharm ; 659: 124250, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38777304

ABSTRACT

The smart oral administration Insulin device has the potential to improve glycemic management. It can reduce the risk of hypoglycemia associated with exogenous Insulin (INS) therapy while also avoiding many of the disadvantages associated with subcutaneous injections. Furthermore, diabetes mellitus (DM) is an endocrine illness characterized by inflammation, and it is critical to minimize the amount of inflammatory markers in diabetic patients while maintaining average blood glucose. In this study, a responsive nanosystem vitamin B12-Fucoidan-Concanavalin A (VB12-FU-ConA NPs) with anti-inflammatory action was developed for smart oral delivery of Insulin. Con A has high sensitivity and strong specificity as a glucose-responsive material. Fucoidan has anti-inflammatory, immunomodulatory, and hypoglycemic functions, and it can bind to Con A to form a reversible complex. Under high glucose conditions, free glucose competitively binds to Con A, which swells the nanocarrier and promotes Insulin release. Furthermore, in the low pH environment of the gastrointestinal tract, positively charged VB12 and anionic fucoidan bind tightly to protect the Insulin wrapped in the carrier, and VB12 can also bind to intestinal epithelial factors to improve transit rate, thereby promoting INS absorption. In vitro tests showed that the release of nanoparticles in hyperglycemic solutions was significantly higher than the drug release in normoglycemic conditions. Oral delivery of the nanosystems dramatically lowered blood glucose levels in type I diabetic mice (T1DM) during in vivo pharmacodynamics, minimizing the risk of hypoglycemia. Blood glucose levels reached a minimum of 8.1 ± 0.4 mmol/L after 8 h. Administering the nanosystem orally notably decreased the serum levels of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) in diabetic mice. The nano delivery system can be degraded and metabolized in the intestinal tract after being taken orally, demonstrating good biodegradability and biosafety. In conclusion, the present study showed that VB12-FU-ConA nanocarriers are expected to be a novel system for rationalizing blood glucose.


Subject(s)
Anti-Inflammatory Agents , Blood Glucose , Diabetes Mellitus, Experimental , Hypoglycemic Agents , Insulin , Polysaccharides , Animals , Polysaccharides/administration & dosage , Polysaccharides/chemistry , Blood Glucose/drug effects , Blood Glucose/analysis , Administration, Oral , Insulin/administration & dosage , Insulin/pharmacokinetics , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/pharmacology , Mice , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/pharmacokinetics , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/blood , Male , Vitamin B 12/administration & dosage , Nanoparticles/administration & dosage , Drug Liberation , Drug Carriers/chemistry , Humans
8.
Int J Pharm ; 659: 124284, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38810934

ABSTRACT

The study aimed to create a low loading, high retention, easier to apply O/W mometasone furoate (MF) cream using a chemical enhancer (CE) approach to provide more options for patients with atopic dermatitis (AD) and to investigate molecular mechanisms of its increased release and retention. A Box-Behnken design determined the optimal formulation based on stability and in vitro skin retention. Evaluations included appearance, rheological properties, irritation, in vivo tissue distribution and pharmacodynamics. Molecular mechanisms of enhanced release were studied using high-speed centrifugation, molecular dynamics and rheology. The interaction between the CE, MF and skin was studied by tape stripping, CLSM, ATR-FTIR and SAXS. The formulation was optimized to contain 0.05% MF and used 10% polyglyceryl-3 oleate (POCC) as the CE. There was no significant difference from Elocon® cream in in vivo retention and pharmacodynamics but increased in vivo retention by 3.14-fold and in vitro release by 1.77-fold compared to the basic formulation. POCC reduced oil phase cohesive energy density, enhancing drug mobility and release. It disrupted skin lipid phases, aiding drug entry and formed hydrogen bonds, prolonging retention. This study highlights POCC as a CE in the cream, offering insights for semi-solid formulation development.


Subject(s)
Drug Liberation , Mometasone Furoate , Skin Cream , Skin , Mometasone Furoate/administration & dosage , Mometasone Furoate/pharmacokinetics , Mometasone Furoate/chemistry , Animals , Skin Cream/administration & dosage , Skin Cream/chemistry , Skin/metabolism , Skin/drug effects , Administration, Cutaneous , Male , Skin Absorption/drug effects , Chemistry, Pharmaceutical/methods , Glycerol/chemistry , Glycerol/analogs & derivatives , Dermatitis, Atopic/drug therapy , Female , Excipients/chemistry , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/pharmacokinetics , Drug Compounding/methods , Oleic Acid/chemistry , Polymers/chemistry
9.
Int J Nanomedicine ; 19: 4217-4234, 2024.
Article in English | MEDLINE | ID: mdl-38766660

ABSTRACT

Introduction: Rheumatoid arthritis (RA) is an inflammatory immune-mediated disease that involves synovitis, cartilage destruction, and even joint damage. Traditional agents used for RA therapy remain unsatisfactory because of their low efficiency and obvious adverse effects. Therefore, we here established RA microenvironment-responsive targeted micelles that can respond to the increase in reactive oxygen species (ROS) levels in the joint and improve macrophage-specific targeting of loaded drugs. Methods: We here prepared ROS-responsive folate-modified curcumin micelles (TK-FA-Cur-Ms) in which thioketal (TK) was used as a ROS-responsive linker for modifying polyethylene glycol 5000 (PEG5000) on the micellar surface. When micelles were in the ROS-overexpressing inflammatory microenvironment, the PEG5000 hydration layer was shed, and the targeting ligand FA was exposed, thereby enhancing cellular uptake by macrophages through active targeting. The targeting, ROS sensitivity and anti-inflammatory properties of the micelles were assessed in vitro. Collagen-induced arthritis (CIA) rats model was utilized to investigate the targeting, expression of serum inflammatory factors and histology change of the articular cartilage by micelles in vivo. Results: TK-FA-Cur-Ms had a particle size of 90.07 ± 3.44 nm, which decreased to 78.87 ± 2.41 nm after incubation with H2O2. The micelles exhibited in vitro targeting of RAW264.7 cells and significantly inhibited inflammatory cytokine levels. Pharmacodynamic studies have revealed that TK-FA-Cur-Ms prolonged the drug circulation and exhibited augmented cartilage-protective and anti-inflammatory effects in vivo. Conclusion: The unique ROS-responsive targeted micelles with targeting, ROS sensitivity and anti-inflammatory properties were successfully prepared and may offer an effective therapeutic strategy against RA.


Subject(s)
Arthritis, Rheumatoid , Curcumin , Folate Receptors, GPI-Anchored , Micelles , Reactive Oxygen Species , Animals , Male , Mice , Rats , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacokinetics , Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/drug therapy , Curcumin/administration & dosage , Curcumin/chemistry , Curcumin/pharmacology , Disease Models, Animal , Drug Carriers/chemistry , Folate Receptors, GPI-Anchored/metabolism , Folic Acid/chemistry , Folic Acid/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Particle Size , Polyethylene Glycols/chemistry , RAW 264.7 Cells , Reactive Oxygen Species/metabolism
10.
Phytother Res ; 38(7): 3660-3694, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38748620

ABSTRACT

Diosmetin, a natural occurring flavonoid, is primarily found in citrus fruits, beans, and other plants. Diosmetin demonstrates a variety of pharmacological activities, including anticancer, antioxidant, anti-inflammatory, antibacterial, metabolic regulation, cardiovascular function improvement, estrogenic effects, and others. The process of literature search was done using PubMed, Web of Science and ClinicalTrials databases with search terms containing Diosmetin, content, anticancer, anti-inflammatory, antioxidant, pharmacological activity, pharmacokinetics, in vivo, and in vitro. The aim of this review is to summarize the in vivo, in vitro and clinical studies of Diosmetin over the last decade, focusing on studies related to its anticancer, anti-inflammatory, and antioxidant activities. It is found that DIO has significant therapeutic effects on skin and cardiovascular system diseases, and its research in pharmacokinetics and toxicology is summarized. It provides the latest information for researchers and points out the limitations of current research and areas that should be strengthened in future research, so as to facilitate the relevant scientific research and clinical application of DIO.


Subject(s)
Antioxidants , Flavonoids , Humans , Flavonoids/pharmacology , Flavonoids/pharmacokinetics , Antioxidants/pharmacology , Antioxidants/pharmacokinetics , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/pharmacokinetics , Cardiovascular Diseases/drug therapy , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/pharmacokinetics
11.
Biomed Chromatogr ; 38(8): e5899, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38797863

ABSTRACT

Nanoliposomes (NLs) are ideal carriers for delivering complex molecules and phytochemical products, but ginger by-products, despite their therapeutic benefits, have poor bioavailability due to their low water solubility and stability. Crude ginger extracts (CGEs) and 6-gingerol were individually encapsulated within NLs for in vitro activity assessment. In vitro evaluation of anti-proliferative and anti-inflammatory properties of encapsulated 6-gingerol and CGE was performed on healthy human periodontal ligament (PDL) fibroblasts and MDA-MB-231 breast cancer cells. Encapsulation efficiency and loading capacity of 6-gingerol reached 25.23% and 2.5%, respectively. NLs were found stable for up to 30 days at 4°C with a gradual load loss of up to 20%. In vitro cytotoxic effect of encapsulated 6-gingerol exceeded 70% in the MDA-MB-231 cell line, in a comparable manner with non-encapsulated 6-gingerol and CGE. The effect of CGE with an IC50 of 3.11 ± 0.39, 7.14 ± 0.80, and 0.82 ± 0.55 µM and encapsulated 6-gingerol on inhibiting IL-8 was evident, indicating its potential anti-inflammatory activity. Encapsulating 6-gingerol within NLs enhanced its stability and facilitated its biological activity. All compounds, including vitamin C, were equivalent at concentrations below 2 mg/mL, with a slight difference in antioxidant activity. The concentrations capable of inhibiting 50% of 2,2-diphenyl-1-picrylhydrazyl (DPPH) substrate were comparable.


Subject(s)
Anti-Inflammatory Agents , Catechols , Fatty Alcohols , Liposomes , Zingiber officinale , Fatty Alcohols/chemistry , Fatty Alcohols/pharmacology , Humans , Catechols/chemistry , Catechols/pharmacology , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacokinetics , Liposomes/chemistry , Cell Line, Tumor , Zingiber officinale/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/pharmacokinetics , Cell Survival/drug effects , Nanoparticles/chemistry , Plant Extracts/chemistry , Plant Extracts/pharmacology , Interleukin-8/metabolism , Cell Proliferation/drug effects
12.
Am J Chin Med ; 52(3): 717-752, 2024.
Article in English | MEDLINE | ID: mdl-38716620

ABSTRACT

Schisandra chinensis (S. chinensis) has a long history as a traditional Chinese medicine that is astringent, beneficial to vital energy, tonifies the kidney, tranquilizes the heart, etc. Significantly, Schisandrol A (SA) is extracted from S. chinensis and shows surprising and satisfactory biological activity, including anti-inflammatory, hepatoprotective, cardiovascular protection, and antitumor properties, among others. SA has a more pronounced protective effect on central damaged nerves among its numerous pharmacological effects, improving neurodegenerative diseases such as Alzheimer's and Parkinson's through the protection of damaged nerve cells and the enhancement of anti-oxidant capacity. Pharmacokinetic studies have shown that SA has a pharmacokinetic profile with a rapid absorption, wide distribution, maximal concentration in the liver, and primarily renal excretion. However, hepatic and intestinal first-pass metabolism can affect SA's bioavailability. In addition, the content of SA, as an index component of S. chinensis Pharmacopoeia, should not be less than 0.40%, and the content of SA in S. chinensis compound formula was determined with the help of high-performance liquid chromatography (HPLC), which is a stable and reliable method, and it can lay a foundation for the subsequent quality control. Therefore, this paper systematically reviews the preparation, pharmacological effects, pharmacokinetic properties, and content determination of SA with the goal of updating and deepening the understanding of SA, as well as providing a theoretical basis for the study of SA at a later stage.


Subject(s)
Cyclooctanes , Lignans , Schisandra , Schisandra/chemistry , Lignans/pharmacokinetics , Cyclooctanes/pharmacokinetics , Humans , Anti-Inflammatory Agents/pharmacokinetics , Animals , Antioxidants/pharmacokinetics , Biological Availability
13.
J Med Chem ; 67(10): 8060-8076, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38722184

ABSTRACT

Interleukin-1 receptor-associated kinase 4 (IRAK4) is a promising therapeutic target in inflammation-related diseases. However, the inhibition of IRAK4 kinase activity may lead to moderate anti-inflammatory efficacy owing to the dual role of IRAK4 as an active kinase and a scaffolding protein. Herein, we report the design, synthesis, and biological evaluation of an efficient and selective IRAK4 proteolysis-targeting chimeric molecule that eliminates IRAK4 scaffolding functions. The most potent compound, LC-MI-3, effectively degraded cellular IRAK4, with a half-maximal degradation concentration of 47.3 nM. LC-MI-3 effectively inhibited the activation of downstream nuclear factor-κB signaling and exerted more potent pharmacological effects than traditional kinase inhibitors. Furthermore, LC-MI-3 exerted significant therapeutic effects in lipopolysaccharide- and Escherichia coli-induced acute and chronic inflammatory skin models compared with kinase inhibitors in vivo. Therefore, LC-MI-3 is a candidate IRAK4 degrader in alternative targeting strategies and advanced drug development.


Subject(s)
Interleukin-1 Receptor-Associated Kinases , Interleukin-1 Receptor-Associated Kinases/antagonists & inhibitors , Interleukin-1 Receptor-Associated Kinases/metabolism , Animals , Humans , Mice , Inflammation/drug therapy , Inflammation/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/therapeutic use , Administration, Oral , Lipopolysaccharides/pharmacology , NF-kappa B/metabolism , NF-kappa B/antagonists & inhibitors , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/pharmacokinetics , Biological Availability , Drug Discovery , Proteolysis/drug effects , Structure-Activity Relationship , Male , Mice, Inbred C57BL
14.
Inflammopharmacology ; 32(3): 1871-1886, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38564091

ABSTRACT

Snow mountain garlic (SMG) is a trans-Himalayan medicinal plant used in the traditional medicine system for several ailments, including inflammatory arthritis. Research studies are insufficient to validate its folk medicinal applications. In the present study, the comparative abundance of its key bioactive phytocompounds, viz., S-allyl-L-cysteine (SAC), alliin, and S-methyl-L-cysteine (SMC) against normal garlic were assessed using the LC-MS/MS-MRM method. In addition, the study also explored the antioxidant and anti-inflammatory potency of crude extract of SMG and purified signature phytocompounds (i.e., SMC, SAC, and alliin) in comparison with normal garlic and dexamethasone in LPS-stimulated RAW264.7 macrophage cells. The LC-MS/MS-MRM study revealed significant differences among SMG and normal garlic, viz., alliin 22.8-fold higher in SMG, and SMC could be detected only in SMG. In the bioassays, SMG extract and purified signature phytocompounds significantly downregulated oxidative damage in activated macrophages, boosting endogenous antioxidants' activity. SMG extract-treated macrophages significantly suppressed NF-κB expression and related inflammatory indicators such as cytokines, COX-2, iNOS, and NO. Notably, the observed anti-inflammatory and antioxidant bioactivities of SMG extract were comparable to signature phytocompounds and dexamethasone. In addition, SAC being uniformly found in SMG and normal garlic, its comparative pharmacokinetics was studied to validate the pharmacodynamic superiority of SMG over normal garlic. Significantly higher plasma concentrations (Cmax), half-life (t1/2), and area under curve (AUC) of SAC following SMG extract administration than normal garlic validated the proposed hypothesis. Thus, the abundance of bioactive phytocompounds and their better pharmacokinetics in SMG extract might be underlying its medicinal merits over normal garlic.


Subject(s)
Anti-Inflammatory Agents , Antioxidants , Garlic , Macrophages , Plant Extracts , Garlic/chemistry , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/pharmacokinetics , Mice , Antioxidants/pharmacology , Antioxidants/pharmacokinetics , RAW 264.7 Cells , Plant Extracts/pharmacology , Plant Extracts/pharmacokinetics , Macrophages/drug effects , Macrophages/metabolism , Tandem Mass Spectrometry/methods , Cysteine/pharmacology , Chromatography, Liquid/methods , Phytochemicals/pharmacology , Phytochemicals/pharmacokinetics , Oxidative Stress/drug effects , Male
15.
J Control Release ; 369: 746-764, 2024 May.
Article in English | MEDLINE | ID: mdl-38599547

ABSTRACT

Acute respiratory distress syndrome (ARDS) is a critical illness characterized by severe lung inflammation. Improving the delivery efficiency and achieving the controlled release of anti-inflammatory drugs at the lung inflammatory site are major challenges in ARDS therapy. Taking advantage of the increased pulmonary vascular permeability and a slightly acidic-inflammatory microenvironment, pH-responsive mineralized nanoparticles based on dexamethasone sodium phosphate (DSP) and Ca2+ were constructed. By further biomimetic modification with M2 macrophage membranes, hybrid mineralized nanovesicles (MM@LCaP) were designed to possess immunomodulatory ability from the membranes and preserve the pH-sensitivity from core nanoparticles for responsive drug release under acidic inflammatory conditions. Compared with healthy mice, the lung/liver accumulation of MM@LCaP in inflammatory mice was increased by around 5.5 times at 48 h after intravenous injection. MM@LCaP promoted the polarization of anti-inflammatory macrophages, calmed inflammatory cytokines, and exhibited a comprehensive therapeutic outcome. Moreover, MM@LCaP improved the safety profile of glucocorticoids. Taken together, the hybrid mineralized nanovesicles-based drug delivery strategy may offer promising ideas for enhancing the efficacy and reducing the toxicity of clinical drugs.


Subject(s)
Anti-Inflammatory Agents , Dexamethasone , Glucocorticoids , Lung , Nanoparticles , Respiratory Distress Syndrome , Animals , Glucocorticoids/administration & dosage , Glucocorticoids/pharmacokinetics , Glucocorticoids/therapeutic use , Dexamethasone/administration & dosage , Dexamethasone/pharmacokinetics , Dexamethasone/therapeutic use , Dexamethasone/analogs & derivatives , Tissue Distribution , Nanoparticles/chemistry , Mice , Respiratory Distress Syndrome/drug therapy , Lung/metabolism , Lung/drug effects , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacokinetics , Macrophages/drug effects , Macrophages/metabolism , Male , Drug Liberation , Pneumonia/drug therapy , Pneumonia/chemically induced , RAW 264.7 Cells , Drug Delivery Systems , Calcium/metabolism , Cytokines/metabolism
16.
Biomed Pharmacother ; 174: 116456, 2024 May.
Article in English | MEDLINE | ID: mdl-38552441

ABSTRACT

Acute lung injury (ALI) is a common and critical respiratory disorder caused by various factors, with viral infection being the leading contributor. Dehydroandrographolide (DAP), a constituent of the Chinese herbal plant Andrographis paniculata, exhibits a range of activities including anti-inflammatory, in vitro antiviral and immune-enhancing effects. This study evaluated the anti-inflammatory effects and pharmacokinetics (PK) profile of DAP in ALI mice induced by intratracheal instillation of Poly(I:C) (PIC). The results showed that oral administration of DAP (10-40 mg/kg) effectively suppressed the increase in lung wet-dry weight ratio, total cells, total protein content, accumulation of immune cells, inflammatory cytokines and neutrophil elastase levels in bronchoalveolar lavage fluid of PIC-treated mice. DAP concentrations, determined by an LC-MS/MS method, in plasma after receiving DAP (20 mg/kg) were unchanged compared to those in normal mice. However, DAP concentrations and relative PK parameters in the lungs were significantly altered in PIC-treated mice, exhibiting a relatively higher maximum concentration, larger AUC, and longer elimination half-life than those in the lungs of normal mice. These results demonstrated that DAP could improve lung edema and inflammation in ALI mice, and suggested that lung injury might influence the PK properties of DAP, leading to increased lung distribution and residence. Our study provides evidence that DAP displays significant anti-inflammatory activity against viral lung injury and is more likely to distribute to damaged lung tissue.


Subject(s)
Acute Lung Injury , Anti-Inflammatory Agents , Bronchoalveolar Lavage Fluid , Diterpenes , Poly I-C , Animals , Acute Lung Injury/drug therapy , Anti-Inflammatory Agents/pharmacokinetics , Anti-Inflammatory Agents/pharmacology , Diterpenes/pharmacokinetics , Diterpenes/pharmacology , Male , Mice , Andrographis/chemistry , Cytokines/metabolism , Lung/drug effects , Lung/metabolism , Lung/pathology , Leukocyte Elastase/metabolism
17.
Drug Deliv Transl Res ; 14(8): 2062-2078, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38517568

ABSTRACT

To improve the therapeutic activity of inhaled glucocorticoids and reduce potential side effects, we designed a formulation combining the advantages of nanoparticles, which have an enhanced uptake by alveolar cells, allow targeted delivery and sustained drug release, as well as limited drug systemic passage, with those of microparticles, which display good alveolar deposition. Herein, a polymer-drug conjugate, poly(malic acid)-budesonide (PMAB), was first synthesized with either 11, 20, 33, or 43 mol% budesonide (drug:polymer from 1:8 to 3:4), the drug creating hydrophobic domains. The obtained conjugates self-assemble into nanoconjugates in water, yielding excellent drug loading of up to 73 wt%, with 80-100 nm diameters. In vitro assays showed that budesonide could be steadily released from the nanoconjugates, and the anti-inflammatory activity was preserved, as evidenced by reduced cytokine production in LPS-activated RAW 264.7 macrophages. Nanoconjugates were then embedded into microparticles through spray-drying with L-leucine, forming nano-embedded microparticles (NEMs). NEMs were produced with an aerodynamic diameter close to 1 µm and a density below 0.1 g.cm-3, indicative of a high alveolar deposition. NEMs spray-dried with the less hydrophobic nanoconjugates, PMAB 1:4, were readily dissolved in simulated lung fluid and were chosen for in vivo experiments to study pharmacokinetics in healthy rats. As it was released in vivo from NEMs, sustained distribution of budesonide was obtained for 48 h in lung tissue, cells, and lining fluid. With high loading rates, modulable release kinetics, and low cytotoxicity, these nanoconjugates delivered by NEMs are promising for the more efficient treatment of pulmonary inflammatory diseases.


Subject(s)
Budesonide , Lung , Nanoconjugates , Animals , Mice , Budesonide/administration & dosage , Budesonide/pharmacokinetics , Budesonide/chemistry , RAW 264.7 Cells , Lung/metabolism , Nanoconjugates/chemistry , Nanoconjugates/administration & dosage , Male , Polymers/chemistry , Polymers/administration & dosage , Malates/chemistry , Malates/administration & dosage , Malates/pharmacokinetics , Administration, Inhalation , Particle Size , Rats , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacokinetics , Drug Liberation , Rats, Sprague-Dawley
18.
Drug Deliv Transl Res ; 14(7): 1820-1838, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38127247

ABSTRACT

Uric acid is the final metabolite in humans. High level of uric acid chronically induces urate deposition, aggravates kidney damage, and concomitantly causes an increase in inflammatory factors. Alleviating acute inflammation and decreasing uric acid levels are the key points in the treatment of inflammatory diseases associated with high uric acid. However, a drug delivery system that combines anti-inflammatory and uric acid reduction functions at the same time remains a challenge to be settled. Here, we designed a nanocrystal-based co-delivery platform, IND Nplex, characterized by loading of indomethacin (IND) and uricase. Compared with free IND or uricase, IND Nplex possessed a better anti-inflammatory effect by restraining the release of inflammation-related factors in vitro. In addition, pharmacokinetic and biodistribution studies revealed that IND Nplex significantly prolonged the retention time in vivo and was more concentrated in the kidney. In acute gouty arthritis model rats, IND Nplex markedly relieved ankle joint swelling and mitigated synovial inflammation. In acute kidney injury model rats, IND Nplex indicated better biocompatibility and significant amelioration of renal fibrosis. Moreover, IND Nplex showed the effect of anti-inflammatory and improved renal function via determination of inflammatory factors and biochemical markers in the serum and kidney. In conclusion, these results indicate that IND Nplex exerts anti-inflammatory activity and uric acid-lowering effect and could become a promising candidate for the treatment of uric acid-related diseases.


Subject(s)
Arthritis, Gouty , Indomethacin , Rats, Sprague-Dawley , Urate Oxidase , Uric Acid , Indomethacin/administration & dosage , Animals , Urate Oxidase/administration & dosage , Urate Oxidase/pharmacokinetics , Urate Oxidase/therapeutic use , Uric Acid/blood , Male , Arthritis, Gouty/drug therapy , Nanoparticles/administration & dosage , Rats , Mice , Inflammation/drug therapy , Tissue Distribution , Acute Kidney Injury/drug therapy , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacokinetics , Anti-Inflammatory Agents/pharmacology , Drug Delivery Systems , Kidney/drug effects , Kidney/metabolism , Humans , RAW 264.7 Cells , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/pharmacology
19.
Clin Pharmacokinet ; 62(9): 1189-1199, 2023 09.
Article in English | MEDLINE | ID: mdl-37598107

ABSTRACT

Triamcinolone acetonide is a glucocorticosteroid used in standard clinical practice for its anti-inflammatory properties. Although it can be given via different routes of administration, the intramuscular route is unique compared with other corticosteroids-its effects remain potent over a longer period of time. We summarize the existing literature on the pharmacokinetic and pharmacodynamic mechanisms of intramuscular triamcinolone acetonide (Kenalog®). The fascinating nature of the purported efficacy of triamcinolone acetonide may be attributed to differing binding mechanisms, low solubility in blood, a low renal clearance rate, and various metabolites and other yet defined effects on skin. The enigma of the purported efficacy of triamcinolone acetonide may lie in the fact that it has a unique nature of having a long-term effect on dermatologic disease using a seemingly low dose compared with other routes of administration and other corticosteroids. Possible reasons for this may be binding differences at the intramuscular site, low solubility due to acetonide esters, a slow rate of absorption from the injected site, and a low renal clearance rate. There is still much to be learned about its mechanism of action, which may be of clinical and therapeutic significance.


Subject(s)
Anti-Inflammatory Agents , Glucocorticoids , Triamcinolone Acetonide , Triamcinolone Acetonide/administration & dosage , Triamcinolone Acetonide/pharmacokinetics , Triamcinolone Acetonide/pharmacology , Injections, Intramuscular , Humans , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacokinetics , Anti-Inflammatory Agents/pharmacology , Glucocorticoids/administration & dosage , Glucocorticoids/pharmacokinetics , Glucocorticoids/pharmacology , Glomerular Filtration Rate
20.
Clin Pharmacol Drug Dev ; 12(11): 1069-1075, 2023 11.
Article in English | MEDLINE | ID: mdl-37246720

ABSTRACT

Celecoxib is a sulfanilamide nonsteroidal anti-inflammatory drug that can selectively inhibit cyclooxygenase-2 to inhibit prostaglandin production, achieving anti-inflammatory and analgesic effects. This study investigated the pharmacokinetics, safety, and bioequivalence of a single oral dose of celecoxib capsule (the test or reference preparation) in healthy volunteers under fasting and fed conditions. A single-center, randomized, open, single-dose, double-cycle crossover self-control design was conducted: 40 healthy volunteers were enrolled in the fasting and fed groups, respectively. A completely randomized method was used, with one group taking the test celecoxib preparation (T) and the other taking the reference celecoxib preparation (R). During the administration period, the safety of the drug was evaluated simultaneously, and venous blood was collected at the corresponding time points. The concentration of celecoxib in plasma was measured by liquid chromatography-tandem mass spectrometry. The main pharmacokinetic parameters were logarithmically converted and analyzed for variance. The 90% confidence interval for the bioavailability of the T compared to the R was calculated using maximum drug plasma concentration, area under the plasma concentration-time curve from time zero to the last quantifiable concentration point, and area under the plasma concentration-time curve from time zero to infinity for a single oral dose in volunteers, and the data obtained were all between 80% and 125%, indicating that the T and R have bioequivalence and good safety during fasting and fed administration.


Subject(s)
Anti-Inflammatory Agents , Celecoxib , East Asian People , Humans , Anti-Inflammatory Agents/pharmacokinetics , Celecoxib/pharmacokinetics , Healthy Volunteers , Therapeutic Equivalency
SELECTION OF CITATIONS
SEARCH DETAIL