Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 66
1.
J Drug Target ; : 1-19, 2024 Apr 29.
Article En | MEDLINE | ID: mdl-38634290

Nanoparticle (NP) functionalization with specific ligands enhances targeted cancer therapy and imaging by promoting receptor recognition and improving cellular uptake. This review focuses on recent research exploring the interaction between cancer cell-expressed chemokine receptor 4 (CXCR4) and ligand-conjugated NPs, utilising small molecules, peptides, and antibodies. Active NP targeting has shown improved tumour targeting and reduced toxicity, enabling precision therapy and diagnosis. However, challenges persist in the clinical translation of targeted NPs due to issues with biological response, tumour accumulation, and maintaining NP quality at an industrial scale. Biological and intratumoral barriers further hinder efficient NP accumulation in tumours, hampering translatability. To address these challenges, the academic community is refocusing efforts on understanding NP biological fate and establishing robust preclinical models. Future studies should investigate NP-body interactions, develop computational models, and identify optimal preclinical models. Establishing central NP research databases and fostering collaboration across disciplines is crucial to expediting clinical translation. Overcoming these hurdles will unlock the transformative potential of CXCR4-ligand-NP conjugates in revolutionising cancer treatment.

2.
ACS Pharmacol Transl Sci ; 7(1): 28-41, 2024 Jan 12.
Article En | MEDLINE | ID: mdl-38230275

Alpha7 nicotinic acetylcholine receptor (α7 nAChR) is an ion-gated calcium channel that plays a significant role in various aspects of cancer pathogenesis, particularly in lung cancer. Preclinical studies have elucidated the molecular mechanism underlying α7 nAChR-associated lung cancer proliferation, chemotherapy resistance, and metastasis. Understanding and targeting this mechanism are crucial for developing therapeutic interventions aimed at disrupting α7 nAChR-mediated cancer progression and improving treatment outcomes. Drug research and discovery have determined natural compounds and synthesized chemical antagonists that specifically target α7 nAChR. However, approved α7 nAChR antagonists for clinical use are lacking, primarily due to challenges related to achieving the desired selectivity, efficacy, and safety profiles required for effective therapeutic intervention. This comprehensive review provided insights into the molecular mechanisms associated with α7 nAChR and its role in cancer progression, particularly in lung cancer. Furthermore, it presents an update on recent evidence about α7 nAChR antagonists and addresses the challenges encountered in drug research and discovery in this field.

3.
ACS Appl Bio Mater ; 6(12): 5426-5441, 2023 Dec 18.
Article En | MEDLINE | ID: mdl-37956113

In this study, we synthesized hollow porous iron oxide nanoparticles (HPIONPs) with surface modifications using polymers, specifically chitosan (Chi), polyethylene glycol (PEG), and alginate (Alg), to improve colloidal stability and biocompatibility. For colloidal stability, Alg-coated HPIONPs maintained size stability up to 24 h, with only an 18% increase, while Chi, PEG, and uncoated HPIONPs showed larger size increases ranging from 64 to 140%. The biocompatibility of polymer-coated HPIONPs was evaluated by assessing their cell viability, genotoxicity, and hemocompatibility. Across tested concentrations from 6.25 to 100 µg/mL, both uncoated and polymer-coated HPIONPs showed minimal cytotoxicity against three normal cell lines: RAW264.7, 3T3-L1, and MCF10A, with cell viability exceeding 80% at the highest concentration. Notably, polymer-coated HPIONPs exhibited nongenotoxicity based on the micronucleus assay and showed hemocompatibility, with only 2-3% hemolysis in mouse blood, in contrast to uncoated HPIONPs which exhibited 4-5%. Furthermore, we evaluated the cytotoxicity of HPIONPs on MDA-MB-231 breast cancer cells after a 2 h exposure to a stationary magnetic field, and the results showed the highest cell death of 38 and 29% when treated with uncoated and polymer-coated HPIONPs at 100 µg/mL, respectively. This phenomenon is attributed to iron catalyzing the Fenton and Haber-Weiss reactions, leading to reactive oxygen species (ROS)-dependent cell death (r ≥ 0.98). In conclusion, the hydrothermal synthesis and subsequent surface modification of HPIONPs with polymers showed improved colloidal stability, nongenotoxicity, and hemocompatibility compared to uncoated HPIONPs while maintaining closely similar levels of cytotoxicity against both normal and cancer cells. This research has paved the way for further exploration of polymer coatings to enhance the overall performance and safety profile of magnetic nanoparticles in delivering anticancer drugs.


Antineoplastic Agents , Chitosan , Mice , Animals , Polymers/chemistry , Porosity , Polyethylene Glycols/chemistry , Chitosan/chemistry , Magnetic Iron Oxide Nanoparticles
4.
MethodsX ; 11: 102318, 2023 Dec.
Article En | MEDLINE | ID: mdl-37608960

The current literature mostly contains relatively vague descriptions of techniques for implementing in vitro magnetic targeting delivery of iron oxide nanoparticles (IONPs), leading to irreproducible processes and incomparable findings. This discrepancy often arises from the varying exposure of IONPs to the non-uniform magnetic field and differences in the concentration of the polymer-coated IONPs. Hence, we meticulously designed and built a system comprising a platform constructed from polyoxymethylene sheets, which securely holds the permanent magnets, and the cell culture plate. We also tailored the preparation process of the IONPs and the in vitro toxicity studies. The inherent characteristics of IONPs are further enhanced by their coating with natural polymers, alginate (Alg) and chitosan (CS).•The design and construction of the platform were carried out using a laser engraving/cutting machine along with graphic design software. The precise locations of the permanent magnets relative to the cell culture plate were determined via a Gaussmeter.•The quantities of the components in the formulation and the method for fabricating the CS/Alg-coated IONPs (CS/Alg-IONPs) were optimized to ensure that the desired physicochemical properties were obtained.•The cultivation and cytotoxicity evaluation of the fabricated CS/Alg-IONPs against MCF-7 breast cancer cells were described.

5.
Int J Pharm ; 640: 123037, 2023 Jun 10.
Article En | MEDLINE | ID: mdl-37172632

Nanotechnology plays an integral role in multimodal analgesia. In this study, we co-encapsulated metformin (Met) and curcumin (Cur) into chitosan/alginate (CTS/ALG) nanoparticles (NPs) at their synergistic drug ratio by applying response surface methodology. The optimized Met-Cur-CTS/ALG-NPs were achieved with Pluronic® F-127 2.33 % (w/v), Met 5.91 mg, and CTS:ALG mass ratio 0.05:1. The prepared Met-Cur-CTS/ALG-NPs had 243 nm particle size, -21.6 mV zeta potential, 32.6 and 44.2 % Met and Cur encapsulations, 19.6 and 6.8 % Met and Cur loading, respectively, and 2.9:1 Met:Cur mass ratio. Met-Cur-CTS/ALG-NPs displayed stability under simulated gastrointestinal (GI) fluid conditions and during storage. In vitro release study of Met-Cur-CTS/ALG-NPs in simulated GI fluids showed sustained release, with Met exhibiting Fickian diffusion and Cur demonstrating non-Fickian diffusion following the Korsmeyer-Peppas model. Met-Cur-CTS/ALG-NPs exhibited increased mucoadhesion and improved cellular uptake in Caco-2 cells. Additionally, Met-Cur-CTS/ALG-NPs exhibited better anti-inflammatory effects in lipopolysaccharide-stimulated RAW 264.7 macrophage and BV-2 microglial cells than the equivalent amount of the Met-Cur physical mixture, indicating a greater ability to modulate peripheral and central immune mechanisms of pain. In the mouse formalin-induced pain model, Met-Cur-CTS/ALG-NPs administered orally exhibited better attenuation of pain-like behaviors and proinflammatory cytokine release compared to the Met-Cur physical mixture. Furthermore, Met-Cur-CTS/ALG-NPs did not induce significant side effects in mice at therapeutic doses. Altogether, the present study establishes a CTS/ALG nano-delivery system for Met-Cur combination against pain with improved efficacy and safety.


Chitosan , Curcumin , Metformin , Nanoparticles , Humans , Mice , Animals , Drug Carriers , Curcumin/pharmacology , Chitosan/pharmacology , Caco-2 Cells , Alginates/pharmacology , Particle Size
6.
Int J Biol Macromol ; 242(Pt 1): 124673, 2023 Jul 01.
Article En | MEDLINE | ID: mdl-37137353

Magnetic drug targeting can be a strategy for effectively delivering phytochemicals in cancer treatment. Here, we demonstrate the benefit of magnetic targeting with superparamagnetic iron oxide nanoparticles for cytotoxicity enhancement of lutein (LUT) against breast cancer cells. Fabrication of LUT-loaded chitosan/alginate iron oxide nanoparticles (LUT-CS/Alg-Fe3O4-NPs) was optimized by a statistical approach using response surface methodology based on the Box-Behnken design. The optimized LUT-CS/Alg-Fe3O4-NPs with a balance among LUT concentration, copolymer coating, and iron ion concentration exhibited controlled size, narrow size distribution, better crystallinity, excellent saturation magnetization, and sustained-release profile. The negligible magnetic coercivity and remanent magnetization confirmed the superparamagnetism of the prepared NPs. The optimized LUT-CS/Alg-Fe3O4-NPs were biocompatible while exhibiting a significantly enhanced cytotoxicity towards breast cancer MCF-7 cells upon exposure to a permanent magnet compared to free LUT with a 4-fold increase, suggesting the potential of LUT-CS/Alg-Fe3O4-NPs as magnetically targeted delivery for breast cancer.


Breast Neoplasms , Chitosan , Nanoparticles , Humans , Female , Breast Neoplasms/drug therapy , Alginates , Lutein
7.
Int J Mol Sci ; 24(9)2023 Apr 29.
Article En | MEDLINE | ID: mdl-37175777

Bacterial meningitis remains one of the most prevalent infectious diseases worldwide. Although advances in medical care have improved mortality and morbidity, neurological complications remain high. Therefore, aside from antibiotics, therapeutic adjuvants targeting neuroinflammation are essential to combat the long-term neuronal sequelae of bacterial meningitis. In the present study, we propose (-)-dendroparishiol as a potential add-on therapy to improve neuroinflammation associated with bacterial meningitis. The biological activity of (-)-dendroparishiol was first predicted by computational analysis and further confirmed in vitro using a cell-based assay with LPS-induced BV-2 microglial cells. Biological pathways involved with (-)-dendroparishiol were identified by applying network pharmacology. Computational predictions of biological activity indicated possible attenuation of several inflammatory processes by (-)-dendroparishiol. In LPS-induced BV-2 microglial cells, (-)-dendroparishiol significantly reduced the expression of inflammatory mediators: iNOS, NO, COX-2, IL-6, and TNF-α. Molecular docking results demonstrated the potential iNOS and COX-2 inhibitory activity of (-)-dendroparishiol. Network pharmacological analysis indicated the plausible role of (-)-dendroparishiol in biological processes involved in oxidative stress and neuroinflammation with enrichment in neuroinflammatory pathways. Overall, this study provides scientific evidence for the potential application of (-)-dendroparishiol in the management of bacterial meningitis-associated neuroinflammation.


Inflammation , Meningitis, Bacterial , Humans , Inflammation/metabolism , Neuroinflammatory Diseases , Lipopolysaccharides/adverse effects , Molecular Docking Simulation , Network Pharmacology , Microglia/metabolism , Meningitis, Bacterial/metabolism , NF-kappa B/metabolism
8.
Molecules ; 28(4)2023 Feb 09.
Article En | MEDLINE | ID: mdl-36838664

A simple and reliable ultra-high-performance liquid chromatographic (UHPLC) method was developed and validated for determination of tetrahydrocurcumin diglutaric acid (TDG) and applied for evaluation of its bioaccessibility. The analytical method was validated to demonstrate as a stability-indicating assay (SIA) according to the ICH Q2(R1) guidelines under various force degradation conditions including thermal degradation, moisture, acid and base hydrolysis, oxidation, and photolysis. The developed chromatographic condition could completely separate all degradants from the analyte of interest. The method linearity was verified in the range of 0.4-12 µg/mL with the coefficient of determination (r2) > 0.995. The accuracy and precision of the method provided %recovery in the range of 98.9-104.2% and %RSD lower than 4.97%, respectively. The limit of detection and quantitation were found to be 0.25 µg/mL and 0.40 µg/mL, respectively. This method has been successfully applied for the bioaccessibility assessment of TDG with the bioaccessibility of TDG approximately four fold greater than THC in simulated gastrointestinal fluid. The validated SIA method can also benefit the quality control of TDG raw materials in pharmaceutical and nutraceutical development.


Curcumin , Chromatography, High Pressure Liquid/methods , Limit of Detection , Drug Stability , Reproducibility of Results
9.
Int J Mol Sci ; 24(4)2023 Feb 10.
Article En | MEDLINE | ID: mdl-36835028

α3ß4 Nicotinic acetylcholine receptor (nAChR) has been recognized as an emerging biomarker for the early detection of drug addiction. Herein, α3ß4 nAChR ligands were designed and synthesized to improve the binding affinity and selectivity of two lead compounds, (S)-QND8 and (S)-T2, for the development of an α3ß4 nAChR tracer. The structural modification was achieved by retaining the key features and expanding the molecular structure with a benzyloxy group to increase the lipophilicity for blood-brain barrier penetration and to extend the ligand-receptor interaction. The preserved key features are a fluorine atom for radiotracer development and a p-hydroxyl motif for ligand-receptor binding affinity. Four (R)- and (S)-quinuclidine-triazole (AK1-AK4) were synthesized and the binding affinity, together with selectivity to α3ß4 nAChR subtype, were determined by competitive radioligand binding assay using [3H]epibatidine as a radioligand. Among all modified compounds, AK3 showed the highest binding affinity and selectivity to α3ß4 nAChR with a Ki value of 3.18 nM, comparable to (S)-QND8 and (S)-T2 and 3069-fold higher affinity to α3ß4 nAChR in comparison to α7 nAChR. The α3ß4 nAChR selectivity of AK3 was considerably higher than those of (S)-QND8 (11.8-fold) and (S)-T2 (294-fold). AK3 was shown to be a promising α3ß4 nAChR tracer for further development as a radiotracer for drug addiction.


Receptors, Nicotinic , Substance-Related Disorders , alpha7 Nicotinic Acetylcholine Receptor , Humans , alpha7 Nicotinic Acetylcholine Receptor/chemistry , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Ligands , Radioligand Assay , Receptors, Nicotinic/metabolism , Substance-Related Disorders/diagnostic imaging , Quinuclidines/chemistry , Quinuclidines/pharmacology , Triazoles/chemistry , Triazoles/pharmacology
10.
Pharmaceutics ; 14(12)2022 Dec 01.
Article En | MEDLINE | ID: mdl-36559173

Favipiravir (FVR) is a repurposed antiviral drug for treating mild to moderate cases of the novel coronavirus disease 2019 (COVID-19). However, its poor solubility and permeability limit its clinical efficacy. To overcome its physicochemical and pharmacokinetic limitations, we statistically designed a mucoadhesive chitosan-alginate nanoparticles (MCS-ALG-NPs) as a new carrier for FVR using response surface methodology, which provided suitable characteristics for transmucosal delivery. The use of mucoadhesive polymers for intranasal administration promotes the residence time and contact of FVR in the mucus membrane. The optimized FVR-MCS-ALG-NPs demonstrated superior mucoadhesion, higher permeation and deposition in the nasal mucosa, and a significant increase in the inhibition of viral replication over 35-fold compared with free FVR. The overall results suggest that MCS-ALG-NPs could be used as an effective mucoadhesive carrier to enhance the activity of FVR against COVID-19.

11.
Polymers (Basel) ; 14(24)2022 Dec 19.
Article En | MEDLINE | ID: mdl-36559930

This study shows the effectiveness of magnetic-guide targeting in the delivery of curcumin diethyl γ-aminobutyrate (CUR-2GE), a prodrug of curcumin (CUR) previously synthesized to overcome unfavorable physicochemical properties of CUR. In this study, chitosan (Ch)-coated iron oxide nanoparticles (Ch-IONPs) were fabricated and optimized using Box-Behnken design-based response surface methodology for delivery of CUR-2GE. Ch was used as a coating material on the nanoparticle surface to avoid aggregation. The optimized condition for preparing Ch-IONPs consisted of using 4 mg Ch fabricated at pH 11 under a reaction temperature of 85 °C. The optimized Ch-IONPs were successfully loaded with CUR-2GE with sufficient loading capacity (1.72 ± 0.01%) and encapsulation efficiency (94.9 ± 0.8%). The obtained CUR-2GE-loaded Ch-IONPs (CUR-2GE-Ch-IONPs) exhibited desirable characteristics including a particle size of less than 50 nm based on TEM images, superparamagnetic property, highly crystalline IONP core, sufficient stability, and sustained-release profile. In the presence of permanent magnets, CUR-2GE-Ch-IONPs significantly increased cellular uptake and cytotoxicity toward MDA-MB-231 with a 12-fold increase in potency compared to free CUR-2GE, indicating the potential of magnetic-field assisted delivery of CUR-2GE-Ch-IONPs for the treatment of triple-negative breast cancer.

12.
Chin Med ; 17(1): 119, 2022 Oct 23.
Article En | MEDLINE | ID: mdl-36274168

BACKGROUND: Curcumin and piperine are major bioactive compounds of Curcuma longa and Piper nigrum, widely consumed as spices and flock medicine. The combinational use of these plants is a common practice in Southeast Asia. Synergism between curcumin and piperine has been found in several animal models but not in periodontal disease and diabetes, and the antinociceptive interaction is still unknown. Hence, the present study aimed to assess the interaction between curcumin and piperine in pain and its potential CNS side effect profile. METHODS: Formalin test and in vitro LPS-stimulated RAW 264.7 macrophage cells were used to assess the synergistic interaction of curcumin and piperine in a mouse model of inflammatory pain. Tail-flick and cold plate tests were applied to determine the antinociceptive synergism between piperine and curcumin. The interaction was determined by applying isobolographic analysis. The potential CNS-side effects of the curcumin and piperine combination were also assessed using LABORAS automated home-cage behavioral analysis. RESULTS: Curcumin alone dose-dependently improved pain-like behaviors in the formalin, tail-flick, and cold plate tests with the ED50 of 71.4, 34.4, and 31.9 mg/kg, respectively. Additionally, piperine exhibited efficacy in the formalin, tail-flick, and cold plate tests with the ED50 of 18.4, 8.1, and 28.1 mg/kg, respectively. The combination of curcumin and piperine (1:1 ED50 ratio) produced synergistic interaction in the formalin, tail-flick, and cold plate tests as assessed significantly lower experimental ED50 values (5.9, 5.2, and 5.5 mg/kg) compared to theoretical ED50 values (44.9, 21.3, and 30.0 mg/kg), isobologram analysis, and interaction index values of 0.13, 0.24 and 0.18, respectively. The synergistic interaction of curcumin and piperine was further confirmed by the efficacy of the combination in LPS-stimulated RAW 264.7 macrophage cells. Curcumin and piperine interacted synergistically, reducing proinflammatory mediators. The combination also demonstrated better compatibility profiles with neuronal cells. Furthermore, the curcumin-piperine combination had no effects on mouse spontaneous locomotor behaviors in LABORAS automated home cage monitoring. CONCLUSION: Overall, the present study demonstrates strong antinociceptive synergism between curcumin and piperine in mouse models with no potential CNS side effects, suggesting its possible use in clinical trials.

13.
ACS Pharmacol Transl Sci ; 5(9): 774-790, 2022 Sep 09.
Article En | MEDLINE | ID: mdl-36110378

Curcumin is a naturally occurring polyphenol compound with potential analgesic effects. It has been shown to improve pain-like behaviors in numerous models of pain. Despite its potential, curcumin exhibits poor physicochemical and pharmacokinetic properties, which hinder its oral therapeutic efficacy. Curcumin diethyl γ-aminobutyrate (CUR-2GE), a carbamate prodrug of curcumin, was designed to overcome these limitations and demonstrated greater anti-neuroinflammatory effects compared to curcumin in vitro. Thus, this study evaluated the effect of CUR-2GE and its parent compound on pain-like behaviors in carrageenan- and LPS-induced mouse models. The possible side effects of CUR-2GE were also assessed by exploring its effects on motor coordination and spontaneous locomotor activity after acute and chronic treatments. The results showed that CUR-2GE improved mechanical and thermal hyperalgesia and locomotor activity to a greater extent than curcumin in carrageenan-induced mice. These results are in line with the ability of CUR-2GE to suppress peripheral inflammation in the paw tissue of carrageenan-induced mice, indicated by a significant decrease in TNF-α and IL-6 expression levels. Similarly, in LPS-induced mice, CUR-2GE improved sickness and pain-like behaviors (exploratory behaviors and long-term locomotor activity) to a greater extent than curcumin. Furthermore, CUR-2GE significantly reduced the level of proinflammatory cytokines in both the plasma and spinal cord tissue of LPS-induced mice, exhibiting significantly higher inhibition than curcumin. Moreover, the motor coordination, and locomotive behaviors of mice were not affected by both acute and chronic administration of CUR-2GE, indicating no potential CNS side effects. Thus, CUR-2GE demonstrated enhanced therapeutic efficacy in mouse models of inflammatory pain without any possible CNS side effects, suggesting its potential to be developed as an analgesic agent against inflammatory pain.

14.
Molecules ; 27(12)2022 Jun 18.
Article En | MEDLINE | ID: mdl-35745046

Oxyresveratrol (OXY) has been reported for its anti-inflammatory activity; however, the pharmaceutical applications of this compound are limited by its physicochemical properties and poor pharmacokinetic profiles. The use of an ester prodrug is a promising strategy to overcome these obstacles. In previous researches, several carboxylate esters of OXY were synthesized and oxyresveratrol tetraacetate (OXY-TAc) was reported to possess anti-melanogenic and anti-skin-aging properties. In this study, in addition to OXY-TAc, two novel ester prodrugs of OXY, oxyresveratrol tetrapropionate (OXY-TPr), and oxyresveratrol tetrabutyrate (OXY-TBu), were synthesized. Results from the Caco-2-permeation assay suggested that synthesized ester prodrugs can improve the membrane-permeation ability of OXY. The OXY-TAc exhibited the most significant profile, then this prodrug was chosen to observe anti-inflammatory activities with lipopolysaccharide (LPS)-stimulated RAW264.7 macrophages. Our results showed that OXY-Tac significantly alleviated secretion of several pro-inflammatory mediators (nitric oxide (NO), interleukin-6 (IL-6), and tumor necrosis factor-alpha (TNF-α)), mitigated expression of enzyme-regulated inflammation (inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2)), and suppressed the MAPK cascades. Interestingly, the observed anti-inflammatory activities of OXY-TAc were more remarkable than those of its parent compound OXY. Taken together, we demonstrated that OXY-TAc improved physicochemical and pharmacokinetic profiles and enhanced the pharmacological effects of OXY. Hence, the results in the present study would strongly support the clinical utilities of OXY-TAc for the treatment of inflammation-related disorders.


Lipopolysaccharides , Prodrugs , Animals , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/pharmacology , Caco-2 Cells , Cyclooxygenase 2/metabolism , Esters/metabolism , Esters/pharmacology , Humans , Inflammation/drug therapy , Inflammation/metabolism , Lipopolysaccharides/metabolism , Lipopolysaccharides/pharmacology , Macrophages , Mice , NF-kappa B/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Plant Extracts , Prodrugs/metabolism , Prodrugs/pharmacology , RAW 264.7 Cells , Stilbenes
15.
J Nat Prod ; 85(7): 1816-1825, 2022 07 22.
Article En | MEDLINE | ID: mdl-35707966

Batatasin III is a stilbenoid compound present in a wide variety of Dendrobium species. Although the pharmacological efficacy of batatasin III has been reported in several disease models, its antinociceptive efficacy and central nervous system (CNS) side effects remain unknown. Thus, this study examined the effects of batatasin III on pain-like behaviors in mouse models of formalin- and lipopolysaccharide (LPS)-induced inflammatory pain. The results revealed a significant antinociceptive effect of batatasin III in both models, as 50 mg/kg batatasin III elicited comparable antinociception as 10 mg/kg indomethacin. Further, the anti-inflammatory effect of batatasin III was assessed in LPS-induced RAW 264.7 macrophages and BV-2 microglial cells. The compound significantly reduced the levels of inflammatory mediators (nitric oxide, TNF-α, and IL-6) in LPS-stimulated cells in a concentration-dependent manner. Following efficacy evaluations, the potential CNS side effects of batatasin III were evaluated using the rotarod test and the Laboratory Animal Behavior Observation, Registration, and Analysis System. Batatasin III-treated mice exhibited comparable forced, spontaneous, and general locomotive behaviors to vehicle-treated mice, indicating no potential CNS side effects. Overall, this study demonstrated the preclinical antinociceptive efficacy and CNS safety of batatasin III, suggesting its potential role in the development of new analgesics.


Dendrobium , Stilbenes , Analgesics/pharmacology , Analgesics/therapeutic use , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Lipopolysaccharides/pharmacology , Mice , Nitric Oxide , Pain/chemically induced , Pain/drug therapy
16.
Sci Rep ; 12(1): 9713, 2022 06 11.
Article En | MEDLINE | ID: mdl-35690654

Metformin is a well-tolerated antidiabetic drug and has recently been repurposed for numerous diseases, including pain. However, a higher dose of metformin is required for effective analgesia, which can potentiate its dose-dependent gastrointestinal side effects. Curcumin is a natural polyphenol and has beneficial therapeutic effects on pain. Curcumin has been used as an analgesic adjuvant with several analgesic drugs, allowing synergistic antinociceptive effects. Nevertheless, whether curcumin can exert synergistic analgesia with metformin is still unknown. In the present study, the nature of curcumin-metformin anti-inflammatory interaction was evaluated in in vitro using lipopolysaccharide-induced RAW 264.7 macrophage and BV-2 microglia cells. In both macrophage and microglia, curcumin effectively potentiates the anti-inflammatory effects of metformin, indicating potential synergistic effects in both peripheral and central pathways of pain. The nature of the interaction between curcumin and metformin was further recapitulated using a mouse model of formalin-induced pain. Coadministration of curcumin and metformin at a 1:1 fixed ratio of their ED50 doses significantly reduced the dose required to produce a 50% effect compared to the theoretically required dose in phase II of the formalin test with a combination index value of 0.24. Besides, the synergistic interaction does not appear to involve severe CNS side effects indicated by no motor alterations, no alterations in short-term and long-term locomotive behaviors, and the general well-being of mice. Our findings suggest that curcumin exerts synergistic anti-inflammation with metformin with no potential CNS adverse effects.


Curcumin , Metformin , Analgesics/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Curcumin/pharmacology , Curcumin/therapeutic use , Humans , Metformin/therapeutic use , Pain/drug therapy
17.
Life Sci ; 303: 120661, 2022 Aug 15.
Article En | MEDLINE | ID: mdl-35643380

AIM: The C-X-C chemokine-receptor type 4 (CXCR4) is an emerging target for cancer drug discovery due to its high expression in cancer cells. The present study aimed to produce CXCR4 overexpressing HEK293T cells for a non-radioactive binding assay as a platform to identify drug candidates targeting CXCR4. MAIN METHODS: HEK293T cells stably expressing human CXCR4 were constructed by transfection of CXCR4 plasmids from the human CXCR4 gene. The CXCR4 overexpressing HEK293T cells were obtained by fluorescence-activated sorting and verified by conducting the competition binding assay of a known CXCR4 inhibitor, AMD3100 (plerixafor), to determine the IC50 value against monoclonal anti-human CD184 (hCD184) antibody tagged with fluorescence probe, phycoerythrin (PE). The non-radioactive binding assay using CXCR4 overexpressing HEK293T cells and PE-anti hCD184 was applied as a platform for identifying the target of natural compounds that exhibited cytotoxicity against cancer cell lines. KEY FINDINGS: The CXCR4 overexpressing HEK293T cells were produced with high expression (99.8%). The IC50 value of plerixafor determined by fluorescence tagged antibody-based competition assay using our developed cells agree with previously reported values using a radioligand binding assay. We observed no significant displacement of bound PE-anti-hCD184 by the test natural compounds which could be due to non-specific binding to other functional targets or organelles, low potency of the natural compounds, or binding to CXCR4 at deeper pockets. SIGNIFICANCE: The verified non-radioactive binding assay can serve as an alternative screening tool for anticancer lead compounds targeting CXCR4 and an essential tool for proof of mechanism study in the drug discovery.


Cyclams , Heterocyclic Compounds , Chemokine CXCL12/metabolism , HEK293 Cells , Hematopoietic Stem Cell Mobilization , Heterocyclic Compounds/pharmacology , Humans , Kidney/metabolism , Receptors, CXCR4/metabolism
18.
Polymers (Basel) ; 14(9)2022 Apr 29.
Article En | MEDLINE | ID: mdl-35567007

Turmeric oil (TO) exhibits various biological activities with limited therapeutic applications due to its instability, volatility, and poor water solubility. Here, we encapsulated TO in chitosan/alginate nanocapsules (CS/Alg-NCs) using o/w emulsification to enhance its physicochemical characteristics, using poloxamer 407 as a non-ionic surfactant. TO-loaded CS/Alg-NCs (TO-CS/Alg-NCs) were prepared with satisfactory features, encapsulation efficiency, release characteristics, and cytotoxicity against breast cancer cells. The average size of the fabricated TO-CS/Alg-NCs was around 200 nm; their distribution was homogenous, and their shapes were spherical, with smooth surfaces. The TO-CS/Alg-NCs showed a high encapsulation efficiency, of 70%, with a sustained release of TO at approximately 50% after 12 h at pH 7.4 and 5.5. The TO-CS/Alg-NCs demonstrated enhanced cytotoxicity against two breast cancer cells, MDA-MB-231 and MCF-7, compared to the unencapsulated TO, suggesting that CS/Alg-NCs are potential nanocarriers for TO and can serve as prospective candidates for in vivo anticancer activity evaluation.

19.
Pharmaceuticals (Basel) ; 15(5)2022 May 18.
Article En | MEDLINE | ID: mdl-35631446

Since the pandemic of severe acute respiratory syndrome coronavirus (SARS-CoV-2) in December 2019, the infection cases have quickly increased by more than 511 million people. The long epidemic outbreak over 28 months has affected health and economies worldwide. An alternative medicine appears to be one choice to alleviate symptoms and reduce mortality during drug shortages. Dendrobium extract is one of the traditional medicines used for COVID-19 infection. Several compounds in Dendrobium sp. had been reported to exert pharmacological activities to treat common COVID-19-related symptoms. Herein, in silico screening of 83 compounds from Dendrobium sp. by using the SARS-CoV-2 spike protein receptor-binding domain (RBD) as a drug target was performed in searching for a new lead compound against SARS-CoV-2 infection. Four hit compounds showing good binding affinity were evaluated for antiviral infection activity. The new lead compound DB36, 5-methoxy-7-hydroxy-9,10-dihydro-1,4-phenanthrenequinone, was identified with the IC50 value of 6.87 ± 3.07 µM. The binding mode revealed that DB36 bound with the spike protein at the host receptor, angiotensin-converting enzyme 2 (ACE2) binding motif, resulted in antiviral activity. This study substantiated the use of Dendrobium extract for the treatment of SARS-CoV-2 infection and has identified new potential chemical scaffolds for further drug development of SARS-CoV-2 entry inhibitors.

20.
Carbohydr Polym ; 288: 119401, 2022 Jul 15.
Article En | MEDLINE | ID: mdl-35450653

Chitosan (Ch)-coated nanostructured lipid carriers (NLCs) have great potential for transdermal delivery with high localization of chemotherapeutics in breast cancer. This study used tetrahydrocurcumin (THC), a primary metabolite of curcumin with enhanced antioxidant and anticancer properties, as a model compound to prepare NLCs. Response surface methodology was employed to optimize THC-loaded Ch-coated NLCs (THC-Ch-NLCs) fabricated by high-shear homogenization. The optimized THC-Ch-NLCs had particle size of 244 ± 18 nm, zeta potential of -17.5 ± 0.5 mV, entrapment efficiency of 76.6 ± 0.2% and drug loading of 0.28 ± 0.01%. In vitro release study of THC-Ch-NLCs showed sustained release following the Korsmeyer-Peppas model with Fickian and non-Fickian diffusion at pH 7.4 and 5.5, respectively. THC-Ch-NLCs demonstrated significantly enhanced in vitro skin permeation, cell uptake, and remarkable cytotoxicity toward MD-MBA-231 breast cancer cells compared to the unencapsulated THC, suggesting Ch-NLCs as potential transdermal nanocarriers of THC for triple-negative breast cancer treatment.


Breast Neoplasms , Chitosan , Curcumin , Nanostructures , Female , Humans , Breast Neoplasms/drug therapy , Chitosan/chemistry , Curcumin/analogs & derivatives , Curcumin/pharmacology , Drug Carriers/chemistry , Lipids/chemistry , Nanostructures/chemistry , Particle Size
...