Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 866
1.
Dalton Trans ; 53(20): 8772-8780, 2024 May 21.
Article En | MEDLINE | ID: mdl-38712840

A series of Ir(III)-naproxen (NPX) conjugates with the molecular formula [Ir(C^N)2bpy(4-CH2ONPX-4'-CH2ONPX)](PF6) (Ir-NPX-1-3) were designed and synthesized, including C^N = 2-phenylpyridine (ppy, Ir-NPX-1), 2-(2-thienyl)pyridine (thpy, Ir-NPX-2) and 2-(2,4-difluorophenyl)pyridine (dfppy, Ir-NPX-3). Cytotoxicity tests showed that Ir-NPX-1-3 exhibited excellent antitumor activity, especially in A549R cells. The cellular uptake experiment showed that the complexes were mainly localized in mitochondria, and induced apoptosis in A549R cells by damaging the structure and function of mitochondria. The main manifestations are a decrease in the mitochondrial membrane potential (MMP), an increase in reactive oxygen species (ROS) levels, and cell cycle arrest. Furthermore, Ir-NPX-1-3 could inhibit the migration and colony formation of cancer cells, demonstrating potential anti-metastatic ability. Finally, the anti-inflammatory and immunological applications of Ir-NPX-1-3 were verified. The downregulation of cyclooxygenase-2 (COX-2) and programmed death-ligand 1 (PD-L1) expression levels and the release of immunogenic cell death (ICD) related signaling molecules such as damage-associated molecular patterns (DAMPs) (cell surface calreticulin (CRT), high mobility group box 1 (HMGB1), and adenosine triphosphate (ATP)) indicate that these Ir(III) -NPX conjugates are novel ICD inducers with synergistic effects in multiple anti-tumor pathways.


Antineoplastic Agents , Coordination Complexes , Iridium , Mitochondria , Naproxen , Iridium/chemistry , Iridium/pharmacology , Naproxen/pharmacology , Naproxen/chemistry , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Mitochondria/drug effects , Mitochondria/metabolism , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Coordination Complexes/chemical synthesis , Animals , Mice , Inflammation/drug therapy , Apoptosis/drug effects , Reactive Oxygen Species/metabolism , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Membrane Potential, Mitochondrial/drug effects , Molecular Structure , Cell Line, Tumor
2.
Int J Mol Sci ; 25(8)2024 Apr 12.
Article En | MEDLINE | ID: mdl-38673856

Immune response to biomaterials, which is intimately related to their surface properties, can produce chronic inflammation and fibrosis, leading to implant failure. This study investigated the development of magnetic nanoparticles coated with silica and incorporating the anti-inflammatory drug naproxen, aimed at multifunctional biomedical applications. The synthesized nanoparticles were characterized using various techniques that confirmed the presence of magnetite and the formation of a silica-rich bioactive glass (BG) layer. In vitro studies demonstrated that the nanoparticles exhibited bioactive properties, forming an apatite surface layer when immersed in simulated body fluid, and biocompatibility with bone cells, with good viability and alkaline phosphatase activity. Naproxen, either free or encapsulated, reduced nitric oxide production, an inflammatory marker, while the BG coating alone did not show anti-inflammatory effects in this study. Overall, the magnetic nanoparticles coated with BG and naproxen showed promise for biomedical applications, especially anti-inflammatory activity in macrophages and in the bone field, due to their biocompatibility, bioactivity, and osteogenic potential.


Coated Materials, Biocompatible , Glass , Magnetite Nanoparticles , Naproxen , Naproxen/pharmacology , Naproxen/chemistry , Glass/chemistry , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Magnetite Nanoparticles/chemistry , Animals , Mice , Humans , Nitric Oxide/metabolism , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Silicon Dioxide/chemistry , Cell Survival/drug effects , RAW 264.7 Cells , Osteogenesis/drug effects
3.
Mol Carcinog ; 63(6): 1188-1204, 2024 Jun.
Article En | MEDLINE | ID: mdl-38506376

Recent preclinical studies have shown that the intake of nonsteroidal anti-inflammatory drugs (NSAIDs) aspirin and naproxen could be an effective intervention strategy against TMPRSS2-ERG fusion-driven prostate tumorigenesis. Herein, as a follow-up mechanistic study, employing TMPRSS2-ERG (fusion) positive tumors and plasma from TMPRSS2-ERG. Ptenflox/flox mice, we profiled the stage specific proteomic changes (focused on inflammatory circulating and prostate tissue/tumor-specific cytokines, chemokines, and growth factors/growth signaling-associated molecules) that contribute to prostate cancer (PCa) growth and progression in the TMPRSS2-ERG fusion-driven mouse model of tumorigenesis. In addition, the association of the protective effects of NSAIDs (aspirin 1400 ppm and naproxen 400 ppm) with the modulation of these specific molecular pathways was determined. A sandwich Elisa based membrane array-proteome profiler identifying 111 distinct signaling molecules was employed. Overall, the plasma and prostate tissue sample analyses identified 54 significant and differentially expressed cytokines, chemokines, and growth factors/growth signaling-associated molecules between PCa afflicted mice (TMPRSS2-ERG. Ptenflox/flox, age-matched noncancerous controls, NSAIDs-supplemented and no-drug controls). Bioinformatic analysis of the array outcomes indicated that the protective effect of NSAIDs was associated with reduced expression of (a) tumor promoting inflammatory molecules (M-CSF, IL-33, CCL22, CCL12, CX3CL1, CHI3L1, and CD93), (b) growth factors- growth signaling-associated molecules (Chemerin, FGF acidic, Flt-3 ligand, IGFBP-5, and PEDF), and (c) tumor microenvironment/stromal remodeling proteins MMP2 and MMP9. Overall, our findings corroborate the pathological findings that protective effects of NSAIDs in TMPSS2-ERG fusion-driven prostate tumorigenesis are associated with antiproliferative and anti-inflammatory effects and possible modulation of the immune cell enriched microenvironment.


Anti-Inflammatory Agents, Non-Steroidal , Aspirin , Naproxen , Prostatic Neoplasms , Animals , Male , Prostatic Neoplasms/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aspirin/pharmacology , Mice , Naproxen/pharmacology , Proteomics/methods , Inflammation/metabolism , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Prostate/pathology , Prostate/metabolism , Prostate/drug effects , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinogenesis/metabolism , Proteome/metabolism , Humans , Cytokines/metabolism , Cytokines/blood
4.
J Int Soc Sports Nutr ; 21(1): 2302046, 2024 Dec.
Article En | MEDLINE | ID: mdl-38198469

BACKGROUND: Non-steroidal anti-inflammatory drugs (NSAIDs) like ibuprofen, flurbiprofen, naproxen sodium, and indomethacin are commonly employed for their pain-relieving and inflammation-reducing qualities. NSAIDs work by blocking COX-1 and/or COX-2, enzymes which play roles in inflammation, fever, and pain. The main difference among NSAIDs lies in their affinity to these enzymes, which in turn, influences prostaglandin secretion, and skeletal muscle growth and regeneration. The current study investigated the effects of NSAIDs on human skeletal muscle cells, focusing on myoblast proliferation, differentiation, and muscle protein synthesis signaling. METHODS: Using human primary muscle cells, we examined the dose-response impact of flurbiprofen (25-200 µM), indomethacin (25-200 µM), ibuprofen (25-200 µM), and naproxen sodium (25-200 µM), on myoblast viability, myotube area, fusion, and prostaglandin production. RESULTS: We found that supraphysiological concentrations of indomethacin inhibited myoblast proliferation (-74 ± 2% with 200 µM; -53 ± 3% with 100 µM; both p < 0.05) compared to control cells and impaired protein synthesis signaling pathways in myotubes, but only attenuated myotube fusion at the highest concentrations (-18 ± 2% with 200 µM, p < 0.05) compared to control myotubes. On the other hand, ibuprofen had no such effects. Naproxen sodium only increased cell proliferation at low concentrations (+36 ± 2% with 25 µM, p < 0.05), and flurbiprofen exhibited divergent impacts depending on the concentration whereby low concentrations improved cell proliferation (+17 ± 1% with 25 µM, p < 0.05) but high concentrations inhibited cell proliferation (-32 ± 1% with 200 µM, p < 0.05). CONCLUSION: Our findings suggest that indomethacin, at high concentrations, may detrimentally affect myoblast proliferation and differentiation via an AKT-dependent mechanism, and thus provide new understanding of NSAIDs' effects on skeletal muscle cell development.


Flurbiprofen , Naproxen , Humans , Naproxen/pharmacology , Ibuprofen/pharmacology , Flurbiprofen/pharmacology , Indomethacin/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Muscle Fibers, Skeletal , Inflammation , Pain , Prostaglandins
5.
Naunyn Schmiedebergs Arch Pharmacol ; 397(3): 1633-1646, 2024 03.
Article En | MEDLINE | ID: mdl-37698622

The aim of this study was the investigation of analgesic and anti-inflammatory activity of naproxen and pioglitazone following intra-plantar injection of carrageenan and assessment of the PPAR-γ receptor involvement in these effects. Rats were intra-plantarly injected with carrageenan (1%, 100 µl) to induce thermal hyperalgesia and paw inflammation. Different groups of rats were pre-treated intraperitoneally with naproxen (1 and 10 mg/kg) or pioglitazone (3 and 10 mg/kg) or GW9662 (a selective PPAR-γ antagonist, 100 µl/paw). The volume of the paw was evaluated using a plethysmometer, and the hot plate test was employed to assess the pain threshold in the animals. Finally, TNF-α, IL-1ß, IL-6, and myeloperoxidase (MPO) activity status were evaluated in the hind paw tissue. Naproxen and pioglitazone demonstrated analgesic and anti-inflammatory activity. Concurrent injection of an ineffective dose of naproxen (1 mg/kg) with an ineffective dose of pioglitazone (3 mg/kg) caused augmented analgesic and anti-inflammatory activity, significantly (p≤0.001 and p≤0.01, respectively). Additionally, intra-plantar injection of GW-9662 before naproxen or pioglitazone significantly suppressed their analgesic (p≤0.001) and anti-inflammatory activity (p≤0.01). Also, naproxen and pioglitazone (10 mg/kg) significantly (p≤0.001) reduced carrageenan-induced MPO activity and TNF-α, IL-6, and IL-1ß releasing. Furthermore, PPAR-γ blockade significantly prevented suppressive effects of naproxen and pioglitazone on the MPO activity and inflammatory cytokines. Pioglitazone significantly increased analgesic and anti-inflammatory effects of naproxen. This study proposes that concurrent treatment with naproxen and pioglitazone may be a substitute for overcome pain and inflammation clinically, in the future, particularly in patients with cardiovascular disorders and diabetes.


Naproxen , Thiazolidinediones , Humans , Rats , Animals , Pioglitazone/pharmacology , Naproxen/pharmacology , Thiazolidinediones/pharmacology , Thiazolidinediones/therapeutic use , Tumor Necrosis Factor-alpha , Interleukin-6 , PPAR gamma , Ligands , Carrageenan , Analgesics/pharmacology , Analgesics/therapeutic use , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Inflammation/chemically induced , Inflammation/drug therapy
6.
Sci Rep ; 13(1): 17498, 2023 10 15.
Article En | MEDLINE | ID: mdl-37840054

Although nonsteroidal antiinflammatory drugs (NSAIDs) are frequently used for fever and pain during pregnancy, their possible interaction with perinatal renal injury induced by preeclampsia (PE) has not been addressed. Here, studies were undertaken in the N(gamma)-nitro-L-arginine methyl ester (L-NAME) PE model to assess the influence of gestational NSAIDs on renal damage in weaning dams. PE-evoked increments and decrements in urine protein and creatinine clearance, respectively, were intensified by celecoxib and weakened by diclofenac or naproxen. Naproxen also improved renal cloudy swelling, necrosis, and reduced glomerular area evoked by PE. The concomitant rises in renal expression of markers of oxidative stress (NOX2/4), extracellular matrix metaloproteinase deposition (MMP9), and prostanoids (PGE2, PGF2α, TXA2) were all more effectively reduced by naproxen compared with celecoxib or diclofenac. Western blotting showed tripled expression of mitogen-activated protein kinases (MAPKs; p-p38, p-JNK1, p-ERK1, p-ERK2) in PE kidneys that was overturned by all NSAIDs, with naproxen producing the largest drop in p-ERK2 expression. The PE-provoked elevation in renal expression of autophagic marker LC3 was reduced by naproxen and diclofenac, but not celecoxib. The data suggests superior effect for naproxen over other NSAIDs in rectifying preeclamptic renal injury and predisposing inflammatory, oxidative, autophagic, and fibrotic signals.


Naproxen , Pre-Eclampsia , Humans , Pregnancy , Female , Rats , Animals , Naproxen/pharmacology , Celecoxib , Diclofenac , Pre-Eclampsia/drug therapy , Weaning , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Vitamins
7.
J Environ Manage ; 348: 119246, 2023 Dec 15.
Article En | MEDLINE | ID: mdl-37820430

Photocatalytic water decontamination has emerged as a highly promising technology for efficient and rapid water treatment, harnessing sustainable solar energy as its driving force. In this study, we prepared visible-light active Bi2S3/CoS2 composites for the degradation of naproxen (NPX) and the inactivation of Escherichia coli (E. coli). The homogeneous dispersion of CoS2 was stably integrated with Bi2S3, resulting in a significant enhancement of the specific surface area, efficient utilization of visible light, and effective separation of photogenerated charge carriers. Consequently, this synergistic photocatalytic system greatly facilitated the successful degradation of NPX and the inactivation of E. coli under visible-light irradiation. Compared to the pure Bi2S3 and CoS2 catalysts, the Bi2S3/CoS2 (1:2) composites displayed significantly enhanced photodegradation activity, achieving 96.46% (k = 0.2847 min-1) degradation of NPX within 90 min and maintaining good recyclability with no significant decline after six successive cycles. Additionally, the photocatalytic inactivation of E. coli results indicated that Bi2S3/CoS2 composites exhibited excellent performance, leading to the inactivation of 7 log10 cfu mL-1 of bacterial cells after 150 min of visible-light exposure. Scanning Electron Microscopy (SEM) and K+ ions leakage tests demonstrated that the destruction of the E. coli cell membrane structure resulted in cell death. The outcomes of this work suggest that Bi2S3/CoS2 composites hold significant potential for treating water contaminated with antibiotic and microbial pollutants.


Escherichia coli , Naproxen , Naproxen/pharmacology , Naproxen/metabolism , Light , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Microscopy, Electron, Scanning , Catalysis
8.
World J Microbiol Biotechnol ; 39(9): 244, 2023 Jul 06.
Article En | MEDLINE | ID: mdl-37407806

Serious infections caused by Pseudomonas aeruginosa are usually related to quorum sensing (QS)-dependent virulence factors. Hence, QS inhibition is a promising approach to overcoming P. aeruginosa infections. This study aimed to investigate the effect of naproxen on biofilm formation and QS-related virulence traits of P. aeruginosa. Furthermore, the anti-QS potential of naproxen was evaluated using real-time PCR and molecular docking analysis. Our findings supported the anti-QS activity of naproxen, as evidenced by down-regulation of the lasI and rhlI genes expression as well as the attenuation of bacterial protease, hemolysin, pyocyanin, biofilm, and motility. Additionally, the high binding affinity of naproxen with QS regulatory proteins was determined in the molecular docking simulation. Altogether, these findings suggest that naproxen has a promising potential in inhibiting QS-associated traits of P. aeruginosa.


Naproxen , Pseudomonas aeruginosa , Naproxen/pharmacology , Molecular Docking Simulation , Quorum Sensing , Biofilms , Virulence Factors/genetics , Virulence Factors/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Bacterial Proteins/metabolism
9.
J Chem Inf Model ; 63(15): 4900-4911, 2023 08 14.
Article En | MEDLINE | ID: mdl-37462219

Our skin constitutes an effective permeability barrier that protects the body from exogenous substances but concomitantly severely limits the number of pharmaceutical drugs that can be delivered transdermally. In topical formulation design, chemical permeation enhancers (PEs) are used to increase drug skin permeability. In vitro skin permeability experiments can measure net effects of PEs on transdermal drug transport, but they cannot explain the molecular mechanisms of interactions between drugs, permeation enhancers, and skin structure, which limits the possibility to rationally design better new drug formulations. Here we investigate the effect of the PEs water, lauric acid, geraniol, stearic acid, thymol, ethanol, oleic acid, and eucalyptol on the transdermal transport of metronidazole, caffeine, and naproxen. We use atomistic molecular dynamics (MD) simulations in combination with developed molecular models to calculate the free energy difference between 11 PE-containing formulations and the skin's barrier structure. We then utilize the results to calculate the final concentration of PEs in skin. We obtain an RMSE of 0.58 log units for calculated partition coefficients from water into the barrier structure. We then use the modified PE-containing barrier structure to calculate the PEs' permeability enhancement ratios (ERs) on transdermal metronidazole, caffeine, and naproxen transport and compare with the results obtained from in vitro experiments. We show that MD simulations are able to reproduce rankings based on ERs. However, strict quantitative correlation with experimental data needs further refinement, which is complicated by significant deviations between different measurements. Finally, we propose a model for how to use calculations of the potential of mean force of drugs across the skin's barrier structure in a topical formulation design.


Molecular Dynamics Simulation , Skin Absorption , Naproxen/metabolism , Naproxen/pharmacology , Caffeine , Metronidazole/metabolism , Metronidazole/pharmacology , Skin , Water/metabolism , Permeability
10.
Plant Commun ; 4(6): 100632, 2023 Nov 13.
Article En | MEDLINE | ID: mdl-37254481

The phytohormone auxin plays central roles in many growth and developmental processes in plants. Development of chemical tools targeting the auxin pathway is useful for both plant biology and agriculture. Here we reveal that naproxen, a synthetic compound with anti-inflammatory activity in humans, acts as an auxin transport inhibitor targeting PIN-FORMED (PIN) transporters in plants. Physiological experiments indicate that exogenous naproxen treatment affects pleiotropic auxin-regulated developmental processes. Additional cellular and biochemical evidence indicates that naproxen suppresses auxin transport, specifically PIN-mediated auxin efflux. Moreover, biochemical and structural analyses confirm that naproxen binds directly to PIN1 protein via the same binding cavity as the indole-3-acetic acid substrate. Thus, by combining cellular, biochemical, and structural approaches, this study clearly establishes that naproxen is a PIN inhibitor and elucidates the underlying mechanisms. Further use of this compound may advance our understanding of the molecular mechanisms of PIN-mediated auxin transport and expand our toolkit in auxin biology and agriculture.


Arabidopsis Proteins , Arabidopsis , Humans , Arabidopsis/metabolism , Naproxen/pharmacology , Naproxen/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Indoleacetic Acids/metabolism , Plants/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/metabolism , NIMA-Interacting Peptidylprolyl Isomerase/metabolism
11.
Front Immunol ; 14: 1162669, 2023.
Article En | MEDLINE | ID: mdl-37207208

Background: Recent clinical trial data from Lynch Syndrome (LS) carriers demonstrated that naproxen administered for 6-months is a safe primary chemoprevention that promotes activation of different resident immune cell types without increasing lymphoid cellularity. While intriguing, the precise immune cell types enriched by naproxen remained unanswered. Here, we have utilized cutting-edge technology to elucidate the immune cell types activated by naproxen in mucosal tissue of LS patients. Methods: Normal colorectal mucosa samples (pre- and post-treatment) from a subset of patients enrolled in the randomized and placebo-controlled 'Naproxen Study' were obtained and subjected to a tissue microarray for image mass cytometry (IMC) analysis. IMC data was processed using tissue segmentation and functional markers to ascertain cell type abundance. Computational outputs were then used to quantitatively compare immune cell abundance in pre- and post-naproxen specimens. Results: Using data-driven exploration, unsupervised clustering identified four populations of immune cell types with statistically significant changes between treatment and control groups. These four populations collectively describe a unique cell population of proliferating lymphocytes within mucosal samples from LS patients exposed to naproxen. Conclusions: Our findings show that daily exposure of naproxen promotes T-cell proliferation in the colonic mucosa, which paves way for developing combination of immunoprevention strategies including naproxen for LS patients.


Antineoplastic Agents , Cancer Vaccines , Colorectal Neoplasms, Hereditary Nonpolyposis , Humans , Naproxen/pharmacology , Immunotherapy , Lymphocytes , Intestinal Mucosa , Chemoprevention
12.
Molecules ; 28(8)2023 Apr 12.
Article En | MEDLINE | ID: mdl-37110629

Naproxen is widely used for anti-inflammatory treatment but it can lead to serious side effects. To improve the anti-inflammatory activity and safety, a novel naproxen derivative containing cinnamic acid (NDC) was synthesized and used in combination with resveratrol. The results showed that the combination of NDC and resveratrol at different ratios have a synergistic anti-inflammatory efficacy in RAW264.7 macrophage cells. It was indicated that the combination of NDC and resveratrol at a ratio of 2:1 significantly inhibited the expression of carbon monoxide (NO), tumor necrosis factor α (TNF-α), interleukin 6 (IL-6), induced nitric oxide synthase (iNOS), cyclooxygenase 2 (COX-2) and reactive oxygen species (ROS) without detectable side effects on cell viability. Further studies revealed that these anti-inflammatory effects were mediated by the activation of nuclear factor kappa-B (NF-κB), mitogen-activated protein kinase (MAPK) and phosphoinositide-3 kinase (PI3K)/protein kinase B (Akt) signaling pathways, respectively. Taken together, these results highlighted the synergistic NDC and resveratrol anti-inflammatory activity that could be further explored as a strategy for the treatment of inflammatory disease with an improved safety profile.


Mitogen-Activated Protein Kinases , NF-kappa B , Animals , Mice , NF-kappa B/metabolism , Mitogen-Activated Protein Kinases/metabolism , Resveratrol/pharmacology , Naproxen/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Signal Transduction , Anti-Inflammatory Agents/pharmacology , RAW 264.7 Cells , Lipopolysaccharides/pharmacology , Nitric Oxide/metabolism , Cyclooxygenase 2/metabolism
13.
Molecules ; 28(8)2023 Apr 13.
Article En | MEDLINE | ID: mdl-37110654

Vulgarin, an eudesmanolide sesquiterpene isolated from Artemisia judaica, was refluxed with iodine to produce two derivatives (1 and 2), which were purified and spectroscopically identified as naproxen methyl ester analogs. The reaction mechanism by which 1 and 2 were formed is explained using a sigmatropic reaction with a 1,3 shift. The scaffold hopping via lactone ring opening enabled the new derivatives of vulgarin (1 and 2) to fit well inside the COX-2 active site with ΔG of -7.73 and -7.58 kcal/mol, respectively, which was better than that of naproxen (ΔG of -7.04 kcal/mol). Moreover, molecular dynamic simulations showed that 1 was able to achieve a faster steady-state equilibrium than naproxen. The novel derivative 1 showed promising cytotoxic activities against HepG-2, HCT-116, MCF-7, and A-549 cancer cell lines compared to those of vulgarin and naproxen.


Antineoplastic Agents , Neoplasms , Humans , Naproxen/pharmacology , Cell Line , Antineoplastic Agents/pharmacology , Cyclooxygenase 2 , Molecular Docking Simulation , Structure-Activity Relationship
14.
Org Biomol Chem ; 21(14): 2968-2975, 2023 04 05.
Article En | MEDLINE | ID: mdl-36938589

Bis-squaramide receptors L1-L4 bearing a dansyl moiety were synthesised and their potential applications as fluorescent probes towards non steroidal anti-inflammatory drugs naproxen and ketoprofen was investigated. A detailed photophysical characterization in CH3CN/DMSO solution (9 : 1 v/v) was conducted and demonstrated that the two macrocyclic receptors L1 and L2 show good sensitivity towards ketoprofen with an ON-OFF fluorescent response, while the two open chain receptors L3 and L4 behave similarly with the three guests considered. DFT theoretical calculations carried out on L2 and L4 as model receptors allowed to propose a possible coordination mode towards the guests. Finally, 1H-NMR spectroscopy in DMSO-d6/0.5% water solution demonstrated that the four receptors interact with the considered guests via H-bonds.


Ketoprofen , Naproxen , Naproxen/pharmacology , Naproxen/chemistry , Ketoprofen/pharmacology , Ketoprofen/chemistry , Dimethyl Sulfoxide , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/chemistry
15.
Int J Cancer ; 152(12): 2567-2579, 2023 06 15.
Article En | MEDLINE | ID: mdl-36752580

Colorectal cancer (CRC) is the second leading cause of cancer-related deaths in the United States. Patients with the genetic disorder Familial Adenomatous Polyposis (FAP) develop hundreds to thousands of polyps that unless removed by prophylactic colectomy will progress to CRC at an early age. Nonsteroidal anti-inflammatory drugs (NSAIDs) and the ω-3 polyunsaturated fatty acid (PUFA) eicosapentaenoic acid (EPA), have been evaluated for their chemopreventive potential in delaying CRC onset in high-risk patients. In our study, we determined whether the NSAID, naproxen, alone or in combination with a chemically-stable EPA analog (TP-252), affects tumor formation in the ApcPirc rat model. When compared to control diet, animals fed naproxen or HD TP-252 had 66% and 82% fewer tumors, respectively. However, animals fed a combination of naproxen and HD TP-252, exhibited a 95% reduction in tumor formation and a 98% reduction in tumor volume, respectively. To elucidate potential mechanisms of tumor protection, a comprehensive, targeted lipidomic analysis was performed on colonic mucosa to determine changes in eicosanoid metabolism. Animals receiving TP-252 alone or in combination with naproxen had significantly reduced mucosal levels of proinflammatory ω-6 eicosanoids (PGE2 , 5-HETE and 14,15-DiHETrE), along with a simultaneous increase in anti-inflammatory EPA-derived ω-3 eicosanoids. A comprehensive lipidomic analysis also uncovered several potential pharmacodynamic (PD) lipid biomarkers, including resolvin E2, 9-HEPE, 12-HEPE and 18-HEPE, that were significantly correlated with tumor protection. Further studies with this drug combination should be focused on dose optimization and the role of EPA-derived lipid mediators in CRC initiation and progression.


Adenomatous Polyposis Coli , Eicosapentaenoic Acid , Rats , Animals , Eicosapentaenoic Acid/pharmacology , Naproxen/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents , Eicosanoids
16.
Biochim Biophys Acta Biomembr ; 1865(3): 184099, 2023 03.
Article En | MEDLINE | ID: mdl-36493856

Non steroidal anti-inflammatory drugs (NSAIDs) are those of the most common over the counter (OTC) medications widely used by millions of people every day. Unfortunately, despite their popularity those drugs can cause serious side effects in the digestive system (ulcers, bleeding, and pain). These inconveniences are caused by the changes in the structures of the outer phospholipid layers of gastric mucus and mucosa. As a result the H+ ions from the stomach acid can pass easily through these natural protective barriers and damage the epithelial cells which causes ulcers and bleeding. Chitosan as a polysaccharide known for its unique biocompatibility, drug delivery possibilities and wound healing effect has been chosen to examine if it can induce the reduction of undesirable effects of naproxen. This paper focuses on the interactions of the naproxen with a model biological membrane with and without the presence of chitosan. Applying the Langmuir technique coupled with the surface potential measurements and the Brewster angle microscope imaging allowed to characterize successfully examined systems in terms of the monolayer compressibility, thickness, stability, electric properties and morphology. The results proved that the presence of naproxen alters the mechanical and electrical properties of the model membrane depending on its surface pressure. Moreover, the addition of chitosan to the lipid-drug system causes significant changes in the properties of the layer, i.e. a reduction of its compressibility, thickness and morphology modification. Nevertheless, chitosan suppresses some changes induced by naproxen such as alteration of the apparent dipole moment and film stability.


Chitosan , Naproxen , Humans , Naproxen/pharmacology , Naproxen/chemistry , Chitosan/chemistry , Ulcer , Water/chemistry , Phospholipids/chemistry
17.
Int J Toxicol ; 42(2): 172-181, 2023.
Article En | MEDLINE | ID: mdl-36537154

Metabolic activation of indirect-acting carcinogens in target organs is a recognized mechanism of carcinogenesis. This study aimed to determine the role of benzo[a]pyrene (BaP) metabolism enzymes lipoxygenase (LOX), cytochrome P4501A1 (CYP1A1), and prostaglandin synthetase (PGS) in the cytotoxicity and DNA damage induced by BaP in the human tracheobronchial epithelial cells (HBECs) using RNA interference strategy and metabolic enzyme inhibitors. Our results showed that in three epithelial cell lines (HBE, HTR-8/SVneo, and HaCat), BaP significantly upregulated 5-LOX protein expression. 15-LOX-2 expression also increased with increasing BaP concentration, but the change was less pronounced than that of 5-LOX. BaP caused significant cytotoxicity, DNA strand breaks, and 8-hydroxy-2'-deoxyguanosine formation in HBE, which was inhibited by 5-LOXshRNA, a specific inhibitor of 5-LOX (AA861), the CYP1A1 inhibitor α-naphthoflavone, and the PGS inhibitor naproxen. The protective effects of 5-LOXshRNA were stronger than AA861, naproxen and α-naphthoflavone. We conclude that BaP may be activated more by 5-LOX than by CYP1A1 and PGS to produce cytotoxicity and DNA damage in HBE.


Benzo(a)pyrene , Cytochrome P-450 CYP1A1 , Humans , Cytochrome P-450 CYP1A1/metabolism , Arachidonate 5-Lipoxygenase/genetics , Naproxen/metabolism , Naproxen/pharmacology , DNA Damage , Epithelial Cells
18.
J Biomol Struct Dyn ; 41(2): 753-763, 2023 02.
Article En | MEDLINE | ID: mdl-34871148

Body's homeostasis is dependent on many factors, such as maintaining balance between free radicals formation and degradation. Human serum albumin (HSA) also plays an important role in homeostasis. The aim of this study was thermodynamic analysis of the interaction between ketoprofen (KET), naproxen (NPX), diclofenac (DIC) and HSA, as well as the effect of drug-albumin binding on HSA antioxidant activity using calorimetric and spectrophotometric techniques. Based on the calorimetric analysis it has been shown that accompanied by hydrophobic interaction drugs-albumin binding is an exoenergetic reaction. All analyzed drugs and HSA showed the ability to react with free radicals such as a radical cation, formed as a result of the reaction between 2,2'-Azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) diammonium salt (ABTS) and potassium persulfate (K2S2O8). Using ABTS assay a synergistic effect of ketoprofen (KET) and naproxen (NPX) on HSA antioxidant activity was observed while the effect of diclofenac (DIC) binding with albumin was probably additive. Because some medications including KET, NPX and DIC belong to over the counter (OTC) non-steroidal anti-inflammatory drugs (NSAIDs), it is necessary to understand their influence on HSA antioxidant activity.Communicated by Ramaswamy H. Sarma.


Ketoprofen , Humans , Ketoprofen/chemistry , Naproxen/pharmacology , Naproxen/chemistry , Naproxen/metabolism , Antioxidants/pharmacology , Serum Albumin, Human , Diclofenac/pharmacology , Diclofenac/chemistry , Serum Albumin/chemistry , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Binding Sites
19.
Int J Neurosci ; 133(2): 159-168, 2023 Feb.
Article En | MEDLINE | ID: mdl-33635748

Aim: Cancer patients undergoing chemotherapy report cognitive changes collectively termed "chemo brain." Neuroinflammation is among the factors believed to contribute to "chemo brain" suggesting a potential beneficial role for anti-inflammatory drugs in cancer patients undergoing chemotherapy. We investigated whether the non-steroidal anti-inflammatory drug naproxen influenced hippocampal inflammation in non-tumor bearing female mice receiving the chemotherapy drug cyclophosphamide (CP).Materials and methods: Intact and ovariectomized C57BL/6 mice were used to examine potential role of ovarian hormones on neuroinflammation. The mice were placed on naproxen (375 ppm) or control diet, and a week later CP (100 mg/kg; i.p.) was administered every 3 days for 2 weeks. We analyzed hippocampal inflammatory biomarkers, anxiety-like behavior, spatial working memory, exploratory behavior, spontaneous locomotor activity and depression-like behavior.Results: CP produced significant effects on anti-inflammatory but not pro-inflammatory biomarkers. However, CP and naproxen in combination produced significant effects on both pro- and anti- inflammatory biomarkers. Naproxen and ovariectomy individually produced significant effects on pro- and anti-inflammatory biomarkers as well. Working memory and depression-like behavior were not significantly influenced by CP, naproxen or ovariectomy individually although CP and ovariectomy produced significant interaction effects on depression-like behavior. Exploratory behavior and locomotor activity showed significant effects of CP, and interaction between CP and naproxen was significant for locomotor activity.Conclusions: Ovariectomy, naproxen and a combination of CP and naproxen upregulate hippocampal pro- and anti- inflammatory biomarkers. None of the factors individually produce significant behavioral changes that could be consistent with chemo brain, although CP and ovariectomy in combination produced significant effects on depression-like behavior, a co-morbidity of chemo brain.


Naproxen , Neuroinflammatory Diseases , Mice , Female , Animals , Naproxen/pharmacology , Naproxen/therapeutic use , Mice, Inbred C57BL , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Hippocampus
20.
Molecules ; 27(20)2022 Oct 14.
Article En | MEDLINE | ID: mdl-36296501

The 4-allyl guaiacol is a natural phenolic molecule that has been widely studied for its antioxidant capacity against reactive-oxygen-species-mediated cellular damage. Therefore, we hypothesized that concomitant use of an antioxidant and NSAID may decrease the risk of gastrointestinal toxicity and make the therapy safer. To address the gastrointestinal toxicity of conventional NSAIDs, a new S-naproxen-4-allyl guaiacol chimera (MAS-1696) was computationally developed, chemically synthesized, and tested for anti-inflammatory effectiveness and gastrointestinal safety. The inhibitory potency of MAS-1696 tested against cyclooxygenase-2 (COX2), 15-lipoxygenase-2 (15-LOX2), and lipoxygenase-5 (5-LOX) in vitro revealed a stronger inhibition of COX2. Furthermore, the MAS-1696 chimera increased the COX selectivity index by 23% as compared to the parent compound naproxen, implying higher efficacy and gastric safety. In vivo data showed that MAS-1696 was less likely to cause gastrointestinal harm than naproxen while also exerting anti-inflammatory and analgesic effects equivalent to or superior to naproxen. In conclusion, MAS-1696 is orally active, bio-labile, and crystalline, making it a medication that may be administered orally.


Gastrointestinal Diseases , Naproxen , Humans , Anti-Inflammatory Agents , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Antioxidants , Arachidonate 15-Lipoxygenase , Cyclooxygenase 2 , Gastrointestinal Diseases/drug therapy , Guaiacol , Naproxen/pharmacology , Naproxen/therapeutic use , Oxygen
...