Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 8.409
Filter
1.
Int Heart J ; 65(3): 475-486, 2024.
Article in English | MEDLINE | ID: mdl-38825493

ABSTRACT

This study aimed to investigate the molecular mechanisms underlying the protective effects of cyclooxygenase (cox) inhibitors against myocardial hypertrophy.Rat H9c2 cardiomyocytes were induced by mechanical stretching. SD rats underwent transverse aortic constriction to induce pressure overload myocardial hypertrophy. Rats were subjected to echocardiography and tail arterial pressure in 12W. qPCR and western blot were used to detect the expression of Notch-related signaling. The inflammatory factors were tested by ELISA in serum, heart tissue, and cell culture supernatant.Compared with control, levels of pro-inflammatory cytokines IL-6, TNF-α, and IL-1ß were increased and anti-inflammatory cytokine IL-10 was reduced in myocardial tissues and serum of rat models. Levels of Notch1 and Hes1 were reduced in myocardial tissues. However, cox inhibitor treatment (aspirin and celecoxib), the improvement of exacerbated myocardial hypertrophy, fibrosis, dysfunction, and inflammation was parallel to the activation of Notch1/Hes1 pathway. Moreover, in vitro experiments showed that, in cardiomyocyte H9c2 cells, application of ~20% mechanical stretching activated inflammatory mediators (IL-6, TNF-α, and IL-1ß) and hypertrophic markers (ANP and BNP). Moreover, expression levels of Notch1 and Hes1 were decreased. These changes were effectively alleviated by aspirin and celecoxib.Cox inhibitors may protect heart from hypertrophy and inflammation possibly via the Notch1/Hes1 signaling pathway.


Subject(s)
Aspirin , Celecoxib , Myocytes, Cardiac , Rats, Sprague-Dawley , Receptor, Notch1 , Signal Transduction , Transcription Factor HES-1 , Animals , Receptor, Notch1/metabolism , Rats , Transcription Factor HES-1/metabolism , Signal Transduction/drug effects , Celecoxib/pharmacology , Aspirin/pharmacology , Aspirin/therapeutic use , Male , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Cyclooxygenase Inhibitors/pharmacology , Cyclooxygenase Inhibitors/therapeutic use , Cardiomegaly/metabolism , Cardiomegaly/prevention & control , Cardiomegaly/etiology , Disease Models, Animal
2.
Planta Med ; 90(7-08): 641-650, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38843802

ABSTRACT

Tropaeolum majus (garden nasturtium) is a plant with relevance in phytomedicine, appreciated not only for its pharmaceutical activities, but also for its beautiful leaves and flowers. Here, we investigated the phytochemical composition of senescent nasturtium leaves. Indeed, we identified yellow chlorophyll catabolites, also termed phylloxanthobilins, which we show to contribute to the bright yellow color of the leaves in the autumn season. Moreover, we isolated and characterized the phylloxanthobilins from T. majus, and report the identification of a pyro-phylloxanthobilin, so far only accessible by chemical synthesis. We show that the phylloxanthobilins contribute to bioactivities of T. majus by displaying strong anti-oxidative effects in vitro and in cellulo, and anti-inflammatory effects as assessed by COX-1 and COX-2 enzyme inhibition, similar to other bioactive ingredients of T. majus, isoquercitrin, and chlorogenic acid. Hence, phylloxanthobilins could play a role in the efficacy of T. majus in the treatment of urinary tract infections, an established indication of T. majus. With the results shown in this study, we aid in the completion of the phytochemical profile of T. majus by identifying additional bioactive natural products as relevant components of this medicinal plant.


Subject(s)
Anti-Inflammatory Agents , Antioxidants , Plant Leaves , Tropaeolum , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Antioxidants/pharmacology , Antioxidants/chemistry , Tropaeolum/chemistry , Plant Leaves/chemistry , Cyclooxygenase 2/metabolism , Cyclooxygenase 1/metabolism , Plant Extracts/pharmacology , Plant Extracts/chemistry , Cyclooxygenase Inhibitors/pharmacology , Cyclooxygenase Inhibitors/chemistry , Humans , Chlorophyll , Phytochemicals/pharmacology , Phytochemicals/isolation & purification , Phytochemicals/chemistry
3.
Eur J Med Chem ; 271: 116397, 2024 May 05.
Article in English | MEDLINE | ID: mdl-38626522

ABSTRACT

In this study, a new series of Isoxazole-carboxamide derivatives were synthesized and characterized via HRMS, 1H-, 13CAPT-NMR, and MicroED. The findings revealed that nearly all of the synthesized derivatives exhibited potent inhibitory activities against both COX enzymes, with IC50 values ranging from 4.1 nM to 3.87 µM. Specifically, MYM1 demonstrated the highest efficacy among the compounds tested against the COX-1, displaying an IC50 value of 4.1 nM. The results showed that 5 compounds possess high COX-2 isozyme inhibitory effects with IC50 value in range 0.24-1.30 µM with COX-2 selectivity indexes (2.51-6.13), among these compounds MYM4 has the lowest IC50 value against COX-2, with selectivity index around 4. Intriguingly, this compound displayed significant antiproliferative effects against CaCo-2, Hep3B, and HeLa cancer cell lines, with IC50 values of 10.22, 4.84, and 1.57 µM, respectively, which was nearly comparable to that of doxorubicin. Compound MYM4 showed low cytotoxic activities on normal cell lines LX-2 and Hek293t with IC50 values 20.01 and 216.97 µM respectively, with safer values than doxorubicin. Furthermore, compound MYM4 was able to induce the apoptosis, suppress the colonization of both HeLa and HepG2 cells. Additionally, the induction of Reactive oxygen species (ROS) production could be the mechanism underlying the apoptotic effect and the cytotoxic activity of the compound. In the 3D multicellular tumor spheroid model, results revealed that MYM4 compound hampered the spheroid formation capacity of Hep3B and HeLa cancer cells. Moreover, the molecular docking of MYM4 compound revealed a high affinity for the COX2 enzyme, with energy scores (S) -7.45 kcal/mol, which were comparable to celecoxib (S) -8.40 kcal/mol. Collectively, these findings position MYM4 as a promising pharmacological candidate as COX inhibitor and anticancer agent.


Subject(s)
Antineoplastic Agents , Cell Proliferation , Cyclooxygenase Inhibitors , Drug Screening Assays, Antitumor , Isoxazoles , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Isoxazoles/pharmacology , Isoxazoles/chemistry , Isoxazoles/chemical synthesis , Structure-Activity Relationship , Cell Proliferation/drug effects , Cyclooxygenase Inhibitors/pharmacology , Cyclooxygenase Inhibitors/chemical synthesis , Cyclooxygenase Inhibitors/chemistry , Molecular Structure , Dose-Response Relationship, Drug , Spheroids, Cellular/drug effects , Models, Molecular , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Cell Line, Tumor
4.
Bioorg Chem ; 147: 107372, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38653152

ABSTRACT

Joining the global demand for the discovery of potent NSAIDs with minimized ulcerogenic effect, new pyrazole clubbed thiazole derivatives 5a-o were designed and synthesized. The new derivatives were initially evaluated for their analgesic activity. Eight compounds 5a, 5c, 5d, 5e, 5f, 5h, 5m, and 5o showed higher activity than Indomethacin (potency = 105-130 % vs. 100 %). Subsequently, they were picked for further evaluation of their anti-inflammatory activity, ulcerogenic liability as well as toxicological studies. Derivatives 5h and 5m showed a potential % edema inhibition after 3 h (79.39 % and 72.12 %, respectively), with a promising safety profile and low ulcer indices (3.80 and 3.20, respectively). The two compounds 5h and 5m were subjected to in vitro COX-1 and COX-2 inhibition assay. The candidate 5h showed nearly equipotent COX-1 inhibition (IC50 = 38.76 nM) compared to the non-selective reference drug Indomethacin (IC50 = 35.72 nM). Compound 5m expressed significant inhibitory activities and a higher COX-2 selectivity index (IC50 = 87.74 nM, SI = 2.05) in comparison with Indomethacin (SI = 0.52), with less selectivity than Celecoxib (SI = 8.31). Simulation docking studies were carried out to gain insights into the binding interaction of compounds 5h and 5m in the vicinity of COX-1 and COX-2 enzymes that illustrated the importance of pyrazole clubbed thiazole core in hydrogen bonding interactions. The thiazole motif of compounds 5h and 5m exhibited a well orientation toward COX-1 Arg120 key residue by hydrogen bonding interactions. Compound 5h revealed an additional arene-cation interaction with Arg120 that could rationalize its superior COX-1 inhibitory activity. Compounds 5h and 5m overlaid the co-crystallized ligand Celecoxib I differently in the active site of COX-2. Compound 5m showed an enhanced accommodation with binding energy of - 6.13 vs. - 1.70 kcal/mol of compounds 5h. The naphthalene ring of compound 5m adopted the Celecoxib I benzene sulfonamide region that is stabilized by hydrogen-arene interactions with the hydrophobic sidechains of the key residues Ser339 and Phe504. Further, the core structure of compound 5m, pyrazole clubbed thiazole, revealed deeper hydrophobic interactions with Ala513, Leu517 and Val509 residues. Finally, a sensitive and accurate UPLC-MS/MS method was developed for the simultaneous estimation of some selected promising pyrazole derivatives in rat plasma. Accordingly, compounds 5h and 5m were suggested to be promising potent analgesic and anti-inflammatory agents with improved safety profiles and a novel COX isozyme modulation activity.


Subject(s)
Analgesics , Anti-Inflammatory Agents, Non-Steroidal , Cyclooxygenase 2 , Edema , Molecular Docking Simulation , Thiazoles , Animals , Male , Mice , Rats , Analgesics/pharmacology , Analgesics/chemistry , Analgesics/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Cyclooxygenase Inhibitors/pharmacology , Cyclooxygenase Inhibitors/chemistry , Cyclooxygenase Inhibitors/chemical synthesis , Dose-Response Relationship, Drug , Drug Discovery , Edema/drug therapy , Edema/chemically induced , Molecular Structure , Pyrazoles/chemistry , Pyrazoles/pharmacology , Pyrazoles/chemical synthesis , Structure-Activity Relationship , Thiazoles/chemistry , Thiazoles/pharmacology , Thiazoles/chemical synthesis
5.
Environ Toxicol Pharmacol ; 108: 104453, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38642625

ABSTRACT

Understanding interactions between legacy and emerging environmental contaminants has important implications for risk assessment, especially when mutagens and carcinogens are involved, whose critical effects are chronic and therefore difficult to predict. The current work aimed to investigate potential interactions between benzo[a]pyrene (B[a]P), a carcinogenic polycyclic aromatic hydrocarbon and legacy pollutant, and diclofenac (DFC), a non-steroidal anti-inflammatory drug and pollutant of emerging concern, and how DFC affects B[a]P toxicity. Exposure to binary mixtures of these chemicals resulted in substantially reduced cytotoxicity in human HepG2 cells compared to single-chemical exposures. Significant antagonistic effects were observed in response to high concentrations of B[a]P in combination with DFC at IC50 and ⅕ IC50. While additive effects were found for levels of intracellular reactive oxygen species, antagonistic mixture effects were observed for genotoxicity. B[a]P induced DNA strand breaks, γH2AX activation, and micronuclei formation at ½ IC50 concentrations or lower, whereas DFC induced only low levels of DNA strand breaks. Their mixture caused significantly lower levels of genotoxicity by all three endpoints compared to those expected based on concentration additivity. In addition, antagonistic mixture effects on CYP1 enzyme activity suggested that the observed reduced genotoxicity of B[a]P was due to its reduced metabolic activation as a result of enzymatic inhibition by DFC. Overall, the findings further support the growing concern that co-exposure to environmental toxicants and their non-additive interactions may be a confounding factor that should not be neglected in environmental and human health risk assessment.


Subject(s)
Benzo(a)pyrene , Carcinogens, Environmental , Diclofenac , Humans , Diclofenac/toxicity , Benzo(a)pyrene/toxicity , Hep G2 Cells , Carcinogens, Environmental/toxicity , Reactive Oxygen Species/metabolism , Cyclooxygenase 1/metabolism , Cell Survival/drug effects , Cyclooxygenase Inhibitors/pharmacology , Cyclooxygenase Inhibitors/toxicity , Cyclooxygenase 2/metabolism , DNA Damage/drug effects , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2 Inhibitors/toxicity , Histones
6.
Neoplasia ; 51: 100991, 2024 05.
Article in English | MEDLINE | ID: mdl-38507887

ABSTRACT

Dihydroartemisinin (DHA) exerts an anti-tumor effect in multiple cancers, however, the molecular mechanism of DHA and whether DHA facilitates the anti-tumor efficacy of cisplatin in non-small cell lung cancer (NSCLC) are unclear. Here, we found that DHA potentiated the anti-tumor effects of cisplatin in NSCLC cells by stimulating reactive oxygen species (ROS)-mediated endoplasmic reticulum (ER) stress, C-Jun-amino-terminal kinase (JNK) and p38 MAPK signaling pathways both in vitro and in vivo. Of note, we demonstrated for the first time that DHA inhibits prostaglandin G/H synthase 1 (PTGS1) expression, resulting in enhanced ROS production. Importantly, silencing PTGS1 sensitized DHA-induced cell death by increasing ROS production and activating ER-stress, JNK and p38 MAPK signaling pathways. In summary, our findings provided new experimental basis and therapeutic prospect for the combined therapy with DHA and cisplatin in some NSCLC patients.


Subject(s)
Artemisinins , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Reactive Oxygen Species , Humans , Apoptosis , Artemisinins/pharmacology , Artemisinins/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Death , Cell Line, Tumor , Cisplatin/pharmacology , Cyclooxygenase 1/metabolism , Lung Neoplasms/pathology , p38 Mitogen-Activated Protein Kinases/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Cyclooxygenase Inhibitors/pharmacology
7.
J Appl Physiol (1985) ; 136(5): 1226-1237, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38545661

ABSTRACT

Cyclooxygenase (COX) products of arachidonic acid metabolism, specifically prostaglandins, play a role in evoking and transmitting the exercise pressor reflex in health and disease. Individuals with type 2 diabetes mellitus (T2DM) have an exaggerated exercise pressor reflex; however, the mechanisms for this exaggerated reflex are not fully understood. We aimed to determine the role played by COX products in the exaggerated exercise pressor reflex in T2DM rats. The exercise pressor reflex was evoked by static muscle contraction in unanesthetized, decerebrate, male, adult University of California Davis (UCD)-T2DM (n = 8) and healthy Sprague-Dawley (n = 8) rats. Changes (Δ) in peak mean arterial pressure (MAP) and heart rate (HR) during muscle contraction were compared before and after intra-arterial injection of indomethacin (1 mg/kg) into the contracting hindlimb. Data are presented as means ± SD. Inhibition of COX activity attenuated the exaggerated peak MAP (Before: Δ32 ± 13 mmHg and After: Δ18 ± 8 mmHg; P = 0.004) and blood pressor index (BPi) (Before: Δ683 ± 324 mmHg·s and After: Δ361 ± 222 mmHg·s; P = 0.006), but not HR (Before: Δ23 ± 8 beats/min and After Δ19 ± 10 beats/min; P = 0.452) responses to muscle contraction in T2DM rats. In healthy rats, COX activity inhibition did not affect MAP, HR, or BPi responses to muscle contraction. Inhibition of COX activity significantly reduced local production of prostaglandin E2 in T2DM and healthy rats. We conclude that peripheral inhibition of COX activity attenuates the pressor response to muscle contraction in T2DM rats, suggesting that COX products partially contribute to the exaggerated exercise pressor reflex in those with T2DM.NEW & NOTEWORTHY We compared the pressor and cardioaccelerator responses to static muscle contraction before and after inhibition of cyclooxygenase (COX) activity within the contracting hindlimb in decerebrate, unanesthetized type 2 diabetic mellitus (T2DM) and healthy rats. The pressor responses to muscle contraction were attenuated after peripheral inhibition of COX activity in T2DM but not in healthy rats. We concluded that COX products partially contribute to the exaggerated pressor reflex in those with T2DM.


Subject(s)
Blood Pressure , Diabetes Mellitus, Type 2 , Heart Rate , Muscle Contraction , Muscle, Skeletal , Rats, Sprague-Dawley , Reflex , Animals , Male , Diabetes Mellitus, Type 2/physiopathology , Diabetes Mellitus, Type 2/metabolism , Muscle Contraction/physiology , Rats , Heart Rate/physiology , Heart Rate/drug effects , Reflex/physiology , Muscle, Skeletal/physiopathology , Blood Pressure/physiology , Blood Pressure/drug effects , Physical Conditioning, Animal/physiology , Indomethacin/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Arterial Pressure/physiology , Prostaglandin-Endoperoxide Synthases/metabolism
8.
Andrology ; 12(4): 899-917, 2024 May.
Article in English | MEDLINE | ID: mdl-37772683

ABSTRACT

BACKGROUND: Acetaminophen and ibuprofen are widely administered to babies due to their presumed safety as over-the-counter drugs. However, no reports exist on the effects of cyclooxygenase inhibitors on undifferentiated spermatogonia and spermatogonial stem cells. Infancy represents a critical period for spermatogonial stem cell formation and disrupting spermatogonial stem cells or their precursors may be associated with infertility and testicular cancer formation. OBJECTIVES: The goal of this study was to examine the molecular and functional impact of cyclooxygenase inhibition and silencing on early steps of undifferentiated spermatogonia (u spg) and spermatogonial stem cell development, to assess the potential reproductive risk of pharmaceutical cyclooxygenase inhibitors. METHODS: The effects of cyclooxygenase inhibition were assessed using the mouse C18-4 undifferentiated juvenile spermatogonial cell line model, previously shown to include cells with spermatogonial stem cell features, by measuring prostaglandins, cell proliferation, and differentiation, using cyclooxygenase 1- and cyclooxygenase 2-selective inhibitors NS398, celecoxib, and FR122047, acetaminophen, and ibuprofen. Cyclooxygenase 1 gene silencing was achieved using a stable short-hairpin RNA approach and clone selection, then assessing gene and protein expression in RNA sequencing, quantitative real-time polymerase chain reaction, and immunofluorescence studies. RESULTS: Cyclooxygenase 2 inhibitors NS398 and celecoxib, as well as acetaminophen, but not ibuprofen, dose-dependently decreased retinoic acid-induced expression of the spg differentiation gene Stra8, while NS398 decreased the spg differentiation marker Kit, suggesting that cyclooxygenase 2 is positively associated with spg differentiation. In contrast, short-hairpin RNA-based cyclooxygenase 1 silencing in C18-4 cells altered cellular morphology and upregulated Stra8 and Kit, implying that cyclooxygenase 1 prevented spg differentiation. Furthermore, RNA sequencing analysis of cyclooxygenase 1 knockdown cells indicated the activation of several signaling pathways including the TGFb, Wnt, and Notch pathways, compared to control C18-4 cells. Notch pathway genes were upregulated by selective cyclooxygenase inhibitors, acetaminophen and ibuprofen. CONCLUSION: We report that cyclooxygenase 1 and 2 differentially regulate undifferentiated spermatogonia/spermatogonial stem cell differentiation. Cyclooxygenases regulate Notch3 expression, with the Notch pathway targeted by PGD2. These data suggest an interaction between the eicosanoid and Notch signaling pathways that may be critical for the development of spermatogonial stem cells and subsequent spermatogenesis, cautioning about using cyclooxygenase inhibitors in infants.


Subject(s)
Nitrobenzenes , Spermatogonia , Sulfonamides , Testicular Neoplasms , Humans , Male , Animals , Mice , Spermatogonia/metabolism , Testicular Neoplasms/metabolism , Cyclooxygenase 1/genetics , Cyclooxygenase 1/metabolism , Cyclooxygenase 1/pharmacology , Cyclooxygenase 2/metabolism , Celecoxib/pharmacology , Celecoxib/metabolism , Ibuprofen/pharmacology , Acetaminophen , Spermatogenesis/physiology , Cell Differentiation/physiology , Cyclooxygenase Inhibitors/pharmacology , RNA/metabolism , Testis/metabolism
9.
Cell Prolif ; 57(5): e13594, 2024 May.
Article in English | MEDLINE | ID: mdl-38155412

ABSTRACT

The study of neurogenesis is essential to understanding fundamental developmental processes and for the development of cell replacement therapies for central nervous system disorders. Here, we designed an in vivo drug screening protocol in developing zebrafish to find new molecules and signalling pathways regulating neurogenesis in the ventral spinal cord. This unbiased drug screen revealed that 4 cyclooxygenase (COX) inhibitors reduced the generation of serotonergic interneurons in the developing spinal cord. These results fitted very nicely with available single-cell RNAseq data revealing that floor plate cells show differential expression of 1 of the 2 COX2 zebrafish genes (ptgs2a). Indeed, several selective COX2 inhibitors and two different morpholinos against ptgs2a reduced the number of serotonergic neurons in the ventral spinal cord and led to locomotor deficits. Single-cell RNAseq data and different pharmacological manipulations further revealed that COX2-floor plate-derived prostaglandin D2 promotes neurogenesis in the developing spinal cord by promoting mitotic activity in progenitor cells. Rescue experiments using a phosphodiesterase-4 inhibitor suggest that intracellular changes in cAMP levels underlie the effects of COX inhibitors on neurogenesis and locomotion. Our study provides compelling in vivo evidence showing that prostaglandin signalling promotes neurogenesis in the ventral spinal cord.


Subject(s)
Cyclooxygenase 2 , Neurogenesis , Spinal Cord , Zebrafish , Animals , Zebrafish/metabolism , Neurogenesis/drug effects , Spinal Cord/metabolism , Spinal Cord/cytology , Spinal Cord/drug effects , Cyclooxygenase 2/metabolism , Cyclooxygenase 2/genetics , Drug Evaluation, Preclinical/methods , Cyclooxygenase 2 Inhibitors/pharmacology , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics , Signal Transduction/drug effects , Cyclooxygenase Inhibitors/pharmacology
10.
Sci Rep ; 13(1): 22730, 2023 12 20.
Article in English | MEDLINE | ID: mdl-38123572

ABSTRACT

Fungal infections of skin including mycoses are one of the most common infections in skin or skins. Mycosis is caused by dermatophytes, non-dermatophyte moulds and yeasts. Various studies show different drugs to treat mycoses, yet there is need to treat it with applied drugs delivery. This study was designed to prepare a bio curcumin (CMN) nanoemulsion (CMN-NEs) for transdermal administration to treat mycoses. The self-nanoemulsification approach was used to prepare a nanoemulsion (NE), utilizing an oil phase consisting of Cremophor EL 100 (Cre EL), glyceryl monooleate (GMO), and polyethylene glycol 5000 (PEG 5000). Particle size (PS), polydispersity index (PDI), zeta potential (ZP), Fourier transform infrared (FTIR) spectrophotometric analysis, and morphological analyses were performed to evaluate the nanoemulsion (NE). The in vitro permeation of CMN was investigated using a modified vertical diffusion cell with an activated dialysis membrane bag. Among all the formulations, a stable, spontaneously produced nanoemulsion was determined with 250 mg of CMN loaded with 10 g of the oil phase. The average droplet size, ZP, and PDI of CMN-NEs were 90.0 ± 2.1 nm, - 7.4 ± 0.4, and 0.171 ± 0.03 mV, respectively. The release kinetics of CMN differed from zero order with a Higuchi release profile as a result of nanoemulsification, which also significantly increased the flux of CMN permeating from the hydrophilic matrix gel. Overall, the prepared nanoemulsion system not only increased the permeability of CMN but also protected it against chemical deterioration. Both CMN-ME (24.0 ± 0.31 mm) and CMN-NE gel (29.6 ± 0.25 mm) had zones of inhibition against Candida albicans that were significantly larger than those of marketed Itrostred gel (21.5 ± 0.34 mm). The prepared CMN-NE improved the bioavailability, better skin penetration, and the CMN-NE gel enhanced the release of CMN from the gel matrix on mycotic patients.


Subject(s)
Curcumin , Mycoses , Humans , Skin Absorption , Curcumin/pharmacology , Curcumin/metabolism , Renal Dialysis , Skin/metabolism , Cyclooxygenase Inhibitors/pharmacology , Emulsions/pharmacology , Mycoses/drug therapy , Mycoses/metabolism
11.
Bioorg Chem ; 141: 106847, 2023 12.
Article in English | MEDLINE | ID: mdl-37722268

ABSTRACT

The anti-inflammatory and analgesic drugs currently used are associated with several adverse effects and found to be highly unsafe for long-term use. Currently, nineteen novel bis-Schiff base derivatives (1-19) of flurbiprofen have been designed, prepared and assessed for in-vivo analgesic, anti-inflammatory and in vivo acute toxicity evaluation. The structures of the acquired compounds were deduced through modern spectroscopic techniques including HR-ESI-MS, 13C-, and 1H NMR. Amongst the series, compounds 7, 9, and 10 attributed potent activities with 93.89, 92.50, and 90.47% decreased edema, respectively compared to flurbiprofen (90.01%), however, compounds 11 and 15 exhibited significant activity of 90.00% decrease. Out of them, fourteen compounds (1-6, 8, 12-14, and 16-19) displayed good activity in the range of 68.96-86.95%. In case of an analgesic study, all the derivatives significantly (p 0.001) increased the pain threshold time particularly compound 7 had the best analgesic effect (24 ± 2.08 s) in comparison with flurbiprofen (21.66 ± 2.02 s) using hot plate test. Similarly, in the acetic acid-induced writhing test, compound 7 determined a potent inhibitory effect (60.47 %) close to flurbiprofen (59.28%). All the synthesized derivatives were found safe up to the dose of 30 mg/kg, in acute toxicity study. On a molecular scale, the synthesized compounds were modeled through a ligand-based pharmacophore study and molecular docking to have insight into the different possible interactions leading to high inhibition levels against the COX-2 enzyme.


Subject(s)
Flurbiprofen , Humans , Flurbiprofen/pharmacology , Flurbiprofen/chemistry , Cyclooxygenase Inhibitors/pharmacology , Molecular Docking Simulation , Cyclooxygenase 2 , Analgesics/pharmacology , Analgesics/therapeutic use , Analgesics/chemistry , Anti-Inflammatory Agents/chemistry , Edema/chemically induced , Edema/drug therapy , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Carrageenan
12.
Molecules ; 28(14)2023 Jul 13.
Article in English | MEDLINE | ID: mdl-37513247

ABSTRACT

Inhibition of cyclooxygenase-2 (COX-2) has been extensively studied as an approach to reduce proinflammatory markers in acute brain diseases, but the anti-neuroinflammatory role of cyclooxygenase-1 (COX-1) inhibition has been rather neglected. We report that m-terphenylamine derivatives are selective COX-1 inhibitors, able to block microglia inflammatory response and elicit a neuroprotective effect. These compounds were synthesized via a three-component reaction of chalcones, ß-ketoesters, and primary amines, followed by hydrolysis/decarboxylation of the ester group. Together with their synthetic intermediates and some urea derivatives, they were studied as inhibitors of COX-1 and COX-2. The m-terphenylamine derivatives, which were selective COX-1 inhibitors, were also analyzed for their ability to block microglia inflammatory and oxidative response. Compound 3b presented an interesting anti-inflammatory and neuroprotective profile by reducing nitrite release, ROS overproduction, and cell death in organotypic hippocampal cultures subjected to LPS. We thus show that COX-1 inhibition is a promising approach to provide enhanced neuroprotection against acute inflammatory processes, which are crucial in the development of a plethora of acute neurodegenerative injuries.


Subject(s)
Microglia , Neuroprotective Agents , Cyclooxygenase 2/metabolism , Neuroprotection , Cyclooxygenase Inhibitors/pharmacology , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/metabolism , Lipopolysaccharides/pharmacology
13.
Molecules ; 28(14)2023 Jul 18.
Article in English | MEDLINE | ID: mdl-37513351

ABSTRACT

Secure and efficient treatment of diverse pain and inflammatory disorders is continually challenging. Although NSAIDs and other painkillers are well-known and commonly available, they are sometimes insufficient and can cause dangerous adverse effects. As yet reported, derivatives of pyrrolo[3,4-d]pyridazinone are potent COX-2 inhibitors with a COX-2/COX-1 selectivity index better than meloxicam. Considering that N-acylhydrazone (NAH) moiety is a privileged structure occurring in many promising drug candidates, we decided to introduce this pharmacophore into new series of pyrrolo[3,4-d]pyridazinone derivatives. The current paper presents the synthesis and in vitro, spectroscopic, and in silico studies evaluating the biological and physicochemical properties of NAH derivatives of pyrrolo[3,4-d]pyridazinone. Novel compounds 5a-c-7a-c were received with high purity and good yields and did not show cytotoxicity in the MTT assay. Their COX-1, COX-2, and 15-LOX inhibitory activities were estimated using enzymatic tests and molecular docking studies. The title N-acylhydrazones appeared to be promising dual COX/LOX inhibitors. Moreover, spectroscopic and computational methods revealed that new compounds form stable complexes with the most abundant plasma proteins-AAG and HSA, but do not destabilize their secondary structure. Additionally, predicted pharmacokinetic and drug-likeness properties of investigated molecules suggest their potentially good membrane permeability and satisfactory bioavailability.


Subject(s)
Cyclooxygenase Inhibitors , Hydrazones , Lipoxygenase Inhibitors , Pyridazines , Pyrroles , Hydrazones/chemical synthesis , Hydrazones/chemistry , Hydrazones/pharmacokinetics , Hydrazones/pharmacology , Cyclooxygenase Inhibitors/chemical synthesis , Cyclooxygenase Inhibitors/chemistry , Cyclooxygenase Inhibitors/pharmacokinetics , Cyclooxygenase Inhibitors/pharmacology , Pyridazines/chemical synthesis , Pyridazines/chemistry , Pyridazines/pharmacokinetics , Pyridazines/pharmacology , Pyrroles/chemical synthesis , Pyrroles/chemistry , Pyrroles/pharmacokinetics , Pyrroles/pharmacology , Humans , Fibroblasts , Computer Simulation , Cell Membrane Permeability , Cell Line
14.
JBRA Assist Reprod ; 27(3): 386-390, 2023 09 12.
Article in English | MEDLINE | ID: mdl-37134015

ABSTRACT

OBJECTIVE: Endogenous prostaglandins are involved in hemostasis, renal excretion of electrolytes, and implicated in dysmenorrhea. Piroxicam and Nitroglycerin are common drugs used in treating dysmenorrhea by inhibiting the cyclooxygenase pathway involved in prostaglandin production. However, studies comparing the effects of these drugs on prostaglandin-modulated hemostasis and renal function are lacking. METHODS: Fifteen female rats (120-160g) were divided into 3 groups (20 per group), namely Control (distilled water, 0.3 mL), Piroxicam treated (3mg/kg) and Nitroglycerin treated (1 mg/kg). Di-estrous phase was confirmed in animals in each group using the Pipette smear method. Treatment was administered for 4 days covering the estrous cycle. Bleeding and clotting time were assessed and blood concentrations of sodium, potassium, urea and platelet counts were evaluated in all phases. Data were analyzed using one-way ANOVA and Newman-Keuls post-hoc test. Statistical significance was considered at p<0.0. RESULTS: The nitroglycerin-treated group showed significant increases in blood potassium during di-estrous while the piroxicam-treated group showed significant increases in blood potassium, urea and clotting time with a significant decrease in sodium levels during di-estrous compared to controls. Results obtained in other phases were not significant compared to controls. CONCLUSIONS: The study showed that Nitroglycerin produces minimum alteration of blood and electrolyte indices compared to piroxicam during di-estrous.


Subject(s)
Nitroglycerin , Piroxicam , Humans , Female , Rats , Animals , Piroxicam/pharmacology , Piroxicam/therapeutic use , Rats, Wistar , Nitroglycerin/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Prostaglandins , Dysmenorrhea , Urea/pharmacology , Sodium , Electrolytes , Potassium
15.
Eur J Pharmacol ; 950: 175751, 2023 Jul 05.
Article in English | MEDLINE | ID: mdl-37116562

ABSTRACT

Cardiac inflammation is easily accompanied by hypoxia, while hypoxia-induced injury and microenvironmental variations limit the efficacy of common anti-inflammatory drugs. In order to effectively attenuate myocardial injury caused by hypoxic and inflammatory injury, we designed and synthesized a kind of anti-inflammatory compounds by coupling cyclooxygenase (COX) and carbonic anhydrase (CA) inhibitors, and evaluated the activity and their mechanism in vitro and in vivo. It was found that these compounds were structurally stable and had two enzymatic inhibition activities. By inhibiting the activity of overexpressed CA under hypoxia, the acidic microenvironment can be regulated to inhibit the hypoxic injury, in which the pH-dependent primary drug resistance can be overcome to improve the anti-inflammatory effect of the COX inhibitor. Consequently, this study provides a new strategy for the treatment of cardiac inflammation accompanied by hypoxia.


Subject(s)
Carbonic Anhydrases , Humans , Cyclooxygenase Inhibitors/pharmacology , Cyclooxygenase Inhibitors/therapeutic use , Carbonic Anhydrase Inhibitors/pharmacology , Carbonic Anhydrase Inhibitors/therapeutic use , Hypoxia/complications , Hypoxia/drug therapy , Anti-Inflammatory Agents , Inflammation/drug therapy , Antigens, Neoplasm
16.
Physiol Rep ; 11(8): e15669, 2023 04.
Article in English | MEDLINE | ID: mdl-37078457

ABSTRACT

Aspirin is one of the most commonly consumed cyclooxygenase (COX)-inhibitors and anti-inflammatory drugs and has been shown to block COX-produced regulators of inflammation and aging skeletal muscle size. We used propensity score matching to compare skeletal muscle characteristics of individuals from the Health ABC study that did not consume aspirin or any other COX-inhibiting drugs (non-consumers, n = 497, 74 ± 3 year, 168 ± 9 cm, 75.1 ± 13.8 kg, 33.1 ± 7.4% body fat, 37% women, 34% black) to those that consumed aspirin daily (and not any other COX-inhibiting drugs) and for at least 1 year (aspirin consumers, n = 515, 74 ± 3 year, 168 ± 9 cm, 76.2 ± 13.6 kg, 33.8 ± 7.1% body fat, 39% women, 30% black, average aspirin consumption: 6 year). Subjects were matched (p > 0.05) based on age, height, weight, % body fat, sex, and race (propensity scores: 0.33 ± 0.09 vs. 0.33 ± 0.09, p > 0.05). There was no difference between non-consumers and aspirin consumers for computed tomography-determined muscle size of the quadriceps (103.5 ± 0.9 vs. 104.9 ± 0.8 cm2 , p > 0.05) or hamstrings (54.6 ± 0.5 vs. 54.9 ± 0.5 cm2 , p > 0.05), or quadriceps muscle strength (111.1 ± 2.0 vs. 111.7 ± 2.0 Nm, p > 0.05). However, muscle attenuation (i.e., density) was higher in the aspirin consumers in the quadriceps (40.9 ± 0.3 vs. 44.4 ± 0.3 Hounsfield unit [HU], p < 0.05) and hamstrings (27.7 ± 0.4 vs. 33.2 ± 0.4 HU, p < 0.05). These cross sectional data suggest that chronic aspirin consumption does not influence age-related skeletal muscle atrophy, but does influence skeletal muscle composition in septuagenarians. Prospective longitudinal investigations remain necessary to better understand the influence of chronic COX regulation on aging skeletal muscle health.


Subject(s)
Aspirin , Muscle, Skeletal , Humans , Female , Male , Aspirin/pharmacology , Cross-Sectional Studies , Prospective Studies , Muscle, Skeletal/physiology , Aging/physiology , Cyclooxygenase Inhibitors/pharmacology
17.
Ann Pharm Fr ; 81(5): 801-813, 2023 Sep.
Article in English | MEDLINE | ID: mdl-36931432

ABSTRACT

BACKGROUND: An array of commercially viable intermediate molecules necessary for the synthesis of a variety of bioactive molecules are chemically synthesized by pyrrolidine and its derivatives, which play a significant role in drug design and development process. AIM: The aim of the present research work was to explore the synthesis of some new pyrrolidine derivatives and to perform their in silico studies and finally evaluation of analgesic and anti-inflammatory activity. OBJECTIVE: The purpose of this study was to synthesis new pyrrolidine derivatives, examine how they affected the COX-1 and COX-2 enzymes computationally, and to screen their in vivo analgesic and anti-inflammatory activity on laboratory animals. METHOD: The new pyrrolidine derivatives were synthesized by condensing N-(3-acetylphenyl)-2-(pyrrolidin-1-yl)acetamide with substituted aniline in ethanol in the presence of catalytic amounts of glacial acetic acid. The structures of novel pyrrolidine derivatives were characterised using IR, NMR, and mass spectroscopy. Several molecular properties of the newly synthesized derivatives were calculated in order to evaluate the nature of the drug-like candidate. A specific reference cyclooxygenase-1 (COX-1) and cyclooxygenase-2 (COX-2) enzyme was used to dock the newly synthesized pyrrolidine derivatives. RESULTS: From the observed data, it was noted that amongst all newly synthesized compounds, A-1 and A-4 exhibited the highest anti-inflammatory and analgesic effects, respectively. CONCLUSION: On the basis of findings of present research, it was concluded that A-1 and A-4 might be utilized as a promising new lead compound for Non-Steroidal Anti-Inflammatory Drug (NSAIDs) development.


Subject(s)
Analgesics , Anti-Inflammatory Agents, Non-Steroidal , Pyrrolidines , Computer Simulation , Pyrrolidines/chemical synthesis , Pyrrolidines/pharmacology , Pyrrolidines/toxicity , Analgesics/chemical synthesis , Analgesics/pharmacology , Analgesics/toxicity , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/toxicity , Male , Female , Animals , Rats , Cyclooxygenase Inhibitors/chemical synthesis , Cyclooxygenase Inhibitors/pharmacology , Cyclooxygenase Inhibitors/toxicity
18.
Molecules ; 28(3)2023 Jan 20.
Article in English | MEDLINE | ID: mdl-36770730

ABSTRACT

Multi-target drugs (MTDs) are emerging alternatives to combination therapies. Since both histone deacetylases (HDACs) and cyclooxygenase-2 (COX-2) are known to be overexpressed in several cancer types, we herein report the design, synthesis, and biological evaluation of a library of dual HDAC-COX inhibitors. The designed compounds were synthesized via an efficient parallel synthesis approach using preloaded solid-phase resins. Biological in vitro assays demonstrated that several of the synthesized compounds possess pronounced inhibitory activities against HDAC and COX isoforms. The membrane permeability and inhibition of cellular HDAC activity of selected compounds were confirmed by whole-cell HDAC inhibition assays and immunoblot experiments. The most promising dual inhibitors, C3 and C4, evoked antiproliferative effects in the low micromolar concentration range and caused a significant increase in apoptotic cells. In contrast to previous reports, the simultaneous inhibition of HDAC and COX activity by dual HDAC-COX inhibitors or combination treatments with vorinostat and celecoxib did not result in additive or synergistic anticancer activities.


Subject(s)
Antineoplastic Agents , Histone Deacetylase Inhibitors , Histone Deacetylase Inhibitors/pharmacology , Cyclooxygenase 2 , Cell Proliferation , Histone Deacetylases , Cyclooxygenase Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Screening Assays, Antitumor
19.
Pharmacol Rep ; 75(2): 376-385, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36788192

ABSTRACT

BACKGROUND: Kidney diseases have become a global health problem, affecting about 15% of adults and being often under-recognized. Immunological system activation was shown to accelerate kidney damage even in inherited disorders. The kynurenine pathway is the main route of tryptophan degradation. A metabolite of kynurenine (KYN), kynurenic acid (KYNA), produced by kynurenine aminotransferases (KATs), was reported to affect fluid and electrolyte balance as a result of natriuresis induction. The accumulation of KYNA was shown in patients with impaired kidney function and its level was related to the degree of kidney damage. Cyclooxygenase (COX) inhibitors are well-known analgesics and most of them demonstrate an anti-inflammatory effect. Their main mechanism of action is prostaglandin synthesis blockade, which is also responsible for their nephrotoxic potential. Since the KYN pathway is known to remain under immunological system control, the purpose of this study was to analyze the effect of 9 COX inhibitors on KYNA production together with KATs' activity in rat kidneys in vitro. METHODS: Experiments were carried out on kidney homogenates in the presence of L-KYN and the selected compound in 6 various concentrations. RESULTS: Among the examined COX inhibitors only acetaminophen did not change KYNA production in rat kidneys in vitro. Additionally, acetaminophen did not affect the activity of KAT I and KAT II, whereas acetylsalicylic acid and ibuprofen inhibited only KAT II. The remaining COX inhibitors decreased the activity of both KATs in rat kidneys in vitro. CONCLUSION: Our study provides novel mechanisms of COX inhibitors action in the kidney, with possible implications for the treatment of kidney diseases.


Subject(s)
Kynurenic Acid , Kynurenine , Rats , Animals , Kynurenine/metabolism , Kynurenic Acid/metabolism , Cyclooxygenase Inhibitors/pharmacology , Acetaminophen/pharmacology , Transaminases/metabolism , Kidney
20.
Molecules ; 28(1)2023 Jan 02.
Article in English | MEDLINE | ID: mdl-36615565

ABSTRACT

Curcumin is a natural ingredient with antioxidant effects, widely studied as a treatment for various types of cancer. However, its effects on ultraviolet radiation have not been fully explored. The effects of single or daily application of 0.1-100 µM curcumin on cell apoptosis in ultraviolet B (UVB)-induced mice were tested using an experimental double-blind posttest design with a control group and two research models: a single application of curcumin before a single UVB exposure and daily application of curcumin for 7 days before a single UVB exposure on the seventh day. Apoptotic cells were counted using a tunnel system kit. The number of apoptotic cells under a single or daily application of curcumin for 7 days was significantly lower than that of the UVB controls (p ≤ 0.05). The number of apoptotic cells decreased with the increasing concentration of curcumin, and the maximum effect was observed at 100 µM. Daily application of topical curcumin was superior in preventing apoptosis (mean apoptotic cell count of 14.86 ± 1.68) compared with a single application (17.46 ± 0.60; p = 0.011). Topical curcumin can act as a potential photoprotective agent in preventing cutaneous malignancies due to UVB radiation. Further studies are warranted, especially in humans.


Subject(s)
Curcumin , Skin Neoplasms , Humans , Mice , Animals , Curcumin/pharmacology , Ultraviolet Rays/adverse effects , Apoptosis , Skin Neoplasms/prevention & control , Antioxidants/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Skin
SELECTION OF CITATIONS
SEARCH DETAIL
...