Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.632
Filtrer
1.
Nat Commun ; 15(1): 6538, 2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39095358

RÉSUMÉ

Muscle invasive bladder cancers (BCs) can be divided into 2 major subgroups-basal/squamous (BASQ) tumors and luminal tumors. Since Pparg has low or undetectable expression in BASQ tumors, we tested the effects of rosiglitazone, Pparg agonist, in a mouse model of BASQ BC. We find that rosiglitazone reduces proliferation while treatment with rosiglitazone plus trametinib, a MEK inhibitor, induces apoptosis and reduces tumor volume by 91% after 1 month. Rosiglitazone and trametinib also induce a shift from BASQ to luminal differentiation in tumors, which our analysis suggests is mediated by retinoid signaling, a pathway known to drive the luminal differentiation program. Our data suggest that rosiglitazone, trametinib, and retinoids, which are all FDA approved, may be clinically active in BASQ tumors in patients.


Sujet(s)
Apoptose , Prolifération cellulaire , Modèles animaux de maladie humaine , Pyridones , Pyrimidinones , Rosiglitazone , Tumeurs de la vessie urinaire , Animaux , Tumeurs de la vessie urinaire/traitement médicamenteux , Tumeurs de la vessie urinaire/anatomopathologie , Tumeurs de la vessie urinaire/génétique , Pyridones/pharmacologie , Pyridones/usage thérapeutique , Pyrimidinones/pharmacologie , Pyrimidinones/usage thérapeutique , Rosiglitazone/pharmacologie , Rosiglitazone/usage thérapeutique , Souris , Apoptose/effets des médicaments et des substances chimiques , Humains , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Invasion tumorale , Femelle , Récepteur PPAR gamma/métabolisme , Récepteur PPAR gamma/agonistes , Thiazolidinediones/pharmacologie , Thiazolidinediones/usage thérapeutique , Différenciation cellulaire/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Rétinoïdes/pharmacologie , Rétinoïdes/usage thérapeutique
2.
Sci Adv ; 10(31): eadn7979, 2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39093975

RÉSUMÉ

We have developed GmPcides from a peptidomimetic dihydrothiazolo ring-fused 2-pyridone scaffold that has antimicrobial activities against a broad spectrum of Gram-positive pathogens. Here, we examine the treatment efficacy of GmPcides using skin and soft tissue infection (SSTI) and biofilm formation models by Streptococcus pyogenes. Screening our compound library for minimal inhibitory (MIC) and minimal bactericidal (MBC) concentrations identified GmPcide PS757 as highly active against S. pyogenes. Treatment of S. pyogenes biofilm with PS757 revealed robust efficacy against all phases of biofilm formation by preventing initial biofilm development, ceasing biofilm maturation and eradicating mature biofilm. In a murine model of S. pyogenes SSTI, subcutaneous delivery of PS757 resulted in reduced levels of tissue damage, decreased bacterial burdens, and accelerated rates of wound healing, which were associated with down-regulation of key virulence factors, including M protein and the SpeB cysteine protease. These data demonstrate that GmPcides show considerable promise for treating S. pyogenes infections.


Sujet(s)
Biofilms , Tests de sensibilité microbienne , Pyridones , Infections des tissus mous , Infections à streptocoques , Streptococcus pyogenes , Streptococcus pyogenes/effets des médicaments et des substances chimiques , Animaux , Infections des tissus mous/traitement médicamenteux , Infections des tissus mous/microbiologie , Biofilms/effets des médicaments et des substances chimiques , Infections à streptocoques/traitement médicamenteux , Infections à streptocoques/microbiologie , Souris , Pyridones/pharmacologie , Pyridones/composition chimique , Antibactériens/pharmacologie , Antibactériens/composition chimique , Modèles animaux de maladie humaine , Thiazoles/pharmacologie , Thiazoles/composition chimique , Dermatoses bactériennes/traitement médicamenteux , Dermatoses bactériennes/microbiologie , Femelle , Cicatrisation de plaie/effets des médicaments et des substances chimiques , Humains
3.
Int J Mol Sci ; 25(13)2024 Jun 29.
Article de Anglais | MEDLINE | ID: mdl-39000295

RÉSUMÉ

Olprinone (OLP) is a selective inhibitor of phosphodiesterase III and is used clinically in patients with heart failure and those undergoing cardiac surgery; however, little is known about the effects of OLP on hepatoprotection. The purpose of this study aimed to determine whether OLP has protective effects in in vivo and in vitro rat models of endotoxin-induced liver injury after hepatectomy and to clarify the mechanisms of action of OLP. In the in vivo model, rats underwent 70% partial hepatectomy and lipopolysaccharide treatment (PH/LPS). OLP administration increased survival by 85.7% and decreased tumor necrosis factor-α, C-X-C motif chemokine ligand 1, and inducible nitric oxide synthase (iNOS) mRNA expression in the livers of rats treated with PH/LPS. OLP also suppressed nuclear translocation and/or DNA binding ability of nuclear factor kappa B (NF-κB). Pathological liver damage induced by PH/LPS was alleviated and neutrophil infiltration was reduced by OLP. Primary cultured rat hepatocytes treated with the pro-inflammatory cytokine interleukin-1ß (IL-1ß) were used as a model of in vitro liver injury. Co-treatment with OLP inhibited dose-dependently IL-1ß-stimulated iNOS induction and NF-κB activation. Our results demonstrate that OLP may partially inhibit the induction of several inflammatory mediators through the suppression of NF-κB and thus prevent liver injury induced by endotoxin after liver resection.


Sujet(s)
Modèles animaux de maladie humaine , Hépatectomie , Hépatocytes , Imidazoles , Facteur de transcription NF-kappa B , Nitric oxide synthase type II , Pyridones , Animaux , Hépatectomie/effets indésirables , Hépatocytes/effets des médicaments et des substances chimiques , Hépatocytes/métabolisme , Rats , Mâle , Pyridones/pharmacologie , Pyridones/usage thérapeutique , Facteur de transcription NF-kappa B/métabolisme , Imidazoles/pharmacologie , Nitric oxide synthase type II/métabolisme , Inhibiteurs de la phosphodiestérase-3/pharmacologie , Inhibiteurs de la phosphodiestérase-3/usage thérapeutique , Interleukine-1 bêta/métabolisme , Lipopolysaccharides/effets indésirables , Lipopolysaccharides/toxicité , Sepsie/traitement médicamenteux , Rat Sprague-Dawley , Cellules cultivées , Facteur de nécrose tumorale alpha/métabolisme , Chimiokine CXCL1/métabolisme , Foie/effets des médicaments et des substances chimiques , Foie/anatomopathologie , Foie/métabolisme
4.
Int J Mol Sci ; 25(13)2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-39000409

RÉSUMÉ

Cardiac fibrosis is a severe outcome of Chagas disease (CD), caused by the protozoan Trypanosoma cruzi. Clinical evidence revealed a correlation between fibrosis levels with impaired cardiac performance in CD patients. Therefore, we sought to analyze the effect of inhibitors of TGF-ß (pirfenidone), p38-MAPK (losmapimod) and c-Jun (SP600125) on the modulation of collagen deposition in cardiac fibroblasts (CF) and in vivo models of T. cruzi chronic infection. Sirius Red/Fast Green dye was used to quantify both collagen expression and total protein amount, assessing cytotoxicity. The compounds were also used to treat C57/Bl6 mice chronically infected with T. cruzi, Brazil strain. We identified an anti-fibrotic effect in vitro for pirfenidone (TGF-ß inhibitor, IC50 114.3 µM), losmapimod (p38 inhibitor, IC50 17.6 µM) and SP600125 (c-Jun inhibitor, IC50 3.9 µM). This effect was independent of CF proliferation since these compounds do not affect T. cruzi-induced host cell multiplication as measured by BrdU incorporation. Assays of chronic infection of mice with T. cruzi have shown a reduction in heart collagen by pirfenidone. These results propose a novel approach to fibrosis therapy in CD, with the prospect of repurposing pirfenidone to prevent the onset of ECM accumulation in the hearts of the patients.


Sujet(s)
Cardiomyopathie associée à la maladie de Chagas , Fibrose , Souris de lignée C57BL , Pyridones , Animaux , Pyridones/pharmacologie , Pyridones/usage thérapeutique , Cardiomyopathie associée à la maladie de Chagas/traitement médicamenteux , Cardiomyopathie associée à la maladie de Chagas/parasitologie , Cardiomyopathie associée à la maladie de Chagas/métabolisme , Cardiomyopathie associée à la maladie de Chagas/anatomopathologie , Souris , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Fibroblastes/parasitologie , Myocarde/anatomopathologie , Myocarde/métabolisme , Collagène/métabolisme , Trypanosoma cruzi/effets des médicaments et des substances chimiques , Humains , Maladie chronique , Facteur de croissance transformant bêta/métabolisme , Modèles animaux de maladie humaine , p38 Mitogen-Activated Protein Kinases/métabolisme , Mâle , Anthracènes
5.
Int J Mol Sci ; 25(13)2024 Jul 06.
Article de Anglais | MEDLINE | ID: mdl-39000539

RÉSUMÉ

Isocitrate dehydrogenase 1 (IDH1) is a necessary enzyme for cellular respiration in the tricarboxylic acid cycle. Mutant isocitrate dehydrogenase 1 (mIDH1) has been detected overexpressed in a variety of cancers. mIDH1 inhibitor ivosidenib (AG-120) was only approved by the Food and Drug Administration (FDA) for marketing, nevertheless, a range of resistance has been frequently reported. In this study, several mIDH1 inhibitors with the common backbone pyridin-2-one were explored using the three-dimensional structure-activity relationship (3D-QSAR), scaffold hopping, absorption, distribution, metabolism, excretion (ADME) prediction, and molecular dynamics (MD) simulations. Comparative molecular field analysis (CoMFA, R2 = 0.980, Q2 = 0.765) and comparative molecular similarity index analysis (CoMSIA, R2 = 0.997, Q2 = 0.770) were used to build 3D-QSAR models, which yielded notably decent predictive ability. A series of novel structures was designed through scaffold hopping. The predicted pIC50 values of C3, C6, and C9 were higher in the model of 3D-QSAR. Additionally, MD simulations culminated in the identification of potent mIDH1 inhibitors, exhibiting strong binding interactions, while the analyzed parameters were free energy landscape (FEL), radius of gyration (Rg), solvent accessible surface area (SASA), and polar surface area (PSA). Binding free energy demonstrated that C2 exhibited the highest binding free energy with IDH1, which was -93.25 ± 5.20 kcal/mol. This research offers theoretical guidance for the rational design of novel mIDH1 inhibitors.


Sujet(s)
Isocitrate dehydrogenases , Simulation de dynamique moléculaire , Relation quantitative structure-activité , Isocitrate dehydrogenases/antagonistes et inhibiteurs , Isocitrate dehydrogenases/composition chimique , Isocitrate dehydrogenases/métabolisme , Isocitrate dehydrogenases/génétique , Humains , Antienzymes/composition chimique , Antienzymes/pharmacologie , Pyridones/composition chimique , Pyridones/pharmacologie
6.
J Agric Food Chem ; 72(28): 15512-15522, 2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-38959331

RÉSUMÉ

Root-knot nematodes pose a serious threat to crops by affecting production and quality. Over a period of time, substantial work has been done toward the development of effective and environmentally benign nematicidal compounds. However, due to the inefficiencies of previously reported synthetics in achieving the target of safe, selective, and effective treatment, it is necessary to develop new efficacious and safer nematicidal agents considering human health and environment on top priority. This work aims to highlight the efficient and convenient l-proline catalyzed synthesis of pyrano[3,2-c]pyridone and their use as potential nematicidal agents. In vitro results of larval mortality and egg hatching inhibition revealed maximum nematicidal activity against Meloidogyne incognita from compounds 15b, 15m, and 15w with LC50 values of 28.8, 46.8, and 49.18 µg/mL at 48 h, respectively. Under similar conditions, pyrano[3,2-c]pyridones derivatives 15b (LC50 = 28.8 µg/mL) was found at par with LC50 (26.92 µg/mL) of commercial nematicide carbofuran. The in vitro results were further validated with in silico studies with the most active compound 15b nematicidal within the binding to the pocket of acetylcholine esterase (AChE). In docking, binding free energy values for compound 15b were found to be -6.90 kcal/mol. Results indicated that pyrano[3,2-c]pyridone derivatives have the potential to control M. incognita.


Sujet(s)
Antihelminthiques antinématodes , Conception de médicament , Simulation de docking moléculaire , Pyridones , Tylenchoidea , Tylenchoidea/effets des médicaments et des substances chimiques , Animaux , Antihelminthiques antinématodes/pharmacologie , Antihelminthiques antinématodes/composition chimique , Antihelminthiques antinématodes/synthèse chimique , Pyridones/composition chimique , Pyridones/pharmacologie , Pyridones/synthèse chimique , Relation structure-activité , Larve/effets des médicaments et des substances chimiques , Larve/croissance et développement , Maladies des plantes/parasitologie , Structure moléculaire
7.
Anal Cell Pathol (Amst) ; 2024: 2751280, 2024.
Article de Anglais | MEDLINE | ID: mdl-38946862

RÉSUMÉ

Background: Biliary atresia (BA) is a devastating congenital disease characterized by inflammation and progressive liver fibrosis. Activation of hepatic stellate cells (HSCs) plays a central role in the pathogenesis of hepatic fibrosis. Our study aimed to investigate the pharmacological effect and potential mechanism of pirfenidone (PFD) and andrographolide (AGP) separately and together on liver fibrosis of BA. Materials and Methods: The bile ducts of male C57BL/6J mice were ligated or had the sham operation. The in vivo effects of PFD and/or AGP on liver fibrosis of BA were evaluated. Human hepatic stellate cells (LX-2) were also treated with PFD and/or AGP in vitro. Results: PFD and/or AGP ameliorates liver fibrosis and inflammation in the mice model of BA, as evidenced by significant downregulated in the accumulation of collagen fibers, hepatic fibrosis markers (α-SMA, collagen I, and collagen IV), and inflammatory markers (IL-1ß, IL-6, and TNF-α). Moreover, compared with monotherapy, these changes are more obvious in the combined treatment of PFD and AGP. Consistent with animal experiments, hepatic fibrosis markers (α-SMA, collagen I, and CTGF) and inflammatory markers (IL-1ß, IL-6, and TNF-α) were significantly decreased in activated LX-2 cells after PFD and/or AGP treatment. In addition, PFD and/or AGP inhibited the activation of HSCs by blocking the TGF-ß/Smad signaling pathway, and the combined treatment of PFD and AGP synergistically inhibited the phosphorylation of Smad2 and Smad3. Conclusion: The combined application of PFD and AGP exerted superior inhibitive effects on HSC activation and liver fibrosis by mediating the TGF-ß/Smad signaling pathway as compared to monotherapy. Therefore, the combination of PFD and AGP may be a promising treatment strategy for liver fibrosis in BA.


Sujet(s)
Diterpènes , Cellules étoilées du foie , Cirrhose du foie , Souris de lignée C57BL , Pyridones , Transduction du signal , Protéines Smad , Facteur de croissance transformant bêta , Cellules étoilées du foie/effets des médicaments et des substances chimiques , Cellules étoilées du foie/métabolisme , Cellules étoilées du foie/anatomopathologie , Animaux , Cirrhose du foie/anatomopathologie , Cirrhose du foie/traitement médicamenteux , Cirrhose du foie/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Diterpènes/pharmacologie , Diterpènes/usage thérapeutique , Mâle , Facteur de croissance transformant bêta/métabolisme , Protéines Smad/métabolisme , Humains , Pyridones/pharmacologie , Lignée cellulaire , Souris , Atrésie des voies biliaires/anatomopathologie , Atrésie des voies biliaires/traitement médicamenteux , Atrésie des voies biliaires/métabolisme , Modèles animaux de maladie humaine , Association de médicaments
8.
Oncol Res ; 32(7): 1197-1207, 2024.
Article de Anglais | MEDLINE | ID: mdl-38948022

RÉSUMÉ

Breast cancer, a predominant global health issue, requires ongoing exploration of new therapeutic strategies. Palbociclib (PAL), a well-known cyclin-dependent kinase (CDK) inhibitor, plays a critical role in breast cancer treatment. While its efficacy is recognized, the interplay between PAL and cellular autophagy, particularly in the context of the RAF/MEK/ERK signaling pathway, remains insufficiently explored. This study investigates PAL's inhibitory effects on breast cancer using both in vitro (MCF7 and MDA-MB-468 cells) and in vivo (tumor-bearing nude mice) models. Aimed at elucidating the impact of PAL on autophagic processes and exploring the potential of combining it with trametinib (TRA), an MEK inhibitor, our research seeks to address the challenge of PAL-induced drug resistance. Our findings reveal that PAL significantly decreases the viability of MCF7 and MDA-MB-468 cells and reduces tumor size in mice while showing minimal cytotoxicity in MCF10A cells. However, PAL also induces protective autophagy, potentially leading to drug resistance via the RAF/MEK/ERK pathway activation. Introducing TRA effectively neutralized this autophagy, enhancing PAL's anti-tumor efficacy. A combination of PAL and TRA synergistically reduced cell viability and proliferation, and in vivo studies showed notable tumor size reduction. In conclusion, the PAL and TRA combination emerges as a promising strategy for overcoming PAL-induced resistance, offering a new horizon in breast cancer treatment.


Sujet(s)
Autophagie , Tumeurs du sein , Pipérazines , Pyridines , Pyridones , Pyrimidinones , Tests d'activité antitumorale sur modèle de xénogreffe , Humains , Animaux , Autophagie/effets des médicaments et des substances chimiques , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Pyridines/pharmacologie , Pyridines/usage thérapeutique , Pyridones/pharmacologie , Pyridones/usage thérapeutique , Femelle , Pyrimidinones/pharmacologie , Pyrimidinones/usage thérapeutique , Souris , Pipérazines/pharmacologie , Pipérazines/usage thérapeutique , Lignée cellulaire tumorale , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Synergie des médicaments , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Souris nude , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Survie cellulaire/effets des médicaments et des substances chimiques , Cellules MCF-7
9.
Chem Biol Interact ; 399: 111146, 2024 Aug 25.
Article de Anglais | MEDLINE | ID: mdl-39002878

RÉSUMÉ

Apixaban is an oral anticoagulant that directly inhibits the target Factor Xa (FXa). In this study, we focused on the in vivo and in vitro effects of adagrasib and asciminib on apixaban metabolism, to discover potential drug-drug interactions (DDI) and explore their inhibitory mechanisms. The levels of apixaban and its metabolite, O-desmethyl-apixaban (M2), were determined by ultra performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS). In vitro evaluation, the maximum half inhibitory concentration (IC50) of adagrasib in rat liver microsomes (RLM) and human liver microsomes (HLM) against apixaban was 7.99 µM and 117.40 µM, respectively. The IC50 value of asciminib against apixaban in RLM and HLM was 4.28 µM and 18.42 µM, respectively. The results of the analysis on inhibition mechanisms showed that adagrasib inhibited the metabolism of apixaban through a non-competitive mechanism, while asciminib inhibited the metabolism of apixaban through a mixed mechanism. Moreover, the interaction of apixaban with adagrasib and asciminib in Sprague-Dawley (SD) rats was also investigated. It was found that the pharmacokinetic characteristics of apixaban were significantly changed when combined with these two antitumor drugs, where AUC(0-t), AUC(0-∞), t1/2, Tmax, and Cmax were increased, while CLz/F was significantly decreased. But both drugs did not appear to affect the metabolism of M2 in a significant way. Consistent results from in vitro and in vivo demonstrated that both adagrasib and asciminib inhibited the metabolism of apixaban. It provided reference data for the future clinical individualization of apixaban.


Sujet(s)
Antinéoplasiques , Microsomes du foie , Pyrazoles , Pyridones , Rat Sprague-Dawley , Animaux , Pyrazoles/pharmacologie , Pyrazoles/métabolisme , Pyridones/pharmacologie , Pyridones/pharmacocinétique , Humains , Microsomes du foie/métabolisme , Rats , Mâle , Antinéoplasiques/pharmacologie , Interactions médicamenteuses , Spectrométrie de masse en tandem , Inhibiteurs du facteur Xa/pharmacologie , Inhibiteurs du facteur Xa/pharmacocinétique , Phénylacétates , Thiophènes
10.
J Nat Prod ; 87(7): 1746-1753, 2024 Jul 26.
Article de Anglais | MEDLINE | ID: mdl-38958274

RÉSUMÉ

Mycoplasma genitalium is a sexually transmitted bacterium associated with urogenital disease syndromes in the US and worldwide. The global rise in drug resistance in M. genitalium necessitates the development of novel drugs to treat this pathogen. To address this need, we have screened extracts from a library of fungal isolates assembled through the University of Oklahoma Citizen Science Soil Collection Program. Analysis of one of the bioactive extracts using bioassay-guided fractionation led to the purification of the compound PF1140 (1) along with a new and several other known pyridones. The N-hydroxy pyridones are generally regarded as siderophores with high binding affinity for iron(III) under physiological conditions. Results from UV-vis absorption spectroscopy-based titration experiments revealed that 1 complexes with Fe3+. As M. genitalium does not utilize iron, we propose that the PF1140-iron complex induces cytotoxicity by facilitating the cellular uptake of iron, which reacts with endogenous hydrogen peroxide to produce toxic hydroxyl radicals.


Sujet(s)
Fer , Mycoplasma genitalium , Mycoplasma genitalium/métabolisme , Mycoplasma genitalium/effets des médicaments et des substances chimiques , Fer/métabolisme , Structure moléculaire , Tests de sensibilité microbienne , Antibactériens/pharmacologie , Antibactériens/composition chimique , Pyridones/pharmacologie , Pyridones/composition chimique , Champignons/composition chimique , Champignons/métabolisme , Composés du fer III/composition chimique , Peroxyde d'hydrogène , Sidérophores/pharmacologie , Sidérophores/composition chimique
11.
J Med Chem ; 67(14): 12366-12385, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39007759

RÉSUMÉ

The dominant role of non-homologous end-joining in the repair of radiation-induced double-strand breaks identifies DNA-dependent protein kinase (DNA-PK) as an excellent target for the development of radiosensitizers. We report the discovery of a new class of imidazo[4,5-c]pyridine-2-one DNA-PK inhibitors. Structure-activity studies culminated in the identification of 78 as a nM DNA-PK inhibitor with excellent selectivity for DNA-PK compared to related phosphoinositide 3-kinase (PI3K) and PI3K-like kinase (PIKK) families and the broader kinome, and displayed DNA-PK-dependent radiosensitization of HAP1 cells. Compound 78 demonstrated robust radiosensitization of a broad range of cancer cells in vitro, displayed high oral bioavailability, and sensitized colorectal carcinoma (HCT116/54C) and head and neck squamous cell carcinoma (UT-SCC-74B) tumor xenografts to radiation. Compound 78 also provided substantial tumor growth inhibition of HCT116/54C tumor xenografts in combination with radiation. Compound 78 represents a new, potent, and selective class of DNA-PK inhibitors with significant potential as radiosensitizers for cancer treatment.


Sujet(s)
DNA-activated protein kinase , Inhibiteurs de protéines kinases , Radiosensibilisants , Humains , DNA-activated protein kinase/antagonistes et inhibiteurs , DNA-activated protein kinase/métabolisme , Animaux , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/composition chimique , Inhibiteurs de protéines kinases/pharmacocinétique , Inhibiteurs de protéines kinases/synthèse chimique , Radiosensibilisants/pharmacologie , Radiosensibilisants/composition chimique , Radiosensibilisants/pharmacocinétique , Radiosensibilisants/synthèse chimique , Relation structure-activité , Souris , Lignée cellulaire tumorale , Imidazoles/pharmacologie , Imidazoles/composition chimique , Imidazoles/synthèse chimique , Imidazoles/pharmacocinétique , Pyridones/pharmacologie , Pyridones/composition chimique , Pyridones/synthèse chimique , Pyridones/pharmacocinétique , Tests d'activité antitumorale sur modèle de xénogreffe , Souris nude , Rats
12.
Int J Nanomedicine ; 19: 5681-5703, 2024.
Article de Anglais | MEDLINE | ID: mdl-38882541

RÉSUMÉ

Introduction: Diabetes mellitus is frequently associated with foot ulcers, which pose significant health risks and complications. Impaired wound healing in diabetic patients is attributed to multiple factors, including hyperglycemia, neuropathy, chronic inflammation, oxidative damage, and decreased vascularization. Rationale: To address these challenges, this project aims to develop bioactive, fast-dissolving nanofiber dressings composed of polyvinylpyrrolidone loaded with a combination of an antibiotic (moxifloxacin or fusidic acid) and anti-inflammatory drug (pirfenidone) using electrospinning technique to prevent the bacterial growth, reduce inflammation, and expedite wound healing in diabetic wounds. Results: The fabricated drug-loaded fibers exhibited diameters of 443 ± 67 nm for moxifloxacin/pirfenidone nanofibers and 488 ± 92 nm for fusidic acid/pirfenidone nanofibers. The encapsulation efficiency, drug loading and drug release studies for the moxifloxacin/pirfenidone nanofibers were found to be 70 ± 3% and 20 ± 1 µg/mg, respectively, for moxifloxacin, and 96 ± 6% and 28 ± 2 µg/mg, respectively, for pirfenidone, with a complete release of both drugs within 24 hours, whereas the fusidic acid/pirfenidone nanofibers were found to be 95 ± 6% and 28 ± 2 µg/mg, respectively, for fusidic acid and 102 ± 5% and 30 ± 2 µg/mg, respectively, for pirfenidone, with a release rate of 66% for fusidic acid and 80%, for pirfenidone after 24 hours. The efficacy of the prepared nanofiber formulations in accelerating wound healing was evaluated using an induced diabetic rat model. All tested formulations showed an earlier complete closure of the wound compared to the controls, which was also supported by the histopathological assessment. Notably, the combination of fusidic acid and pirfenidone nanofibers demonstrated wound healing acceleration on day 8, earlier than all tested groups. Conclusion: These findings highlight the potential of the drug-loaded nanofibrous system as a promising medicated wound dressing for diabetic foot applications.


Sujet(s)
Antibactériens , Bandages , Pied diabétique , Libération de médicament , Acide fusidique , Moxifloxacine , Nanofibres , Pyridones , Cicatrisation de plaie , Pied diabétique/traitement médicamenteux , Pied diabétique/thérapie , Nanofibres/composition chimique , Animaux , Moxifloxacine/administration et posologie , Moxifloxacine/pharmacologie , Moxifloxacine/composition chimique , Moxifloxacine/pharmacocinétique , Cicatrisation de plaie/effets des médicaments et des substances chimiques , Antibactériens/composition chimique , Antibactériens/pharmacologie , Antibactériens/administration et posologie , Antibactériens/pharmacocinétique , Pyridones/composition chimique , Pyridones/pharmacologie , Pyridones/pharmacocinétique , Pyridones/administration et posologie , Acide fusidique/administration et posologie , Acide fusidique/pharmacologie , Acide fusidique/composition chimique , Acide fusidique/pharmacocinétique , Rats , Mâle , Diabète expérimental , Povidone/composition chimique , Rat Sprague-Dawley
13.
Nat Commun ; 15(1): 4893, 2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38849340

RÉSUMÉ

Amyotrophic lateral sclerosis (ALS) is a debilitating motor neuron disease and lacks effective disease-modifying treatments. This study utilizes a comprehensive multiomic approach to investigate the early and sex-specific molecular mechanisms underlying ALS. By analyzing the prefrontal cortex of 51 patients with sporadic ALS and 50 control subjects, alongside four transgenic mouse models (C9orf72-, SOD1-, TDP-43-, and FUS-ALS), we have uncovered significant molecular alterations associated with the disease. Here, we show that males exhibit more pronounced changes in molecular pathways compared to females. Our integrated analysis of transcriptomes, (phospho)proteomes, and miRNAomes also identified distinct ALS subclusters in humans, characterized by variations in immune response, extracellular matrix composition, mitochondrial function, and RNA processing. The molecular signatures of human subclusters were reflected in specific mouse models. Our study highlighted the mitogen-activated protein kinase (MAPK) pathway as an early disease mechanism. We further demonstrate that trametinib, a MAPK inhibitor, has potential therapeutic benefits in vitro and in vivo, particularly in females, suggesting a direction for developing targeted ALS treatments.


Sujet(s)
Sclérose latérale amyotrophique , Modèles animaux de maladie humaine , Système de signalisation des MAP kinases , Souris transgéniques , Sclérose latérale amyotrophique/génétique , Sclérose latérale amyotrophique/traitement médicamenteux , Sclérose latérale amyotrophique/métabolisme , Humains , Femelle , Animaux , Mâle , Souris , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Pyridones/pharmacologie , Pyridones/usage thérapeutique , Protéine FUS de liaison à l'ARN/métabolisme , Protéine FUS de liaison à l'ARN/génétique , Cortex préfrontal/métabolisme , Transcriptome , Superoxide dismutase-1/génétique , Superoxide dismutase-1/métabolisme , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Adulte d'âge moyen , microARN/génétique , microARN/métabolisme , Protéine C9orf72/génétique , Protéine C9orf72/métabolisme , Caractères sexuels , Sujet âgé , Facteurs sexuels , Pyrimidinones
14.
Biomed Pharmacother ; 176: 116896, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38876049

RÉSUMÉ

Idiopathic pulmonary fibrosis (IPF) is a severe disability due to progressive lung dysfunction. IPF has long been viewed as a non-immune form of pulmonary fibrosis, but nowadays it is accepted that a chronic inflammatory response can exacerbate fibrotic patterns. IL-1-like cytokines and ATP are highly detected in the lung and broncho-alveolar lavage fluid of IPF patients. Because ATP binds the purinergic receptor P2RX7 involved in the release of IL-1-like cytokines, we aimed to understand the role of P2RX7 in IPF. PBMCs from IPF patients were treated with nintedanib or pirfenidone in the presence of ATP. Under these conditions, PBMCs still released IL-1-like cytokines and the pro-fibrotic TGFß. Bulk and scRNAseq demonstrated that lung tissues of IPF patients had higher levels of P2RX7, especially on macrophages, which were correlated to T cell activity and inflammatory response with a TGFBI and IL-10 signature. A subcluster of macrophages in IPF lung tissues had 2055 genes that were not in common with the other subclusters, and that were involved in metabolic and PDGF, FGF and VEGF associated pathways. These data confirmed what observed on circulating cells that, although treated with anti-fibrotic agents, nintedanib or pirfenidone, they were still able to release IL-1 cytokines and the fibrogenic TGFß. In conclusion, these data imply that because nintedanib and pirfenidone do not block ATP-induced IL-1-like cytokines and TGFß induced during P2RX7 activation, it is plausible to consider P2RX7 on circulating cells and/or tissue biopsies as potential pharmacological tool for IPF patients.


Sujet(s)
Adénosine triphosphate , Fibrose pulmonaire idiopathique , Indoles , Pyridones , Récepteurs purinergiques P2X7 , Humains , Fibrose pulmonaire idiopathique/traitement médicamenteux , Fibrose pulmonaire idiopathique/métabolisme , Fibrose pulmonaire idiopathique/anatomopathologie , Pyridones/pharmacologie , Pyridones/usage thérapeutique , Indoles/pharmacologie , Indoles/usage thérapeutique , Adénosine triphosphate/métabolisme , Récepteurs purinergiques P2X7/métabolisme , Mâle , Poumon/effets des médicaments et des substances chimiques , Poumon/anatomopathologie , Poumon/métabolisme , Femelle , Cytokines/métabolisme , Sujet âgé , Agranulocytes/effets des médicaments et des substances chimiques , Agranulocytes/métabolisme , Adulte d'âge moyen , Facteur de croissance transformant bêta/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques
15.
J Zoo Wildl Med ; 55(2): 313-321, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38875188

RÉSUMÉ

High pathogenicity avian influenza is an acute zoonotic disease with high mortality in birds caused by a high pathogenicity avian influenza virus (HPAIV). Recently, HPAIV has rapidly spread worldwide and has killed many wild birds, including endangered species. Baloxavir marboxil (BXM), an anti-influenza agent used for humans, was reported to reduce mortality and virus secretion from HPAIV-infected chickens (Gallus domesticus, order Galliformes) at a dosage of ≥2.5 mg/kg when administered simultaneously with viral challenge. Application of this treatment to endangered birds requires further information on potential avian-specific toxicity caused by repeated exposure to BXM over the long term. To obtain information of potential avian-specific toxicity, a 4-wk oral repeated-dose study of BXM was conducted in chickens (n = 6 or 7 per group), which are commonly used as laboratory avian species. The study was conducted in reference to the human pharmaceutical guidelines for nonclinical repeated-dose drug toxicity studies to evaluate systemic toxicity and exposure. No adverse changes were observed in any organs examined, and dose proportional increases in systemic exposure to active pharmaceutical ingredients were noted from 12.5 to 62.5 mg/kg per day. BXM showed no toxicity to chickens at doses of up to 62.5 mg/kg per day, at which systemic exposure was approximately 71 times higher than systemic exposure at 2.5 mg/kg, the reported efficacious dosage amount, in HPAIV-infected chickens. These results also suggest that BXM could be considered safe for treating HPAIV-infected endangered birds due to its high safety margin compared with the efficacy dose. The data in this study could contribute to the preservation of endangered birds by using BXM as a means of protecting biodiversity.


Sujet(s)
Antiviraux , Poulets , Dibenzothiépines , Morpholines , Pyridones , Triazines , Animaux , Triazines/administration et posologie , Dibenzothiépines/administration et posologie , Administration par voie orale , Antiviraux/administration et posologie , Antiviraux/pharmacologie , Morpholines/administration et posologie , Morpholines/pharmacologie , Pyridones/administration et posologie , Pyridones/pharmacologie , Pyridines/administration et posologie , Thiépines/administration et posologie , Thiépines/pharmacologie , Mâle , Grippe chez les oiseaux/traitement médicamenteux , Femelle , Oxazines , Hydroxy-butyrates/administration et posologie
16.
Bioorg Med Chem Lett ; 109: 129849, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-38876177

RÉSUMÉ

Clinical studies have shown that inhibitors of bromodomain and extra-terminal domain (BET) proteins, particularly BRD4, have antitumor activity and efficacy. The BET protein has two domains, BD1 and BD2, and we previously focused on BD1 and reported orally bioavailable BD1-selective inhibitors. In this study, we obtained a BD1 inhibitor, a more potent and highly selective pyrazolopyridone derivative 13a, and confirmed its in vivo efficacy.


Sujet(s)
Pyridones , Humains , Administration par voie orale , Relation structure-activité , Animaux , Pyridones/composition chimique , Pyridones/pharmacologie , Pyridones/synthèse chimique , Pyridones/pharmacocinétique , Pyrazoles/composition chimique , Pyrazoles/pharmacologie , Pyrazoles/synthèse chimique , Découverte de médicament , Facteurs de transcription/antagonistes et inhibiteurs , Facteurs de transcription/métabolisme , Structure moléculaire , Protéines du cycle cellulaire/antagonistes et inhibiteurs , Protéines du cycle cellulaire/métabolisme , Souris , Domaines protéiques , Relation dose-effet des médicaments , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Antinéoplasiques/synthèse chimique , Rats , Protéines contenant un bromodomaine
17.
Eur J Pharmacol ; 977: 176721, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-38851561

RÉSUMÉ

Underactive bladder (UAB), characterized by a complex set of symptoms with few treatment options, can significantly reduce the quality of life of affected people. UAB is characterized by hyperplasia and fibrosis of the bladder wall as well as decreased bladder compliance. Pirfenidone is a powerful anti-fibrotic agent that inhibits the progression of fibrosis in people with idiopathic pulmonary fibrosis. In the current study, we evaluated the efficacy of pirfenidone in the treatment of bladder fibrosis in a UAB rat model. UAB was induced by crushing damage to nerve bundles in the major pelvic ganglion. Forty-two days after surgery, 1 mL distilled water containing pirfenidone (100, 300, or 500 mg/kg) was orally administered once every 2 days for a total of 10 times for 20 days to the rats in the pirfenidone-treated groups. Crushing damage to the nerve bundles caused voiding dysfunction, resulting in increased bladder weight and the level of fibrous related factors in the bladder, leading to UAB symptoms. Pirfenidone treatment improved urinary function, increased bladder weight and suppressed the expression of fibrosis factors. The results of this experiment suggest that pirfenidone can be used to ameliorate difficult-to-treat urological conditions such as bladder fibrosis. Therefore, pirfenidone treatment can be considered an option to improve voiding function in patient with incurable UAB.


Sujet(s)
Fibrose , Pyridones , Rat Sprague-Dawley , Hypoactivité vésicale , Vessie urinaire , Miction , Animaux , Pyridones/pharmacologie , Pyridones/usage thérapeutique , Vessie urinaire/effets des médicaments et des substances chimiques , Vessie urinaire/anatomopathologie , Vessie urinaire/physiopathologie , Rats , Miction/effets des médicaments et des substances chimiques , Hypoactivité vésicale/traitement médicamenteux , Hypoactivité vésicale/physiopathologie , Hypoactivité vésicale/étiologie , Modèles animaux de maladie humaine , Femelle , Mâle
18.
Cells ; 13(12)2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38920644

RÉSUMÉ

Hepatocellular carcinoma (HCC) development is associated with altered modifications in DNA methylation, changing transcriptional regulation. Emerging evidence indicates that DNA methyltransferase 1 (DNMT1) plays a key role in the carcinogenesis process. This study aimed to investigate how pirfenidone (PFD) modifies this pathway and the effect generated by the association between c-Myc expression and DNMT1 activation. Rats F344 were used for HCC development using 50 mg/kg of diethylnitrosamine (DEN) and 25 mg/kg of 2-Acetylaminofluorene (2-AAF). The HCC/PFD group received simultaneous doses of 300 mg/kg of PFD. All treatments lasted 12 weeks. On the other hand, HepG2 cells were used to evaluate the effects of PFD in restoring DNA methylation in the presence of the inhibitor 5-Aza. Histopathological, biochemical, immunohistochemical, and western blot analysis were carried out and our findings showed that PFD treatment reduced the amount and size of tumors along with decreased Glipican-3, ß-catenin, and c-Myc expression in nuclear fractions. Also, this treatment improved lipid metabolism by modulating PPARγ and SREBP1 signaling. Interestingly, PFD augmented DNMT1 and DNMT3a protein expression, which restores global methylation, both in our in vivo and in vitro models. In conclusion, our results suggest that PFD could slow down HCC development by controlling DNA methylation.


Sujet(s)
Carcinome hépatocellulaire , DNA (Cytosine-5-)-methyltransferase 1 , Méthylation de l'ADN , Antigène nucléaire de prolifération cellulaire , Pyridones , Animaux , DNA (Cytosine-5-)-methyltransferase 1/métabolisme , DNA (Cytosine-5-)-methyltransferase 1/génétique , Méthylation de l'ADN/effets des médicaments et des substances chimiques , Méthylation de l'ADN/génétique , Pyridones/pharmacologie , Rats , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/métabolisme , Humains , Cellules HepG2 , Antigène nucléaire de prolifération cellulaire/métabolisme , Mâle , Rats de lignée F344 , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/anatomopathologie , Tumeurs du foie/génétique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , N-Éthyl-N-nitroso-éthanamine , Tumeurs expérimentales du foie/traitement médicamenteux , Tumeurs expérimentales du foie/anatomopathologie , Tumeurs expérimentales du foie/métabolisme , Tumeurs expérimentales du foie/génétique
19.
Acta Chim Slov ; 71(2): 264-287, 2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-38919094

RÉSUMÉ

Combined in silico strategy for molecular mechanisms exploration of a series 3H-thiazolo[4,5-b]pyridin-2-ones exhibiting strong anti-exudative action through QSAR analysis, molecular docking and pharmacophore modelling is reported. GA-ML technique was used for QSAR models generation with 2D autocorrelation descriptors. One- and two-parameter regressions revealed that certain structural patterns or heteroatoms contribute mutually to the anti-exudative activity potentiation. Possible action mechanisms were discovered through flexible docking simulations with cyclooxygenase pathway enzymes (COX-1, COX-2, mPGES-1). Docking results indicated the possibility of stable complexes formation with the effective docking scores and proper orientation of ligands within the enzymes active sites. Pharmacophore modelling was carried out using protein-ligand interaction fingerprints methodology. Two- and three-centre 3D pharmacophore queries were constructed. Their analysis indicated the functionality of bicyclic thiazolopyridine scaffold proved by the steric placement of heteroatoms in the corresponding pharmacophore centres.


Sujet(s)
Simulation de docking moléculaire , Relation quantitative structure-activité , Thiazoles , Thiazoles/composition chimique , Thiazoles/pharmacologie , Simulation numérique , Pyridones/pharmacologie , Pyridones/composition chimique , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/composition chimique , Pyridines/pharmacologie , Pyridines/composition chimique , Cyclooxygenase 2/métabolisme , Inflammation/traitement médicamenteux
20.
Molecules ; 29(12)2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38930946

RÉSUMÉ

Cisplatin, a platinum-based chemotherapeutic, is effective against various solid tumors, but its use is often limited by its nephrotoxic effects. This study evaluated the protective effects of trametinib, an FDA-approved selective inhibitor of mitogen-activated protein kinase kinase 1/2 (MEK1/2), against cisplatin-induced acute kidney injury (AKI) in mice. The experimental design included four groups, control, trametinib, cisplatin, and a combination of cisplatin and trametinib, each consisting of eight mice. Cisplatin was administered intraperitoneally at a dose of 20 mg/kg to induce kidney injury, while trametinib was administered via oral gavage at 3 mg/kg daily for three days. Assessments were conducted 72 h after cisplatin administration. Our results demonstrate that trametinib significantly reduces the phosphorylation of MEK1/2 and extracellular signal-regulated kinase 1/2 (ERK1/2), mitigated renal dysfunction, and ameliorated histopathological abnormalities. Additionally, trametinib significantly decreased macrophage infiltration and the expression of pro-inflammatory cytokines in the kidneys. It also lowered lipid peroxidation by-products, restored the reduced glutathione/oxidized glutathione ratio, and downregulated NADPH oxidase 4. Furthermore, trametinib significantly inhibited both apoptosis and necroptosis in the kidneys. In conclusion, our data underscore the potential of trametinib as a therapeutic agent for cisplatin-induced AKI, highlighting its role in reducing inflammation, oxidative stress, and tubular cell death.


Sujet(s)
Atteinte rénale aigüe , Cisplatine , Modèles animaux de maladie humaine , Inflammation , Stress oxydatif , Pyridones , Pyrimidinones , Animaux , Cisplatine/effets indésirables , Atteinte rénale aigüe/induit chimiquement , Atteinte rénale aigüe/traitement médicamenteux , Atteinte rénale aigüe/métabolisme , Atteinte rénale aigüe/anatomopathologie , Pyridones/pharmacologie , Stress oxydatif/effets des médicaments et des substances chimiques , Souris , Pyrimidinones/pharmacologie , Inflammation/traitement médicamenteux , Inflammation/induit chimiquement , Inflammation/métabolisme , Mâle , Mort cellulaire/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Tubules rénaux/anatomopathologie , Tubules rénaux/effets des médicaments et des substances chimiques , Tubules rénaux/métabolisme , Peroxydation lipidique/effets des médicaments et des substances chimiques , Cytokines/métabolisme , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE