Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.461
Filtrer
1.
Drug Deliv ; 31(1): 2380538, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39044468

RÉSUMÉ

Rheumatoid arthritis (RA) is a chronic inflammatory joint disease accompanied by energy depletion and accumulation of reactive oxygen species (ROS). Inorganic nanoparticles (NPs) offer great promise for the treatment of RA because they mostly have functions beyond being drug carriers. However, conventional nanomaterials become coated with a protein corona (PC) or lose their cargo prematurely in vivo, reducing their therapeutic efficacy. To avoid these problems, we loaded methotrexate (MTX) into hollow structured manganese dioxide nanoparticles (H-MnO2 NPs), then coated them with a 'pseudo-corona' of human serum albumin (HSA) at physiological concentrations to obtain HSA-MnO2@MTX NPs. Efficacy of MTX, MnO2@MTX, and HSA-MnO2@MTX NPs was compared in vitro and in vivo. Compared to MnO2@MTX, HSA-coated NPs were taken up better by lipopolysaccharide-activated RAW264.7 and were more effective at lowering levels of pro-inflammatory cytokines and preventing ROS accumulation. HSA-MnO2@MTX NPs were also more efficient at blocking the proliferation and migration of fibroblast-like synoviocytes from rats with collagen-induced arthritis. In this rat model, HSA-MnO2@MTX NPs showed better biodistribution than other treatments, specifically targeting the ankle joint. Furthermore, HSA-MnO2@MTX NPs reduced swelling in the paw, regulated pro-inflammatory cytokine production, and limited cartilage degradation and signs of inflammation. These results establish the therapeutic potential of HSA-MnO2@MTX NPs against RA.


Sujet(s)
Polyarthrite rhumatoïde , Vecteurs de médicaments , Composés du manganèse , Méthotrexate , Nanoparticules , Oxydes , Espèces réactives de l'oxygène , Sérum-albumine humaine , Animaux , Composés du manganèse/composition chimique , Oxydes/composition chimique , Polyarthrite rhumatoïde/traitement médicamenteux , Souris , Rats , Nanoparticules/composition chimique , Sérum-albumine humaine/composition chimique , Humains , Méthotrexate/pharmacologie , Méthotrexate/administration et posologie , Méthotrexate/pharmacocinétique , Méthotrexate/composition chimique , Espèces réactives de l'oxygène/métabolisme , Cellules RAW 264.7 , Vecteurs de médicaments/composition chimique , Mâle , Arthrite expérimentale/traitement médicamenteux , Distribution tissulaire , Antirhumatismaux/pharmacologie , Antirhumatismaux/administration et posologie , Antirhumatismaux/pharmacocinétique , Rat Sprague-Dawley , Cytokines/métabolisme
2.
Molecules ; 29(13)2024 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-38999143

RÉSUMÉ

OBJECTIVE: To elucidate the chemical profile of Xanthocerais lignum's extracts of different polarities and their impact on rheumatoid arthritis (RA), we identified anti-RA markers and predicted their action mechanisms. METHODS: A collagen-induced arthritis rat model was established, and UPLC-Q-Exactive Orbitrap MS technology was employed to analyze and identify the chemical constituents within the alcohol extract of Xanthocerais lignum and its various extraction fractions, as well as their translocation into the bloodstream. Serum spectrum-effect correlation analysis was utilized to elucidate the pharmacodynamic material basis of Xanthocerais lignum against RA and to screen for Q-Markers. Finally, the potential anti-RA mechanisms of the Q-Markers were predicted through compound-target interaction data and validated using molecular docking techniques. RESULTS: We identified 71 compounds, with flavan-3-ols and flavanones as key components. Of these, 36 were detected in the bloodstream, including 17 original and 19 metabolized forms. Proanthocyanidin A2, dihydroquercetin, catechin, and epicatechin (plus glucuronides) showed potential anti-RA activity. These compounds, acting as Q-Markers, may modulate ERK, NF-κB, HIF-1α, and VEGF in the HIF-1 pathway. CONCLUSIONS: This research clarifies Xanthocerais lignum's pharmacodynamic material basis against RA, identifies 4 Q-Markers, and offers insights into their mechanisms, aiding quality assessment and lead compound development for RA treatment.


Sujet(s)
Polyarthrite rhumatoïde , Marqueurs biologiques , Simulation de docking moléculaire , Extraits de plantes , Animaux , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/sang , Polyarthrite rhumatoïde/métabolisme , Rats , Marqueurs biologiques/sang , Extraits de plantes/composition chimique , Extraits de plantes/pharmacologie , Arthrite expérimentale/traitement médicamenteux , Arthrite expérimentale/sang , Arthrite expérimentale/métabolisme , Mâle , Modèles animaux de maladie humaine , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Antirhumatismaux/pharmacologie , Antirhumatismaux/composition chimique , Antirhumatismaux/usage thérapeutique , Facteur de croissance endothéliale vasculaire de type A/sang , Facteur de croissance endothéliale vasculaire de type A/métabolisme
3.
Clin Exp Rheumatol ; 42(7): 1387-1397, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38976290

RÉSUMÉ

OBJECTIVES: The imbalance between apoptosis and proliferation in fibroblast-like synoviocytes (FLSs) plays a key role in the pathogenesis of rheumatoid arthritis (RA). This study aims to investigate the potential of all-trans retinoic acid (ATRA) as a supplementary therapeutic agent alongside methotrexate (MTX) for RA, by examining its ability to inhibit synovial cell proliferation and enhance apoptosis through the ROS-JNK signalling pathway. METHODS: The viability, apoptosis, and autophagy levels of human rheumatoid arthritis fibroblast-like synovial cells (HFLS-RA) were evaluated, while ROS generation was measured through the DCFH-DA fluorescence microplate assay. Western blotting was used to analyse the expression levels of JNK signalling pathway-related proteins. To assess therapeutic potential in vivo, a collagen-induced arthritis (CIA) model was established in Wistar rats. RESULTS: Small doses of MTX did not significantly affect the viability of HFLS-RAs or induce apoptosis. However, when ATRA was added to the treatment, the therapy markedly inhibited cell proliferation and induced apoptosis and excessive autophagy. Mechanistically, ATRA activated the ROS/JNK signalling pathway in HFLS-RAs. ROS scavengers and JNK inhibitors significantly attenuated ATRA-induced apoptosis and autophagy. In vivo, the combination therapy demonstrated a remarkable enhancement of the anti-arthritic efficacy in CIA rats. CONCLUSIONS: The ability of ATRA to inhibit proliferation in RA FLSs through autophagy and apoptosis underscores its potential as a supplementary therapeutic agent alongside MTX for RA, particularly when compared to the limited impact of MTX on these processes. This combined strategy holds promise for enhancing therapeutic outcomes and warrants further investigation in the management of RA.


Sujet(s)
Apoptose , Arthrite expérimentale , Polyarthrite rhumatoïde , Autophagie , Prolifération cellulaire , Méthotrexate , Rat Wistar , Espèces réactives de l'oxygène , Cellules synoviales , Trétinoïne , Trétinoïne/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/anatomopathologie , Polyarthrite rhumatoïde/métabolisme , Méthotrexate/pharmacologie , Autophagie/effets des médicaments et des substances chimiques , Animaux , Humains , Arthrite expérimentale/traitement médicamenteux , Arthrite expérimentale/anatomopathologie , Arthrite expérimentale/métabolisme , Espèces réactives de l'oxygène/métabolisme , Cellules synoviales/effets des médicaments et des substances chimiques , Cellules synoviales/anatomopathologie , Cellules synoviales/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Association de médicaments , Antirhumatismaux/pharmacologie , Membrane synoviale/effets des médicaments et des substances chimiques , Membrane synoviale/anatomopathologie , Membrane synoviale/métabolisme , Mâle , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Rats , Lignée cellulaire
4.
Clin Exp Pharmacol Physiol ; 51(8): e13906, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38965677

RÉSUMÉ

In this study, we wanted to investigate the effectiveness of combining disease-modifying anti-rheumatic drugs (DMARD) with hyperbaric oxygen therapy (HBOT) in reducing inflammation in a rheumatoid arthritis (RA) model using rats. We divided 56 male Sprague-Dawley rats into seven groups and induced RA using complete Freund's adjuvant. Some groups received HBOT, whereas others were given etanercept or leflunomide. We started the treatment on the 10th day after inducing RA and continued it for 18 days. To evaluate the effectiveness of the treatments, we measured paw swelling and used X-rays to examine the joints before and after the treatment. We also analysed the levels of two inflammatory markers, tumour necrosis factor (TNF)-α and interleukin (IL)-1ß, using an enzyme-linked immunosorbent assay. Additionally, we conducted histological analysis and assessed the expressions of anti-IL-1ß and anti-TNF-α antibodies. All the treatment groups showed a significant decrease in arthritis scores, paw swelling and levels of TNF-α and IL-1ß. The X-ray images revealed improvements in joint structure, and the histopathological analysis showed reduced inflammation and collagen abnormalities. Combining DMARD with HBOT had similar effects to individual therapies, suggesting a cost-effective and potentially safer approach for improving outcomes in rats with RA.


Sujet(s)
Antirhumatismaux , Polyarthrite rhumatoïde , Oxygénation hyperbare , Interleukine-1 bêta , Rat Sprague-Dawley , Facteur de nécrose tumorale alpha , Animaux , Oxygénation hyperbare/méthodes , Mâle , Antirhumatismaux/usage thérapeutique , Antirhumatismaux/pharmacologie , Polyarthrite rhumatoïde/thérapie , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/anatomopathologie , Polyarthrite rhumatoïde/métabolisme , Rats , Interleukine-1 bêta/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Modèles animaux de maladie humaine , Étanercept/usage thérapeutique , Étanercept/pharmacologie , Arthrite expérimentale/thérapie , Arthrite expérimentale/anatomopathologie , Arthrite expérimentale/traitement médicamenteux , Arthrite expérimentale/métabolisme , Léflunomide/usage thérapeutique , Léflunomide/pharmacologie
5.
Sci Rep ; 14(1): 15226, 2024 07 02.
Article de Anglais | MEDLINE | ID: mdl-38956271

RÉSUMÉ

This study aims to identify factors influencing the alleviation of knee joint symptoms in patients with rheumatoid arthritis treated with biologic or target synthetic disease-modifying antirheumatic drugs (b/tsDMARDs). Among 2321 patients who started b/tsDMARDs between 2010 and 2023, we focused on 295 patients who had knee swelling or tenderness at the initiation of b/tsDMARDs and continued b/tsDMARDs at least 3 months, with recorded knee symptoms 6 months later. Symptom relief after 6 months was 78.2% for interleukin 6 (IL-6) inhibitors, 68.6% for Janus kinase (JAK) inhibitors, 65.8% for tumor necrosis factor (TNF) inhibitors, and 57.6% for cytotoxic T lymphocyte-associated antigen-4-Ig (CTLA4-Ig). The initial use of b/tsDMARDs and the use of IL-6 inhibitors in comparison to CTLA4-Ig emerged as a significant factor associated with the improvement of knee joint symptoms. Among 141 patients who underwent knee radiography at baseline and two years later, the deterioration in knee joint radiographs was 7.7% for IL-6 inhibitors, 6.3% for JAK inhibitors, 21.9% for TNF inhibitors, and 25.9% for CTLA4-Ig. The use of IL-6 inhibitors was a significant factor associated with the improvement of knee joint symptoms and the inhibition of joint destruction compared to CTLA4-Ig.


Sujet(s)
Abatacept , Antirhumatismaux , Polyarthrite rhumatoïde , Interleukine-6 , Inhibiteurs du facteur de nécrose tumorale , Humains , Polyarthrite rhumatoïde/traitement médicamenteux , Femelle , Mâle , Interleukine-6/antagonistes et inhibiteurs , Interleukine-6/métabolisme , Adulte d'âge moyen , Abatacept/usage thérapeutique , Antirhumatismaux/usage thérapeutique , Antirhumatismaux/pharmacologie , Inhibiteurs du facteur de nécrose tumorale/usage thérapeutique , Sujet âgé , Articulation du genou/imagerie diagnostique , Articulation du genou/anatomopathologie , Articulation du genou/effets des médicaments et des substances chimiques , Adulte , Inhibiteurs des Janus kinases/usage thérapeutique , Inhibiteurs des Janus kinases/pharmacologie , Résultat thérapeutique , Facteur de nécrose tumorale alpha/antagonistes et inhibiteurs
6.
Front Immunol ; 15: 1352251, 2024.
Article de Anglais | MEDLINE | ID: mdl-38840915

RÉSUMÉ

Objective: Antigen-presenting dendritic cells (DCs) and monocytes play an essential role in rheumatoid arthritis (RA) pathogenesis, however, their tolerogenic potential remains unclear. Herein, the tolerogenic profiles of DCs are characterized in treatment-naïve RA patients to determine their role to inflammatory arthritis management. Methods: Thirty-six treatment-naïve RA patients were enrolled, of which 62% were non-responders to methotrexate (MTX) monotherapy based on disease activity score (DAS) after 6-months of therapy. DC and monocyte subset frequencies, activation (CD40, CD86, CD209 expression), and tolerogenic profile (intracellular indoleamine-2,3-dioxygenase [IDO1] and cytotoxic T lymphocyte antigen 4 [CTLA-4] expression) were examined in the baseline peripheral blood by multicolor flow-cytometry. Soluble CTLA-4 (sCTLA-4) levels in plasma were measured. Results: DC subsets were decreased in RA compared to healthy controls (HC), and the frequency of conventional DCs (cDC) inversely correlated with inflammatory markers and improvement in disease activity. CD141+ cDC1s were the major IDO1-expressing cells. IDO1+cDC1s were reduced in RA patients compared to HC. The baseline frequency of IDO1+cDC1s inversely correlated with improvement in disease activity. CTLA-4 expression in CD1c+ cDC2s and monocytes was lower in RA patients compared to HC. Moreover, MTX-responders had a significantly lower frequency of IDO1+cDC1 cells and higher level of sCTLA-4 in the plasma compared to MTX non-responders. There was a strong predictive association of low IDO1+cDC1 cells, low sCTLA-4 and non-response to MTX. Conclusions: Our findings reveal altered DC and monocytes immunophenotypes that are associated with RA pathology and treatment response. The frequencies of tolerogenic IDO1+cDC1s and the low level of sCTLA-4 are strongly associated with MTX non-responsiveness and therapeutic outcome. These results suggest that investigation of the association IDO1+cDC1 and sCTLA-4 with response to treatment may be more generalizable to other autoimmune diseases.


Sujet(s)
Polyarthrite rhumatoïde , Antigène CTLA-4 , Cellules dendritiques , Indoleamine-pyrrole 2,3,-dioxygenase , Méthotrexate , Humains , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/immunologie , Polyarthrite rhumatoïde/sang , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Indoleamine-pyrrole 2,3,-dioxygenase/métabolisme , Méthotrexate/usage thérapeutique , Méthotrexate/pharmacologie , Femelle , Mâle , Adulte d'âge moyen , Adulte , Antirhumatismaux/usage thérapeutique , Antirhumatismaux/pharmacologie , Sujet âgé , Monocytes/immunologie , Monocytes/métabolisme , Résultat thérapeutique , Marqueurs biologiques
7.
Sci Rep ; 14(1): 12935, 2024 06 05.
Article de Anglais | MEDLINE | ID: mdl-38839973

RÉSUMÉ

The inhibition of tumor necrosis factor (TNF)-α trimer formation renders it inactive for binding to its receptors, thus mitigating the vicious cycle of inflammation. We designed a peptide (PIYLGGVFQ) that simulates a sequence strand of human TNFα monomer using a series of in silico methods, such as active site finding (Acsite), protein-protein interaction (PPI), docking studies (GOLD and Flex-X) followed by molecular dynamics (MD) simulation studies. The MD studies confirmed the intermolecular interaction of the peptide with the TNFα. Fluorescence-activated cell sorting and fluorescence microscopy revealed that the peptide effectively inhibited the binding of TNF to the cell surface receptors. The cell culture assays showed that the peptide significantly inhibited the TNFα-mediated cell death. In addition, the nuclear translocation of the nuclear factor kappa B (NFκB) was significantly suppressed in the peptide-treated A549 cells, as observed in immunofluorescence and gel mobility-shift assays. Furthermore, the peptide protected against joint damage in the collagen-induced arthritis (CIA) mouse model, as revealed in the micro focal-CT scans. In conclusion, this TNFα antagonist would be helpful for the prevention and repair of inflammatory bone destruction and subsequent loss in the mouse model of CIA as well as human rheumatoid arthritis (RA) patients. This calls upon further clinical investigation to utilize its potential effect as an antiarthritic drug.


Sujet(s)
Peptides , Facteur de nécrose tumorale alpha , Humains , Facteur de nécrose tumorale alpha/métabolisme , Facteur de nécrose tumorale alpha/antagonistes et inhibiteurs , Animaux , Souris , Peptides/pharmacologie , Peptides/composition chimique , Arthrite expérimentale/traitement médicamenteux , Arthrite expérimentale/métabolisme , Arthrite expérimentale/anatomopathologie , Simulation de docking moléculaire , Cellules A549 , Simulation de dynamique moléculaire , Facteur de transcription NF-kappa B/métabolisme , Facteur de transcription NF-kappa B/antagonistes et inhibiteurs , Mâle , Antirhumatismaux/pharmacologie , Antirhumatismaux/composition chimique , Antirhumatismaux/usage thérapeutique , Liaison aux protéines , Modèles animaux de maladie humaine
8.
J Control Release ; 371: 498-515, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38849090

RÉSUMÉ

Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease characterized by synovial inflammation and inflammatory cellular infiltration. Functional cells in the RA microenvironment (RAM) are composed of activated immune cells and effector cells. Activated immune cells, including macrophages, neutrophils, and T cells, can induce RA. Effector cells, including synoviocytes, osteoclasts, and chondrocytes, receiving inflammatory stimuli, exacerbate RA. These functional cells, often associated with the upregulation of surface-specific receptor proteins and significant homing effects, can secrete pro-inflammatory factors and interfere with each other, thereby jointly promoting the progression of RA. Recently, some nanomedicines have alleviated RA by targeting and modulating functional cells with ligand modifications, while other nanoparticles whose surfaces are camouflaged by membranes or extracellular vesicles (EVs) of these functional cells target and attack the lesion site for RA treatment. When ligand-modified nanomaterials target specific functional cells to treat RA, the functional cells are subjected to attack, much like the intended targets. When functional cell membranes or EVs are modified onto nanomaterials to deliver drugs for RA treatment, functional cells become the attackers, similar to arrows. This study summarized how diversified functional cells serve as targets or arrows by engineered nanoparticles to treat RA. Moreover, the key challenges in preparing nanomaterials and their stability, long-term efficacy, safety, and future clinical patient compliance have been discussed here.


Sujet(s)
Polyarthrite rhumatoïde , Nanomédecine , Nanoparticules , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/immunologie , Humains , Nanomédecine/méthodes , Animaux , Nanoparticules/administration et posologie , Systèmes de délivrance de médicaments , Antirhumatismaux/administration et posologie , Antirhumatismaux/pharmacologie , Antirhumatismaux/usage thérapeutique , Vésicules extracellulaires
9.
Expert Opin Investig Drugs ; 33(7): 671-676, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38879822

RÉSUMÉ

INTRODUCTION: Polymyalgia rheumatica (PMR) is an inflammatory rheumatic disorder characterized by pain and stiffness in the shoulder and pelvic girdles, constitutional symptoms, and elevated acute-phase reactants. Glucocorticoids (GCs) remain the first-choice treatment for PMR, but relapses are common. Identification of steroid-sparing agents is therefore of utmost importance. AREAS COVERED: The efficacy of conventional immunosuppressive drugs is controversial. The use of interleukin (IL)-6 receptor inhibitors proved to be effective and safe in treating PMR patients. Currently, there are 12 ongoing clinical trials exploring potential treatments such as leflunomide, low-dose IL-2, rituximab, abatacept, secukinumab, Janus kinase inhibitors, and selective inhibitors like SPI-62 and ABBV 154. EXPERT OPINION: The high efficacy of IL-6 R receptor inhibitors as well as the numerous drug trials currently recruiting suggest that several therapeutic options will be available in the near future. Accurate diagnosis and early stratification of PMR patients according to the giant cell arteritis-PMR Spectrum Disease 'GPSD' and potential risk factors for relapsing disease or GC-related adverse events are crucial to identify patients who would benefit most from GC-sparing agents. The development of internationally accepted definitions for remission and relapse is urgently needed. Early referral strategies to specialist settings would improve disease stratification and personalized treatment.


Sujet(s)
Développement de médicament , Médicaments en essais cliniques , Glucocorticoïdes , Rhumatisme inflammatoire des ceintures , Humains , Rhumatisme inflammatoire des ceintures/traitement médicamenteux , Médicaments en essais cliniques/pharmacologie , Médicaments en essais cliniques/effets indésirables , Glucocorticoïdes/administration et posologie , Glucocorticoïdes/pharmacologie , Glucocorticoïdes/effets indésirables , Immunosuppresseurs/pharmacologie , Immunosuppresseurs/effets indésirables , Immunosuppresseurs/administration et posologie , Immunosuppresseurs/usage thérapeutique , Antirhumatismaux/pharmacologie , Antirhumatismaux/effets indésirables , Antirhumatismaux/administration et posologie , Récepteurs à l'interleukine-6/antagonistes et inhibiteurs , Récidive , Animaux , Facteurs de risque
10.
Pharmacol Res Perspect ; 12(4): e1230, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38940379

RÉSUMÉ

This study provides a detailed understanding of the preclinical pharmacokinetics and metabolism of ELP-004, an osteoclast inhibitor in development for the treatment of bone erosion. Current treatments for arthritis, including biological disease-modifying antirheumatic drugs, are not well-tolerated in a substantial subset of arthritis patients and are expensive; therefore, new treatments are needed. Pharmacokinetic parameters of ELP-004 were tested with intravenous, oral, and subcutaneous administration and found to be rapidly absorbed and distributed. We found that ELP-004 was non-mutagenic, did not induce chromosome aberrations, non-cardiotoxic, and had minimal off-target effects. Using in vitro hepatic systems, we found that ELP-004 is primarily metabolized by CYP1A2 and CYP2B6 and predicted metabolic pathways were identified. Finally, we show that ELP-004 inhibits osteoclast differentiation without suppressing overall T-cell function. These preclinical data will inform future development of an oral compound as well as in vivo efficacy studies in mice.


Sujet(s)
Ostéoclastes , Animaux , Souris , Ostéoclastes/effets des médicaments et des substances chimiques , Mâle , Évaluation préclinique de médicament , Femelle , Souris de lignée C57BL , Administration par voie orale , Humains , Différenciation cellulaire/effets des médicaments et des substances chimiques , Lymphocytes T/effets des médicaments et des substances chimiques , Lymphocytes T/métabolisme , Antirhumatismaux/pharmacologie , Antirhumatismaux/pharmacocinétique , Antirhumatismaux/administration et posologie
11.
J Med Chem ; 67(12): 9869-9895, 2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-38888047

RÉSUMÉ

Rheumatoid arthritis (RA) is a chronic autoimmune disease. Targeting NLRP3 inflammasome, specifically its interaction with NEK7 via the LRR domain of NLRP3, is a promising therapeutic strategy. Our research aimed to disrupt this interaction by focusing on the LRR domain. Through virtual screening, we identified five compounds with potent anti-inflammatory effects and ideal LRR binding affinity. Lead compound C878-1943 underwent structural optimization, yielding pyridoimidazole derivatives with different anti-inflammatory activities. Compound I-19 from the initial series effectively inhibited caspase-1 and IL-1ß release in an adjuvant-induced arthritis (AIA) rat model, significantly reducing joint swelling and spleen/thymus indices. To further enhance potency and extend in vivo half-life, a second series including II-8 was developed, demonstrating superior efficacy and longer half-life. Both I-19 and II-8 bind to the LRR domain, inhibiting NLRP3 inflammasome activation. These findings introduce novel small molecule inhibitors targeting the LRR domain of NLRP3 protein and disrupt NLRP3-NEK7 interaction, offering a novel approach for RA treatment.


Sujet(s)
Arthrite expérimentale , Polyarthrite rhumatoïde , Kinases apparentées à NIMA , Protéine-3 de la famille des NLR contenant un domaine pyrine , Protéine-3 de la famille des NLR contenant un domaine pyrine/antagonistes et inhibiteurs , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Kinases apparentées à NIMA/antagonistes et inhibiteurs , Kinases apparentées à NIMA/métabolisme , Animaux , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/métabolisme , Humains , Rats , Arthrite expérimentale/traitement médicamenteux , Découverte de médicament , Relation structure-activité , Mâle , Inflammasomes/métabolisme , Inflammasomes/antagonistes et inhibiteurs , Simulation de docking moléculaire , Antirhumatismaux/pharmacologie , Antirhumatismaux/composition chimique , Antirhumatismaux/synthèse chimique , Antirhumatismaux/usage thérapeutique
12.
Inflamm Res ; 73(8): 1371-1391, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38879731

RÉSUMÉ

Rheumatoid arthritis (RA) is a chronic, systemic inflammatory disorder characterized by joint destruction due to synovial hypertrophy and the infiltration of inflammatory cells. Despite substantial progress in RA treatment, challenges persist, including suboptimal treatment responses and adverse effects associated with current therapies. This study investigates the anti-rheumatic capabilities of the newly identified multi-protein kinase inhibitor, KMU-11342, aiming to develop innovative agents targeting RA. In this study, we synthesized the novel multi-protein kinase inhibitor KMU-11342, based on indolin-2-one. We assessed its cardiac electrophysiological safety using the Langendorff system in rat hearts and evaluated its toxicity in zebrafish in vivo. Additionally, we examined the anti-rheumatic effects of KMU-11342 on human rheumatoid arthritis fibroblast-like synoviocytes (RA-FLS), THP-1 cells, and osteoclastogenesis in RAW264.7 cells. KMU-11342 demonstrated the ability to inhibit LPS-induced chemokine inhibition and the upregulation of pro-inflammatory cytokines, cyclooxygenase-2, inducible nitric oxide synthase, p-IKKα/ß, p-NF-κB p65, and the nuclear translocation of NF-κB p65 in RA-FLS. It effectively suppressed the upregulation of NLR family pyrin domain containing 3 (NLRP3) and caspase-1 cleavage. Furthermore, KMU-11342 hindered the activation of osteoclast differentiation factors such as RANKL-induced TRAP, cathepsin K, NFATc-1, and c-Fos in RAW264.7 cells. KMU-11342 mitigates LPS-mediated inflammatory responses in THP-1 cells by inhibiting the activation of NLRP3 inflammasome. Notably, KMU-11342 exhibited minimal cytotoxicity in vivo and electrophysiological cardiotoxicity ex vivo. Consequently, KMU-11342 holds promise for development as a therapeutic agent in RA treatment.


Sujet(s)
Antirhumatismaux , Polyarthrite rhumatoïde , Cellules synoviales , Danio zébré , Animaux , Humains , Souris , Polyarthrite rhumatoïde/traitement médicamenteux , Cellules RAW 264.7 , Cellules synoviales/effets des médicaments et des substances chimiques , Antirhumatismaux/pharmacologie , Antirhumatismaux/usage thérapeutique , Rats , Mâle , Cytokines/métabolisme , Cellules THP-1 , Indoles/pharmacologie , Indoles/usage thérapeutique , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Rat Sprague-Dawley
13.
Medicina (Kaunas) ; 60(6)2024 May 22.
Article de Anglais | MEDLINE | ID: mdl-38929463

RÉSUMÉ

Background and Objective: Hydroxychloroquine sulfate (HCQ) is a lysosomotropic agent administered in systemic lupus erythematosus and rheumatoid arthritis that has fewer toxic effects than chloroquine. However, HCQ may still be responsible for retinal toxicity. In this study, we observed structural changes in the retinas of experimental rats after prolonged exposure to HCQ. Matherials and Methods: We investigated several aspects regarding retinal changes, at both the histopathological and ultrastructural levels. We used 96 male albino Wistar rats distributed into four equal groups (n = 24 per group): the first three groups were treated with different doses of HCQ (50, 100, and 200 mg/kg HCQ, injected intraperitoneally in a single dose daily), and the last group (the control group, n = 24) was treated with saline solution administered in the same way (0.4 mL of saline solution). The treated groups received HCQ daily for 4 months, and every month, six animals from each group were sacrificed to assess retinal changes. The eyes were examined via optical (OM) and electronic microscopy (EM). Statistical analysis was deployed, and results regarding retinal morpho-photometry were acquired. Results: We observed structural retinal changes in both high and low doses of HCQ; while high doses determined a significant thinning of the retina, lower doses caused retinal thickening. Morphological retinal changes upon exposure to HCQ are believed to be caused by accumulated HCQ in lysosomes found in retinal ganglion cells and in the inner nuclear and photoreceptor cell layers. Such changes were most evident in the group receiving HCQ intraperitoneally in doses of 100 mg/kg for a longer period (4 months). Conclusions: The present study highlights histopathological and ultrastructural retinal changes induced by chronic HCQ administration, which were strongly connected to the dosage and period of exposure.


Sujet(s)
Hydroxychloroquine , Rat Wistar , Rétine , Hydroxychloroquine/usage thérapeutique , Hydroxychloroquine/pharmacologie , Hydroxychloroquine/effets indésirables , Animaux , Rats , Rétine/effets des médicaments et des substances chimiques , Rétine/ultrastructure , Rétine/anatomopathologie , Mâle , Antirhumatismaux/usage thérapeutique , Antirhumatismaux/pharmacologie
14.
J Ethnopharmacol ; 332: 118377, 2024 Oct 05.
Article de Anglais | MEDLINE | ID: mdl-38782307

RÉSUMÉ

ETHNOPHARMACOLOGICAL RELEVANCE: The Tibetan medicine Ganlu Formula, as a classic prescription, is widely used across the Qinghai-Tibet Plateau area of China, which has a significant effect on relieving the course of rheumatoid arthritis (RA). However, the active compounds and underlying mechanisms of Ganlu Formula in RA treatment remain largely unexplored. AIM OF THE STUDY: This study aimed to elucidate the active substances and potential mechanisms of the ethyl acetate extract of Ganlu Formula ethyl acetate extract (GLEE) in the treatment of RA. MATERIALS AND METHODS: Ultra-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF-MS) was utilized to analyze and identify the chemical constituents within GLEE. Discovery Studio molecular virtual docking technology was utilized to dock the interaction of GLEE with inflammation-related pathway proteins. The GLEE gene library was obtained by transcriptome sequencing. Collagen-induced arthritic(CIA) rats were utilized to assess the antiarthritic efficacy of GLEE. Micro-CT imaging was employed to visualize the rat paw, and ultrasound imaging revealed knee joint effusion. Evaluation of synovial tissue pathological changes was conducted through hematoxylin-eosin staining and saffranine solid green staining, while immunohistochemical staining was employed to assess NLRP3 expression along with inflammatory markers. Immunofluorescence staining was utilized to identify M1 macrophages. RESULTS: Metabolomic analysis via UPLC-Q-TOF-MS identified 28 potentially bioactive compounds in GLEE, which interacted with the active sites of key proteins such as NLRP3, NF-κB, and STAT3 through hydrogen bonds, C-H bonds, and electrostatic attractions. In vitro analyses demonstrated that GLEE significantly attenuated NLRP3 inflammasome activation and inhibited the polarization of bone marrow-derived macrophages (BMDMs) towards the M1 phenotype. In vivo, GLEE not only prevented bone mineral density (BMD) loss but also reduced ankle swelling in CIA rats. Furthermore, it decreased the expression of the NLRP3 inflammasome and curtailed the release of inflammatory mediators within the knee joint. CONCLUSION: GLEE effectively mitigated inflammatory responses in both blood and knee synovial membranes of CIA rats, potentially through the down-regulation of the NLRP3/Caspase-1/IL-1ß signaling pathway and reduction in M1 macrophage polarization.


Sujet(s)
Arthrite expérimentale , Macrophages , Protéine-3 de la famille des NLR contenant un domaine pyrine , Animaux , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Arthrite expérimentale/traitement médicamenteux , Arthrite expérimentale/anatomopathologie , Rats , Mâle , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Extraits de plantes/pharmacologie , Extraits de plantes/composition chimique , Médicaments issus de plantes chinoises/pharmacologie , Médicaments issus de plantes chinoises/composition chimique , Simulation de docking moléculaire , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/composition chimique , Polyarthrite rhumatoïde/traitement médicamenteux , Rat Sprague-Dawley , Souris , Antirhumatismaux/pharmacologie , Antirhumatismaux/isolement et purification , Antirhumatismaux/composition chimique , Acétates
15.
Bioorg Chem ; 149: 107499, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38815476

RÉSUMÉ

Janus Kinase 3 (JAK3) is important for the signaling transduction of cytokines in immune cells and is identified as potential target for treatment of rheumatoid arthritis (RA). Recently, we designed and synthesized two JAK3 inhibitors J1b and J1f, which featured with high selectivity but mild bioactivity. Therefore, in present study the structure was optimized to increase the potency. As shown in the results, most of the compounds synthesized showed stronger inhibitory activities against JAK3 in contrast to the lead compounds, among which 9a was the most promising candidate because it had the most potent effect in ameliorating carrageenan-induced inflammation of mice and exhibited low acute in vivo toxicity (MTD > 2 g/kg). Further analysis revealed that 9a was highly selective to JAK3 (IC50 = 0.29 nM) with only minimal effect on other JAK members (>3300-fold) and those kinases bearing a thiol in a position analogous to that of Cys909 in JAK3 (>150-fold). Meanwhile, the selectivity of JAK3 was also confirmed by PBMC stimulation assay, in which 9a irreversibly bound to JAK3 and robustly inhibited the signaling transduction with mild suppression on other JAKs. Moreover, it was showed that 9a could remarkably inhibited the proliferation of lymphocytes in response to concanavalin A and significantly mitigate disease severity in collagen induced arthritis. Therefore, present data indicate that compound 9a is a selective JAK3 inhibitor and could be a promising candidate for clinical treatment of RA.


Sujet(s)
Polyarthrite rhumatoïde , Janus kinase 3 , Inhibiteurs de protéines kinases , Pyrimidines , Janus kinase 3/antagonistes et inhibiteurs , Janus kinase 3/métabolisme , Polyarthrite rhumatoïde/traitement médicamenteux , Animaux , Pyrimidines/composition chimique , Pyrimidines/pharmacologie , Pyrimidines/synthèse chimique , Humains , Relation structure-activité , Souris , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/composition chimique , Inhibiteurs de protéines kinases/synthèse chimique , Structure moléculaire , Relation dose-effet des médicaments , Pyrroles/composition chimique , Pyrroles/pharmacologie , Pyrroles/synthèse chimique , Carragénane , Mâle , Arthrite expérimentale/traitement médicamenteux , Arthrite expérimentale/induit chimiquement , Antirhumatismaux/pharmacologie , Antirhumatismaux/composition chimique , Antirhumatismaux/synthèse chimique , Simulation de docking moléculaire
16.
Drugs ; 84(5): 587-598, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38703349

RÉSUMÉ

Although several biological disease-modifying antirheumatic drugs (bDMARDs), including interleukin (IL)-17A inhibitors, are approved for psoriatic arthritis, the treatment of this disease remains suboptimal. Bimekizumab (Bimzelx®), a dual IL-17A and IL-17F inhibitor, is approved in the EU, Great Britain and Japan for the treatment of psoriatic arthritis. In pivotal phase 3 clinical trials in patients who were bDMARD-naïve or previously had an inadequate response or intolerance to tumour necrosis factor (TNF) α inhibitors, bimekizumab improved the signs and symptoms of psoriatic arthritis across a range of joint, skin, radiographic and patient-reported outcomes compared with placebo, including the proportion of patients achieving a ≥ 50% response in the American College of Rheumatology criteria. Phase 2 clinical trial data have shown that responses are maintained up to 3 years. Bimekizumab was generally well tolerated in patients with psoriatic arthritis, with a safety profile consistent with that in other approved indications. The most common adverse events included nasopharyngitis, upper respiratory tract infection, oral candidiasis, headache and diarrhoea. In conclusion, bimekizumab extends the treatment options available to patients with psoriatic arthritis.


Psoriatic arthritis is a chronic, painful, inflammatory condition that can involve peripheral and axial joints, skin and nails, and can lead to structural joint damage. Several disease-modifying antirheumatic drugs (DMARDs), including biologicals (bDMARDs) that target various inflammatory cytokines, are approved for psoriatic arthritis. However, many patients do not respond or lose response to these treatments. Bimekizumab (Bimzelx®), an inhibitor of both interleukin (IL)-17A and IL-17F, is approved for the treatment of psoriatic arthritis in the EU, Great Britain and Japan. In clinical trials, bimekizumab improved the signs and symptoms of psoriatic arthritis across key disease domains compared with placebo, including the proportion of patients achieving ≥ 50% response in the American College of Rheumatology criteria. Bimekizumab was efficacious whether patients were naïve to bDMARDs or previously had an inadequate response to or did not tolerate tumour necrosis factor (TNF) α inhibitors. Clinical responses were sustained long-term (up to 3 years). Bimekizumab was generally well tolerated, with the most common adverse events being infections (such as nasopharyngitis, upper respiratory tract infection and oral candidiasis), headache and diarrhoea. In conclusion, bimekizumab extends the treatment options available to patients with psoriatic arthritis.


Sujet(s)
Anticorps monoclonaux humanisés , Antirhumatismaux , Arthrite psoriasique , Interleukine-17 , Humains , Arthrite psoriasique/traitement médicamenteux , Anticorps monoclonaux humanisés/usage thérapeutique , Anticorps monoclonaux humanisés/effets indésirables , Anticorps monoclonaux humanisés/pharmacologie , Interleukine-17/antagonistes et inhibiteurs , Antirhumatismaux/usage thérapeutique , Antirhumatismaux/effets indésirables , Antirhumatismaux/pharmacologie
17.
Expert Opin Investig Drugs ; 33(7): 661-670, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38698301

RÉSUMÉ

INTRODUCTION: Rheumatoid arthritis (RA) is an autoimmune disorder with a characteristic chronic inflammation of the synovium that may lead to the destruction of the joints in untreated patients. Interestingly, despite the availability of several effective treatments, many patients do not achieve remission or low disease activity or may experience disease relapse.Following the above unmet needs, bispecific antibodies (BsAbs) have emerged as a new approach to improve the disease's treatment. BsAbs are designed to simultaneously target two different proteins involved in RA pathogenesis, leading to enhanced efficacy and reduced side effects compared to traditional monoclonal antibodies (mAbs). AREAS COVERED: In this review, we discuss the development of BsAbs for RA treatment, including their mechanism of action, efficacy, and safety profile. We also deal with the challenges and future directions in this field. EXPERT OPINION: BsAbs show promise in preclinical and clinical evaluations for treating RA. Further research is needed to optimize design and dosage and identify ideal patient groups. BsAbs can benefit disease management and improve outcomes of RA patients.


Sujet(s)
Anticorps bispécifiques , Antirhumatismaux , Polyarthrite rhumatoïde , Développement de médicament , Humains , Anticorps bispécifiques/pharmacologie , Anticorps bispécifiques/administration et posologie , Anticorps bispécifiques/effets indésirables , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/immunologie , Animaux , Antirhumatismaux/pharmacologie , Antirhumatismaux/effets indésirables , Antirhumatismaux/administration et posologie , Antirhumatismaux/usage thérapeutique , Anticorps monoclonaux/pharmacologie , Anticorps monoclonaux/administration et posologie , Anticorps monoclonaux/effets indésirables , Médicaments en essais cliniques/pharmacologie , Médicaments en essais cliniques/administration et posologie , Médicaments en essais cliniques/effets indésirables
18.
J Ethnopharmacol ; 332: 118286, 2024 Oct 05.
Article de Anglais | MEDLINE | ID: mdl-38723919

RÉSUMÉ

ETHNOPHARMACOLOGICAL RELEVANCE: Di-Long (Pheretima vulgaris) is a classic animal sourced traditional Chinese medicine. It has been used for the treatment of joint inflammation and arthralgia for over two thousand years due to its effects of Tong-Luo-Zhi-Tong (dredging collaterals and alleviating pain). Our previous study showed that Chinese medicine Di-Long has significant anti-rheumatoid arthritis (RA) effects. AIM OF THE STUDY: Considering Di-Long as a potential source of active compounds with specific anti-RA therapeutic effects, this research was to obtain the anti-RA target-specific active fraction from Di-Long extracts (DL), and to further explore the chemical basis and verify the anti-RA mechanism of this active fraction. MATERIALS AND METHODS: Transcriptomic was applied to obtain the main anti-RA targets of DL on human RA fibroblast-like synoviocytes (FLS) and validated by qPCR. The target-corresponding active fraction was isolated from DL by ethanol precipitation and gel chromatography, and analyzed by nanoliter chromatography-mass spectrometry. Anti-RA effects of this active fraction was investigated by collagen-induced arthritis (CIA) in mice, and anti-RA mechanisms were verified in cocultured model of rat FLS and peripheral blood lymphocytes. RESULTS: We confirmed that CXCL10/CXCR3 was the main anti-RA target of DL. The active fraction - A (2182 - 890 Da) was isolated from DL based on its CXCL10 inhibiting effects in RA-FLS. Fraction A contains 195 peptides (192 were newly discovered), 26 of which might be bioactive and were considered to be the chemical basis of its anti-RA effects. Fraction A significantly ameliorated the joint destruction and overall inflammation in CIA mice, and downregulated CXCR3 expression in mice joint. Fraction A inhibited the chemotaxis of Th-cells in rat peripheral blood lymphocytes towards the TNF-α-induced rat FLS through CXCL10/CXCR3 pathway. CONCLUSIONS: Our work indicated that active fraction from DL containing small peptides exhibits promising therapeutic effects for RA through inhibiting CXCL10/CXCR3 chemotaxis.


Sujet(s)
Antirhumatismaux , Arthrite expérimentale , Polyarthrite rhumatoïde , Chimiokine CXCL10 , Chimiotaxie , Récepteurs CXCR3 , Membrane synoviale , Animaux , Récepteurs CXCR3/métabolisme , Chimiokine CXCL10/métabolisme , Arthrite expérimentale/traitement médicamenteux , Polyarthrite rhumatoïde/traitement médicamenteux , Mâle , Antirhumatismaux/pharmacologie , Antirhumatismaux/isolement et purification , Rats , Humains , Chimiotaxie/effets des médicaments et des substances chimiques , Membrane synoviale/effets des médicaments et des substances chimiques , Membrane synoviale/métabolisme , Souris , Souris de lignée DBA , Médicaments issus de plantes chinoises/pharmacologie , Médicaments issus de plantes chinoises/composition chimique , Cellules synoviales/effets des médicaments et des substances chimiques , Cellules synoviales/métabolisme
19.
Expert Opin Investig Drugs ; 33(6): 591-600, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38696223

RÉSUMÉ

INTRODUCTION: IL-17 has been described as a pro-inflammatory cytokine that is relevant in the seronegative spondylarthritides with IL-17 targeted therapies being licensed for their treatment.There is evidence to demonstrate that IL-17 is found in RA joints and contributes to the pro-inflammatory cascade. This results in synovial hyperplasia and osteoclastogenesis thus causing joint destruction and bony erosions. AREAS COVERED: This review article summarizes trials that have studied the use of IL-17 targeted therapies in RA patients who have failed conventional synthetic disease-modifying therapy (C-DMARDS) and biologic DMARDS. EXPERT OPINION: The trials that have studied IL-17 inhibitors in RA patients have only shown a modest improvement in disease activity. In several trials, the primary endpoint was not achieved whilst in others, when comparing with existing licensed biologics for RA, did not demonstrate any superiority.Tissue Necrosis Factor-alpha (TNF-α) likely plays more of a pivotal role in the pathogenesis of RA with IL-17 having a synergistic effect. Therefore, in our opinion, IL-17 inhibitors as an independent therapy for RA are less likely to provide a cost-effective benefit. There may be scope to potentially combine it with TNF-α-inhibitors (TNF-i), but this requires further research especially with the potential concerns related to increased immunosuppression.


Sujet(s)
Antirhumatismaux , Polyarthrite rhumatoïde , Interleukine-17 , Humains , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/physiopathologie , Interleukine-17/antagonistes et inhibiteurs , Antirhumatismaux/pharmacologie , Antirhumatismaux/administration et posologie , Antirhumatismaux/effets indésirables , Animaux , Facteur de nécrose tumorale alpha/antagonistes et inhibiteurs , Produits biologiques/pharmacologie , Produits biologiques/administration et posologie , Thérapie moléculaire ciblée , Analyse coût-bénéfice
20.
J Drug Target ; 32(6): 707-723, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38652489

RÉSUMÉ

Background: Rheumatoid arthritis (RA) is a systemic autoimmune disease that significantly impacts the quality of life of those affected. Owing to the complex pathophysiology of RA, it is not possible for any singular treatment to entirely impede the progression of the disease. Hence, the current study aimed to adopt a holistic and synergistic approach towards the management of RA by means of a co-delivery strategy involving methotrexate (MTH), a conventional slow-acting anti-rheumatic drug, and baicalin (BCN), a bioactive phytochemical using a transethosomal (TRS) gel formulation.Purpose: The present study aims to evaluate the potential benefits of administering MTH and BCN in nanoparticulate form, which may lead to improved stability and solubility, as well as enhanced penetration into the arthritic tissues of interest.Methods and results: The MTH-BCN-TRS that were synthesised exhibited small particle size of 151.3 nm and polydispersity index of 0.125, as well as a favourable zeta potential of -32.22 mV. Additional assessments were conducted, including a pharmacokinetic analysis, TEM, skin permeation analysis and confocal microscopy. According to the Confocal laser scanning microscopy (CLSM) study, the formulated MTH-BCN-TRS gel exhibited superior MTH and BCN permeation through the skin layers when compared to the MTH-BCN suspension gel. The MTT experiment on Raw 264.7 and SW982 cell lines revealed a considerable reduction (p < .05) in the IC50 value of the MTH-BCN-TRS formulation (9.2 mM and 43.2 mM, respectively) in comparison to the drug suspension. According to the findings of the in vivo study, it was found that the MTH-BCN-TRS gel exhibits significantly promising anti-arthritic properties when compared to the conventional diclofenac gel. This was demonstrated through histopathological studies and radiographic analysis. Furthermore, skin irritation investigation on Wistar albino rats confirmed that the formulated MTH-BCN-TRS is a safe option for topical treatment on the skin. The present study has confirmed that the formulated TRS vesicles are a valuable carrier for the transdermal delivery of MTH and BCN, which may be used for the management of rheumatoid arthritis.


Sujet(s)
Administration par voie cutanée , Antirhumatismaux , Polyarthrite rhumatoïde , Flavonoïdes , Méthotrexate , Absorption cutanée , Méthotrexate/administration et posologie , Méthotrexate/pharmacocinétique , Méthotrexate/pharmacologie , Méthotrexate/composition chimique , Animaux , Flavonoïdes/administration et posologie , Flavonoïdes/pharmacologie , Flavonoïdes/pharmacocinétique , Polyarthrite rhumatoïde/traitement médicamenteux , Antirhumatismaux/administration et posologie , Antirhumatismaux/pharmacocinétique , Antirhumatismaux/pharmacologie , Rats , Taille de particule , Mâle , Souris , Nanoparticules/composition chimique , Rat Wistar , Systèmes de délivrance de médicaments , Liposomes , Cellules RAW 264.7
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE