Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 393
Filtrer
1.
Immun Inflamm Dis ; 12(8): e1361, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39092772

RÉSUMÉ

BACKGROUND: Temporomandibular joint osteoarthritis (TMJOA) is a degenerative cartilage disease. 17ß-estradiol (E2) aggravates the pathological process of TMJOA; however, the mechanisms of its action have not been elucidated. Thus, we investigate the influence of E2 on the cellular biological behaviors of synoviocytes and the molecular mechanisms. METHODS: Primary fibroblast-like synoviocytes (FLSs) isolated from rats were treated with TNF-α to establish cell model, and phenotypes were evaluated using cell counting kit-8, EdU, Tanswell, enzyme-linked immunosorbent assay, and quantitative real-time PCR (qPCR). The underlying mechanism of E2, FTO-mediated NLRC5 m6A methylation, was assessed using microarray, methylated RNA immunoprecipitation, qPCR, and western blot. Moreover, TMJOA-like rat model was established by intra-articular injection of monosodium iodoacetate (MIA), and bone morphology and pathology were assessed using micro-CT and H&E staining. RESULTS: The results illustrated that E2 facilitated the proliferation, migration, invasion, and inflammation of TNF-α-treated FLSs. FTO expression was downregulated in TMJOA and was reduced by E2 in FLSs. Knockdown of FTO promoted m6A methylation of NLRC5 and enhanced NLRC5 stability by IGF2BP1 recognition. Moreover, E2 promoted TMJ pathology and condyle remodeling, and increased bone mineral density and trabecular bone volume fraction, which was rescued by NLRC5 knockdown. CONCLUSION: E2 promoted the progression of TMJOA.


Sujet(s)
Alpha-ketoglutarate-dependent dioxygenase FTO , Oestradiol , Arthrose , Animaux , Rats , Oestradiol/pharmacologie , Alpha-ketoglutarate-dependent dioxygenase FTO/métabolisme , Alpha-ketoglutarate-dependent dioxygenase FTO/génétique , Arthrose/métabolisme , Arthrose/anatomopathologie , Arthrose/génétique , Évolution de la maladie , Cellules synoviales/métabolisme , Cellules synoviales/effets des médicaments et des substances chimiques , Cellules synoviales/anatomopathologie , Rat Sprague-Dawley , Modèles animaux de maladie humaine , Articulation temporomandibulaire/anatomopathologie , Articulation temporomandibulaire/métabolisme , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique , Cellules cultivées , Mâle , Adénosine/métabolisme , Adénosine/analogues et dérivés , Prolifération cellulaire/effets des médicaments et des substances chimiques
2.
Int J Rheum Dis ; 27(8): e15282, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39091178

RÉSUMÉ

OBJECTIVE: To investigate the impact of IGJ on the proliferation, inflammation, and motility of rheumatoid arthritis (RA) fibroblast-like synoviocytes and elucidate the underlying mechanism. METHODS: The expression of IGJ RA fibroblast-like synoviocytes was assessed using immunoblot and qPCR. Cell growth was evaluated using CCK-8 and FCM assays. The effects on inflammatory response were determined by ELISA and immunoblot assays. Cell motility was assessed using transwell and immunoblot assays. The mechanism was further confirmed using immunoblot assays. RESULTS: IGJ expression was found to be elevated in fibroid synovial cells of RA. IGJ ablation inhibited the growth of MH7A cells and suppressed the inflammatory response. Knockdown of IGJ also blocked cell motility. Mechanically, the knockdown of IGJ suppressed the NF-κB axis in MH7A cells. CONCLUSION: IGJ suppresses RA in fibroblast-like synoviocytes via NF-κB pathway.


Sujet(s)
Polyarthrite rhumatoïde , Mouvement cellulaire , Prolifération cellulaire , Fibroblastes , Facteur de transcription NF-kappa B , Transduction du signal , Cellules synoviales , Humains , Cellules synoviales/métabolisme , Cellules synoviales/anatomopathologie , Cellules synoviales/effets des médicaments et des substances chimiques , Polyarthrite rhumatoïde/anatomopathologie , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/génétique , Polyarthrite rhumatoïde/immunologie , Facteur de transcription NF-kappa B/métabolisme , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Cellules cultivées , Lignée cellulaire , Hyaluronoglucosaminidase
3.
Zhongguo Zhong Yao Za Zhi ; 49(11): 2906-2919, 2024 Jun.
Article de Chinois | MEDLINE | ID: mdl-39041150

RÉSUMÉ

Rheumatoid arthritis(RA) is a condition in which the joints are in a weakly acidic environment. In RA, RA fibroblastlike synoviocytes( RAFLS) in the joints become abnormally activated and secrete a large amount of matrix metalloproteinases(MMPs), and the receptor protein CD44 on the cell membrane is specifically upregulated. Xuetongsu(XTS), an active ingredient in the Tujia ethnomedicine Xuetong, is known to inhibit the proliferation of RAFLS. However, its development and utilization have been limited due to poor targeting ability. A biomimetic XTS-Prussian blue nanoparticles(PB NPs) drug delivery system called THMPX NPs which can target CD44 was constructed in this study. The surface of THMPX NPs was modified with hyaluronic acid(HA) and a long chain of triglycerol monostearate(TGMS) and 3-aminobenzeneboronic acid(PBA)(PBA-TGMS). The overexpressed MMPs and H+ in inflammatory RAFLS can synergistically cleave the PBA-TGMS on the surface of the nanoparticles, exposing HA to interact with CD44. This allows THMPX NPs to accumulate highly in RAFLS, and upon near-infrared light irradiation, generate heat and release XTS, thereby inhibiting the proliferation and migration of RAFLS. Characterization revealed that THMPX NPs were uniform cubes with a diameter of(190. 3±4. 7) nm and an average potential of(-15. 3± 2. 3) m V. Upon near-infrared light irradiation for 5 min, the temperature of THMPX NPs reached 41. 5 ℃, indicating MMPs and H+-triggered drug release. Safety assessments showed that THMPX NPs had a hemolysis rate of less than 4% and exhibited no cytotoxicity against normal RAW264. 7 and human fibroblast-like synoviocytes(HFLS). In vitro uptake experiments demonstrated the significant targeting ability of THMPX NPs to RAFLS. Free radical scavenging experiments revealed excellent free radical clearance capacity of THMPX NPs, capable of removing reactive oxygen species in RAFLS. Cell counting kit-8 and scratch assays demonstrated that THMPX NPs significantly suppressed the viability and migratory ability of RAFLS. This study provides insights into the development of innovative nanoscale targeted drugs from traditional ethnic medicines for RA treatment.


Sujet(s)
Mouvement cellulaire , Prolifération cellulaire , Matrix metalloproteinases , Nanoparticules , Prolifération cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des radiations , Nanoparticules/composition chimique , Humains , Mouvement cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des radiations , Matrix metalloproteinases/métabolisme , Matrix metalloproteinases/génétique , Hexacyanoferrates II/composition chimique , Concentration en ions d'hydrogène , Cellules synoviales/effets des médicaments et des substances chimiques , Cellules synoviales/effets des radiations , Cellules synoviales/métabolisme , Lasers , Antigènes CD44/métabolisme , Antigènes CD44/génétique , Médicaments issus de plantes chinoises/composition chimique , Médicaments issus de plantes chinoises/pharmacologie , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/métabolisme
4.
Zhongguo Zhong Yao Za Zhi ; 49(11): 3081-3094, 2024 Jun.
Article de Chinois | MEDLINE | ID: mdl-39041168

RÉSUMÉ

The effect and mechanism of Huangqin Qingre Chubi Capsules(HQC) on rheumatoid arthritis(RA) were studied.Seventy male SPF rats were randomly divided into normal group, model group, low-(0. 18 g·kg~(-1)), middle-(0. 36 g·kg~(-1)), and high-(0. 72 g·kg~(-1)) dose groups of HQC, methotrexate group(MTX, 0. 75 mg·kg~(-1)), and negative control group(NC group, model +saline). Adjuvant arthritis fibroblast-like synoviocytes(AA-FLS) were divided into normal group, model group, low-, middle-, and high-dose groups of HQC, and negative control group. RT-qPCR and Western blot were used to detect the m RNA and protein expressions of METTL3, SFRP4, ß-catenin, CCND1, c-Myc, MMP3, and fibronectin. The protein expression of MMP3 and ß-catenin was detected by immunofluorescence. The gene expression level of METTL3 on AA-FLS was knocked down to further examine the expression of each gene. ELISA measured the levels of IL-1ß, IL-6, and IL-8. The results showed that compared with the normal group, rats in the model group found redness and swelling in their limbs and significantly increased joint swelling. Compared with the model group, the joint swelling degree of each treatment group significantly decreased(P<0. 05). The paw retraction threshold and body weight mass index both significantly increased(P<0. 05). METTL3 was highly expressed on AA and negatively correlated with the expression of SFRP4. After treatment, the m RNA and protein expression of METTL3, ß-catenin, CCND1, c-Myc, fibronectin, and MMP3 were significantly decreased on AA-FLS(P< 0. 05). Compared with the model group, knocking down METTL3 resulted in reduced m RNA and protein expression of ß-catenin, CCND1, c-Myc, fibronectin, and MMP3(P< 0. 05). At the same time, the m RNA and protein expressions of ß-catenin, CCND1, c-Myc, fibronectin, and MMP3 in the HQC+METTL3 knockdown group were significantly lower than those in the METTL3 knockdown group(P<0. 05). HQC could reduce the levels of IL-1ß, IL-6, and IL-8 to varying degrees(P<0. 05). The results indicate that HQC has a significant improvement effect on arthritis in AA rats. The expression of METTL3 is significantly increased in synovial tissue and AA-FLS of AA rats, which may be a potential target for the diagnosis and treatment of RA. HQC improves RA through the METTL3-SFRP4/Wnt/ß-catenin signaling pathway and has significant antiinflammatory and anti-rheumatic effects.


Sujet(s)
Polyarthrite rhumatoïde , Capsules , Médicaments issus de plantes chinoises , Voie de signalisation Wnt , bêta-Caténine , Animaux , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/génétique , Médicaments issus de plantes chinoises/administration et posologie , Médicaments issus de plantes chinoises/pharmacologie , Rats , Mâle , Voie de signalisation Wnt/effets des médicaments et des substances chimiques , bêta-Caténine/métabolisme , bêta-Caténine/génétique , Methyltransferases/génétique , Methyltransferases/métabolisme , Humains , Rat Sprague-Dawley , Cellules synoviales/effets des médicaments et des substances chimiques , Cellules synoviales/métabolisme , Protéines proto-oncogènes
5.
Discov Med ; 36(186): 1441-1452, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39054715

RÉSUMÉ

BACKGROUND: Synovial inflammation plays a crucial role in osteoarthritis (OA). Gastrodin (GAS), an active ingredient derived from the Gastrodia elata Blume rhizome, possesses antioxidant and anti-inflammatory pharmacological effects. This research aimed to evaluate the function and molecular mechanism of GAS on human fibroblast-like synoviocytes of osteoarthritis (HFLS-OA) induced by interleukin (IL)-1ß. METHODS: The impact of GAS on the viability of IL-1ß-treated HFLS-OA cells was assessed using the cell counting kit-8 (CCK-8). Quantitative real-time reverse transcription PCR (qRT-PCR) was employed to detect changes in IL-8, IL-6, monocyte chemotactic protein-1 (MCP-1), tumor necrosis factor (TNF)-α, and Gremlin-1 mRNA expression in each group. Corresponding kits were utilized to measure the catalase (CAT) and superoxide dismutase (SOD) activities, as well as the nitric oxide (NO) level. Western blot analysis was conducted to examine the expression of extracellular matrix degradation-associated proteins and nuclear factor kappa-B (NF-κB) pathway-correlated proteins in each group. RESULTS: GAS significantly promoted the proliferation of IL-1ß-induced HFLS-OA cells and concurrently down-regulated Gremlin-1 mRNA expression (p < 0.05). Through the down-regulation of Gremlin-1 expression, GAS exhibited the following effects: decreased IL-8, IL-6, and TNF-α mRNA expression, as well as NO levels (p < 0.05); increased SOD and CAT activities (p < 0.05); down-regulated matrix metallopeptidase 13 (MMP-13) and MMP-1 protein expression levels (p < 0.01); and up-regulated collagen II protein expression level (p < 0.01) in IL-1ß-treated HFLS-OA cells. Additionally, GAS decreased phospho-inhibitory kappa B (p-IκB)/IκB, phospho-inhibitory kappa B kinase (p-IKK)/IKK, and p-p65/p65 ratios in IL-1ß-induced HFLS-OA cells by inhibiting Gremlin-1 expression (p < 0.01). CONCLUSION: GAS demonstrates a positive impact on inflammation, oxidative stress, and extracellular matrix degradation in IL-1ß-mediated HFLS-OA cells. This effect is achieved by suppressing Gremlin-1 expression and reducing NF-κB pathway activity.


Sujet(s)
Alcools benzyliques , Matrice extracellulaire , Glucosides , Inflammation , Interleukine-1 bêta , Facteur de transcription NF-kappa B , Stress oxydatif , Cellules synoviales , Humains , Glucosides/pharmacologie , Interleukine-1 bêta/métabolisme , Alcools benzyliques/pharmacologie , Facteur de transcription NF-kappa B/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Inflammation/métabolisme , Inflammation/anatomopathologie , Matrice extracellulaire/métabolisme , Matrice extracellulaire/effets des médicaments et des substances chimiques , Cellules synoviales/effets des médicaments et des substances chimiques , Cellules synoviales/métabolisme , Cellules synoviales/anatomopathologie , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Arthrose/anatomopathologie , Arthrose/métabolisme , Arthrose/traitement médicamenteux , Protéines et peptides de signalisation intercellulaire
7.
Cell Biochem Funct ; 42(5): e4091, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38973151

RÉSUMÉ

The intron retention (IR) is a phenomenon utilized by cells to allow diverse fates at the same mRNA, leading to a different pattern of synthesis of the same protein. In this study, we analyzed the modulation of phosphoinositide-specific phospholipase C (PI-PLC) enzymes by Harpagophytum procumbens extract (HPE) in synoviocytes from joins of osteoarthritis (OA) patients. In some samples, the PI-PLC γ1 isoform mature mRNA showed the IR and, in these synoviocytes, the HPE treatment increased the phenomenon. Moreover, we highlighted that as a consequence of IR, a lower amount of PI-PLC γ1 was produced. The decrease of PI-PLC γ1 was associated with the decrease of metalloprotease-3 (MMP-3), and MMP-13, and ADAMTS-5 after HPE treatment. The altered expression of MMPs is a hallmark of the onset and progression of OA, thus substances able to decrease their expression are very desirable. The interesting outcomes of this study are that 35% of analyzed synovial tissues showed the IR phenomenon in the PI-PLC γ1 mRNA and that the HPE treatment increased this phenomenon. For the first time, we found that the decrease of PI-PLC γ1 protein in synoviocytes interferes with MMP production, thus affecting the pathways involved in the MMP expression. This finding was validated by the silencing of PI-PLC γ1 in synoviocytes where the IR phenomenon was not present. Our results shed new light on the biochemical mechanisms involved in the degrading enzyme production in the joint of OA patients, suggesting a new therapeutic target and highlighting the importance of personalized medicine.


Sujet(s)
Fibroblastes , Introns , Phospholipase C gamma , ARN messager , Humains , ARN messager/métabolisme , ARN messager/génétique , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Phospholipase C gamma/métabolisme , Phospholipase C gamma/génétique , Cellules cultivées , Arthrose/métabolisme , Arthrose/anatomopathologie , Membrane synoviale/métabolisme , Membrane synoviale/cytologie , Membrane synoviale/effets des médicaments et des substances chimiques , Matrix metalloproteinase 3/métabolisme , Matrix metalloproteinase 3/génétique , Protéine ADAMTS5/métabolisme , Protéine ADAMTS5/génétique , Cellules synoviales/métabolisme , Cellules synoviales/effets des médicaments et des substances chimiques , Matrix Metalloproteinase 13/métabolisme , Matrix Metalloproteinase 13/génétique
8.
J Nanobiotechnology ; 22(1): 383, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38951875

RÉSUMÉ

The characteristic features of the rheumatoid arthritis (RA) microenvironment are synovial inflammation and hyperplasia. Therefore, there is a growing interest in developing a suitable therapeutic strategy for RA that targets the synovial macrophages and fibroblast-like synoviocytes (FLSs). In this study, we used graphene oxide quantum dots (GOQDs) for loading anti-arthritic sinomenine hydrochloride (SIN). By combining with hyaluronic acid (HA)-inserted hybrid membrane (RFM), we successfully constructed a new nanodrug system named HA@RFM@GP@SIN NPs for target therapy of inflammatory articular lesions. Mechanistic studies showed that this nanomedicine system was effective against RA by facilitating the transition of M1 to M2 macrophages and inhibiting the abnormal proliferation of FLSs in vitro. In vivo therapeutic potential investigation demonstrated its effects on macrophage polarization and synovial hyperplasia, ultimately preventing cartilage destruction and bone erosion in the preclinical models of adjuvant-induced arthritis and collagen-induced arthritis in rats. Metabolomics indicated that the anti-arthritic effects of HA@RFM@GP@SIN NPs were mainly associated with the regulation of steroid hormone biosynthesis, ovarian steroidogenesis, tryptophan metabolism, and tyrosine metabolism. More notably, transcriptomic analyses revealed that HA@RFM@GP@SIN NPs suppressed the cell cycle pathway while inducing the cell apoptosis pathway. Furthermore, protein validation revealed that HA@RFM@GP@SIN NPs disrupted the excessive growth of RAFLS by interfering with the PI3K/Akt/SGK/FoxO signaling cascade, resulting in a decline in cyclin B1 expression and the arrest of the G2 phase. Additionally, considering the favorable biocompatibility and biosafety, these multifunctional nanoparticles offer a promising therapeutic approach for patients with RA.


Sujet(s)
Polyarthrite rhumatoïde , Prolifération cellulaire , Graphite , Macrophages , Morphinanes , Boîtes quantiques , Cellules synoviales , Morphinanes/pharmacologie , Morphinanes/composition chimique , Animaux , Boîtes quantiques/composition chimique , Boîtes quantiques/usage thérapeutique , Polyarthrite rhumatoïde/traitement médicamenteux , Cellules synoviales/effets des médicaments et des substances chimiques , Cellules synoviales/métabolisme , Graphite/composition chimique , Graphite/pharmacologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Rats , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Mâle , Arthrite expérimentale/traitement médicamenteux , Arthrite expérimentale/anatomopathologie , Rat Sprague-Dawley , Souris , Humains , Cellules RAW 264.7 , Acide hyaluronique/composition chimique , Acide hyaluronique/pharmacologie
9.
PLoS One ; 19(7): e0302119, 2024.
Article de Anglais | MEDLINE | ID: mdl-39083495

RÉSUMÉ

Rheumatoid arthritis (RA) is an autoimmune disease characterized by joint inflammation and swelling. Several studies have demonstrated that RA fibroblast-like synovial cells (RA-FLS) play an important role in RA pathogenesis. Activated RA-FLS contribute to synovial inflammation by secreting inflammatory cytokines including interleukin (IL)-1ß, IL-6 and tumor necrosis factor-α. LMT-28 is derivative of oxazolidone and exerts anti-inflammatory effects on RA via IL-6 signaling pathway regulation. LMT-28 also regulates T cell differentiation in RA condition. However, the effect of LMT-28 on the migration and invasion of RA-FLS remains unknown. Kaempferol has been reported to have pharmacological effects on various diseases, such as inflammatory diseases, autoimmune diseases, and cancer. Additionally, kaempferol has been reported to inhibit RA-FLS migration and invasion, but it is not known about the therapeutic mechanism including molecular mechanism such as receptor. The present study aimed to investigate the synergistic effects of the combined treatment of LMT-28 and kaempferol on RA-FLS activation and RA pathogenesis in mouse model. LMT-28 and kaempferol co-administration inhibited RA disease severity and histological collapse in the joint tissues of CIA mice, as well as downregulated the levels of pro-inflammatory cytokines in mouse serum. Additionally, the combined treatment inhibited excessive differentiation of T helper 17 cells and osteoclasts. Furthermore, compared with single treatments, combined treatment showed enhanced inhibitory effects on the hyperactivation of IL-6-induced signaling pathway in RA-FLS. Combined treatment also inhibited RA-FLS cell proliferation, migration, and invasion and suppressed the expression of matrix metalloproteinase in RA-FLS. Furthermore, we confirmed that the combined treatment inhibited chondrocyte proliferation, migration, and invasion. In conclusion, our results suggest that the combined treatment of LMT-28 and kaempferol exerts a synergistic effect on the RA development via the regulation of IL-6-induced hyperactivation of RA-FLS. Furthermore, this study suggests that combination therapies can be an effective therapeutic option for arthritis.


Sujet(s)
Anti-inflammatoires , Arthrite expérimentale , Kaempférols , Animaux , Kaempférols/pharmacologie , Kaempférols/usage thérapeutique , Kaempférols/administration et posologie , Souris , Arthrite expérimentale/traitement médicamenteux , Arthrite expérimentale/anatomopathologie , Arthrite expérimentale/métabolisme , Anti-inflammatoires/pharmacologie , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/anatomopathologie , Polyarthrite rhumatoïde/métabolisme , Souris de lignée DBA , Modèles animaux de maladie humaine , Mâle , Mouvement cellulaire/effets des médicaments et des substances chimiques , Interleukine-6/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cytokines/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Différenciation cellulaire/effets des médicaments et des substances chimiques , Association de médicaments , Cellules synoviales/effets des médicaments et des substances chimiques , Cellules synoviales/métabolisme
10.
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
11.
Adv Rheumatol ; 64(1): 46, 2024 06 07.
Article de Anglais | MEDLINE | ID: mdl-38849923

RÉSUMÉ

BACKGROUND: Fibroblast-like synoviocytes (FLSs) are involved in osteoarthritis (OA) pathogenesis through pro-inflammatory cytokine production. TAK-242, a TLR4 blocker, has been found to have a significant impact on the gene expression profile of pro-inflammatory cytokines such as IL1-ß, IL-6, TNF-α, and TLR4, as well as the phosphorylation of Ikßα, a regulator of the NF-κB signaling pathway, in OA-FLSs. This study aims to investigate this effect because TLR4 plays a crucial role in inflammatory responses. MATERIALS AND METHODS: Ten OA patients' synovial tissues were acquired, and isolated FLSs were cultured in DMEM in order to assess the effectiveness of TAK-242. The treated FLSs with TAK-242 and Lipopolysaccharides (LPS) were analyzed for the mRNA expression level of IL1-ß, IL-6, TNF-α, and TLR4 levels by Real-Time PCR. Besides, we used western blot to assess the protein levels of Ikßα and pIkßα. RESULTS: The results represented that TAK-242 effectively suppressed the gene expression of inflammatory cytokines IL1-ß, IL-6, TNF-α, and TLR4 which were overexpressed upon LPS treatment. Additionally, TAK-242 inhibited the phosphorylation of Ikßα which was increased by LPS treatment. CONCLUSION: According to our results, TAK-242 shows promising inhibitory effects on TLR4-mediated inflammatory responses in OA-FLSs by targeting the NF-κB pathway. TLR4 inhibitors, such as TAK-242, may be useful therapeutic agents to reduce inflammation and its associated complications in OA patients, since traditional and biological treatments may not be adequate for all of them.


Sujet(s)
Cytokines , Interleukine-1 bêta , Interleukine-6 , Lipopolysaccharides , Facteur de transcription NF-kappa B , Transduction du signal , Sulfonamides , Cellules synoviales , Récepteur de type Toll-4 , Facteur de nécrose tumorale alpha , Humains , Transduction du signal/effets des médicaments et des substances chimiques , Cellules synoviales/effets des médicaments et des substances chimiques , Cellules synoviales/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Sulfonamides/pharmacologie , Sulfonamides/usage thérapeutique , Récepteur de type Toll-4/métabolisme , Cytokines/métabolisme , Interleukine-6/métabolisme , Interleukine-1 bêta/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Lipopolysaccharides/pharmacologie , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Arthrose/métabolisme , Arthrose/traitement médicamenteux , Cellules cultivées , Phosphorylation , ARN messager/métabolisme , Mâle , Femelle , Adulte d'âge moyen
12.
Int Immunopharmacol ; 137: 112394, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-38852517

RÉSUMÉ

BACKGROUND: Ferroptosis is a distinct iron-dependent non-apoptotic type of programmed cell death that is implicated in the pathophysiology of rheumatoid arthritis (RA). Although asiatic acid (AA) is documented to have significant anti-inflammatory effects in various diseases, it is not known whether it can regulate RA via ferroptosis. METHODS: The effects of AA on rheumatoid arthritis fibroid-like synoviocytes (RA-FLS) were assessed in vitro, and a rat model of type II collagen-induced arthritis (CIA) was established to evaluate the effectiveness of AA treatment in vivo. RESULTS: AA significantly reduced both viability and colony formation in cultured RA-FLS, while increasing the levels of reactive oxygen species (ROS), ferrous iron (Fe2+), malondialdehyde (MDA), and lactate dehydrogenase (LDH), as well as the expression of COX2. Furthermore, AA induced ferroptosis in RA-FLS by promoting Fe2+ accumulation through downregulation of the expression of Keap1 and FTH1 and upregulation of Nrf2 and HMOX1. In vivo, AA treatment was found to reduce toe swelling and the arthritis score in CIA rats, as well as relieve inflammation and ankle damage and significantly upregulate the expression of Nrf2 and HMOX1 in the synovial fluid. CONCLUSION: Treatment with AA significantly reduced the viability of RA-FLS and triggered ferroptosis by promoting accumulation of Fe2+via the Nrf2-HMOX1 pathway, and was effective in relieving inflammation in CIA model rats. These findings suggest that the use of AA may be a promising strategy for the clinical treatment of RA.


Sujet(s)
Arthrite expérimentale , Polyarthrite rhumatoïde , Ferroptose , Facteur-2 apparenté à NF-E2 , Triterpènes pentacycliques , Transduction du signal , Cellules synoviales , Animaux , Ferroptose/effets des médicaments et des substances chimiques , Polyarthrite rhumatoïde/traitement médicamenteux , Facteur-2 apparenté à NF-E2/métabolisme , Triterpènes pentacycliques/usage thérapeutique , Triterpènes pentacycliques/pharmacologie , Arthrite expérimentale/traitement médicamenteux , Humains , Rats , Transduction du signal/effets des médicaments et des substances chimiques , Cellules synoviales/effets des médicaments et des substances chimiques , Cellules synoviales/métabolisme , Mâle , Cellules cultivées , Anti-inflammatoires/usage thérapeutique , Anti-inflammatoires/pharmacologie , Heme oxygenase-1/métabolisme , Rat Sprague-Dawley , Espèces réactives de l'oxygène/métabolisme , Heme oxygenase (decyclizing)
13.
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
14.
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
15.
Clin Immunol ; 264: 110255, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38763433

RÉSUMÉ

Fibroblast-like synoviocytes (FLS) play critical roles in rheumatoid arthritis (RA). Itaconate (ITA), an endogenous metabolite derived from the tricarboxylic acid (TCA) cycle, has attracted attention because of its anti-inflammatory, antiviral, and antimicrobial effects. This study evaluated the effect of ITA on FLS and its potential to treat RA. ITA significantly decreased FLS proliferation and migration in vitro, as well as mitochondrial oxidative phosphorylation and glycolysis measured by an extracellular flux analyzer. ITA accumulates metabolites including succinate and citrate in the TCA cycle. In rats with type II collagen-induced arthritis (CIA), intra-articular injection of ITA reduced arthritis and bone erosion. Irg1-deficient mice lacking the ability to produce ITA had more severe arthritis than control mice in the collagen antibody-induced arthritis. ITA ameliorated CIA by inhibiting FLS proliferation and migration. Thus, ITA may be a novel therapeutic agent for RA.


Sujet(s)
Arthrite expérimentale , Polyarthrite rhumatoïde , Mouvement cellulaire , Prolifération cellulaire , Fibroblastes , Succinates , Cellules synoviales , Animaux , Cellules synoviales/effets des médicaments et des substances chimiques , Cellules synoviales/métabolisme , Mouvement cellulaire/effets des médicaments et des substances chimiques , Arthrite expérimentale/traitement médicamenteux , Arthrite expérimentale/métabolisme , Arthrite expérimentale/anatomopathologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Succinates/pharmacologie , Rats , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Mâle , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/métabolisme , Souris , Souris knockout , Cellules cultivées , Souris de lignée DBA , Cycle citrique/effets des médicaments et des substances chimiques
16.
Zhongguo Zhong Yao Za Zhi ; 49(7): 1947-1955, 2024 Apr.
Article de Chinois | MEDLINE | ID: mdl-38812207

RÉSUMÉ

This study aims to decipher the mechanism of sinomenine in inhibiting platelet-derived growth factor/platelet-derived growth factor receptor(PDGF/PDGFR) signaling pathway in rheumatoid arthritis-fibroblast-like synoviocyte(RA-FLS) migration induced by neutrophil extracellular traps(NETs). RA-FLS was isolated from the synovial tissue of 3 RA patients and cultured. NETs were extracted from the peripheral venous blood of 4 RA patients and 4 healthy control(HC). RA-FLS was classified into control group, HC-NETs group, RA-NETs group, RA-NETs+sinomenine group and RA-NETs+sinomenine+CP-673451 group. RNA-sequencing(RNA-seq) was conducted to identify the differentially expressed genes between HC-NETs and RA-NETs groups. Sangerbox was used to perform the Gene Ontology(GO) function and the Kyoto Encyclopedia of Genes and Genomes(KEGG) pathway enrichment. Cytoscape was employed to build the protein-protein interaction(PPI) network. AutoDock Vina and PyMOL were used for molecular docking of sinomenine with PDGFß and PDGFRß. The cell proliferation and migration were determined by the cell counting kit-8(CCK-8) and cell scratch assay, respectively. Western blot was employed to determine the protein level of PDGFRß. Real-time quantitative polymerase chain reaction(RT-qPCR) was carried out to determine the mRNA levels of matrix metalloproteinases(MMPs). The results revealed that neutrophils in RA patients were more likely to produce NETs. Compared with HC-NETs group, RA-NETs group showed up-regulated expression of PDGFß and PDGFRß. Compared with control group, RA-NETs group showed increased cell proliferation and migration and up-regulated protein level of PDGFRß and mRNA levels of PDGFß, PDGFRß, MMP1, MMP3, and MMP9(P<0.05). Compared with RA-NETs group, RA-NETs+sinomenine group presented decreased cell proliferation and migration and down-regulated protein and mRNA level of PDGFRß and mRNA levels of MMP1, MMP3, and MMP9(P<0.05). Compared with RA-NETs+sinomenine group, the proliferation ability of RA-NETs+sinomenine+CP-673451 group decreased(P<0.05). The findings prove that sinomenine reduces the RA-NETs-induced RA-FLS migration by inhibiting PDGF/PDGFR signaling pathway, thus mitigating RA.


Sujet(s)
Polyarthrite rhumatoïde , Mouvement cellulaire , Morphinanes , Facteur de croissance dérivé des plaquettes , Transduction du signal , Cellules synoviales , Humains , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/génétique , Polyarthrite rhumatoïde/métabolisme , Mouvement cellulaire/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Morphinanes/pharmacologie , Cellules synoviales/effets des médicaments et des substances chimiques , Cellules synoviales/métabolisme , Facteur de croissance dérivé des plaquettes/génétique , Facteur de croissance dérivé des plaquettes/métabolisme , Récepteurs aux facteurs de croissance dérivés des plaquettes/génétique , Récepteurs aux facteurs de croissance dérivés des plaquettes/métabolisme , Granulocytes neutrophiles/effets des médicaments et des substances chimiques , Granulocytes neutrophiles/métabolisme , Mâle , Femelle , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme
17.
Bone Res ; 12(1): 31, 2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-38782893

RÉSUMÉ

Rheumatoid arthritis (RA) is an autoimmune disease. Early studies hold an opinion that gut microbiota is environmentally acquired and associated with RA susceptibility. However, accumulating evidence demonstrates that genetics also shape the gut microbiota. It is known that some strains of inbred laboratory mice are highly susceptible to collagen-induced arthritis (CIA), while the others are resistant to CIA. Here, we show that transplantation of fecal microbiota of CIA-resistant C57BL/6J mice to CIA-susceptible DBA/1J mice confer CIA resistance in DBA/1J mice. C57BL/6J mice and healthy human individuals have enriched B. fragilis than DBA/1J mice and RA patients. Transplantation of B. fragilis prevents CIA in DBA/1J mice. We identify that B. fragilis mainly produces propionate and C57BL/6J mice and healthy human individuals have higher level of propionate. Fibroblast-like synoviocytes (FLSs) in RA are activated to undergo tumor-like transformation. Propionate disrupts HDAC3-FOXK1 interaction to increase acetylation of FOXK1, resulting in reduced FOXK1 stability, blocked interferon signaling and deactivation of RA-FLSs. We treat CIA mice with propionate and show that propionate attenuates CIA. Moreover, a combination of propionate with anti-TNF etanercept synergistically relieves CIA. These results suggest that B. fragilis or propionate could be an alternative or complementary approach to the current therapies.


Sujet(s)
Arthrite expérimentale , Polyarthrite rhumatoïde , Microbiome gastro-intestinal , Histone deacetylases , Souris de lignée C57BL , Cellules synoviales , Animaux , Humains , Mâle , Souris , Arthrite expérimentale/anatomopathologie , Arthrite expérimentale/métabolisme , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/anatomopathologie , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/microbiologie , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Facteurs de transcription Forkhead/métabolisme , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Histone deacetylases/métabolisme , Souris de lignée DBA , Transduction du signal/effets des médicaments et des substances chimiques , Cellules synoviales/métabolisme , Cellules synoviales/effets des médicaments et des substances chimiques , Cellules synoviales/anatomopathologie
18.
Gene ; 920: 148538, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-38710294

RÉSUMÉ

To explore whether Fengshi Liuhe Decoction (FLD) alleviates rheumatoid arthritis (RA) via the Fzd6/NF-κB signaling axis. We used real-time quantitative PCR (qPCR) and western blotting (WB) to determine the genes of the frizzled (Fzd) protein 1- Fzd protein 10 that are significantly differentially expressed between normal rat fibroblast-like synoviocyte (FLS) and collagen II-induced arthritis (CIA) rat FLS. Next, we used enzyme-linked immunosorbent assay (ELISA) to evaluate the levels of inflammatory factors in cell culture supernatant to determine the ability of FLD to ameliorate RA. Finally, we employed WB to detect the key gene expression in protein levels of the Fzd6/NF-κB signaling axis among normal rat FLS, CIA rat FLS, and FLD-treated CIA rat FLS. Our results showed that Fzd6 expression was significantly higher in CIA rat FLS at both the mRNA and protein levels than in normal rat FLS. FLD was found to downregulate Fzd6 and inflammatory factors, including COX-2, IL-8, and TNF-α, at both the mRNA and protein levels. FLD was also found to downregulate the total protein levels of Fzd6 and the NF-κB signaling pathway key gene phosphorylation of p-p65/p65 and p-IκBα/IκBα. Moreover, FLD inhibited the nuclear translocation of NF-κB p65 in CIA rat FLS. FLD can alleviate inflammation of CIA rat FLS via the Fzd6/NF-κB signaling axis.


Sujet(s)
Polyarthrite rhumatoïde , Médicaments issus de plantes chinoises , Récepteurs Frizzled , Facteur de transcription NF-kappa B , Transduction du signal , Animaux , Transduction du signal/effets des médicaments et des substances chimiques , Rats , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/génétique , Facteur de transcription NF-kappa B/métabolisme , Récepteurs Frizzled/métabolisme , Récepteurs Frizzled/génétique , Médicaments issus de plantes chinoises/pharmacologie , Médicaments issus de plantes chinoises/usage thérapeutique , Arthrite expérimentale/traitement médicamenteux , Arthrite expérimentale/métabolisme , Arthrite expérimentale/génétique , Cellules synoviales/métabolisme , Cellules synoviales/effets des médicaments et des substances chimiques , Mâle , Cellules cultivées
19.
Eur J Pharmacol ; 972: 176551, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38570082

RÉSUMÉ

Fibroblast-like synoviocytes (FLS) play an important role in rheumatoid arthritis (RA)-related swelling and bone damage. Therefore, novel targets for RA therapy in FLS are urgently discovered for improving pathologic phenomenon, especially joint damage and dyskinesia. Here, we suggested that pyruvate kinase M2 (PKM2) in FLS represented a pharmacological target for RA treatment by antimalarial drug artemisinin (ART). We demonstrated that ART selectively inhibited human RA-FLS and rat collagen-induced arthritis (CIA)-FLS proliferation and migration without observed toxic effects. In particular, the identification of targets revealed that PKM2 played a crucial role as a primary regulator of the cell cycle, leading to the heightened proliferation of RA-FLS. ART exhibited a direct interaction with PKM2, resulting in an allosteric modulation that enhances the lactylation modification of PKM2. This interaction further promoted the binding of p300, ultimately preventing the nuclear translocation of PKM2 and inducing cell cycle arrest at the S phase. In vivo, ART obviously suppressed RA-mediated synovial hyperplasia, bone damage and inflammatory response to further improve motor behavior in CIA-rats. Taken together, these findings indicate that directing interventions towards PKM2 in FLS could offer a hopeful avenue for pharmaceutical treatments of RA through the regulation of cell cycle via PKM2 lactylation.


Sujet(s)
Polyarthrite rhumatoïde , Prolifération cellulaire , Cellules synoviales , Cellules synoviales/effets des médicaments et des substances chimiques , Cellules synoviales/métabolisme , Cellules synoviales/anatomopathologie , Polyarthrite rhumatoïde/anatomopathologie , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/métabolisme , Animaux , Prolifération cellulaire/effets des médicaments et des substances chimiques , Humains , Rats , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Pyruvate kinase/métabolisme , , Mâle , Hormones thyroïdiennes/métabolisme , Arthrite expérimentale/anatomopathologie , Arthrite expérimentale/traitement médicamenteux , Arthrite expérimentale/métabolisme , Mouvement cellulaire/effets des médicaments et des substances chimiques , Thérapie moléculaire ciblée , Protéines membranaires/métabolisme , Protéines de transport/métabolisme , Bibliothèques de petites molécules/pharmacologie , Bibliothèques de petites molécules/composition chimique
20.
Biomater Adv ; 160: 213853, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38636119

RÉSUMÉ

Patients with rheumatoid arthritis (RA) often have one or more painfuljoints despite adequate medicine. Local drug delivery to the synovial cavity bids for high drug concentration with minimal systemic adverse effects. However, anti-RA drugs show short half-lives in inflamed joints after intra-articular delivery. To improve the therapeutic efficacy, it is essential to ensure that a drug is only released from the formulation when it is needed. In this work, we developed an intelligent "Self-actuating" drug delivery system where Disease-modifying anti-rheumatic Drug (DMARD) methotrexate is incorporated within a matrix intended to be injected directly into joints. This formulation has the property to sense the need and release medication only when joints are inflamed in response to inflammatory enzyme Matrix metalloproteinases (MMP). These enzymes are important proteases in RA pathology, and several MMP are present in augmented levels in synovial fluid and tissues. A high level of MMP present in synovial tissues of RA patients would facilitate the release of drugs in response and ascertain controlled drug release. The formulation is designed to be stable within the joint environment, but to dis-assemble in response to inflammation. The synthesized enzyme-responsive methotrexate (Mtx) encapsulated micron-sized polymer-lipid hybrid hydrogel microspheres (Mtx-PLHM) was physiochemically characterized and tested in synovial fluid, Human Fibroblast like synoviocytes (h-FLS) (derived from RA patients) and a rat arthritic animal model. Mtx-PLHM can self-actuate and augment the release of Mtx drug upon contact with either exogenously added MMP or endogenous MMP present in the synovial fluid of patients with RA. The drug release from the prepared formulation is significantly amplified to several folds in the presence of MMP-2 and MMP-9 enzymes. In the rat arthritic model, Mtx-PLHM showed promising therapeutic results with the significant alleviation of RA symptoms through decrease in joint inflammation, swelling, bone erosion, and joint damage examined by X-ray analysis, histopathology and immune-histology. This drug delivery system would be nontoxic as it releases more drug only during the period of exacerbation of inflammation. This will simultaneously protect patients from unwanted side effects when the disease is inactive and lower the need for repeated joint injections.


Sujet(s)
Antirhumatismaux , Polyarthrite rhumatoïde , Préparations à action retardée , Hydrogels , Méthotrexate , Microsphères , Cellules synoviales , Animaux , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/anatomopathologie , Humains , Méthotrexate/pharmacologie , Méthotrexate/usage thérapeutique , Méthotrexate/composition chimique , Méthotrexate/administration et posologie , Hydrogels/composition chimique , Cellules synoviales/effets des médicaments et des substances chimiques , Cellules synoviales/métabolisme , Cellules synoviales/anatomopathologie , Rats , Antirhumatismaux/pharmacologie , Antirhumatismaux/administration et posologie , Antirhumatismaux/usage thérapeutique , Antirhumatismaux/pharmacocinétique , Libération de médicament , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Mâle , Inflammation/traitement médicamenteux , Inflammation/anatomopathologie , Matrix metalloproteinases/métabolisme , Synovie/effets des médicaments et des substances chimiques , Synovie/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE