Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.453
Filtrar
1.
Ter Arkh ; 96(7): 690-694, 2024 Jul 30.
Artículo en Ruso | MEDLINE | ID: mdl-39106512

RESUMEN

AIM: Evaluation in real clinical practice of the effectiveness and safety of levilimab therapy in patients with highly and moderately active rheumatoid arthritis (RA). MATERIALS AND METHODS: A prospective observational study (6 months) involving 35 patients with RA (29 women and 6 men, mean age 53.17±13.2 years) who were treated at the Ochapovsky Regional Clinic Hospital of Krasnodar Region. All patients included in the study were prescribed the drug levilimab (Ilsira). RESULTS: After 1 month of observation, there was a decrease in the clinical and laboratory activity of the process in the form of a decrease in the number of painful joints - 17.0 (14.0; 20.0) vs 8.0 (6.0; 10.0); p=0.000001, number of swollen joints - 3.0 (2.0; 4.0) vs 0.0 (0.0; 0.0); p=0.000002, reduction in pain intensity according to visual analog scale - 60.0 (60.0; 70.0) mm vs 30.0 (20.0; 40.0) mm (p=0.000001). Also, by the end of the first month of therapy, there was a decrease in clinical activity indices DAS28-ESR by 43%, SDAI by 60%, CDAI by 55%. Positive dynamics of laboratory parameters were noted - a decrease in erythrocyte sedimentation rate by 76%, a decrease in C-reactive protein level by 98%. By the 6th month of therapy, a decrease in RF by 36% and ACCP by 11% was recorded, but the dynamics of these indicators did not reach statistical significance. By the end of 4 weeks of treatment, 24 (68.6%) patients showed an increase in the level of total blood cholesterol - 5.1 (3.91; 6.0) mmol/L vs 6.1 (4.99; 7.07) mmol/L (p=0.000006), while 11 (45.8%) patients from this group had initially elevated cholesterol levels (6.4±0.6 mmol/L). In 5 (14.3%) patients, an increase in alanine aminotransferase (ALT) was recorded in the 4th week - 17.0 (11.0; 25.0) U/L vs 32.0 (22.0; 43.0) U/L (p=0.000062) and aspartate aminotransferase (AST) - 19.0 (14.0; 24.0) U/L vs 25.0 (18.0; 36.0) U/L (p=0.000171), in 1 (2.85%) of the patient, an increase in ALT and AST above normal was noted (ALT 144 U/L, AST 52 U/L), which required discontinuation of levilimab. In 2 (5.7%) patients, by the end of the 4th week a decrease in the absolute number of neutrophils was registered - 3.2 (2.6; 4.0)×10E9/L vs 2.3 (2.0; 2.5)×10E9/L (p=0.002), which did not require discontinuation of treatment, since the number of cells remained more than 1×10E9/L. During treatment with levilimab 162 mg subcutaneously once a week, the proportion of patients taking prednisolone decreased from 46% at the start of therapy to 11% at the end of 6 months of therapy. CONCLUSION: Levilimab is a highly effective drug for the treatment of patients with highly and moderately active RA and has a favorable tolerability and safety profile.


Asunto(s)
Artritis Reumatoide , Humanos , Artritis Reumatoide/tratamiento farmacológico , Masculino , Femenino , Persona de Mediana Edad , Resultado del Tratamiento , Estudios Prospectivos , Adulto , Antirreumáticos/administración & dosificación , Antirreumáticos/efectos adversos , Antirreumáticos/farmacología , Antirreumáticos/uso terapéutico , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anciano , Índice de Severidad de la Enfermedad
2.
Drug Des Devel Ther ; 18: 3523-3545, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39135759

RESUMEN

Purpose: Sinomenine (SIN) is commonly used in Traditional Chinese Medicine (TCM) as a respected remedy for rheumatoid arthritis (RA). Nevertheless, the therapeutic mechanism of SIN in RA remains incompletely understood. This study aimed to delve into the molecular mechanism of SIN in the treatment of RA. Methods: The potential targets of SIN were predicted using the TCMSP server, STITCH database, and SwissTarget Prediction. Differentially expressed genes (DEGs) in RA were obtained from the GEO database. Enrichment analyses and molecular docking were conducted to explore the potential mechanism of SIN in the treatment of RA. In vitro and in vivo studies were conducted to validate the intervention effects of SIN on rheumatoid arthritis, as determined through network pharmacology analyses. Results: A total of 39 potential targets associated with the therapeutic effects of SIN in RA were identified. Enrichment analysis revealed that these potential targets are primarily enriched in PI3K-Akt signaling pathway, and the molecular docking suggests that SIN may act on specific proteins in the pathway. Experimental results have shown that exposure to SIN inhibits cytokine secretion, promotes apoptosis, reduces metastasis and invasion, and blocks the activation of the PI3K-Akt signaling pathway in RA fibroblast-like synoviocytes (RA-FLS). Moreover, SIN treatment alleviated arthritis-related symptoms and regulated the differentiation of CD4+ T cells in the spleen of collagen-induced arthritis (CIA) mice. Conclusion: By utilizing network pharmacology, molecular modeling, and in vitro/in vivo validation, this study demonstrates that SIN can alleviate RA by inhibiting the PI3K-Akt signaling pathway. These findings enhance the understanding of the therapeutic mechanisms of SIN in RA, offering a stronger theoretical foundation for its future clinical application.


Asunto(s)
Artritis Reumatoide , Simulación del Acoplamiento Molecular , Morfinanos , Farmacología en Red , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Morfinanos/farmacología , Morfinanos/química , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Ratones , Animales , Transducción de Señal/efectos de los fármacos , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo , Masculino , Antirreumáticos/farmacología , Antirreumáticos/química , Células Cultivadas , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/patología , Artritis Experimental/metabolismo , Ratones Endogámicos DBA
3.
Int J Mol Sci ; 25(15)2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-39125897

RESUMEN

Rheumatoid arthritis (RA) is a highly prevalent autoimmune disorder. The pathogenesis of the disease is complex and involves various cellular populations, including fibroblast-like synoviocytes, macrophages, and T cells, among others. Identification of signalling pathways and molecules that actively contribute to the development of the disease is crucial to understanding the mechanisms involved in the chronic inflammatory environment present in affected joints. Recent studies have demonstrated that the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway regulates the behaviour of immune cells and contributes to the progression of RA. Several JAK inhibitors, such as tofacitinib, baricitinib, upadacitinib, and filgocitinib, have been developed, and their efficacy and safety in patients with RA have been comprehensively investigated in a number of clinical trials. Consequently, JAK inhibitors have been approved and registered as a treatment for patients with RA. In this review, we discuss the involvement of JAK/STAT signalling in the pathogenesis of RA and summarise the potential beneficial effects of JAK inhibitors in cells implicated in the pathogenesis of the disease. Moreover, we present the most important phase 3 clinical trials that evaluated the use of these agents in patients.


Asunto(s)
Artritis Reumatoide , Inhibidores de las Cinasas Janus , Quinasas Janus , Humanos , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/inmunología , Artritis Reumatoide/metabolismo , Inhibidores de las Cinasas Janus/uso terapéutico , Inhibidores de las Cinasas Janus/farmacología , Quinasas Janus/metabolismo , Quinasas Janus/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Factores de Transcripción STAT/metabolismo , Animales , Antirreumáticos/uso terapéutico , Antirreumáticos/farmacología
4.
Int J Biol Sci ; 20(10): 3725-3741, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39113703

RESUMEN

The probability of cardiovascular events has been reported lower in rheumatoid arthritis (RA) patients treated with leflunomide. However, the anti-atherosclerotic and cardiovascular protective effects and metabolism of leflunomide are not explored. In this study, we assessed the potential benefits of leflunomide on atherosclerosis and revealed the underlying mechanism. ApoE-/- mice were fed a western diet (WD) alone or supplemented with leflunomide (20 mg/kg, oral gavage, once per day) for 12 weeks. Samples of the aorta, heart, liver, serum, and macrophages were collected. We found that leflunomide significantly reduced lesion size in both en-face aortas and aortic root in WD-fed ApoE-/- mice. Leflunomide also obviously improved dyslipidemia, reduced hepatic lipid content, and improved disorders of glucose and lipid metabolism in vivo. RNA-Seq results showed that leflunomide effectively regulated the genes' expression involved in the lipid metabolism pathway. Importantly, leflunomide significantly increased the phosphorylation levels of AMPKα and acetyl-CoA carboxylase (ACC) in vivo. Furthermore, leflunomide and its active metabolite teriflunomide suppressed lipid accumulation in free fatty acid (FFA)-induced AML12 cells and improved endothelial dysfunction in palmitic acid (PA)-induced HUVECs through activating AMPK signaling and inhibiting dihydroorotate dehydrogenase (DHODH) signaling pathway. We present evidence that leflunomide and teriflunomide ameliorate atherosclerosis by regulating lipid metabolism and endothelial dysfunction. Our findings suggest a promising use of antirheumatic small-molecule drugs leflunomide and teriflunomide for the treatment of atherosclerosis and related cardiovascular diseases (CVDs).


Asunto(s)
Antirreumáticos , Aterosclerosis , Dihidroorotato Deshidrogenasa , Leflunamida , Metabolismo de los Lípidos , Transducción de Señal , Animales , Leflunamida/uso terapéutico , Leflunamida/farmacología , Aterosclerosis/metabolismo , Aterosclerosis/tratamiento farmacológico , Ratones , Metabolismo de los Lípidos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Dihidroorotato Deshidrogenasa/metabolismo , Antirreumáticos/farmacología , Antirreumáticos/uso terapéutico , Humanos , Proteínas Quinasas Activadas por AMP/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/metabolismo , Masculino , Ratones Endogámicos C57BL , Células Endoteliales de la Vena Umbilical Humana/metabolismo
5.
Clin Exp Pharmacol Physiol ; 51(8): e13906, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38965677

RESUMEN

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.


Asunto(s)
Antirreumáticos , Artritis Reumatoide , Oxigenoterapia Hiperbárica , Interleucina-1beta , Ratas Sprague-Dawley , Factor de Necrosis Tumoral alfa , Animales , Oxigenoterapia Hiperbárica/métodos , Masculino , Antirreumáticos/uso terapéutico , Antirreumáticos/farmacología , Artritis Reumatoide/terapia , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/patología , Artritis Reumatoide/metabolismo , Ratas , Interleucina-1beta/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Modelos Animales de Enfermedad , Etanercept/uso terapéutico , Etanercept/farmacología , Artritis Experimental/terapia , Artritis Experimental/patología , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/metabolismo , Leflunamida/uso terapéutico , Leflunamida/farmacología
6.
Drug Deliv ; 31(1): 2380538, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39044468

RESUMEN

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.


Asunto(s)
Artritis Reumatoide , Portadores de Fármacos , Compuestos de Manganeso , Metotrexato , Nanopartículas , Óxidos , Especies Reactivas de Oxígeno , Albúmina Sérica Humana , Animales , Compuestos de Manganeso/química , Óxidos/química , Artritis Reumatoide/tratamiento farmacológico , Ratones , Ratas , Nanopartículas/química , Albúmina Sérica Humana/química , Humanos , Metotrexato/farmacología , Metotrexato/administración & dosificación , Metotrexato/farmacocinética , Metotrexato/química , Especies Reactivas de Oxígeno/metabolismo , Células RAW 264.7 , Portadores de Fármacos/química , Masculino , Artritis Experimental/tratamiento farmacológico , Distribución Tisular , Antirreumáticos/farmacología , Antirreumáticos/administración & dosificación , Antirreumáticos/farmacocinética , Ratas Sprague-Dawley , Citocinas/metabolismo
7.
Clin Exp Rheumatol ; 42(7): 1387-1397, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38976290

RESUMEN

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.


Asunto(s)
Apoptosis , Artritis Experimental , Artritis Reumatoide , Autofagia , Proliferación Celular , Metotrexato , Ratas Wistar , Especies Reactivas de Oxígeno , Sinoviocitos , Tretinoina , Tretinoina/farmacología , Apoptosis/efectos de los fármacos , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/patología , Artritis Reumatoide/metabolismo , Metotrexato/farmacología , Autofagia/efectos de los fármacos , Animales , Humanos , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/patología , Artritis Experimental/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Sinoviocitos/efectos de los fármacos , Sinoviocitos/patología , Sinoviocitos/metabolismo , Proliferación Celular/efectos de los fármacos , Quimioterapia Combinada , Antirreumáticos/farmacología , Membrana Sinovial/efectos de los fármacos , Membrana Sinovial/patología , Membrana Sinovial/metabolismo , Masculino , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratas , Línea Celular
8.
Sci Rep ; 14(1): 15226, 2024 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-38956271

RESUMEN

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.


Asunto(s)
Abatacept , Antirreumáticos , Artritis Reumatoide , Interleucina-6 , Inhibidores del Factor de Necrosis Tumoral , Humanos , Artritis Reumatoide/tratamiento farmacológico , Femenino , Masculino , Interleucina-6/antagonistas & inhibidores , Interleucina-6/metabolismo , Persona de Mediana Edad , Abatacept/uso terapéutico , Antirreumáticos/uso terapéutico , Antirreumáticos/farmacología , Inhibidores del Factor de Necrosis Tumoral/uso terapéutico , Anciano , Articulación de la Rodilla/diagnóstico por imagen , Articulación de la Rodilla/patología , Articulación de la Rodilla/efectos de los fármacos , Adulto , Inhibidores de las Cinasas Janus/uso terapéutico , Inhibidores de las Cinasas Janus/farmacología , Resultado del Tratamiento , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
9.
Molecules ; 29(13)2024 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-38999143

RESUMEN

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.


Asunto(s)
Artritis Reumatoide , Biomarcadores , Simulación del Acoplamiento Molecular , Extractos Vegetales , Animales , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/sangre , Artritis Reumatoide/metabolismo , Ratas , Biomarcadores/sangre , Extractos Vegetales/química , Extractos Vegetales/farmacología , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/sangre , Artritis Experimental/metabolismo , Masculino , Modelos Animales de Enfermedad , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Antirreumáticos/farmacología , Antirreumáticos/química , Antirreumáticos/uso terapéutico , Factor A de Crecimiento Endotelial Vascular/sangre , Factor A de Crecimiento Endotelial Vascular/metabolismo
10.
Expert Opin Investig Drugs ; 33(8): 815-827, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38967534

RESUMEN

INTRODUCTION: Pulmonary involvement is one of the most common extra-articular manifestations of rheumatoid arthritis (RA), a systemic inflammatory disease characterized by joint swelling and tenderness. All lung compartments can be interested in the course of RA, including parenchyma, airways, and, more rarely, pleura and vasculature. AREAS COVERED: The aim of this paper is to review the main RA lung manifestations, focusing on pathogenesis, clinical and therapeutic issues of RA-related interstitial lung disease (ILD). Despite an increasing number of studies in the last years, pathogenesis of RA-ILD remains largely debated and the treatment of RA patients with lung involvement is still challenging in these patients. EXPERT OPINION: Management of RA-ILD is largely based on expert-opinion. Due to the broad clinical manifestations, including both joints and pulmonary involvement, multidisciplinary discussion, including rheumatologist and pulmonologist, is essential, not only for diagnosis, but also to evaluate the best therapeutic approach and follow-up. In fact, the coexistence of different lung manifestations may influence the treatment response and safety. The identification of biomarkers and risk-factors for an early identification of RA patients at risk of developing ILD remains a need that still needs to be fulfilled, and that will require further investigation in the next years.


Asunto(s)
Antirreumáticos , Artritis Reumatoide , Enfermedades Pulmonares Intersticiales , Humanos , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/fisiopatología , Enfermedades Pulmonares Intersticiales/tratamiento farmacológico , Enfermedades Pulmonares Intersticiales/fisiopatología , Enfermedades Pulmonares Intersticiales/etiología , Antirreumáticos/farmacología , Antirreumáticos/efectos adversos , Antirreumáticos/administración & dosificación , Drogas en Investigación/farmacología , Factores de Riesgo , Animales , Biomarcadores/metabolismo
11.
J Ethnopharmacol ; 334: 118524, 2024 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-38971344

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: As a traditional Chinese medicine, the flower of Rhododendron molle G. Don (RMF) is record in the Chinese pharmacopoeia, and is commonly utilized for treating rheumatoid arthritis (RA) in clinical practice. However, its precise mechanisms necessitate further exploration. AIM OF THE STUDY: To expound the effective components, targets, metabolites, and pathways participated in RMF's anti-RA effects by metabolomics integrated network pharmacology. MATERIALS AND METHODS: CIA rats were intragastric administered RMF for 2 weeks, following which the therapeutic effects were comprehensively evaluated. Serum metabolomics was adopted to investigate the differential metabolites (DEMs). UHPLC-Q-Exactive-MS method was applied to identify the components of RMF, and then network pharmacology was utilize to select the component-RA-targets. Molecular docking and Western blotting were utilized to validate the key targets. RESULTS: RA symptoms were alleviated by RMF through the inhibition secretion of pro-inflammatory factors IL-1ß, IL-6 and TNF-α, along with relief in bone destruction observed in CIA rats. Four targets, namely AKR1B1, TPH1, CYP1A1, and CYP1A2, were identified, along with their corresponding metabolites, namely D-glucose, D-mannose, L-tryptophan, 11-deoxycorticosterone, and 17α-hydroxyprogesterone. These were found to be involved in three key metabolic pathways: steroid hormone biosynthesis, tryptophan metabolism, and galactose metabolism. Additionally, five significant anti-RA active components were identified from RMF, including Rhodojaponin (Rj)-Ⅱ, Rj-Ⅲ, Rj-Ⅴ, Rj-Ⅵ, and quercetin. CONCLUSIONS: The anti-RA mechanisms of RMF were investigated in this study, focusing on active components, upstream targets, and downstream metabolites. These findings lay a foundation for the clinical practice and drug development of RMF.


Asunto(s)
Artritis Experimental , Artritis Reumatoide , Flores , Metabolómica , Farmacología en Red , Rhododendron , Animales , Rhododendron/química , Flores/química , Artritis Reumatoide/tratamiento farmacológico , Ratas , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/metabolismo , Masculino , Antirreumáticos/farmacología , Antirreumáticos/aislamiento & purificación , Simulación del Acoplamiento Molecular , Medicamentos Herbarios Chinos/farmacología , Ratas Sprague-Dawley , Extractos Vegetales/farmacología
12.
Inflamm Res ; 73(8): 1371-1391, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38879731

RESUMEN

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.


Asunto(s)
Antirreumáticos , Artritis Reumatoide , Sinoviocitos , Pez Cebra , Animales , Humanos , Ratones , Artritis Reumatoide/tratamiento farmacológico , Células RAW 264.7 , Sinoviocitos/efectos de los fármacos , Antirreumáticos/farmacología , Antirreumáticos/uso terapéutico , Ratas , Masculino , Citocinas/metabolismo , Células THP-1 , Indoles/farmacología , Indoles/uso terapéutico , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Ratas Sprague-Dawley
13.
J Med Chem ; 67(12): 9869-9895, 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38888047

RESUMEN

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.


Asunto(s)
Artritis Experimental , Artritis Reumatoide , Quinasas Relacionadas con NIMA , Proteína con Dominio Pirina 3 de la Familia NLR , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Quinasas Relacionadas con NIMA/antagonistas & inhibidores , Quinasas Relacionadas con NIMA/metabolismo , Animales , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/metabolismo , Humanos , Ratas , Artritis Experimental/tratamiento farmacológico , Descubrimiento de Drogas , Relación Estructura-Actividad , Masculino , Inflamasomas/metabolismo , Inflamasomas/antagonistas & inhibidores , Simulación del Acoplamiento Molecular , Antirreumáticos/farmacología , Antirreumáticos/química , Antirreumáticos/síntesis química , Antirreumáticos/uso terapéutico
14.
Pharmacol Res Perspect ; 12(4): e1230, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38940379

RESUMEN

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.


Asunto(s)
Osteoclastos , Animales , Ratones , Osteoclastos/efectos de los fármacos , Masculino , Evaluación Preclínica de Medicamentos , Femenino , Ratones Endogámicos C57BL , Administración Oral , Humanos , Diferenciación Celular/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Antirreumáticos/farmacología , Antirreumáticos/farmacocinética , Antirreumáticos/administración & dosificación
15.
Sci Rep ; 14(1): 12935, 2024 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-38839973

RESUMEN

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.


Asunto(s)
Péptidos , Factor de Necrosis Tumoral alfa , Humanos , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Animales , Ratones , Péptidos/farmacología , Péptidos/química , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/metabolismo , Artritis Experimental/patología , Simulación del Acoplamiento Molecular , Células A549 , Simulación de Dinámica Molecular , FN-kappa B/metabolismo , FN-kappa B/antagonistas & inhibidores , Masculino , Antirreumáticos/farmacología , Antirreumáticos/química , Antirreumáticos/uso terapéutico , Unión Proteica , Modelos Animales de Enfermedad
16.
Front Immunol ; 15: 1352251, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38840915

RESUMEN

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.


Asunto(s)
Artritis Reumatoide , Antígeno CTLA-4 , Células Dendríticas , Indolamina-Pirrol 2,3,-Dioxigenasa , Metotrexato , Humanos , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/inmunología , Artritis Reumatoide/sangre , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Metotrexato/uso terapéutico , Metotrexato/farmacología , Femenino , Masculino , Persona de Mediana Edad , Adulto , Antirreumáticos/uso terapéutico , Antirreumáticos/farmacología , Anciano , Monocitos/inmunología , Monocitos/metabolismo , Resultado del Tratamiento , Biomarcadores
17.
Medicina (Kaunas) ; 60(6)2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38929463

RESUMEN

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.


Asunto(s)
Hidroxicloroquina , Ratas Wistar , Retina , Hidroxicloroquina/uso terapéutico , Hidroxicloroquina/farmacología , Hidroxicloroquina/efectos adversos , Animales , Ratas , Retina/efectos de los fármacos , Retina/ultraestructura , Retina/patología , Masculino , Antirreumáticos/uso terapéutico , Antirreumáticos/farmacología
18.
J Control Release ; 371: 498-515, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38849090

RESUMEN

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.


Asunto(s)
Artritis Reumatoide , Nanomedicina , Nanopartículas , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/inmunología , Humanos , Nanomedicina/métodos , Animales , Nanopartículas/administración & dosificación , Sistemas de Liberación de Medicamentos , Antirreumáticos/administración & dosificación , Antirreumáticos/farmacología , Antirreumáticos/uso terapéutico , Vesículas Extracelulares
19.
Expert Opin Investig Drugs ; 33(7): 671-676, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38879822

RESUMEN

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.


Asunto(s)
Desarrollo de Medicamentos , Drogas en Investigación , Glucocorticoides , Polimialgia Reumática , Humanos , Polimialgia Reumática/tratamiento farmacológico , Drogas en Investigación/farmacología , Drogas en Investigación/efectos adversos , Glucocorticoides/administración & dosificación , Glucocorticoides/farmacología , Glucocorticoides/efectos adversos , Inmunosupresores/farmacología , Inmunosupresores/efectos adversos , Inmunosupresores/administración & dosificación , Inmunosupresores/uso terapéutico , Antirreumáticos/farmacología , Antirreumáticos/efectos adversos , Antirreumáticos/administración & dosificación , Receptores de Interleucina-6/antagonistas & inhibidores , Recurrencia , Animales , Factores de Riesgo
20.
Bioorg Chem ; 149: 107499, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38815476

RESUMEN

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.


Asunto(s)
Artritis Reumatoide , Janus Quinasa 3 , Inhibidores de Proteínas Quinasas , Pirimidinas , Janus Quinasa 3/antagonistas & inhibidores , Janus Quinasa 3/metabolismo , Artritis Reumatoide/tratamiento farmacológico , Animales , Pirimidinas/química , Pirimidinas/farmacología , Pirimidinas/síntesis química , Humanos , Relación Estructura-Actividad , Ratones , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/síntesis química , Estructura Molecular , Relación Dosis-Respuesta a Droga , Pirroles/química , Pirroles/farmacología , Pirroles/síntesis química , Carragenina , Masculino , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/inducido químicamente , Antirreumáticos/farmacología , Antirreumáticos/química , Antirreumáticos/síntesis química , Simulación del Acoplamiento Molecular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...