Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 10.967
Filtrar
1.
Clin Exp Hypertens ; 46(1): 2366270, 2024 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-38864268

RESUMO

OBJECTIVE: To elucidate the underlying mechanism by which the proliferation and migration abilities of human umbilical cord mesenchymal stem cells (hUC-MSCs) determine their therapeutic efficacy in rheumatoid arthritis treatment. METHODS: The DBA/1J mice were utilized to establish a collagen-induced RA (CIA) mouse model and to validate the therapeutic efficacy of hUC-MSCs transfected with CD151 siRNA. RNA-seq, QT-PCR and western blotting were utilized to evaluate the mRNA and protein levels of the PI3K/AKT pathway, respectively. RESULTS: IFN-γ significantly enhanced the proliferation and migration abilities of hUC-MSCs, up-regulating the expression of CD151, a gene related to cell proliferation and migration. Effective inhibition of this effect was achieved through CD151 siRNA treatment. However, IFN-γ did not affect hUC-MSCs differentiation or changes in cell surface markers. Additionally, transplantation of CD151-interfered hUC-MSCs (siRNA-CD151-hUC-MSCs) resulted in decreased colonization in the toes of CIA mice and worse therapeutic effects compared to empty vector treatment (siRNA-NC-hUC-MSCs). CONCLUSION: IFN-γ facilitates the proliferation and migration of hUC-MSCs through the CD151/PI3K/AKT pathway. The therapeutic efficacy of siRNA-CD151-hUC-MSCs was found to be inferior to that of siRNA-NC-hUC-MSCs.


Assuntos
Artrite Reumatoide , Movimento Celular , Proliferação de Células , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Camundongos Endogâmicos DBA , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais , Animais , Artrite Reumatoide/terapia , Artrite Reumatoide/metabolismo , Camundongos , Células-Tronco Mesenquimais/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transplante de Células-Tronco Mesenquimais/métodos , Fosfatidilinositol 3-Quinases/metabolismo , Humanos , Interferon gama/metabolismo , Cordão Umbilical/citologia , Artrite Experimental/terapia , Artrite Experimental/metabolismo , Masculino
2.
JCI Insight ; 9(11)2024 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-38855867

RESUMO

In rheumatoid arthritis, inflammatory mediators extravasate from blood into joints via gaps between endothelial cells (ECs), but the contribution of ECs is not known. Sphingosine 1-phosphate receptor 1 (S1PR1), widely expressed on ECs, maintains the vascular barrier. Here, we assessed the contribution of vascular integrity and EC S1PR1 signaling to joint damage in mice exposed to serum-induced arthritis (SIA). EC-specific deletion of S1PR1 or pharmacological blockade of S1PR1 promoted vascular leak and amplified SIA, whereas overexpression of EC S1PR1 or treatment with an S1PR1 agonist delayed SIA. Blockade of EC S1PR1 induced membrane metalloproteinase-dependent cleavage of vascular endothelial cadherin (VE-cadherin), a principal adhesion molecule that maintains EC junctional integrity. We identified a disintegrin and a metalloproteinase domain 10 (ADAM10) as the principal VE-cadherin "sheddase." Mice expressing a stabilized VE-cadherin construct had decreased extravascular VE-cadherin and vascular leakage in response to S1PR1 blockade, and they were protected from SIA. Importantly, patients with active rheumatoid arthritis had decreased circulating S1P and microvascular expression of S1PR1, suggesting a dysregulated S1P/S1PR1 axis favoring vascular permeability and vulnerability. We present a model in which EC S1PR1 signaling maintains homeostatic vascular barrier function by limiting VE-cadherin shedding mediated by ADAM10 and suggest this signaling axis as a therapeutic target in inflammatory arthritis.


Assuntos
Proteína ADAM10 , Antígenos CD , Artrite Experimental , Artrite Reumatoide , Caderinas , Células Endoteliais , Receptores de Esfingosina-1-Fosfato , Animais , Caderinas/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo , Receptores de Esfingosina-1-Fosfato/genética , Camundongos , Artrite Experimental/metabolismo , Artrite Experimental/patologia , Antígenos CD/metabolismo , Antígenos CD/genética , Células Endoteliais/metabolismo , Humanos , Artrite Reumatoide/metabolismo , Artrite Reumatoide/patologia , Artrite Reumatoide/genética , Proteína ADAM10/metabolismo , Proteína ADAM10/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Transdução de Sinais , Camundongos Knockout , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Masculino , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Lisofosfolipídeos/metabolismo , Permeabilidade Capilar , Feminino
3.
BMC Musculoskelet Disord ; 25(1): 447, 2024 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-38844896

RESUMO

BACKGROUND: Although various anti-inflammatory medicines are widely recommended for osteoarthritis (OA) treatment, no significantly clinical effect has been observed. This study aims to examine the effects of vitamin B6, a component that has been reported to be capable of alleviating inflammation and cell death in various diseases, on cartilage degeneration in OA. METHODS: Collagen-induced arthritis (CIA) mice model were established and the severity of OA in cartilage was determined using the Osteoarthritis Research Society International (OARSI) scoring system. The mRNA and protein levels of indicators associated with extracellular matrix (ECM) metabolism, apoptosis and inflammation were detected. The effect of vitamin B6 (VB6) on the mice were assessed using HE staining and masson staining. The apoptosis rate of cells was assessed using TdT-mediated dUTP nick end labeling. RESULTS: Our results showed a trend of improved OARSI score in mice treated with VB6, which remarkably inhibited the hyaline cartilage thickness, chondrocyte disordering, and knees hypertrophy. Moreover, the VB6 supplementation reduced the protein expression of pro-apoptosis indicators, including Bax and cleaved caspase-3 and raised the expression level of anti-apoptosis marker Bcl-2. Importantly, VB6 improved ECM metabolism in both in vivo and in vitro experiments. CONCLUSIONS: This study demonstrated that VB6 alleviates OA through regulating ECM metabolism, inflammation and apoptosis in chondrocytes and CIA mice. The findings in this study provide a theoretical basis for targeted therapy of OA, and further lay the theoretical foundation for studies of mechanisms of VB6 in treating OA.


Assuntos
Apoptose , Artrite Experimental , Condrócitos , Inflamação , Osteoartrite , Vitamina B 6 , Animais , Apoptose/efeitos dos fármacos , Camundongos , Vitamina B 6/farmacologia , Vitamina B 6/uso terapêutico , Osteoartrite/tratamento farmacológico , Osteoartrite/patologia , Osteoartrite/metabolismo , Artrite Experimental/tratamento farmacológico , Artrite Experimental/patologia , Artrite Experimental/metabolismo , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Inflamação/patologia , Masculino , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Condrócitos/patologia , Camundongos Endogâmicos DBA , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Matriz Extracelular/metabolismo , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/patologia , Cartilagem Articular/efeitos dos fármacos , Cartilagem Articular/patologia , Cartilagem Articular/metabolismo
4.
Int J Immunopathol Pharmacol ; 38: 3946320241260635, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38831558

RESUMO

BACKGROUND: Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disorder mainly affecting joints, yet the systemic inflammation can influence other organs and tissues. The objective of this study was to unravel the ameliorative capability of Ondansetron (O) or ß-sitosterol (BS) against inflammatory reactions and oxidative stress that complicates Extra-articular manifestations (EAM) in liver, kidney, lung, and heart of arthritic and arthritic irradiated rats. METHODS: This was accomplished by exposing adjuvant-induced arthritis (AIA) rats to successive weekly fractions of total body γ-irradiation (2 Gray (Gy)/fraction once per week for four weeks, up to a total dose of 8 Gy). Arthritic and/or arthritic irradiated rats were either treated with BS (40 mg/kg b.wt. /day, orally) or O (2 mg/kg) was given ip) or were kept untreated as model groups. RESULTS: Body weight changes, paw circumference, oxidative stress indices, inflammatory response biomarkers, expression of Janus kinase-2 (JAK-2), Signal transducer and activator of transcription 3 (STAT3), high mobility group box1 (HMGB1), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), as well as pro- and anti-inflammatory mediators in the target organs, besides histopathological examination of ankle joints and extra-articular tissues. Treatment of arthritic and/or arthritic irradiated rats with BS or O powerfully alleviated changes in body weight gain, paw swelling, oxidative stress, inflammatory reactions, and histopathological degenerative alterations in articular and non-articular tissues. CONCLUSION: The obtained data imply that BS or O improved the articular and EAM by regulating oxidative and inflammatory indices in arthritic and arthritic irradiated rats.


Assuntos
Artrite Experimental , Rim , Fígado , Pulmão , Ondansetron , Estresse Oxidativo , Sitosteroides , Animais , Sitosteroides/farmacologia , Pulmão/efeitos dos fármacos , Pulmão/patologia , Pulmão/metabolismo , Pulmão/efeitos da radiação , Artrite Experimental/patologia , Artrite Experimental/tratamento farmacológico , Artrite Experimental/metabolismo , Rim/efeitos dos fármacos , Rim/patologia , Rim/metabolismo , Rim/efeitos da radiação , Estresse Oxidativo/efeitos dos fármacos , Ratos , Fígado/efeitos dos fármacos , Fígado/patologia , Fígado/metabolismo , Fígado/efeitos da radiação , Masculino , Ondansetron/farmacologia , Proteína HMGB1/metabolismo , Coração/efeitos dos fármacos , Coração/efeitos da radiação , Miocárdio/patologia , Miocárdio/metabolismo , Inflamação/patologia , Inflamação/metabolismo , Anti-Inflamatórios/farmacologia , Fator de Transcrição STAT3/metabolismo , Ratos Wistar
5.
Cell Mol Biol (Noisy-le-grand) ; 70(6): 73-77, 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38836679

RESUMO

GABBR1 receptors have been implicated in the progression of rheumatoid arthritis (RA), and p38 MAP kinase (MAPK) was shown to be downregulated by GABA and result in unchecked production of pro-inflammatory cytokine. GABBR1 is a member of GABA receptors, and it is known to be upregulated and plays a vital role in RA. Glucocorticoids are efficient therapeutics in rheumatoid arthritis (RA) and are known to regulate GABA actions; therefore, we intended to investigate the potential of glucocorticoids in RA concerning the potential pathway GABBR1/MAPK. Joint specimens were obtained from collagen-induced arthritis mouse model. A double-blind semi-quantitative analysis of vascularity, cell infiltration, as well as lining thickness by help of a 4-point scale setting was used to assess joint inflammation. Expression of GABBR1 and p38 was evaluated immunohistochemically. In vitro peripheral blood (PB), synovial fluid (SF), and mononuclear cells (MCs) were acquired from RA mice. Western blotting was used for detecting expression of GABBR1 and p38 proteins. The presence of high levels of GABBR1 and p38 was prevalent in RA joints relative to healthy joints and related to the inflammation level. Glucocorticoid treatment alters GABBR1 along with p38 protein expression in joints while reducing joint inflammation. Ex vivo and in vitro assays revealed glucocorticoids have a direct impact on p38, such as the decreased GABBR1 expression level after dexamethasone incubation with SFMC. GABBR1 together with p38 expression in RA joints depends on local inflammation and can be targeted by glucocorticoids.


Assuntos
Artrite Experimental , Artrite Reumatoide , Glucocorticoides , Proteínas Quinases p38 Ativadas por Mitógeno , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/metabolismo , Artrite Reumatoide/patologia , Animais , Glucocorticoides/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Artrite Experimental/tratamento farmacológico , Artrite Experimental/metabolismo , Artrite Experimental/patologia , Camundongos , Masculino , Articulações/patologia , Articulações/efeitos dos fármacos , Articulações/metabolismo , Camundongos Endogâmicos DBA , Líquido Sinovial/metabolismo , Líquido Sinovial/efeitos dos fármacos , Microambiente Celular/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/efeitos dos fármacos , Modelos Animais de Doenças
6.
Int J Mol Sci ; 25(11)2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38892051

RESUMO

Dietary supplementation with n-3 polyunsaturated fatty acids (PUFA) has been found to be beneficial in rodent rheumatoid arthritis models and human trials. However, the molecular targets of n-3 PUFAs and their beneficial effects on rheumatoid arthritis are under-researched. Free fatty acid receptor 4 (FFA4, also known as GPR120) is a receptor for n-3 PUFA. We aim to investigate whether FFA4 activation reduces collagen-induced rheumatoid arthritis (CIA) by using an FFA4 agonist, compound A (CpdA), in combination with DBA-1J Ffa4 gene wild-type (WT) and Ffa4 gene knock-out (KO) mice. CIA induced an increase in the arthritis score, foot edema, synovial hyperplasia, pannus formation, proteoglycan loss, cartilage damage, and bone erosion, whereas the administration of CpdA significantly suppressed those increases in Ffa4 WT mice but not Ffa4 gene KO mice. CIA increased mRNA expression levels of pro-inflammatory Th1/Th17 cytokines, whereas CpdA significantly suppressed those increases in Ffa4 WT mice but not Ffa4 gene KO mice. CIA induced an imbalance between Th1/Th17 and Treg cells, whereas CpdA rebalanced them in spleens from Ffa4 WT mice but not Ffa4 gene KO mice. In SW982 synovial cells, CpdA reduced the LPS-induced increase in pro-inflammatory cytokine levels. In summary, the present results suggest that the activation of FFA4 in immune and synovial cells could suppress the characteristics of rheumatoid arthritis and be an adjuvant therapy.


Assuntos
Artrite Experimental , Camundongos Knockout , Receptores Acoplados a Proteínas G , Linfócitos T Reguladores , Células Th1 , Células Th17 , Animais , Artrite Experimental/patologia , Artrite Experimental/imunologia , Artrite Experimental/metabolismo , Artrite Experimental/tratamento farmacológico , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Células Th17/imunologia , Células Th17/metabolismo , Células Th17/efeitos dos fármacos , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/agonistas , Camundongos , Células Th1/imunologia , Células Th1/metabolismo , Células Th1/efeitos dos fármacos , Camundongos Endogâmicos DBA , Artrite Reumatoide/metabolismo , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/imunologia , Artrite Reumatoide/patologia , Masculino , Citocinas/metabolismo
7.
Int J Mol Sci ; 25(11)2024 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-38892148

RESUMO

The primary emphasis of photoimmunology is the impact of nonionizing radiation on the immune system. With the development of terahertz (THz) and sub-terahertz (sub-THz) technology, the biological effects of this emerging nonionizing radiation, particularly its influence on immune function, remain insufficiently explored but are progressively attracting attention. Here, we demonstrated that 0.1 sub-THz radiation can modulate the immune system and alleviate symptoms of arthritis in collagen-induced arthritis (CIA) mice through a nonthermal manner. The application of 0.1 sub-THz irradiation led to a decrease in proinflammatory factors within the joints and serum, reducing the levels of blood immune cells and the quantity of splenic CD4+ T cells. Notably, 0.1 sub-THz irradiation restored depleted Treg cells in CIA mice and re-established the Th17/Treg equilibrium. These findings suggested that sub-THz irradiation plays a crucial role in systemic immunoregulation. Further exploration of its immune modulation mechanisms revealed the anti-inflammatory properties of 0.1 sub-THz on LPS-stimulated skin keratinocytes. Through the reduction in NF-κB signaling and NLRP3 inflammasome activation, 0.1 sub-THz irradiation effectively decreased the production of inflammatory factors and immune-active substances, including IL-1ß and PGE2, in HaCaT cells. Consequently, 0.1 sub-THz irradiation mitigated the inflammatory response and contributed to the maintenance of immune tolerance in CIA mice. This research provided significant new evidence supporting the systemic impacts of 0.1 sub-THz radiation, particularly on the immune system. It also enhanced the field of photoimmunology and offered valuable insights into the potential biomedical applications of 0.1 sub-THz radiation for treating autoimmune diseases.


Assuntos
Artrite Experimental , Animais , Artrite Experimental/imunologia , Artrite Experimental/radioterapia , Artrite Experimental/patologia , Camundongos , Radiação Terahertz , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Masculino , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Inflamassomos/metabolismo , Inflamassomos/imunologia , NF-kappa B/metabolismo , Camundongos Endogâmicos DBA , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/efeitos da radiação , Humanos , Transdução de Sinais/efeitos da radiação , Queratinócitos/efeitos da radiação , Queratinócitos/imunologia , Queratinócitos/metabolismo
8.
Immun Inflamm Dis ; 12(6): e1322, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38888462

RESUMO

INTRODUCTION: Triptolide (TP), a natural product derived from the herbal medicine Tripterygium wilfordii, exhibits potent immunosuppressive activity. However, the mechanisms underlying its effects in rheumatoid arthritis remain incompletely understood. METHODS: Collagen-induced arthritis (CIA) model was induced in Sprague-Dawley rats by immunization with bovine type II collagen, and TP was administrated as treatment. The therapeutic effect of TP was evaluated based on paw swelling, histopathology, and serum levels of inflammatory factors. Exosomes isolated from rat serum were characterized by transmission electron microscopy, dynamic light scattering, and western blot analysis. Proteomic profiling of exosomes was analyzed by direct DIA quantitative proteomics analysis. Gene ontology and the Kyoto Encyclopedia of Genes and Genomes databases were employed for enrichment analysis related to molecular function, biological processes, and signaling pathways. Western blot analysis was used to analyze differentially expressed proteins. RESULTS: TP treatment ameliorated arthritic phenotypes in CIA rats as evidenced by reduced arthritis score, paw swelling, pathological injury severity scores, and serum levels of inflammatory cytokines. The proteomic analysis revealed that TP treatment significantly inhibited complement and coagulation cascades, interleukin-17 signaling pathway, and cholesterol metabolism, which were reactivated in CIA rats. Importantly, lipocalin 2 (LCN2) and myeloperoxidase (MPO) levels were markedly upregulated in the CIA group but suppressed upon TP administration. Furthermore, in synovial tissues, LCN2 and MPO expression levels were also elevated in the CIA group but decreased following TP treatment. CONCLUSION: Our findings demonstrate that TP alleviates CIA, possibly through modulation of exosomal LCN2 and MPO proteins.


Assuntos
Artrite Experimental , Diterpenos , Compostos de Epóxi , Exossomos , Fenantrenos , Proteômica , Ratos Sprague-Dawley , Animais , Compostos de Epóxi/farmacologia , Compostos de Epóxi/uso terapêutico , Fenantrenos/farmacologia , Fenantrenos/uso terapêutico , Diterpenos/farmacologia , Diterpenos/uso terapêutico , Artrite Experimental/tratamento farmacológico , Artrite Experimental/metabolismo , Artrite Experimental/imunologia , Ratos , Proteômica/métodos , Exossomos/metabolismo , Masculino , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/imunologia , Artrite Reumatoide/metabolismo , Transdução de Sinais/efeitos dos fármacos , Modelos Animais de Doenças
9.
Sci Rep ; 14(1): 12935, 2024 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-38839973

RESUMO

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.


Assuntos
Peptídeos , Fator de Necrose Tumoral alfa , Humanos , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Animais , Camundongos , Peptídeos/farmacologia , Peptídeos/química , Artrite Experimental/tratamento farmacológico , Artrite Experimental/metabolismo , Artrite Experimental/patologia , Simulação de Acoplamento Molecular , Células A549 , Simulação de Dinâmica Molecular , NF-kappa B/metabolismo , NF-kappa B/antagonistas & inibidores , Masculino , Antirreumáticos/farmacologia , Antirreumáticos/química , Antirreumáticos/uso terapêutico , Ligação Proteica , Modelos Animais de Doenças
10.
Int Immunopharmacol ; 136: 112383, 2024 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-38843642

RESUMO

The treatment of autoimmune and inflammatory diseases often requires targeting multiple pathogenic pathways. KYS202004A is a novel bispecific fusion protein designed to antagonize TNF-α and IL-17A, pivotal in the pathophysiology of autoimmune and inflammatory diseases. Our initial efforts focused on screening for optimal structure by analyzing expression levels, purity, and binding capabilities. The binding affinity of KYS202004A to TNF-α and IL-17A was evaluated using SPR. In vitro, we assessed the inhibitory capacity of KYS202004A on cytokine-induced CXCL1 expression in HT29 cells. In vivo, its efficacy was tested using a Collagen-Induced Arthritis (CIA) model in transgenic human-IL-17A mice and an imiquimod-induced psoriasis model in cynomolgus monkeys. KYS202004A demonstrated significant inhibition of IL-17A and TNF-α signaling pathways, outperforming the efficacy of monotherapeutic agents ixekizumab and etanercept in reducing CXCL1 expression in vitro and ameliorating disease markers in vivo. In the CIA model, KYS202004A significantly reduced clinical symptoms, joint destruction, and serum IL-6 concentrations. The psoriasis model revealed that KYS202004A, particularly at a 2  mg/kg dose, was as effective as the combination of ixekizumab and etanercept. This discovery represents a significant advancement in treating autoimmune and inflammatory diseases, offering a dual-targeted therapeutic approach with enhanced efficacy over current monotherapies.


Assuntos
Artrite Experimental , Interleucina-17 , Macaca fascicularis , Psoríase , Proteínas Recombinantes de Fusão , Fator de Necrose Tumoral alfa , Animais , Interleucina-17/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Humanos , Psoríase/tratamento farmacológico , Psoríase/imunologia , Psoríase/induzido quimicamente , Proteínas Recombinantes de Fusão/uso terapêutico , Proteínas Recombinantes de Fusão/farmacologia , Artrite Experimental/tratamento farmacológico , Artrite Experimental/imunologia , Camundongos , Quimiocina CXCL1/metabolismo , Quimiocina CXCL1/genética , Células HT29 , Doenças Autoimunes/tratamento farmacológico , Doenças Autoimunes/imunologia , Camundongos Transgênicos , Modelos Animais de Doenças , Anticorpos Biespecíficos/uso terapêutico , Anticorpos Biespecíficos/farmacologia , Masculino , Avaliação Pré-Clínica de Medicamentos , Imiquimode , Anti-Inflamatórios/uso terapêutico , Anti-Inflamatórios/farmacologia , Camundongos Endogâmicos DBA
11.
Arthritis Res Ther ; 26(1): 121, 2024 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-38879555

RESUMO

BACKGROUND: Janus kinase (JAK) inhibitors, such as baricitinib, are widely used to treat rheumatoid arthritis (RA). Clinical studies show that baricitinib is more effective at reducing pain than other similar drugs. Here, we aimed to elucidate the molecular mechanisms underlying the pain relief conferred by baricitinib, using a mouse model of arthritis. METHODS: We treated collagen antibody-induced arthritis (CAIA) model mice with baricitinib, celecoxib, or vehicle, and evaluated the severity of arthritis, histological findings of the spinal cord, and pain-related behaviours. We also conducted RNA sequencing (RNA-seq) to identify alterations in gene expression in the dorsal root ganglion (DRG) following baricitinib treatment. Finally, we conducted in vitro experiments to investigate the direct effects of baricitinib on neuronal cells. RESULTS: Both baricitinib and celecoxib significantly decreased CAIA and improved arthritis-dependent grip-strength deficit, while only baricitinib notably suppressed residual tactile allodynia as determined by the von Frey test. CAIA induction of inflammatory cytokines in ankle synovium, including interleukin (IL)-1ß and IL-6, was suppressed by treatment with either baricitinib or celecoxib. In contrast, RNA-seq analysis of the DRG revealed that baricitinib, but not celecoxib, restored gene expression alterations induced by CAIA to the control condition. Among many pathways changed by CAIA and baricitinib treatment, the interferon-alpha/gamma, JAK-signal transducer and activator of transcription 3 (STAT3), and nuclear factor kappa B (NF-κB) pathways were considerably decreased in the baricitinib group compared with the celecoxib group. Notably, only baricitinib decreased the expression of colony-stimulating factor 1 (CSF-1), a potent cytokine that causes neuropathic pain through activation of the microglia-astrocyte axis in the spinal cord. Accordingly, baricitinib prevented increases in microglia and astrocytes caused by CAIA. Baricitinib also suppressed JAK/STAT3 pathway activity and Csf1 expression in cultured neuronal cells. CONCLUSIONS: Our findings demonstrate the effects baricitinib has on the DRG in relation to ameliorating both inflammatory and neuropathic pain.


Assuntos
Artrite Experimental , Azetidinas , Gânglios Espinais , Interleucina-6 , Janus Quinases , Neuralgia , Purinas , Pirazóis , Fator de Transcrição STAT3 , Transdução de Sinais , Sulfonamidas , Animais , Azetidinas/farmacologia , Azetidinas/uso terapêutico , Sulfonamidas/farmacologia , Sulfonamidas/uso terapêutico , Pirazóis/farmacologia , Pirazóis/uso terapêutico , Fator de Transcrição STAT3/metabolismo , Purinas/farmacologia , Artrite Experimental/metabolismo , Artrite Experimental/tratamento farmacológico , Gânglios Espinais/metabolismo , Gânglios Espinais/efeitos dos fármacos , Neuralgia/tratamento farmacológico , Neuralgia/metabolismo , Janus Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Camundongos , Interleucina-6/metabolismo , Masculino , Neurônios/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/patologia , Camundongos Endogâmicos DBA , Inflamação/metabolismo , Inflamação/tratamento farmacológico , Inibidores de Janus Quinases/farmacologia , Inibidores de Janus Quinases/uso terapêutico
12.
Adv Rheumatol ; 64(1): 44, 2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38816873

RESUMO

OBJECTIVES: Research has demonstrated that obesity may be associated with rheumatoid arthritis (RA). In addition, gut microbiota and its metabolites contribute to the occurrence and development of RA and obesity. However, the mechanism by which obesity affects RA remains unclear. In this study, we aimed to investigate whether gut microbiota and their metabolites alter the effects of high fat diet (HFD) on the severity of collagen-induced arthritis (CIA) in mice. METHODS: Briefly, mice were divided into normal group (N), CIA model group (C), HFD group (T), and HFD CIA group (CT). Hematoxylin and Eosin staining(HE) and Safranin O-fast green staining were conducted, and levels of blood lipid and inflammatory cytokines were measured. 16S rDNA sequencing technique and liquid chromatography-mass spectrometry (LC-MS)-based metabolomics were performed to explore changes in the microbiota structure to further reveal the pathomechanism of HFD on CIA. RESULTS: HFD aggravated the severity of CIA in mice. The CT group had the highest proportion of microbial abundance of Blautia, Oscillibacter, Ruminiclostridium-9, and Lachnospiraceae UCG 006 at the genus level, but had a lower proportion of Alistipes. Additionally, the fecal metabolic phenotype of the combined CT group shows significant changes, with differential metabolites enriched in 9 metabolic pathways, including primary bile acid biosynthesis, arginine biosynthesis, sphingolipid metabolism, purine metabolism, linoleic acid metabolism, oxytocin signaling pathway, aminoacyl-tRNA biosynthesis, the pentose phosphate pathway, and sphingolipid signaling pathway. Correlation analysis revealed that some of the altered gut microbiota genera were strongly correlated with changes in fecal metabolites, total cholesterol (TC), triglyceride (TG), and inflammatory cytokine levels. CONCLUSIONS: This study shows that HFD may aggravate inflammatory reaction in CIA mice by altering the gut microbiota and metabolic pathways.


Assuntos
Artrite Experimental , Dieta Hiperlipídica , Microbioma Gastrointestinal , Animais , Dieta Hiperlipídica/efeitos adversos , Microbioma Gastrointestinal/fisiologia , Camundongos , Artrite Experimental/microbiologia , Artrite Experimental/metabolismo , Citocinas/metabolismo , Masculino , Índice de Gravidade de Doença , Obesidade/metabolismo , Obesidade/microbiologia , Modelos Animais de Doenças
13.
Innate Immun ; 30(2-4): 66-78, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38780369

RESUMO

Rheumatoid arthritis (RA) is a chronic disease characterized by joint inflammation and severe disability. However, there is a lack of safe and effective drugs for treating RA. In our previous study, we discovered that myricetin (MC) and celecoxib have a synergistic effect in the treatment of RA. We conducted in vitro and in vivo experiments to further investigate the effects and mechanisms of action of MC. Our findings demonstrated that MC treatment effectively reduced the release of neutrophil extracellular traps (NETs) and alleviated the inflammatory response in RA. Mechanistic studies showed that MC prevents the entry of PADI4 and MPO into the cell nucleus, thereby protecting DNA from decondensation. In a rat arthritis model, MC improved histological changes in ankle joints and suppressed NET-related signaling factors. In conclusion, MC protects the ankle joints against arthritis by inhibiting MPO and PADI4, thereby reducing NET release. The pharmacological mechanism of MC in RA involves the inhibition of NET release.


Assuntos
Artrite Reumatoide , Modelos Animais de Doenças , Armadilhas Extracelulares , Flavonoides , Neutrófilos , Peroxidase , Proteína-Arginina Desiminase do Tipo 4 , Animais , Armadilhas Extracelulares/efeitos dos fármacos , Armadilhas Extracelulares/metabolismo , Artrite Reumatoide/tratamento farmacológico , Ratos , Flavonoides/farmacologia , Proteína-Arginina Desiminase do Tipo 4/metabolismo , Humanos , Neutrófilos/imunologia , Neutrófilos/efeitos dos fármacos , Peroxidase/metabolismo , Desiminases de Arginina em Proteínas/metabolismo , Artrite Experimental/tratamento farmacológico , Masculino , Ratos Sprague-Dawley , Celecoxib/farmacologia , Celecoxib/uso terapêutico , Células Cultivadas , Índice de Gravidade de Doença
14.
J Med Chem ; 67(11): 9495-9515, 2024 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-38780432

RESUMO

We describe the discovery of a thioester-containing glucocorticoid receptor modulator (GRM) payload and the corresponding antibody-drug conjugate (ADC). Payload 6 was designed for rapid hepatic inactivation to minimize systemic exposure of nonconjugated GRM. Mouse PK indicated that 6 is cleared 10-fold more rapidly than a first-generation GRM payload, resulting in 10-fold lower exposure and 3-fold decrease in Cmax. The anti-mTNF conjugate ADC5 fully inhibited inflammation in mouse contact hypersensitivity with minimal effects on corticosterone, a biomarker for systemic GRM effects, at doses up to and including 100 mg/kg. Concomitant inhibition of P1NP suggests potential delivery to cells involved in the remodeling of bone, which may be a consequence of TNF-targeting or bystander payload effects. Furthermore, ADC5 fully suppressed inflammation in collagen-induced arthritis mouse model after one 10 mg/kg dose for 21 days. The properties of the anti-hTNF conjugate were suitable for liquid formulation and may enable subcutaneous dosing.


Assuntos
Artrite Experimental , Corticosterona , Imunoconjugados , Fator de Necrose Tumoral alfa , Animais , Artrite Experimental/tratamento farmacológico , Artrite Experimental/metabolismo , Camundongos , Imunoconjugados/farmacologia , Imunoconjugados/química , Imunoconjugados/farmacocinética , Imunoconjugados/uso terapêutico , Corticosterona/sangue , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Receptores de Glucocorticoides/metabolismo , Receptores de Glucocorticoides/antagonistas & inibidores , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Glucocorticoides/farmacologia , Humanos , Masculino , Modelos Animais de Doenças
15.
Clin Immunol ; 264: 110255, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38763433

RESUMO

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.


Assuntos
Artrite Experimental , Artrite Reumatoide , Movimento Celular , Proliferação de Células , Fibroblastos , Succinatos , Sinoviócitos , Animais , Sinoviócitos/efeitos dos fármacos , Sinoviócitos/metabolismo , Movimento Celular/efeitos dos fármacos , Artrite Experimental/tratamento farmacológico , Artrite Experimental/metabolismo , Artrite Experimental/patologia , Proliferação de Células/efeitos dos fármacos , Succinatos/farmacologia , Ratos , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Masculino , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/metabolismo , Camundongos , Camundongos Knockout , Células Cultivadas , Camundongos Endogâmicos DBA , Ciclo do Ácido Cítrico/efeitos dos fármacos
16.
J Ethnopharmacol ; 332: 118377, 2024 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-38782307

RESUMO

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


Assuntos
Artrite Experimental , Macrófagos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Animais , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Artrite Experimental/tratamento farmacológico , Artrite Experimental/patologia , Ratos , Masculino , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Extratos Vegetais/farmacologia , Extratos Vegetais/química , Medicamentos de Ervas Chinesas/farmacologia , Medicamentos de Ervas Chinesas/química , Simulação de Acoplamento Molecular , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/química , Artrite Reumatoide/tratamento farmacológico , Ratos Sprague-Dawley , Camundongos , Antirreumáticos/farmacologia , Antirreumáticos/isolamento & purificação , Antirreumáticos/química , Acetatos
17.
J Autoimmun ; 146: 103235, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38696926

RESUMO

Soluble components secreted by Tfh cells are critical for the germinal center responses. In this study, we investigated whether Tfh cells could regulate the B-cell response by releasing small extracellular vesicles (sEVs). Our results showed that Tfh cells promote B-cell differentiation and antibody production through sEVs and that CD40L plays a crucial role in Tfh-sEVs function. In addition, increased Tfh-sEVs were found in mice with collagen-induced arthritis (CIA). Adoptive transfer of Tfh cells significantly exacerbated the severity of CIA; however, the effect of Tfh cells on exacerbating the CIA process was significantly diminished after inhibiting sEVs secretion. Moreover, the levels of plasma Tfh-like-sEVs and CD40L expression on Tfh-like-sEVs in RA patients were significantly higher than those in healthy subjects. In summary, Tfh cell-derived sEVs can enhance the B-cell response, and exacerbate the procession of autoimmune arthritis.


Assuntos
Artrite Experimental , Linfócitos B , Vesículas Extracelulares , Células T Auxiliares Foliculares , Animais , Artrite Experimental/imunologia , Vesículas Extracelulares/imunologia , Vesículas Extracelulares/metabolismo , Camundongos , Linfócitos B/imunologia , Linfócitos B/metabolismo , Humanos , Células T Auxiliares Foliculares/imunologia , Masculino , Artrite Reumatoide/imunologia , Diferenciação Celular/imunologia , Ativação Linfocitária/imunologia , Transferência Adotiva , Ligante de CD40/metabolismo , Ligante de CD40/imunologia , Centro Germinativo/imunologia , Centro Germinativo/metabolismo , Índice de Gravidade de Doença , Feminino
18.
J Ethnopharmacol ; 332: 118286, 2024 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-38723919

RESUMO

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.


Assuntos
Antirreumáticos , Artrite Experimental , Artrite Reumatoide , Quimiocina CXCL10 , Quimiotaxia , Receptores CXCR3 , Membrana Sinovial , Animais , Receptores CXCR3/metabolismo , Quimiocina CXCL10/metabolismo , Artrite Experimental/tratamento farmacológico , Artrite Reumatoide/tratamento farmacológico , Masculino , Antirreumáticos/farmacologia , Antirreumáticos/isolamento & purificação , Ratos , Humanos , Quimiotaxia/efeitos dos fármacos , Membrana Sinovial/efeitos dos fármacos , Membrana Sinovial/metabolismo , Camundongos , Camundongos Endogâmicos DBA , Medicamentos de Ervas Chinesas/farmacologia , Medicamentos de Ervas Chinesas/química , Sinoviócitos/efeitos dos fármacos , Sinoviócitos/metabolismo
19.
Bone Res ; 12(1): 31, 2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-38782893

RESUMO

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.


Assuntos
Artrite Experimental , Artrite Reumatoide , Microbioma Gastrointestinal , Histona Desacetilases , Camundongos Endogâmicos C57BL , Sinoviócitos , Animais , Humanos , Masculino , Camundongos , Artrite Experimental/patologia , Artrite Experimental/metabolismo , Artrite Reumatoide/metabolismo , Artrite Reumatoide/patologia , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/microbiologia , Fibroblastos/metabolismo , Fibroblastos/efeitos dos fármacos , Fatores de Transcrição Forkhead/metabolismo , Microbioma Gastrointestinal/efeitos dos fármacos , Histona Desacetilases/metabolismo , Camundongos Endogâmicos DBA , Transdução de Sinais/efeitos dos fármacos , Sinoviócitos/metabolismo , Sinoviócitos/efeitos dos fármacos , Sinoviócitos/patologia
20.
Bioorg Chem ; 149: 107499, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38815476

RESUMO

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.


Assuntos
Artrite Reumatoide , Janus Quinase 3 , Inibidores de Proteínas Quinases , Pirimidinas , Janus Quinase 3/antagonistas & inibidores , Janus Quinase 3/metabolismo , Artrite Reumatoide/tratamento farmacológico , Animais , Pirimidinas/química , Pirimidinas/farmacologia , Pirimidinas/síntese química , Humanos , Relação Estrutura-Atividade , Camundongos , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/síntese química , Estrutura Molecular , Relação Dose-Resposta a Droga , Pirróis/química , Pirróis/farmacologia , Pirróis/síntese química , Carragenina , Masculino , Artrite Experimental/tratamento farmacológico , Artrite Experimental/induzido quimicamente , Antirreumáticos/farmacologia , Antirreumáticos/química , Antirreumáticos/síntese química , Simulação de Acoplamento Molecular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA