Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 20.522
Filtrer
1.
Cell Mol Biol (Noisy-le-grand) ; 70(6): 73-77, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38836679

RÉSUMÉ

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.


Sujet(s)
Arthrite expérimentale , Polyarthrite rhumatoïde , Glucocorticoïdes , p38 Mitogen-Activated Protein Kinases , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/anatomopathologie , Animaux , Glucocorticoïdes/pharmacologie , p38 Mitogen-Activated Protein Kinases/métabolisme , Arthrite expérimentale/traitement médicamenteux , Arthrite expérimentale/métabolisme , Arthrite expérimentale/anatomopathologie , Souris , Mâle , Articulations/anatomopathologie , Articulations/effets des médicaments et des substances chimiques , Articulations/métabolisme , Souris de lignée DBA , Synovie/métabolisme , Synovie/effets des médicaments et des substances chimiques , Microenvironnement cellulaire/effets des médicaments et des substances chimiques , Agranulocytes/métabolisme , Agranulocytes/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine
2.
Int J Biol Sci ; 20(8): 2922-2942, 2024.
Article de Anglais | MEDLINE | ID: mdl-38904021

RÉSUMÉ

Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease characterized by synovial inflammation and the production of autoantibodies. Previous studies have indicated an association between high-salt diets (HSD) and an increased risk of RA, yet the underlying mechanisms remain unclear. Macrophage pyroptosis, a pro-inflammatory form of cell death, plays a pivotal role in RA. In this study, we demonstrate that HSD exacerbates the severity of arthritis in collagen-induced arthritis (CIA) mice, correlating with macrophage infiltration and inflammatory lesions. Given the significant alterations observed in macrophages from CIA mice subjected to HSD, we specifically investigate the impact of HSD on macrophage responses in the inflammatory milieu of RA. In our in vitro experiments, pretreatment with NaCl enhances LPS-induced pyroptosis in RAW.264.7 and THP-1 cells through the p38 MAPK/NF-κB signaling pathway. Subsequent experiments reveal that Slc6a12 inhibitors and SGK1 silencing inhibit sodium-induced activation of macrophage pyroptosis and the p38 MAPK/NF-κB signaling pathway, whereas overexpression of the SGK1 gene counteracts the effect of sodium on macrophages. In conclusion, our findings verified that high salt intake promotes the progression of RA and provided a detailed elucidation of the activation of macrophage pyroptosis induced by sodium transportation through the Slc6a12 channel.


Sujet(s)
Polyarthrite rhumatoïde , Macrophages , Protein-Serine-Threonine Kinases , Pyroptose , Animaux , Souris , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/anatomopathologie , Macrophages/métabolisme , Pyroptose/effets des médicaments et des substances chimiques , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Chlorure de sodium/pharmacologie , Cellules RAW 264.7 , Humains , Mâle , Protéines précoces immédiates/métabolisme , Protéines précoces immédiates/génétique , Arthrite expérimentale/métabolisme , Transduction du signal , p38 Mitogen-Activated Protein Kinases/métabolisme , Souris de lignée DBA
3.
BMC Musculoskelet Disord ; 25(1): 447, 2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38844896

RÉSUMÉ

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.


Sujet(s)
Apoptose , Arthrite expérimentale , Chondrocytes , Inflammation , Arthrose , Vitamine B6 , Animaux , Apoptose/effets des médicaments et des substances chimiques , Souris , Vitamine B6/pharmacologie , Vitamine B6/usage thérapeutique , Arthrose/traitement médicamenteux , Arthrose/anatomopathologie , Arthrose/métabolisme , Arthrite expérimentale/traitement médicamenteux , Arthrite expérimentale/anatomopathologie , Arthrite expérimentale/métabolisme , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Inflammation/anatomopathologie , Mâle , Chondrocytes/effets des médicaments et des substances chimiques , Chondrocytes/métabolisme , Chondrocytes/anatomopathologie , Souris de lignée DBA , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique , Matrice extracellulaire/métabolisme , Matrice extracellulaire/effets des médicaments et des substances chimiques , Matrice extracellulaire/anatomopathologie , Cartilage articulaire/effets des médicaments et des substances chimiques , Cartilage articulaire/anatomopathologie , Cartilage articulaire/métabolisme
4.
Clin Exp Hypertens ; 46(1): 2366270, 2024 Dec 31.
Article de Anglais | MEDLINE | ID: mdl-38864268

RÉSUMÉ

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.


Sujet(s)
Polyarthrite rhumatoïde , Mouvement cellulaire , Prolifération cellulaire , Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses , Souris de lignée DBA , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Transduction du signal , Animaux , Polyarthrite rhumatoïde/thérapie , Polyarthrite rhumatoïde/métabolisme , Souris , Cellules souches mésenchymateuses/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Transplantation de cellules souches mésenchymateuses/méthodes , Phosphatidylinositol 3-kinases/métabolisme , Humains , Interféron gamma/métabolisme , Cordon ombilical/cytologie , Arthrite expérimentale/thérapie , Arthrite expérimentale/métabolisme , Mâle
5.
Int Immunopharmacol ; 136: 112383, 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-38843642

RÉSUMÉ

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.


Sujet(s)
Arthrite expérimentale , Interleukine-17 , Macaca fascicularis , Psoriasis , Protéines de fusion recombinantes , Facteur de nécrose tumorale alpha , Animaux , Interleukine-17/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Humains , Psoriasis/traitement médicamenteux , Psoriasis/immunologie , Psoriasis/induit chimiquement , Protéines de fusion recombinantes/usage thérapeutique , Protéines de fusion recombinantes/pharmacologie , Arthrite expérimentale/traitement médicamenteux , Arthrite expérimentale/immunologie , Souris , Chimiokine CXCL1/métabolisme , Chimiokine CXCL1/génétique , Cellules HT29 , Maladies auto-immunes/traitement médicamenteux , Maladies auto-immunes/immunologie , Souris transgéniques , Modèles animaux de maladie humaine , Anticorps bispécifiques/usage thérapeutique , Anticorps bispécifiques/pharmacologie , Mâle , Évaluation préclinique de médicament , Imiquimod , Anti-inflammatoires/usage thérapeutique , Anti-inflammatoires/pharmacologie , Souris de lignée DBA
6.
Int J Mol Sci ; 25(11)2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38892051

RÉSUMÉ

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.


Sujet(s)
Arthrite expérimentale , Souris knockout , Récepteurs couplés aux protéines G , Lymphocytes T régulateurs , Lymphocytes auxiliaires Th1 , Cellules Th17 , Animaux , Arthrite expérimentale/anatomopathologie , Arthrite expérimentale/immunologie , Arthrite expérimentale/métabolisme , Arthrite expérimentale/traitement médicamenteux , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme , Cellules Th17/immunologie , Cellules Th17/métabolisme , Cellules Th17/effets des médicaments et des substances chimiques , Récepteurs couplés aux protéines G/métabolisme , Récepteurs couplés aux protéines G/génétique , Récepteurs couplés aux protéines G/agonistes , Souris , Lymphocytes auxiliaires Th1/immunologie , Lymphocytes auxiliaires Th1/métabolisme , Lymphocytes auxiliaires Th1/effets des médicaments et des substances chimiques , Souris de lignée DBA , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/immunologie , Polyarthrite rhumatoïde/anatomopathologie , Mâle , Cytokines/métabolisme
7.
Int J Mol Sci ; 25(11)2024 May 29.
Article de Anglais | MEDLINE | ID: mdl-38892148

RÉSUMÉ

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.


Sujet(s)
Arthrite expérimentale , Animaux , Arthrite expérimentale/immunologie , Arthrite expérimentale/radiothérapie , Arthrite expérimentale/anatomopathologie , Souris , Rayonnement térahertz , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique , Mâle , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Inflammasomes/métabolisme , Inflammasomes/immunologie , Facteur de transcription NF-kappa B/métabolisme , Souris de lignée DBA , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/effets des radiations , Humains , Transduction du signal/effets des radiations , Kératinocytes/effets des radiations , Kératinocytes/immunologie , Kératinocytes/métabolisme
8.
Arthritis Res Ther ; 26(1): 121, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38879555

RÉSUMÉ

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.


Sujet(s)
Arthrite expérimentale , Azétidines , Ganglions sensitifs des nerfs spinaux , Interleukine-6 , Janus kinases , Névralgie , Purines , Pyrazoles , Facteur de transcription STAT-3 , Transduction du signal , Sulfonamides , Animaux , Azétidines/pharmacologie , Azétidines/usage thérapeutique , Sulfonamides/pharmacologie , Sulfonamides/usage thérapeutique , Pyrazoles/pharmacologie , Pyrazoles/usage thérapeutique , Facteur de transcription STAT-3/métabolisme , Purines/pharmacologie , Arthrite expérimentale/métabolisme , Arthrite expérimentale/traitement médicamenteux , Ganglions sensitifs des nerfs spinaux/métabolisme , Ganglions sensitifs des nerfs spinaux/effets des médicaments et des substances chimiques , Névralgie/traitement médicamenteux , Névralgie/métabolisme , Janus kinases/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Souris , Interleukine-6/métabolisme , Mâle , Neurones/métabolisme , Neurones/effets des médicaments et des substances chimiques , Neurones/anatomopathologie , Souris de lignée DBA , Inflammation/métabolisme , Inflammation/traitement médicamenteux , Inhibiteurs des Janus kinases/pharmacologie , Inhibiteurs des Janus kinases/usage thérapeutique
9.
J Ethnopharmacol ; 332: 118286, 2024 Oct 05.
Article de Anglais | MEDLINE | ID: mdl-38723919

RÉSUMÉ

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


Sujet(s)
Antirhumatismaux , Arthrite expérimentale , Polyarthrite rhumatoïde , Chimiokine CXCL10 , Chimiotaxie , Récepteurs CXCR3 , Membrane synoviale , Animaux , Récepteurs CXCR3/métabolisme , Chimiokine CXCL10/métabolisme , Arthrite expérimentale/traitement médicamenteux , Polyarthrite rhumatoïde/traitement médicamenteux , Mâle , Antirhumatismaux/pharmacologie , Antirhumatismaux/isolement et purification , Rats , Humains , Chimiotaxie/effets des médicaments et des substances chimiques , Membrane synoviale/effets des médicaments et des substances chimiques , Membrane synoviale/métabolisme , Souris , Souris de lignée DBA , Médicaments issus de plantes chinoises/pharmacologie , Médicaments issus de plantes chinoises/composition chimique , Cellules synoviales/effets des médicaments et des substances chimiques , Cellules synoviales/métabolisme
10.
Int J Nanomedicine ; 19: 4411-4427, 2024.
Article de Anglais | MEDLINE | ID: mdl-38774028

RÉSUMÉ

Background: Rheumatoid arthritis (RA) is a chronic and systemic autoimmune disease characterized by synovial inflammation and joint destruction. Despite progress in RA therapy, it remains difficult to achieve long-term remission in RA patients. Phosphodiesterase 3B (Pde3b) is a member of the phosphohydrolyase family that are involved in many signal transduction pathways. However, its role in RA is yet to be fully addressed. Methods: Studies were conducted in arthritic DBA/1 mice, a suitable mouse strain for collagen-induced rheumatoid arthritis (CIA), to dissect the role of Pde3b in RA pathogenesis. Next, RNAi-based therapy with Pde3b siRNA-loaded liposomes was assessed in a CIA model. To study the mechanism involved, we investigated the effect of Pde3b knockdown on macrophage polarization and related signaling pathway. Results: We demonstrated that mice with CIA exhibited upregulated Pde3b expression in macrophages. Notably, intravenous administration of liposomes loaded with Pde3b siRNA promoted the macrophage anti-inflammatory program and alleviated CIA in mice, as indicated by the reduced inflammatory response, synoviocyte infiltration, and bone and cartilage erosion. Mechanistic study revealed that depletion of Pde3b increased cAMP levels, by which it enhanced PKA-CREB-C/EBPß pathway to transcribe the expression of anti-inflammatory program-related genes. Conclusion: Our results support that Pde3b is involved in the pathogenesis of RA, and Pde3b siRNA-loaded liposomes might serve as a promising therapeutic approach against RA.


Sujet(s)
Arthrite expérimentale , Polyarthrite rhumatoïde , Cyclic Nucleotide Phosphodiesterases, Type 3 , Thérapie génétique , Liposomes , Macrophages , Souris de lignée DBA , Petit ARN interférent , Animaux , Liposomes/composition chimique , Liposomes/administration et posologie , Cyclic Nucleotide Phosphodiesterases, Type 3/génétique , Cyclic Nucleotide Phosphodiesterases, Type 3/métabolisme , Polyarthrite rhumatoïde/génétique , Polyarthrite rhumatoïde/thérapie , Polyarthrite rhumatoïde/induit chimiquement , Souris , Arthrite expérimentale/génétique , Arthrite expérimentale/prévention et contrôle , Arthrite expérimentale/thérapie , Macrophages/effets des médicaments et des substances chimiques , Petit ARN interférent/génétique , Petit ARN interférent/administration et posologie , Thérapie génétique/méthodes , Mâle , Transduction du signal/effets des médicaments et des substances chimiques
11.
Sci Rep ; 14(1): 10096, 2024 05 02.
Article de Anglais | MEDLINE | ID: mdl-38698014

RÉSUMÉ

Pou6f2 is a genetic connection between central corneal thickness (CCT) in the mouse and a risk factor for developing primary open-angle glaucoma. POU6F2 is also a risk factor for several conditions in humans, including glaucoma, myopia, and dyslexia. Recent findings demonstrate that POU6F2-positive retinal ganglion cells (RGCs) comprise a number of RGC subtypes in the mouse, some of which also co-stain for Cdh6 and Hoxd10. These POU6F2-positive RGCs appear to be novel of ON-OFF directionally selective ganglion cells (ooDSGCs) that do not co-stain with CART or SATB2 (typical ooDSGCs markers). These POU6F2-positive cells are sensitive to damage caused by elevated intraocular pressure. In the DBA/2J mouse glaucoma model, heavily-labeled POU6F2 RGCs decrease by 73% at 8 months of age compared to only 22% loss of total RGCs (labeled with RBPMS). Additionally, Pou6f2-/- mice suffer a significant loss of acuity and spatial contrast sensitivity along with an 11.4% loss of total RGCs. In the rhesus macaque retina, POU6F2 labels the large parasol ganglion cells that form the magnocellular (M) pathway. The association of POU6F2 with the M-pathway may reveal in part its role in human glaucoma, myopia, and dyslexia.


Sujet(s)
Dyslexie , Glaucome , Myopie , Cellules ganglionnaires rétiniennes , Animaux , Humains , Souris , Modèles animaux de maladie humaine , Dyslexie/génétique , Dyslexie/métabolisme , Dyslexie/anatomopathologie , Glaucome/anatomopathologie , Glaucome/métabolisme , Glaucome/génétique , Pression intraoculaire , Souris de lignée DBA , Souris knockout , Myopie/anatomopathologie , Myopie/métabolisme , Myopie/génétique , Cellules ganglionnaires rétiniennes/anatomopathologie , Cellules ganglionnaires rétiniennes/métabolisme , Facteurs de risque
12.
Int Immunopharmacol ; 136: 112264, 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-38810308

RÉSUMÉ

BACKGROUND: Chemotaxis and trafficking of dendritic cells (DCs) induced by cytokine receptors are crucial steps in rheumatoid arthritis (RA) pathogenesis. C-C chemokine receptor type 5 (CCR5) plays a key role in DC movement and has been implicated in multitudinous inflammatory and immunology diseases. Thus, targeting CCR5 to suppress DC chemotaxis is considered as a potential strategy for the management of RA. METHODS: Herein, we first synthesized a new hybrid named CT3-1 which based on artesunate and isatin. Besides, we studied the regulating effectiveness of CT3-1 on bone marrow-derived DCs (BMDCs) and on collagen-induced arthritis (CIA) through RNA-seq analysis, cell function experiments in vitro and mice model in vivo. RESULTS: The results shown that CT3-1 mainly reduced CCR5 expression of immature BMDCs and importantly inhibited immature BMDC migration induced by CCR5 in vitro, with no or minor influence on other functions of DCs, such as phagocytosis and maturation. In the mouse model, CT3-1 relieved arthritis severity and inhibited CIA development. Furthermore, CT3-1 intervention decreased the expression of CCR5 in DCs and reduced the proportion of DCs in the peripheral blood of CIA mice. CONCLUSIONS: Our findings suggest that CCR5-induced chemotaxis and trafficking of immature DCs are important in RA. Targeting CCR5 and inhibiting immature DC chemotaxis may provide a novel choice for the treatment of RA and other similar autoimmune diseases. Moreover, we synthesized a new hybrid compound CT3-1 that could inhibit immature DC trafficking and effectively relieve RA by directly reducing the CCR5 expression of immature DCs.


Sujet(s)
Artésunate , Arthrite expérimentale , Polyarthrite rhumatoïde , Chimiotaxie , Cellules dendritiques , Récepteurs CCR5 , Animaux , Cellules dendritiques/effets des médicaments et des substances chimiques , Cellules dendritiques/immunologie , Récepteurs CCR5/métabolisme , Arthrite expérimentale/traitement médicamenteux , Arthrite expérimentale/immunologie , Chimiotaxie/effets des médicaments et des substances chimiques , Artésunate/pharmacologie , Artésunate/usage thérapeutique , Souris , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/immunologie , Souris de lignée DBA , Mâle , Cellules cultivées , Humains
13.
J Autoimmun ; 146: 103238, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38754239

RÉSUMÉ

BACKGROUND: Women are more likely to develop autoimmune diseases than men. Contribution from microchimerism (Mc) has been proposed, as women naturally acquire Mc from more sources than men because of pregnancy. Women with Rheumatoid Arthritis (RA) who lack RA-associated HLA alleles have been found to harbor Mc with RA-associated HLA alleles in higher amounts than healthy women in prior work. However, an immunological impact of Mc remains to be elucidated. OBJECTIVES: To test the hypothesis that Mc with RA-risk associated HLA alleles can result in the production of RA-associated autoantibodies, when host genetic risk is absent. METHODS: DBA/2 mice are unable to produce RA-specific anti-citrullinated autoantibodies (ACPAs) after immunization with the enzyme peptidyl arginine deiminase (PAD) in a previously developed model. DBA/2 females were mated with C57BL/6 males humanized to express HLA-DR4, which is associated with RA-risk and production of ACPAs, to evaluate DR4+ fetal Mc contribution. Next, DBA/2 females born of heterozygous DR4+/- mothers were evaluated for DR4+ Mc of maternal or littermate origin. Finally, DBA/2 females from DR4+/- mothers were crossed with DR4+ males, to evaluate the contribution of any Mc source to ACPA production. RESULTS: After PAD immunization, between 20 % and 43 % of DBA/2 females (otherwise unable to produce ACPAs) had detectable ACPAs (CCP2 kit) after exposure to sources of Mc with RA-associated HLA alleles, compared to 0 % of unmated/unexposed DBA/2 females. Further the microchimeric origin of the autoantibodies was confirmed by detecting a C57BL/6-specific immunoglobulin isotype in the DBA/2 response. CONCLUSION: Our study demonstrates that Mc cells can produce "autoantibodies" and points to a role of Mc in the biology of autoimmune diseases, including RA.


Sujet(s)
Polyarthrite rhumatoïde , Autoanticorps , Chimérisme , Souris de lignée DBA , Polyarthrite rhumatoïde/immunologie , Animaux , Souris , Femelle , Autoanticorps/immunologie , Mâle , Humains , Modèles animaux de maladie humaine , Allèles , Souris de lignée C57BL , Anticorps anti-protéines citrullinées/immunologie , Grossesse
14.
PLoS One ; 19(5): e0304601, 2024.
Article de Anglais | MEDLINE | ID: mdl-38820310

RÉSUMÉ

Both clinical and animal studies demonstrated that seizure-induced respiratory arrest (S-IRA) contributes importantly to sudden unexpected death in epilepsy (SUDEP). It has been shown that enhancing serotonin (5-HT) function relieves S-IRA in animal models of SUDEP, including DBA/1 mice. Direct activation of 5-HT3 and 5-HT4 receptors suppresses S-IRA in DBA/1 mice, indicating that these receptors are involved in S-IRA. However, it remains unknown if other subtypes of 5-HT receptors are implicated in S-IRA in DBA/1 mice. In this study, we investigated the action of an agonist of the 5-HT1A (8-OH-DPAT), 5-HT2A (TCB-2), 5-HT2B (BW723C86), 5-HT2C (MK-212), 5-HT6 (WAY-208466) and 5-HT7 (LP-211) receptor on S-IRA in DBA/1 mice. An agonist of the 5-HT receptor or a vehicle was intraperitoneally administered 30 min prior to acoustic simulation, and the effect of each drug/vehicle on the incidence of S-IRA was videotaped for offline analysis. We found that the incidence of S-IRA was significantly reduced by TCB-2 at 10 mg/kg (30%, n = 10; p < 0.01, Fisher's exact test) but was not altered by other agonists compared with the corresponding vehicle controls in DBA/1 mice. Our data demonstrate that 5-HT2A receptors are implicated in S-IRA, and 5-HT1A, 5-HT2B, 5-HT2C, 5-HT6 and 5-HT7 receptors are not involved in S-IRA in DBA/1 mice.


Sujet(s)
Souris de lignée DBA , Récepteurs sérotoninergiques , Crises épileptiques , Animaux , Récepteurs sérotoninergiques/métabolisme , Crises épileptiques/métabolisme , Souris , Mâle , Agonistes des récepteurs de la sérotonine/pharmacologie , Mort subite et inexpliquée en épilepsie/étiologie , Modèles animaux de maladie humaine
15.
Bone Res ; 12(1): 31, 2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-38782893

RÉSUMÉ

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


Sujet(s)
Arthrite expérimentale , Polyarthrite rhumatoïde , Microbiome gastro-intestinal , Histone deacetylases , Souris de lignée C57BL , Cellules synoviales , Animaux , Humains , Mâle , Souris , Arthrite expérimentale/anatomopathologie , Arthrite expérimentale/métabolisme , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/anatomopathologie , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/microbiologie , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Facteurs de transcription Forkhead/métabolisme , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Histone deacetylases/métabolisme , Souris de lignée DBA , Transduction du signal/effets des médicaments et des substances chimiques , Cellules synoviales/métabolisme , Cellules synoviales/effets des médicaments et des substances chimiques , Cellules synoviales/anatomopathologie
16.
Clin Immunol ; 264: 110255, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38763433

RÉSUMÉ

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


Sujet(s)
Arthrite expérimentale , Polyarthrite rhumatoïde , Mouvement cellulaire , Prolifération cellulaire , Fibroblastes , Succinates , Cellules synoviales , Animaux , Cellules synoviales/effets des médicaments et des substances chimiques , Cellules synoviales/métabolisme , Mouvement cellulaire/effets des médicaments et des substances chimiques , Arthrite expérimentale/traitement médicamenteux , Arthrite expérimentale/métabolisme , Arthrite expérimentale/anatomopathologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Succinates/pharmacologie , Rats , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Mâle , Polyarthrite rhumatoïde/traitement médicamenteux , Polyarthrite rhumatoïde/métabolisme , Souris , Souris knockout , Cellules cultivées , Souris de lignée DBA , Cycle citrique/effets des médicaments et des substances chimiques
17.
Cell Commun Signal ; 22(1): 230, 2024 Apr 16.
Article de Anglais | MEDLINE | ID: mdl-38627796

RÉSUMÉ

OBJECTIVE: Recurrent pregnancy loss (RPL) patients have higher absolute numbers of decidual natural killer (dNK) cells with elevated intracellular IFN-γ levels leading to a pro-inflammatory cytokine milieu, which contributes to RPL pathogenesis. The main objective of this study was twofold: first to explore the regulatory effects and mechanisms of villus-derived exosomes (vEXOs) from induced abortion patients or RPL patients at the level of intracellular IFN-γ in dNK cells; second to determine the validity of application of vEXOs in the treatment of unexplained RPL (uRPL) through in vitro experiments and mouse models. METHODS: Exosomes were isolated from villus explants by ultracentrifugation, co-cultured with dNK cells, and purified by enzymatic digestion and magnetically activated cell sorting. Flow cytometry, enzyme-linked immunosorbent assays, and RT-qPCR were used to determine IFN-γ levels. Comparative miRNA analysis of vEXOs from induced abortion (IA) and uRPL patients was used to screen potential candidates involved in dNK regulation, which was further confirmed by luciferase reporter assays. IA-vEXOs were electroporated with therapeutic miRNAs and encapsulated in a China Food and Drug Administration (CFDA)-approved hyaluronate gel (HA-Gel), which has been used as a clinical biomaterial in cell therapy for > 30 years. In vivo tracking was performed using 1,1-dioctadecyl-3,3,3,3-tetramethylindotricarbocyaine iodide (DiR) labelling. Tail-vein and uterine horn injections were used to evaluate therapeutic effects of the engineered exosomes in an abortion-prone mouse model (CBA/J × DBA/2 J). Placental growth was evaluated based on placental weight. IFN-γ mRNA levels in mouse placentas were measured by RT-qPCR. RESULTS: IFN-γ levels were significantly higher in dNK cells of uRPL patients than in IA patients. Both uRPL-vEXOs and IA-vEXOs could be efficiently internalized by dNK cells, whereas uRPL-vEXOs could not reduce the expression of IFN-γ by dNK cells as much as IA-vEXOs. Mechanistically, miR-29a-3p was delivered by vEXOs to inhibit IFN-γ production by binding to the 3' UTR of IFN-γ mRNA in dNK cells. For in vivo treatment, application of the HA-Gel effectively prolonged the residence time of vEXOs in the uterine cavity via sustained release. Engineered vEXOs loaded with miR-29a-3p reduced the embryo resorption rate in RPL mice with no signs of systemic toxicity. CONCLUSION: Our study provides the first evidence that villi can regulate dNK cell production of IFN-γ via exosome-mediated transfer of miR-29a-3p, which deepens our understanding of maternal-fetal immune tolerance for pregnancy maintenance. Based on this, we developed a new strategy to mix engineered vEXOs with HA-Gel, which exhibited good therapeutic effects in mice with uRPL and could be used for potential clinical applications in uRPL treatment.


Sujet(s)
Avortement provoqué , Avortement spontané , microARN , Animaux , Femelle , Humains , Souris , Grossesse , Avortement spontané/génétique , Avortement spontané/métabolisme , Caduques/métabolisme , Interféron gamma/métabolisme , Cellules tueuses naturelles , Souris de lignée CBA , Souris de lignée DBA , microARN/génétique , microARN/métabolisme , Placenta/métabolisme , ARN messager/métabolisme
18.
Int J Mol Sci ; 25(8)2024 Apr 10.
Article de Anglais | MEDLINE | ID: mdl-38673758

RÉSUMÉ

Animal tumors serve as reasonable models for human cancers. Both human and animal tumors often reveal triplet EPR signals of nitrosylhemoglobin (HbNO) as an effect of nitric oxide formation in tumor tissue, where NO is complexed by Hb. In search of factors determining the appearance of nitrosylhemoglobin (HbNO) in solid tumors, we compared the intensities of electron paramagnetic resonance (EPR) signals of various iron-nitrosyl complexes detectable in tumor tissues, in the presence and absence of excess exogenous iron(II) and diethyldithiocarbamate (DETC). Three types of murine tumors, namely, L5178Y lymphoma, amelanotic Cloudman S91 melanoma, and Ehrlich carcinoma (EC) growing in DBA/2 or Swiss mice, were used. The results were analyzed in the context of vascularization determined histochemically using antibodies to CD31. Strong HbNO EPR signals were found in melanoma, i.e., in the tumor with a vast amount of a hemorrhagic necrosis core. Strong Fe(DETC)2NO signals could be induced in poorly vascularized EC. In L5178Y, there was a correlation between both types of signals, and in addition, Fe(RS)2(NO)2 signals of non-heme iron-nitrosyl complexes could be detected. We postulate that HbNO EPR signals appear during active destruction of well-vascularized tumor tissue due to hemorrhagic necrosis. The presence of iron-nitrosyl complexes in tumor tissue is biologically meaningful and defines the evolution of complicated tumor-host interactions.


Sujet(s)
Acide diéthyl-dithiocarbamique , Hémoglobines , Monoxyde d'azote , Animaux , Monoxyde d'azote/métabolisme , Acide diéthyl-dithiocarbamique/pharmacologie , Acide diéthyl-dithiocarbamique/composition chimique , Souris , Hémoglobines/métabolisme , Hémoglobines/composition chimique , Spectroscopie de résonance de spin électronique/méthodes , Piégeage de spin/méthodes , Néovascularisation pathologique/métabolisme , Lignée cellulaire tumorale , Modèles animaux de maladie humaine , Souris de lignée DBA , Composés du fer II/composition chimique
19.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167171, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38631411

RÉSUMÉ

Patients with advanced chronic kidney disease (CKD) have elevated circulating calcium × phosphate product levels and exhibit soft tissue calcification. Besides the cardiovascular system, calcification is commonly observed in the cornea in CKD patients on hemodialysis. Cardiovascular calcification is a cell-mediated, highly regulated process, and we hypothesized that a similar regulatory mechanism is implicated in corneal calcification with the involvement of corneal epithelial cells (CECs). We established a mouse model of CKD-associated corneal calcification by inducing CKD in DBA/2J mice with an adenine and high phosphate diet. CKD was associated with aorta and corneal calcification as detected by OsteoSense staining and corneal Ca measurement (1.67-fold elevation, p < 0.001). In vitro, excess phosphate and Ca induced human CEC calcification in a dose-dependent and synergistic manner, without any influence on cell viability. High phosphate and Ca-containing osteogenic medium (OM; 2.5 mmol/L excess phosphate and 0.6 mmol/L excess Ca over control) increased the protein expression of Runx2 and induced its nuclear translocation. OM increased the expression of the bone-specific Ca-binding protein osteocalcin (130-fold increase, p < 0.001). Silencing of Runx2 attenuated OM-induced CEC calcification. Immunohistology revealed upregulation of Runx2 and overlapping between the Runx2 and the Alizarin red positive areas of calcification in the cornea of CKD mice. This work sheds light on the mechanism of CKD-induced corneal calcification and provides tools to test calcification inhibitors for the prevention of this detrimental process.


Sujet(s)
Calcinose , Calcium , Sous-unité alpha 1 du facteur CBF , Ostéoblastes , Phosphates , Insuffisance rénale chronique , Animaux , Sous-unité alpha 1 du facteur CBF/métabolisme , Sous-unité alpha 1 du facteur CBF/génétique , Insuffisance rénale chronique/anatomopathologie , Insuffisance rénale chronique/métabolisme , Insuffisance rénale chronique/complications , Souris , Humains , Ostéoblastes/métabolisme , Ostéoblastes/anatomopathologie , Phosphates/métabolisme , Calcium/métabolisme , Calcinose/anatomopathologie , Calcinose/métabolisme , Épithélium antérieur de la cornée/anatomopathologie , Épithélium antérieur de la cornée/métabolisme , Mâle , Souris de lignée DBA , Cellules épithéliales/métabolisme , Cellules épithéliales/anatomopathologie , Modèles animaux de maladie humaine , Phénotype
20.
Front Immunol ; 15: 1385085, 2024.
Article de Anglais | MEDLINE | ID: mdl-38650931

RÉSUMÉ

The biosynthesis of C-reactive protein (CRP) in the liver is increased in inflammatory diseases including rheumatoid arthritis. Previously published data suggest a protective function of CRP in arthritis; however, the mechanism of action of CRP remains undefined. The aim of this study was to evaluate the effects of human CRP on the development of collagen-induced arthritis (CIA) in mice which is an animal model of autoimmune inflammatory arthritis. Two CRP species were employed: wild-type CRP which binds to aggregated IgG at acidic pH and a CRP mutant which binds to aggregated IgG at physiological pH. Ten CRP injections were given on alternate days during the development of CIA. Both wild-type and mutant CRP reduced the incidence of CIA, that is, reduced the number of mice developing CIA; however, CRP did not affect the severity of the disease in arthritic mice. The serum levels of IL-17, IL-6, TNF-α, IL-10, IL-2 and IL-1ß were measured: both wild-type and mutant CRP decreased the level of IL-17 and IL-6 but not of TNF-α, IL-10, IL-2 and IL-1ß. These data suggest that CRP recognizes and binds to immune complexes, although it was not clear whether CRP functioned in its native pentameric or in its structurally altered pentameric form in the CIA model. Consequently, ligand-complexed CRP, through an as-yet undefined mechanism, directly or indirectly, inhibits the production of IL-17 and eventually protects against the initiation of the development of arthritis. The data also suggest that IL-17, not TNF-α, is critical for the development of autoimmune inflammatory arthritis.


Sujet(s)
Arthrite expérimentale , Protéine C-réactive , Interleukine-17 , Facteur de nécrose tumorale alpha , Animaux , Arthrite expérimentale/immunologie , Arthrite expérimentale/sang , Protéine C-réactive/métabolisme , Interleukine-17/sang , Souris , Facteur de nécrose tumorale alpha/sang , Humains , Mâle , Souris de lignée DBA , Modèles animaux de maladie humaine , Polyarthrite rhumatoïde/immunologie , Polyarthrite rhumatoïde/sang
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...