Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 11 de 11
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Ann Rheum Dis ; 81(5): 644-652, 2022 05.
Article de Anglais | MEDLINE | ID: mdl-35144926

RÉSUMÉ

OBJECTIVE: To comparatively analyse the aberrant affinity maturation of the antinuclear and rheumatoid factor (RF) B cell repertoires in blood and tissues of patients with Sjögren's syndrome (SjS) using an integrated omics workflow. METHODS: Peptide sequencing of anti-Ro60, anti-Ro52, anti-La and RF was combined with B cell repertoire analysis at the DNA, RNA and single cell level in blood B cell subsets, affected salivary gland and extranodal marginal zone lymphomas of mucosa-associated lymphoid tissue (MALT) of patients with SjS. RESULTS: Affected tissues contained anti-Ro60, anti-Ro52, anti-La and RF clones as a small part of a polyclonal infiltrate. Anti-Ro60, anti-La and anti-Ro52 clones outnumbered RF clones. MALT lymphoma tissues contained monoclonal RF expansions. Autoreactive clones were not selected from a restricted repertoire in a circulating B cell subset. The antinuclear antibody (ANA) repertoires displayed similar antigen-dependent and immunoglobulin (Ig) G1-directed affinity maturation. RF clones displayed antigen-dependent, IgM-directed and more B cell receptor integrity-dependent affinity maturation. This coincided with extensive intra-clonal diversification in RF-derived lymphomas. Regeneration of clinical disease manifestations after rituximab coincided with large RF clones, which not necessarily belonged to the lymphoma clone, that displayed continuous affinity maturation and intra-clonal diversification. CONCLUSION: The ANA and RF repertoires in patients with SjS display tissue-restricted, antigen-dependent and divergent affinity maturation. Affinity maturation of RF clones deviates further during RF clone derived lymphomagenesis and during regeneration of the autoreactive repertoire after temporary disruption by rituximab. These data give insight into the molecular mechanisms of autoreactive inflammation in SjS, assist MALT lymphoma diagnosis and allow tracking its response to rituximab.


Sujet(s)
Lymphome B de la zone marginale , Protéogénomique , Syndrome de Gougerot-Sjögren , Lymphocytes B/immunologie , Lymphocytes B/métabolisme , Humains , Immunoglobuline G/immunologie , Facteur rhumatoïde/métabolisme , Rituximab/usage thérapeutique , Syndrome de Gougerot-Sjögren/immunologie
2.
Rheumatology (Oxford) ; 58(3): 536-546, 2019 03 01.
Article de Anglais | MEDLINE | ID: mdl-30508140

RÉSUMÉ

OBJECTIVE: To investigate the role of AXL, a member of the anti-inflammatory TYRO3, AXL MER (TAM) receptor family, in arthritis. METHODS: KRN serum transfer arthritis was induced in Axl-/- and wild-type mice. Knee and ankle joints were scored macro- and microscopically. Synovial gene and protein expression of Axl was determined in naïve and TGF-ß1-overexpressing joints. AXL expression was determined in M1-like or M2-like macrophages and RA synovium. Human macrophages, fibroblasts and synovial micromasses were stimulated with TGF-ß1 or the AXL inhibitor R428. RESULTS: Ankle joints of Axl-/- mice showed exacerbated arthritis pathology, whereas no effect of Axl gene deletion was observed on gonarthritis pathology. To explain this spatial difference, we examined the synovium of naïve mice. In contrast to the knee, the ankle synovial cells prominently expressed AXL. Moreover, the M2-like macrophage phenotype was the dominant cell type in the naïve ankle joint. Human M2-like macrophages expressed higher levels of AXL and blocking AXL increased their inflammatory response. In the murine ankle synovium, gene expression of Tgfb1 was increased and Tgb1 correlated with Axl. Moreover, TGFB1 and AXL expression also correlated in human RA synovium. In human macrophages and synovial micromasses, TGF-ß1 enhanced AXL expression. Moreover, TGF-ß1 overexpression in naïve murine knee joints induced synovial AXL expression. CONCLUSION: Differences in synovial AXL expression are in accordance with the observation that AXL dampens arthritis in ankle, but not in knee joints. We provide evidence that the local differences in AXL expression could be due to TGF-ß1, and suggest similar pathways operate in RA synovium.


Sujet(s)
Arthrite expérimentale/métabolisme , Protéines proto-oncogènes/métabolisme , Récepteurs à activité tyrosine kinase/métabolisme , Membrane synoviale/métabolisme , Facteur de croissance transformant bêta-1/métabolisme , Animaux , Articulation talocrurale/métabolisme , Arthrite expérimentale/génétique , Fibroblastes/métabolisme , Humains , Articulation du genou/métabolisme , Macrophages/métabolisme , Souris , Souris knockout , Protéines proto-oncogènes/génétique , Récepteurs à activité tyrosine kinase/génétique , Axl Receptor Tyrosine Kinase
3.
ALTEX ; 36(1): 18-28, 2019.
Article de Anglais | MEDLINE | ID: mdl-30303512

RÉSUMÉ

Therapeutic agents that are used by patients with rheumatic and musculoskeletal diseases were originally developed and tested in animal models, and although retrospective studies show a limited predictive value, it could be explained by the fact that there are no good in vitro alternatives. In this study, we developed a 3-dimensional synovial membrane model made of either human primary synovial cell suspensions or a mix of primary fibroblast-like synoviocytes and CD14+ mononuclear cells. We analyzed the composition of the mature micromasses by immunohistochemical staining and flow cytometry and show that the outer surface forms a lining layer consisting out of fibroblast-like and macrophage-like cells, reflecting the in vivo naïve synovial membrane. To recreate the affected synovial membrane in rheumatoid arthritis (RA), the micromasses were exposed to the pro-inflammatory cytokine Tumor Necrosis Factor Alpha (TNF-α). This led to increased pro-inflammatory cytokine expression and production and to hyperplasia of the membrane. To recreate the synovial membrane in osteoarthritis (OA), the micromasses were exposed to Transforming Growth Factor Beta (TGF-ß). This led to fibrosis-like changes of the membrane, including increased Alpha Smooth Muscle Actin and increased expression of fibrosis-related genes PLOD2 and COL1A1. Interestingly, the macrophages in the micromass showed phenotypic plasticity as prolonged TNF-α or TGF-ß stimulation strongly reduced the occurrence of Cluster of Differentiation 163-positive M2-like macrophages. We showed the plasticity of the micromasses as a synovial model for studying RA and OA pathology and propose that the synovial lining micromass system can be a good alternative for testing drugs.


Sujet(s)
Fibroblastes/physiologie , Agranulocytes/physiologie , Membrane synoviale/anatomopathologie , Structures d'échafaudage tissulaires , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Humains , Imagerie tridimensionnelle , Inflammation/induit chimiquement , Inflammation/métabolisme , Antigènes CD14/métabolisme
4.
PeerJ ; 6: e4771, 2018.
Article de Anglais | MEDLINE | ID: mdl-29868252

RÉSUMÉ

OBJECTIVE: Tumor necrosis factor-inducible gene 6 (TSG-6) has anti-inflammatory and chondroprotective effects in mouse models of inflammatory arthritis. Because cartilage damage and inflammation are also observed in osteoarthritis (OA), we determined the effect of viral overexpression of TSG-6 in experimental osteoarthritis. METHODS: Bone marrow-derived cells were differentiated to multinucleated osteoclasts in the presence of recombinant TSG-6 or after transduction with a lentiviral TSG-6 expression vector. Multi-nucleated osteoclasts were analyzed after tartrate resistant acid phosphatase staining and resorption activity was determined on dentin slices. Collagenase-induced osteoarthritis (CIOA) was induced in C57BL/6 mice after intra-articular injection of an adenoviral TSG-6 or control luciferase expression vector. Inflammation-related protease activity was measured using bioluminescent Prosense probes. After a second adenovirus injection, cartilage damage was assessed in histological sections stained with Safranin-O. Ectopic bone formation was scored in X-ray images of the affected knees. RESULTS: TSG-6 did not inhibit the formation of multi-nucleated osteoclasts, but caused a significant reduction in the resorption activity on dentin slices. Adenoviral TSG-6 gene therapy in CIOA could not reduce the cartilage damage compared to the luciferase control virus and no significant difference in inflammation-related protease activity was noted between the TSG-6 and control treated group. Instead, X-ray analysis and histological analysis revealed the presence of ectopic bone formation in the TSG-6 treated group. CONCLUSION: Gene therapy based on the expression of TSG-6 could not provide cartilage protection in experimental osteoarthritis, but instead resulted in increased ectopic bone formation.

5.
Front Immunol ; 9: 742, 2018.
Article de Anglais | MEDLINE | ID: mdl-29706963

RÉSUMÉ

Objective: Rheumatoid arthritis (RA) is a chronic and progressive joint disease. It appears that anti-inflammatory feedback mechanisms that could restrain joint inflammation and restore homeostasis are insufficient to perform this control. In this study, we investigated the contribution of the MER tyrosine kinase-mediated anti-inflammatory response on arthritis and whether targeting MER could be a valid approach to treat RA. Methods: KRN serum transfer arthritis (KRN STA) was induced in either Mertk-deficient mice or in mice that adenovirally overexpressed Pros1. Human synovial micromasses were treated with MER-specific antibodies or PROS1. Collagen-induced arthritis (CIA) mice were treated with MER-specific agonistic antibodies or by viral overexpression of Pros1. Results: Mertk-/- mice showed exacerbated arthritis pathology, whereas Pros1 overexpression diminished joint pathology in KRN STA. Human synovial micromasses challenged with MER-specific antibodies enhanced the secretion of inflammatory cytokines, whereas stimulating MER with PROS1 reduced the secretion of these cytokines, confirming the protective role of MER. Next, we treated CIA mice with MER-specific agonistic antibodies, and this unexpectedly resulted in exacerbated arthritis pathology. This was associated with increased numbers of apoptotic cells in their knee joints and higher serum levels of interleukin (IL)-16C, a cytokine released by secondary necrotic neutrophils. Apoptotic cell numbers and IL-16C levels were enhanced during arthritis in Mertk-/- mice and reduced in Pros1-overexpressing mice. Conclusion: MER plays a protective role during joint inflammation and activating MER by its ligand PROS1 ameliorates disease. Treatment of mice with MER receptor agonistic antibodies is deleterious due to its counterproductive effect of blocking efferocytosis in the arthritic joint.


Sujet(s)
Arthrite expérimentale/immunologie , Polyarthrite rhumatoïde/immunologie , Protéines de transport/physiologie , c-Mer Tyrosine kinase/physiologie , Animaux , Arthrite expérimentale/anatomopathologie , Polyarthrite rhumatoïde/anatomopathologie , Protéines de liaison au calcium , Lignée cellulaire , Cytokines/immunologie , Modèles animaux de maladie humaine , Femelle , Humains , Articulation du genou/immunologie , Articulation du genou/anatomopathologie , Mâle , Souris de lignée C57BL , Souris de lignée DBA , Souris knockout , Membrane synoviale/immunologie
6.
PLoS One ; 11(11): e0167076, 2016.
Article de Anglais | MEDLINE | ID: mdl-27870898

RÉSUMÉ

Osteoarthritis (OA) is a degenerative joint disease affecting cartilage and is the most common form of arthritis worldwide. One third of OA patients have severe synovitis and less than 10% have no evidence of synovitis. Moreover, synovitis is predictive for more severe disease progression. This offers a target for therapy but more research on the pathophysiological processes in the synovial tissue of these patients is needed. Functional studies performed with synovial tissue will be more approachable when this material, that becomes available by joint replacement surgery, can be stored for later use. We set out to determine the consequences of slow-freezing of human OA synovial tissue. Therefore, we validated a method that can be applied in every routine laboratory and performed a comparative study of five cryoprotective agent (CPA) solutions. To determine possible deleterious cryopreservation-thaw effects on viability, the synovial tissue architecture, metabolic activity, RNA quality, expression of cryopreservation associated stress genes, and expression of OA characteristic disease genes was studied. Furthermore, the biological activity of the cryopreserved tissue was determined by measuring cytokine secretion induced by the TLR ligands lipopolysaccharides and Pam3Cys. Compared to non frozen synovium, no difference in cell and tissue morphology could be identified in the conditions using the CS10, standard and CryoSFM CPA solution for cryopreservation. However, we observed significantly lower preservation of tissue morphology with the Biofreeze and CS2 media. The other viability assays showed trends in the same direction but were not sensitive enough to detect significant differences between conditions. In all assays tested a clearly lower viability was detected in the condition in which synovium was frozen without CPA solution. This detailed analysis showed that OA synovial tissue explants can be cryopreserved while maintaining the morphology, viability and phenotypical response after thawing, offering enhanced opportunities for human in vitro studies.


Sujet(s)
Cryoconservation/méthodes , Arthrose , Membrane synoviale , Femelle , Régulation de l'expression des gènes , Humains , Mâle , Arthrose/métabolisme , Arthrose/anatomopathologie , Membrane synoviale/métabolisme , Membrane synoviale/anatomopathologie
7.
Arthritis Res Ther ; 18: 186, 2016 08 12.
Article de Anglais | MEDLINE | ID: mdl-27519904

RÉSUMÉ

BACKGROUND: Gene therapy has the potential to provide long-term production of therapeutic proteins in the joints of osteoarthritis (OA) patients. The objective of this study was to analyse the therapeutic potential of disease-inducible expression of anti-inflammatory interleukin-10 (IL-10) in the three-dimensional micromass model of the human synovial membrane. METHODS: Synovial tissue samples from OA patients were digested and the cells were mixed with Matrigel to obtain 3D micromasses. The CXCL10 promoter combined with the firefly luciferase reporter in a lentiviral vector was used to determine the response of the CXCL10 promoter to tumour necrosis factor alpha (TNF-α), interleukin-1ß (IL-1ß) and lipopolysaccharide (LPS). The effects of recombinant IL-10 on gene expression were determined by quantitative PCR. The production of IL-10 from the CXCL10p-IL10 vector and the effects on pro-inflammatory cytokine production were assessed by multiplex ELISA. RESULTS: Micromasses made from whole synovial membrane cell suspensions form a distinct surface composition containing macrophage and fibroblast-like synoviocytes thus mimicking the synovial lining. This lining can be transduced by lentiviruses and allow CXCL-10 promoter-regulated transgene expression. Adequate amounts of IL-10 transgene were produced after stimulation with pro-inflammatory factors able to reduce the production of synovial IL-1ß and IL-6. CONCLUSIONS: Synovial micromasses are a suitable model to test disease-regulated gene therapy approaches and the CXCL10p-IL10 vector might be a good candidate to decrease the inflammatory response implicated in the pathogenesis of OA.


Sujet(s)
Thérapie génétique/méthodes , Interleukine-10/biosynthèse , Arthrose/immunologie , Membrane synoviale/immunologie , Techniques de culture de tissus/méthodes , Chimiokine CXCL10/génétique , Test ELISA , Femelle , Cytométrie en flux , Vecteurs génétiques , Humains , Interleukine-10/immunologie , Lentivirus , Mâle , Microscopie confocale , Arthrose/métabolisme , Régions promotrices (génétique) , Réaction de polymérisation en chaine en temps réel , Membrane synoviale/métabolisme
8.
Hum Gene Ther ; 27(3): 244-54, 2016 Mar.
Article de Anglais | MEDLINE | ID: mdl-26711533

RÉSUMÉ

Disease-inducible promoters for the treatment of rheumatoid arthritis (RA) have the potential to provide regulated expression of therapeutic proteins in arthritic joints. In this study, we set out to identify promoters of human genes that are upregulated during RA and are suitable to drive the expression of relevant amounts of anti-inflammatory interleukin (IL)-10. Microarray analysis of RA synovial biopsies compared with healthy controls yielded a list of 22 genes upregulated during RA. Of these genes, CXCL10 showed the highest induction in lipopolysaccharide-stimulated synovial cells. The CXCL10 promoter was obtained from human cDNA and cloned into a lentiviral vector carrying firefly luciferase to determine the promoter inducibility in primary synovial cells and in THP-1 cells. The promoter activation was strongest 8-12 hr after stimulation with the proinflammatory cytokine tumor necrosis factor (TNF)-α and was reinducible after 96 hr. In addition, the CXCL10 promoter showed a significant response to RA patient serum, compared with sera from healthy individuals. The luciferase gene was replaced with IL-10 to determine the therapeutic properties of the CXCL10p-IL10 lentiviral vector. Primary synovial cells transduced with CXCL10p-IL10 showed a great increase in IL-10 production after stimulation, which reduced the release of proinflammatory cytokines TNF-α and IL-1ß. We conclude that the selected proximal promoter of the CXCL10 gene responds to inflammatory mediators present in the serum of patients with RA and that transduction with the lentiviral CXCL10p-IL10 vector reduces inflammatory cytokine production by primary synovial cells from patients with RA. CXCL10 promoter-regulated IL-10 overexpression can thus provide disease-inducible local gene therapy suitable for RA.


Sujet(s)
Polyarthrite rhumatoïde/génétique , Chimiokine CXCL10/génétique , Régulation de l'expression des gènes , Interleukine-10/génétique , Régions promotrices (génétique) , Polyarthrite rhumatoïde/métabolisme , Polyarthrite rhumatoïde/thérapie , Lignée cellulaire , Cytokines/métabolisme , Analyse de profil d'expression de gènes , Vecteurs génétiques/génétique , Humains , Interleukine-10/métabolisme , Lentivirus/génétique , Synovie/métabolisme , Transgènes
9.
Mol Nutr Food Res ; 59(9): 1701-12, 2015 Sep.
Article de Anglais | MEDLINE | ID: mdl-26047123

RÉSUMÉ

SCOPE: This study shows the effect of bovine milk derived extracellular vesicles (BMEVs) on spontaneous polyarthritis in IL-1Ra-deficient mice and collagen-induced arthritis. METHODS AND RESULTS: BMEVs were isolated from semi-skimmed milk by ultracentrifugation and the particle size was around 100 nm by dynamic light scattering and electron microscopy. BMEVs expressed exosome marker CD63, immunoregulatory microRNA's (miR-30a, -223, -92a), and milk-specific beta-casein and beta-lactoglobulin mRNA. In vitro, PKH-67-labeled BMEVs were taken up by RAW264.7, splenocytes, and intestinal cells as determined by flow cytometry and confocal microscopy. IL-1Ra(-/-) mice received BMEVs by daily oral gavage starting at wk 5 till 15 after birth and collagen-induced arthritis mice via their drinking water starting 1 wk before immunization till day 40. Macroscopically, BMEV treatment delayed the onset of arthritis and histology showed diminished cartilage pathology and bone marrow inflammation in both models. BMEV treatment also reduced the serum levels of MCP-1 and IL-6 and their production by splenic cells. BMEV treatment diminished the anticollagen IgG2a levels, which was accompanied by reduced splenic Th1 (Tbet) and Th17 (RORγT) mRNA. CONCLUSION: This is the first report that oral delivery of BMEVs ameliorates experimental arthritis and this warrants further research to determine whether this beneficial effect can be seen in rheumatoid arthritis patients.


Sujet(s)
Arthrite expérimentale/thérapie , Vésicules extracellulaires/métabolisme , Lait/composition chimique , Administration par voie orale , Animaux , Caséines/génétique , Caséines/métabolisme , Bovins , Lignée cellulaire tumorale , Chimiokine CCL2/sang , Collagène/toxicité , Exosomes/génétique , Exosomes/métabolisme , Marqueurs génétiques , Immunoglobuline G/sang , Antagoniste du récepteur à l'interleukine-1/déficit , Antagoniste du récepteur à l'interleukine-1/génétique , Antagoniste du récepteur à l'interleukine-1/métabolisme , Interleukine-6/sang , Lactoglobulines/génétique , Lactoglobulines/métabolisme , Souris , microARN/génétique , microARN/métabolisme , Membre-3 du groupe F de la sous-famille-1 de récepteurs nucléaires/génétique , Membre-3 du groupe F de la sous-famille-1 de récepteurs nucléaires/métabolisme , Taille de particule , ARN messager/génétique , ARN messager/métabolisme , Rate/cytologie , Rate/métabolisme , Antigène CD63/génétique , Antigène CD63/métabolisme
10.
PLoS One ; 10(3): e0121123, 2015.
Article de Anglais | MEDLINE | ID: mdl-25822997

RÉSUMÉ

SCOPE: Extracellular vesicles, including exosomes, have been identified in all biological fluids and rediscovered as an important part of the intercellular communication. Breast milk also contains extracellular vesicles and the proposed biological function is to enhance the antimicrobial defense in newborns. It is, however, unknown whether extracellular vesicles are still present in commercial milk and, more importantly, whether they retained their bioactivity. Here, we characterize the extracellular vesicles present in semi-skimmed cow milk available for consumers and study their effect on T cells. METHODS AND RESULTS: Extracellular vesicles from commercial milk were isolated and characterized. Milk-derived extracellular vesicles contained several immunomodulating miRNAs and membrane protein CD63, characteristics of exosomes. In contrast to RAW 267.4 derived extracellular vesicles the milk-derived extracellular vesicles were extremely stable under degrading conditions, including low pH, boiling and freezing. Milk-derived extracellular vesicles were easily taken up by murine macrophages in vitro. Furthermore, we found that they can facilitate T cell differentiation towards the pathogenic Th17 lineage. Using a (CAGA)12-luc reporter assay we showed that these extracellular vesicles carried bioactive TGF-ß, and that anti-TGF-ß antibodies blocked Th17 differentiation. CONCLUSION: Our findings show that commercial milk contains stable extracellular vesicles, including exosomes, and carry immunoregulatory cargo. These data suggest that the extracellular vesicles present in commercial cow milk remains intact in the gastrointestinal tract and exert an immunoregulatory effect.


Sujet(s)
Industrie laitière/normes , Vésicules extracellulaires/métabolisme , Lait/composition chimique , Lait/immunologie , Cellules Th17/immunologie , Facteur de croissance transformant bêta/métabolisme , Animaux , Anticorps/immunologie , Bovins , Différenciation cellulaire/immunologie , Femelle , Luciferases , Macrophages/métabolisme , Souris , Microscopie électronique à transmission , Nanoparticules , Réaction de polymérisation en chaine en temps réel , Statistique non paramétrique , Antigène CD63/métabolisme , Facteur de croissance transformant bêta/immunologie
11.
Ann Rheum Dis ; 74(11): 2084-91, 2015 Nov.
Article de Anglais | MEDLINE | ID: mdl-25028707

RÉSUMÉ

OBJECTIVES: Rheumatoid arthritis is a chronic destructive autoimmune disease, but the course is unpredictable in individual patients. An attractive treatment would provide a disease-regulated therapy that offers personalised drug delivery. Therefore, we expressed the anti-inflammatory interleukin-10 (IL-10) gene under the control of inflammation-dependent promoters in a mouse model of arthritis. METHODS: Proximal promoters of S100a8, Cxcl1, Mmp13, Saa3, IL-1b and Tsg6 were selected by whole-genome expression analysis of inflamed synovial tissues from arthritic mice. Mice were injected intraarticularly in knee joints with lentiviral vectors expressing a luciferase reporter or the therapeutic protein IL-10 under control of the Saa3 or Mmp13 promoter. After 4 days, arthritis was induced by intraarticular injection of streptococcal cell walls (SCW). At different time points after arthritis induction, in vivo bioluminescent imaging was performed and knee joints were dissected for histological and RNA analysis. RESULTS: The disease-regulated promoter-luciferase reporter constructs showed different activation profiles during the course of the disease. The Saa3 and Mmp13 promoters were significantly induced at day 1 or day 4 after arthritis induction respectively and selected for further research. Overexpression of IL-10 using these two disease-inducible promoters resulted in less synovitis and markedly diminished cartilage proteoglycan depletion and in upregulation of IL-1Ra and SOCS3 gene expression. CONCLUSIONS: Our study shows that promoters of genes that are expressed locally during arthritis can be candidates for disease-regulated overexpression of biologics into arthritic joints, as shown for IL-10 in SCW arthritis. The disease-inducible approach might be promising for future tailor-made local gene therapy in arthritis.


Sujet(s)
Arthrite expérimentale/thérapie , Cartilage articulaire/métabolisme , Thérapie génétique , Interleukine-10 , Protéoglycanes/métabolisme , ARN messager/métabolisme , Membrane synoviale/immunologie , Synovite/thérapie , Animaux , Arthrite expérimentale/immunologie , Arthrite expérimentale/anatomopathologie , Polyarthrite rhumatoïde , Paroi cellulaire/immunologie , Expression des gènes , Antagoniste du récepteur à l'interleukine-1/génétique , Mâle , Matrix Metalloproteinase 13/génétique , Souris , Souris de lignée C57BL , Régions promotrices (génétique) , Protéine amyloïde A sérique/génétique , Streptococcus/immunologie , Protéine-3 suppressive de la signalisation des cytokine , Protéines SOCS/génétique , Membrane synoviale/anatomopathologie , Synovite/immunologie , Synovite/anatomopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...