Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 178
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Rheumatology (Oxford) ; 58(6): 1065-1074, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30649473

RESUMEN

OBJECTIVES: Previously, we have shown the involvement of Wnt-activated protein Wnt-1-induced signaling protein 1 (WISP1) in the development of OA in mice. Here, we aimed to characterize the relation between WISP1 expression and human OA and its regulatory epigenetic determinants. METHODS: Preserved and lesioned articular cartilage from end-stage OA patients and non-OA-diagnosed individuals was collected. WISP1 expression was determined using immunohistochemistry and damage was classified using Mankin scoring. RNA expression and DNA methylation were assessed in silico from genome-wide datasets (microarray analysis and RNA sequencing, and 450 k-methylationarrays, respectively). Effects of WISP1 were tested in pellet cultures of primary human chondrocytes. RESULTS: WISP1 expression in cartilage of OA patients was increased compared with non-OA-diagnosed controls and, within OA patients, WISP1 was even higher in lesioned compared with preserved regions, with expression strongly correlating with Mankin score. In early symptomatic OA patients with disease progression, higher synovial WISP1 expression was observed as compared with non-progressors. Notably, increased WISP1 expression was inversely correlated with methylation levels of a positional CpG-dinucleotide (cg10191240), with lesioned areas showing strong hypomethylation for this CpG as compared with preserved cartilage. Additionally, we observed that methylation levels were allele-dependent for an intronic single-nucleotide polymorphism nearby cg10191240. Finally, addition of recombinant WISP1 to pellets of primary chondrocytes strongly inhibited deposition of extracellular matrix as reflected by decreased pellet circumference, proteoglycan content and decreased expression of matrix components. CONCLUSION: Increased WISP1 expression is found in lesioned human articular cartilage, and appears epigenetically regulated via DNA methylation. In vitro assays suggest that increased WISP1 is detrimental for cartilage integrity.


Asunto(s)
Proteínas CCN de Señalización Intercelular/metabolismo , Cartílago Articular/metabolismo , Osteoartritis de la Rodilla/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Condrocitos/metabolismo , Metilación de ADN , Epigénesis Genética , Humanos , Articulación de la Rodilla/metabolismo
2.
Rheumatology (Oxford) ; 58(3): 536-546, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30508140

RESUMEN

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.


Asunto(s)
Artritis Experimental/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Membrana Sinovial/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Articulación del Tobillo/metabolismo , Artritis Experimental/genética , Fibroblastos/metabolismo , Humanos , Articulación de la Rodilla/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Tirosina Quinasa del Receptor Axl
3.
Rheumatology (Oxford) ; 57(4): 737-747, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29361119

RESUMEN

Objectives: RA is a chronic autoimmune disease leading to progressive destruction of cartilage and bone. RA patients show elevated IL-22 levels and the amount of IL-22-producing Th cells positively correlates with the extent of erosive disease, suggesting a role for this cytokine in RA pathogenesis. The purpose of this study was to determine the feasibility of SPECT/CT imaging with 111In-labelled anti-fibroblast activation protein antibody (28H1) to monitor the therapeutic effect of neutralizing IL-22 in experimental arthritis. Methods: Mice (six mice/group) with CIA received anti-IL-22 or isotype control antibodies. To monitor therapeutic effects after treatment, SPECT/CT images were acquired 24 h after injection of 111In-28H1. Imaging results were compared with macroscopic, histologic and radiographic arthritis scores. Results: Neutralizing IL-22 before CIA onset effectively prevented arthritis development, reaching a disease incidence of only 50%, vs 100% in the control group. SPECT imaging showed significantly lower joint tracer uptake in mice treated early with anti-IL-22 antibodies compared with the control-treated group. Reduction of disease activity in those mice was confirmed by macroscopic, histological and radiographic pathology scores. However, when treatment was initiated in a later phase of CIA, progression of joint pathology could not be prevented. Conclusion: These findings suggest that IL-22 plays an important role in CIA development, and neutralizing this cytokine seems an attractive new strategy in RA treatment. Most importantly, SPECT/CT imaging with 111In-28H1 can be used to specifically monitor therapy responses, and is potentially more sensitive in disease monitoring than the gold standard method of macroscopic arthritis scoring.


Asunto(s)
Artritis/diagnóstico por imagen , Cartílago Articular/diagnóstico por imagen , Gelatinasas/genética , Regulación de la Expresión Génica , Interleucinas/genética , Proteínas de la Membrana/genética , ARN Mensajero/genética , Serina Endopeptidasas/genética , Tomografía Computarizada por Tomografía Computarizada de Emisión de Fotón Único/métodos , Animales , Artritis/tratamiento farmacológico , Artritis/genética , Cartílago Articular/efectos de los fármacos , Cartílago Articular/metabolismo , Colágeno/toxicidad , Modelos Animales de Enfermedad , Endopeptidasas , Gelatinasas/biosíntesis , Inmunohistoquímica , Interleucinas/biosíntesis , Masculino , Proteínas de la Membrana/biosíntesis , Ratones , Ratones Endogámicos DBA , Reacción en Cadena en Tiempo Real de la Polimerasa , Serina Endopeptidasas/biosíntesis , Membrana Sinovial/metabolismo , Membrana Sinovial/patología , Interleucina-22
4.
J Cell Physiol ; 232(1): 225-33, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27138291

RESUMEN

The general consensus is that milk promotes bone growth and density because is a source of calcium and contains components that enhance intestinal calcium uptake or directly affect bone metabolism. In this study, we investigated the effect of bovine-derived milk 100,000 g pellet (P100), which contains nanoparticles (<220 nm) including extracellular vesicles, on osteoclast differentiation and bone resorption. Bone marrow-derived osteoclast precursor cells were differentiated into osteoclasts by M-CSF and RANKL (control) and in the presence of milk P100. Milk P100 treatment until day 4 increased the number of TRAP-positive mononuclear cells and small (≤5 nuclei) osteoclasts. The number of large (≥6 nuclei) osteoclasts remained the same. These alterations were associated with increased expression of TRAP, NFATc1, and c-Fos. Cells seeded in a calcium-phosphate coated plate or bone slices showed reduced resorption area when exposed to milk P100 during the differentiation phase and even after osteoclast formation. Interestingly, milk P100 treatment enhanced Cathepsin K expression but reduced Carbonic Anhydrase 2 gene expression. Moreover, intracellular acid production was also decreased by milk P100 treatment. Oral delivery of milk P100 to female DBA1/J mice for 7 weeks did not alter bone area; however, increased osteoclast number and area in tibia without changes in serum RANKL and CTX-I levels. We showed for the first time the effect of milk P100 on osteoclast differentiation both in vitro and in vivo and found that milk P100 increased the formation of small osteoclasts but this does not lead to more bone resorption probably due to reduced acid secretion. J. Cell. Physiol. 232: 225-233, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Resorción Ósea/tratamiento farmacológico , Diferenciación Celular/efectos de los fármacos , Leche/metabolismo , Nanopartículas/administración & dosificación , Osteoclastos/metabolismo , Animales , Resorción Ósea/metabolismo , Fosfatos de Calcio/farmacología , Diferenciación Celular/fisiología , Ratones Endogámicos C57BL , Factores de Transcripción NFATC/metabolismo , Osteoclastos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Transducción de Señal/efectos de los fármacos
5.
N Engl J Med ; 370(5): 433-43, 2014 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-24350901

RESUMEN

BACKGROUND: Plasmacytoid dendritic cells have been implicated in the pathogenesis of systemic sclerosis through mechanisms beyond the previously suggested production of type I interferon. METHODS: We isolated plasmacytoid dendritic cells from healthy persons and from patients with systemic sclerosis who had distinct clinical phenotypes. We then performed proteome-wide analysis and validated these observations in five large cohorts of patients with systemic sclerosis. Next, we compared the results with those in patients with systemic lupus erythematosus, ankylosing spondylitis, and hepatic fibrosis. We correlated plasma levels of CXCL4 protein with features of systemic sclerosis and studied the direct effects of CXCL4 in vitro and in vivo. RESULTS: Proteome-wide analysis and validation showed that CXCL4 is the predominant protein secreted by plasmacytoid dendritic cells in systemic sclerosis, both in circulation and in skin. The mean (±SD) level of CXCL4 in patients with systemic sclerosis was 25,624±2652 pg per milliliter, which was significantly higher than the level in controls (92.5±77.9 pg per milliliter) and than the level in patients with systemic lupus erythematosus (1346±1011 pg per milliliter), ankylosing spondylitis (1368±1162 pg per milliliter), or liver fibrosis (1668±1263 pg per milliliter). CXCL4 levels correlated with skin and lung fibrosis and with pulmonary arterial hypertension. Among chemokines, only CXCL4 predicted the risk and progression of systemic sclerosis. In vitro, CXCL4 down-regulated expression of transcription factor FLI1, induced markers of endothelial-cell activation, and potentiated responses of toll-like receptors. In vivo, CXCL4 induced the influx of inflammatory cells and skin transcriptome changes, as in systemic sclerosis. CONCLUSIONS: Levels of CXCL4 were elevated in patients with systemic sclerosis and correlated with the presence and progression of complications, such as lung fibrosis and pulmonary arterial hypertension. (Funded by the Dutch Arthritis Association and others.).


Asunto(s)
Células Dendríticas/metabolismo , Factor Plaquetario 4/sangre , Esclerodermia Sistémica/sangre , Adulto , Animales , Biomarcadores/sangre , Citocinas/metabolismo , Hipertensión Pulmonar Primaria Familiar , Femenino , Humanos , Hipertensión Pulmonar/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Factor Plaquetario 4/metabolismo , Proteoma , Fibrosis Pulmonar/sangre , ARN Mensajero/metabolismo , Esclerodermia Sistémica/etiología , Piel/patología
6.
Am J Pathol ; 185(7): 1970-80, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25976248

RESUMEN

Proteins from the Wnt signaling pathway are very important for joint development. Curiously, osteoarthritis (OA) is thought to be a recapitulation of developmental processes. Various members of the Wnt signaling pathway are overexpressed in the synovium during experimental OA. Here, we investigated the potency of specific Wnt proteins, when expressed in the synovium, to induce OA pathology. We overexpressed Wnt5a, Wnt8a, Wnt16, and WISP1 in the synovium using adenoviral vectors. We determined whether overexpression resulted in OA pathology by histology, and we measured whether Wnt signaling led to increased protease activity in the joint. Synovial overexpression of Wnt8a and Wnt16 led to canonical Wnt signaling in the cartilage, whereas overexpression of Wnt5a did not. Canonical Wnt signaling increased protease activity and induced cartilage damage shortly after overexpression. Specific blocking of the canonical Wnt signaling pathway with Dickkopf-1 reduced the Wnt-signaling-induced cartilage damage. By contrast, the noncanonical signaling Wnt5a did not cause cartilage lesions. Overexpression of WISP1, a downstream protein of canonical Wnt signaling, resulted in increased cartilage damage. In conclusion, our data show that canonical Wnts and WISP1, which we found overexpressed in the synovium during experimental OA, may conduce to OA pathology.


Asunto(s)
Proteínas CCN de Señalización Intercelular/metabolismo , Cartílago Articular/patología , Osteoartritis/patología , Péptido Hidrolasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , Animales , Artritis Experimental , Cartílago Articular/metabolismo , Línea Celular , Humanos , Rodilla/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Osteoartritis/metabolismo , Membrana Sinovial/metabolismo , Membrana Sinovial/patología , Proteínas Wnt/genética
7.
Rheumatology (Oxford) ; 55(1): 16-24, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26231344

RESUMEN

There is increasing evidence that low-density lipoprotein (LDL) cholesterol plays a role in the pathology of OA. Specifically, oxidized LDL (oxLDL), which has been shown to play an essential role during development of atherosclerosis, could be involved in processes such as synovial inflammation, cartilage destruction and bone deformations. OxLDL can activate synovial cells such as macrophages, endothelial cells and synovial fibroblasts, resulting in release of growth factors, MMP and pro-inflammatory cytokines. In this review article, we discuss the role of LDL and oxLDL in OA joint pathology and share our viewpoint of possible mechanisms by which these proteins could influence the development and progression of OA. The proposed theory could provide insight into the aetiopathology of OA and give rise to new potential treatments.


Asunto(s)
Cartílago Articular/metabolismo , LDL-Colesterol/metabolismo , Lipoproteínas LDL/metabolismo , Osteoartritis/metabolismo , Membrana Sinovial/metabolismo , Cartílago Articular/patología , Citocinas/metabolismo , Humanos , Osteoartritis/patología , Oxidación-Reducción
8.
Rheumatology (Oxford) ; 55(9): 1536-47, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26667213

RESUMEN

The Wnt signalling pathway is gaining increasing attention in the field of joint pathologies, attributable to its role in the development and homeostasis of the tissues found in the joint, including bone and cartilage. Imbalance in this pathway has been implicated in the development and progression of OA, and interference with the pathway might therefore depict an effective treatment strategy. Though offering multiple opportunities, it is yet to be decided which starting point will bring forth the most promising results. The complexity of the pathway and its interaction with other pathways (such as the TGF-ß signalling pathway, which also has a central role in the maintenance of joint homeostasis) means that acting directly on proteins in this signalling cascade entails a high risk of undesired side effects. Therefore, interference with Wnt-induced proteins, such as WISP1, might be an overall more effective and safer therapeutic approach to inhibit the pathological events that take place during OA.


Asunto(s)
Proteínas CCN de Señalización Intercelular/fisiología , Osteoartritis/etiología , Proteínas Proto-Oncogénicas/fisiología , Factor de Crecimiento Transformador beta/fisiología , Proteínas Wnt/fisiología , Vía de Señalización Wnt/fisiología , Enfermedades de los Cartílagos/etiología , Enfermedades de los Cartílagos/fisiopatología , Comunicación Celular/fisiología , Núcleo Celular/fisiología , Condrocitos/fisiología , Citoplasma/fisiología , Homeostasis/fisiología , Humanos , Osteoartritis/fisiopatología , Receptor Cross-Talk/fisiología
9.
J Immunol ; 192(9): 4103-11, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24683190

RESUMEN

Increasing epidemiologic evidence supports a link between periodontitis and rheumatoid arthritis. The actual involvement of periodontitis in the pathogenesis of rheumatoid arthritis and the underlying mechanisms remain, however, poorly understood. We investigated the influence of concomitant periodontitis on clinical and histopathologic characteristics of T cell-mediated experimental arthritis and evaluated modulation of type II collagen (CII)-reactive Th cell phenotype as a potential mechanism. Repeated oral inoculations of periodontal pathogens Porphyromonas gingivalis and Prevotella nigrescens induced periodontitis in mice, as evidenced by alveolar bone resorption. Interestingly, concurrent periodontitis induced by both bacteria significantly aggravated the severity of collagen-induced arthritis. Exacerbation of arthritis was characterized by increased arthritic bone erosion, whereas cartilage damage remained unaffected. Both P. gingivalis and P. nigrescens skewed the CII-specific T cell response in lymph nodes draining arthritic joints toward the Th17 phenotype without affecting Th1. Importantly, the levels of IL-17 induced by periodontal pathogens in CII-specific T cells directly correlated with the intensity of arthritic bone erosion, suggesting relevance in pathology. Furthermore, IL-17 production was significantly correlated with periodontal disease-induced IL-6 in lymph node cell cultures. The effects of the two bacteria diverged in that P. nigrescens, in contrast to P. gingivalis, suppressed the joint-protective type 2 cytokines, including IL-4. Further in vitro studies showed that the Th17 induction strongly depended on TLR2 expression on APCs and was highly promoted by IL-1. Our data provide evidence of the involvement of periodontitis in the pathogenesis of T cell-driven arthritis through induction of Ag-specific Th17 response.


Asunto(s)
Artritis Experimental/complicaciones , Artritis Experimental/inmunología , Artritis Reumatoide/inmunología , Enfermedades Periodontales/complicaciones , Enfermedades Periodontales/inmunología , Animales , Artritis Experimental/patología , Artritis Reumatoide/complicaciones , Artritis Reumatoide/patología , Interleucina-1/inmunología , Ratones , Ratones Endogámicos BALB C , Enfermedades Periodontales/microbiología , Células Th17/inmunología , Receptor Toll-Like 2/inmunología
10.
Ann Rheum Dis ; 74(11): 2084-91, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25028707

RESUMEN

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.


Asunto(s)
Artritis Experimental/terapia , Cartílago Articular/metabolismo , Terapia Genética , Interleucina-10 , Proteoglicanos/metabolismo , ARN Mensajero/metabolismo , Membrana Sinovial/inmunología , Sinovitis/terapia , Animales , Artritis Experimental/inmunología , Artritis Experimental/patología , Artritis Reumatoide , Pared Celular/inmunología , Expresión Génica , Proteína Antagonista del Receptor de Interleucina 1/genética , Masculino , Metaloproteinasa 13 de la Matriz/genética , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Proteína Amiloide A Sérica/genética , Streptococcus/inmunología , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/genética , Membrana Sinovial/patología , Sinovitis/inmunología , Sinovitis/patología
11.
Mol Imaging ; 13: 1-10, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24881106

RESUMEN

Rheumatoid arthritis (RA) and osteoarthritis (OA) are serologically and clinically distinctive, but at the local level, both diseases have many molecular pathways in common. In vivo molecular imaging can unravel the local pathologic processes involved in both diseases. In this study, we investigated matrix metalloproteinase (MMP) and cathepsin activity during cartilage destruction, in an RA and an OA mouse model, using biophotonic imaging of substrate-based probes. Mice with collagen-induced arthritis (CIA) or destabilization of the medial meniscus (DMM) were imaged using near-infrared fluorescent probes, activated by several cathepsins or MMPs. Fluorescence signal intensity was compared to synovial gene expression, histology, and cartilage staining of a neoepitope of aggrecan cleaved by MMPs with the amino acids DIPEN. Increased cathepsin and MMP activity was seen during CIA, whereas the DMM model only showed increased MMP activity. DIPEN expression was seen only during CIA. A possible explanation can be differences in gene expressions; MMP3 and -13, known to produce DIPEN neoepitopes, were upregulated in the CIA model, whereas MMP12, known to be involved in elastin degradation and chemokine inhibition, was upregulated in the DMM model. Thus, molecular imaging showed no cathepsin activity at the time of cartilage damage in the DMM model, whereas both cathepsins and MMPs are active in the CIA model during disease progression.


Asunto(s)
Artritis Reumatoide/metabolismo , Catepsinas/análisis , Metaloproteinasas de la Matriz/análisis , Imagen Molecular/métodos , Osteoartritis/metabolismo , Animales , Artritis Experimental/metabolismo , Catepsinas/metabolismo , Muerte Celular , Condrocitos/citología , Condrocitos/metabolismo , Colágeno Tipo II/efectos adversos , Colágeno Tipo II/inmunología , Masculino , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Rodilla de Cuadrúpedos/química , Rodilla de Cuadrúpedos/metabolismo , Rodilla de Cuadrúpedos/patología
12.
Cell Tissue Res ; 355(1): 163-71, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24192939

RESUMEN

Lysyl hydroxylase 2b (LH2b) is known to increase pyridinoline cross-links, making collagen less susceptible to enzymatic degradation. Previously, we observed a relationship between LH2b and osteoarthritis-related fibrosis in murine knee joint. For this study, we investigate if transforming growth factor-beta (TGF-ß) and connective tissue growth factor (CTGF) regulate procollagen-lysine, 2-oxoglutarate 5-dioxygenase 2 (PLOD2) (gene encoding LH2b) and LH2b expression differently in osteoarthritic human synovial fibroblasts (hSF). Furthermore, we investigate via which TGF-ß route (Smad2/3P or Smad1/5/8P) LH2b is regulated, to explore options to inhibit LH2b during fibrosis. To answer these questions, fibroblasts were isolated from knee joints of osteoarthritis patients. The hSF were stimulated with TGF-ß with or without a kinase inhibitor of ALK4/5/7 (SB-505124) or ALK1/2/3/6 (dorsomorphin). TGF-ß, CTGF, constitutively active (ca)ALK1 and caALK5 were adenovirally overexpressed in hSF. The gene expression levels of PLOD1/2/3, CTGF and COL1A1 were analyzed with Q-PCR. LH2 protein levels were determined with western blot. As expected, TGF-ß induced PLOD2/LH2 expression in hSF, whereas CTGF did not. PLOD1 and PLOD3 were not affected by either TGF-ß or CTGF. SB-505124 prevented the induction of TGF-ß-induced PLOD2, CTGF and COL1A1. Surprisingly, dorsomorphin completely blocked the induction of CTGF and COL1A1, whereas TGF-ß-induced PLOD2 was only slightly reduced. Overexpression of caALK5 in osteoarthritic hSF significantly induced PLOD2/LH2 expression, whereas caALK1 had no effect. We showed, in osteoarthritic hSF, that TGF-ß induced PLOD2/LH2 via ALK5 Smad2/3P. This elevation of LH2b in osteoarthritic hSF makes LH2b an interesting target to interfere with osteoarthritis-related persistent fibrosis.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Fibroblastos/metabolismo , Osteoartritis/metabolismo , Procolágeno-Lisina 2-Oxoglutarato 5-Dioxigenasa/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Células Cultivadas , Fibroblastos/patología , Fibrosis/genética , Fibrosis/metabolismo , Fibrosis/patología , Regulación de la Expresión Génica , Humanos , Osteoartritis/genética , Osteoartritis/patología , Procolágeno-Lisina 2-Oxoglutarato 5-Dioxigenasa/metabolismo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/genética , Transducción de Señal , Proteína Smad1/metabolismo , Proteína Smad2/metabolismo
13.
Arthritis Rheum ; 65(10): 2583-93, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23860661

RESUMEN

OBJECTIVE: Previous studies have demonstrated a protective role of Toll-like receptor 2 (TLR-2) and a proinflammatory function of TLR-4 during chronic T cell-driven arthritis. The involvement of TLRs in T cell-independent arthritic processes, however, remains unclear. This study was undertaken to determine the functional significance of TLR-2 and TLR-4 in T cell-independent immune complex-driven arthritis. METHODS: Serum-transfer arthritis was induced in wild-type and TLR-deficient mice by intraperitoneal injections of arthritogenic K/BxN mouse serum. Arthritis was assessed macroscopically and by histologic analysis. The influence of TLR-2 on macrophage cytokine profile, Fcγ receptor (FcγR) expression, and response to immune complexes was determined. RESULTS: While TLR-4, unexpectedly, did not play any significant role, TLR-2 deficiency accelerated the onset and markedly increased the severity of acute immune complex-driven arthritis in mice. TLR-2 deficiency resulted in a substantial increase in joint inflammation, bone erosion, and cartilage pathology, indicating a protective function of TLR-2 in passive FcγR-driven disease. Ex vivo study of the macrophage inflammatory phenotype revealed increased production of tumor necrosis factor α (TNFα) and interleukin-6 (IL-6) despite similar levels of IL-10, along with a significant increase in FcγR-specific response, in TLR-2-/- mouse macrophages early in the disease. Although distinct FcγR messenger RNA expression was not affected, cell surface protein expression of the inhibitory FcγRIIB in TLR-2-/- naive primary macrophages was specifically diminished, resulting in a higher proinflammatory response. Accordingly, TLR-2 stimulation specifically up-regulated FcγRIIB, but not the activating FcγR, on macrophages. CONCLUSION: TLR-2 regulates acute immune complex-driven arthritis by controlling macrophage FcγR response. Our findings indicate that the protective role of TLR-2 is extended beyond its previously described role in promoting Treg cells during T cell-mediated arthritis.


Asunto(s)
Complejo Antígeno-Anticuerpo/fisiología , Artritis Experimental/fisiopatología , Receptores de IgG/fisiología , Transducción de Señal/fisiología , Receptor Toll-Like 2/fisiología , Animales , Artritis Experimental/inmunología , Artritis Experimental/patología , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Técnicas In Vitro , Interleucina-6/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Suero/inmunología , Índice de Severidad de la Enfermedad , Receptor Toll-Like 2/deficiencia , Receptor Toll-Like 2/genética , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/fisiología , Factor de Necrosis Tumoral alfa/metabolismo
14.
J Immunol ; 189(7): 3767-76, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22942425

RESUMEN

Salivary glands in patients with Sjögren's syndrome (SS) develop ectopic lymphoid structures (ELS) characterized by B/T cell compartmentalization, the formation of high endothelial venules, follicular dendritic cell networks, functional B cell activation with expression of activation-induced cytidine deaminase, as well as local differentiation of autoreactive plasma cells. The mechanisms that trigger ELS formation, autoimmunity, and exocrine dysfunction in SS are largely unknown. In this article, we present a novel model of inducible ectopic lymphoid tissue formation, breach of humoral self-tolerance, and salivary hypofunction after delivery of a replication-deficient adenovirus-5 in submandibular glands of C57BL/6 mice through retrograde excretory duct cannulation. In this model, inflammation rapidly and consistently evolves from diffuse infiltration toward the development of SS-like periductal lymphoid aggregates within 2 wk from AdV delivery. These infiltrates progressively acquire ELS features and support functional GL7(+)/activation-induced cytidine deaminase(+) germinal centers. Formation of ELS is preceded by ectopic expression of lymphoid chemokines CXCL13, CCL19, and lymphotoxin-ß, and is associated with development of anti-nuclear Abs in up to 75% of mice. Finally, reduction in salivary flow was observed over 3 wk post-AdV infection, consistent with exocrine gland dysfunction as a consequence of the inflammatory response. This novel model has the potential to unravel the cellular and molecular mechanisms that regulate ELS formation and their role in exocrine dysfunction and autoimmunity in SS.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Glándulas Exocrinas/fisiopatología , Tejido Linfoide/inmunología , Tejido Linfoide/patología , Sialadenitis/patología , Animales , Enfermedades Autoinmunes/fisiopatología , Modelos Animales de Enfermedad , Glándulas Exocrinas/inmunología , Glándulas Exocrinas/patología , Tejido Linfoide/fisiopatología , Ratones , Ratones Endogámicos C57BL , Estructura Terciaria de Proteína , Sialadenitis/inmunología , Sialadenitis/fisiopatología
15.
Proc Natl Acad Sci U S A ; 108(12): 4962-7, 2011 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-21383200

RESUMEN

Different splice variants of the proinflammatory cytokine IL-32 are found in various tissues; their putative differences in biological function remain unknown. In the present study, we report that IL-32γ is the most active isoform of the cytokine. Splicing to one less active IL-32ß appears to be a salvage mechanism to reduce inflammation. Adenoviral overexpression of IL-32γ (AdIL-32γ) resulted in exclusion of the IL-32γ-specific exon in vitro as well as in vivo, primarily leading to expression of IL-32ß mRNA and protein. Splicing of the IL-32γ-specific exon was prevented by single-nucleotide mutation, which blocked recognition of the splice site by the spliceosome. Overexpression of splice-resistant IL-32γ in THP1 cells or rheumatoid arthritis (RA) synovial fibroblasts resulted in a greater induction of proinflammatory cytokines such as IL-1ß, compared with IL-32ß. Intraarticular introduction of IL-32γ in mice resulted in joint inflammation and induction of several mediators associated with joint destruction. In RA synovial fibroblasts, overexpression of primarily IL-32ß showed minimal secretion and reduced cytokine production. In contrast, overexpression of splice-resistant IL-32γ in RA synovial fibroblasts exhibited marked secretion of IL-32γ. In RA, we observed increased IL-32γ expression compared with osteoarthritis synovial tissue. Furthermore, expression of TNFα and IL-6 correlated significantly with IL-32γ expression in RA, whereas this was not observed for IL-32ß. These data reveal that naturally occurring IL-32γ can be spliced into IL-32ß, which is a less potent proinflammatory mediator. Splicing of IL-32γ into IL-32ß is a safety switch in controlling the effects of IL-32γ and thereby reduces chronic inflammation.


Asunto(s)
Empalme Alternativo/inmunología , Artritis Reumatoide/inmunología , Fibroblastos/inmunología , Interleucinas/inmunología , Membrana Sinovial/inmunología , Células 3T3 , Adenoviridae , Empalme Alternativo/genética , Animales , Artritis Reumatoide/genética , Artritis Reumatoide/metabolismo , Artritis Reumatoide/patología , Citocinas/genética , Citocinas/inmunología , Citocinas/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patología , Células HeLa , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Interleucinas/genética , Interleucinas/metabolismo , Ratones , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , Isoformas de Proteínas/metabolismo , Sitios de Empalme de ARN/genética , Membrana Sinovial/metabolismo , Membrana Sinovial/patología
16.
J Biol Chem ; 287(8): 5733-43, 2012 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-22203669

RESUMEN

IL-32 can be expressed in several isoforms. The amino acid sequences of the major IL-32 isoforms were used to predict the secondary and tertiary protein structure by I-TASSER software. The secondary protein structure revealed coils and α-helixes, but no ß sheets. Furthermore, IL-32 contains an RGD motif, which potentially activates procaspase-3 intracellular and or binds to integrins. Mutation of the RGD motif did not result in inhibition of the IL-32ß- or IL-32γ-induced cytotoxicity mediated through caspase-3. Although IL-32α interacted with the extracellular part of αVß3 and αVß6 integrins, only the αVß3 binding was inhibited by small RGD peptides. Additionally, IL-32ß was able to bind to αVß3 integrins, whereas this binding was not inhibited by small RGD peptides. In addition to the IL-32/integrin interactions, we observed that IL-32 is also able to interact with intracellular proteins that are involved in integrin and focal adhesion signaling. Modeling of IL-32 revealed a distinct α-helix protein resembling the focal adhesion targeting region of focal adhesion kinase (FAK). Inhibition of FAK resulted in modulation of the IL-32ß- or IL-32γ-induced cytotoxicity. Interestingly, IL-32α binds to paxillin without the RGD motif being involved. Finally, FAK inhibited IL-32α/paxillin binding, whereas FAK also could interact with IL-32α, demonstrating that IL-32 is a member of the focal adhesion protein complex. This study demonstrates for the first time that IL-32 binds to the extracellular domain of integrins and to intracellular proteins like paxillin and FAK, suggesting a dual role for IL-32 in integrin signaling.


Asunto(s)
Quinasa 1 de Adhesión Focal/metabolismo , Adhesiones Focales/metabolismo , Interleucinas/química , Interleucinas/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Antígenos de Neoplasias/metabolismo , Caspasa 3/metabolismo , Muerte Celular/efectos de los fármacos , Biología Computacional , Activación Enzimática/efectos de los fármacos , Adhesiones Focales/efectos de los fármacos , Células HEK293 , Humanos , Integrina alfaVbeta3/metabolismo , Integrinas/metabolismo , Interleucinas/genética , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Oligopéptidos/farmacología , Paxillin/metabolismo , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Transducción de Señal/efectos de los fármacos , Programas Informáticos
17.
Ann Rheum Dis ; 72(2): 278-85, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22918932

RESUMEN

OBJECTIVE: To investigate the role of Fcγ receptors (FcγRs) in osteoclastogenesis and osteoclast function. METHODS: Bone destruction was analysed in arthritic knee joints of several FcγR-knockout mouse strains. Unfractionated bone marrow cells were differentiated in vitro towards osteoclasts in the absence or presence of immune complexes (ICs) and stimulated thereafter for 24 h with tumour necrosis factor α (TNFα) or lipopolysaccharide (LPS). In addition, mature osteoclasts were stimulated with ICs. Experiments were analysed for osteoclast formation, bone resorption and the expression of FcγRs and osteoclast markers. RESULTS: Bone destruction was significantly increased in arthritic knee joints of FcγRIIB-deficient mice. All FcγR classes were highly expressed on osteoclast precursors. Expression of the inhibitory FcγRIIB was similar on mature osteoclasts compared to macrophages, whereas activating FcγR levels were significantly lower. IC stimulation of mature osteoclasts did not affect their number or their bone resorptive capacity. ICs significantly inhibited differentiation of unfractionated bone marrow cells towards osteoclasts, bone resorption and expression of osteoclast markers. In the presence of ICs, osteoclastogenesis of FcγRIIB(-/-) precursors and bone resorption remained inhibited. In contrast, ICs could not inhibit osteoclast formation or bone resorption of FcRγ-chain(-/-) precursors. When IC-inhibited osteoclastogenesis was followed by stimulation with TNFα or LPS, the inhibitory effects of ICs were overruled. CONCLUSION: Activating FcγRs mediate IC-induced inhibition of osteoclastogenesis, which might be overruled in the presence of proinflammatory mediators. This suggests that the balance of FcγR-mediated inflammation, through proinflammatory cytokine production, as well as the direct inhibitory effect of ICs on osteoclastogenesis determines the net effect on bone loss.


Asunto(s)
Complejo Antígeno-Anticuerpo/inmunología , Artritis Experimental/inmunología , Células Progenitoras Mieloides/inmunología , Osteoclastos/inmunología , Receptores de IgG/inmunología , Animales , Artritis Experimental/metabolismo , Artritis Experimental/patología , Artritis Reumatoide/inmunología , Artritis Reumatoide/metabolismo , Artritis Reumatoide/patología , Resorción Ósea/inmunología , Resorción Ósea/metabolismo , Resorción Ósea/patología , Diferenciación Celular/inmunología , Modelos Animales de Enfermedad , Citometría de Flujo , Articulación de la Rodilla/inmunología , Articulación de la Rodilla/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Progenitoras Mieloides/citología , Células Progenitoras Mieloides/metabolismo , Osteoclastos/citología , Osteoclastos/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de IgG/metabolismo
18.
Ann Rheum Dis ; 72(8): 1407-15, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23291389

RESUMEN

OBJECTIVE: A prominent role of Toll-like receptor 4 (TLR4) in arthritis is emerging. TLR4 is functional in immune cells and stromal cells. The aim was to investigate the involvement of TLR4 in bone marrow (BM)-derived and resident cells in arthritis. METHODS: Reciprocal sex-mismatched BM transplantation was performed between IL-1Ra(-/-)TLR4(+/+) and IL-1Ra(-/-)TLR4(-/-) double knockout animals in Balb/c background. Arthritis was assessed macroscopically and by histopathology. Immunity was evaluated by splenic cytokine production and flow cytometry in draining lymph node (DLN) cells. RESULTS: Arthritis progression was reduced to a similar extent in animals lacking TLR4 on BM-derived, resident cells or both. Histology revealed that joint inflammation was partially TLR4-dependent in either BM-derived or resident cells. TLR4 plays an additive role in BM-derived and resident cells in promoting cartilage erosion. By contrast, TLR4 was equally important in BM-derived and resident cells in mediating bone erosion. Systemically, TLR4 in both BM-derived and resident cells contributed to IL-17 production by splenic T-cells, whereas in the DLNs of arthritic joints this was not the case. Interestingly, in DLN, the dominant cells producing IL-17 were CD4 negative, and cell numbers were determined by TLR4 in the BM-derived cells. CONCLUSIONS: TLR4 is necessary in both BM-derived and resident cells for full-blown joint swelling, inflammation and bone erosion. Furthermore, TLR4 on BM-derived and tissue-resident cells show an additive effect in cartilage destruction. Interestingly, TLR4 on BM-derived and tissue-resident cells are both required for IL-17 production in spleen, but only in BM-derived cells in DLN.


Asunto(s)
Artritis Experimental/metabolismo , Células de la Médula Ósea/metabolismo , Rodilla de Cuadrúpedos/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Artritis Experimental/inmunología , Artritis Experimental/patología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/patología , Trasplante de Médula Ósea , Cartílago Articular/patología , Femenino , Interleucina-17/metabolismo , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Bazo/metabolismo , Rodilla de Cuadrúpedos/inmunología , Rodilla de Cuadrúpedos/patología
19.
Rheumatology (Oxford) ; 52(5): 806-14, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23353647

RESUMEN

OBJECTIVE: One of the disease hallmarks of RA is progressive cartilage and bone destruction in the joints. The exact mechanism underlying this disease process is largely unknown. Nod1, an intracellular pattern recognition receptor expressed by the innate immune system, has been previously shown to display anti-inflammatory effects in experimental arthritis. Furthermore, an insertion/deletion polymorphism in NOD1 has been demonstrated to modulate cytokine responses of immune cells. In this study, the effect of the insertion/deletion polymorphism in NOD1 on RA susceptibility and severity was assessed. METHODS: Ex vivo stimulation of primary immune cells and osteoclasts with microbial triggers was performed to measure cytokine responses and osteoclast-specific gene expression in relation to the NOD1 genotype. In total, 1047 RA patients from two centres were genotyped for the NOD1 polymorphism and compared with 431 healthy controls. Clinical scores of joint inflammation and destruction were correlated with the NOD1 genotype. RESULTS: Functional analysis revealed increased production of pro-inflammatory cytokines in cells from individuals bearing the NOD1 +32656 insertion allele. Furthermore, osteoclast bone resorption activity was elevated, as reflected by increased expression of the lysosomal protease cathepsin K. However, the insertion allele of the NOD1 +32656 polymorphism was not associated with either susceptibility to, or clinical parameters of, inflammation or bone destruction in RA patients. CONCLUSION: These findings demonstrate that the NOD1 polymorphism modulates pro-inflammatory cytokine responses induced through Toll-like receptor or Nod-like receptor ligands. Nevertheless, these effects of genetic variation in NOD1 appear to be redundant in RA susceptibility and severity.


Asunto(s)
Artritis Reumatoide/genética , Citocinas/metabolismo , Predisposición Genética a la Enfermedad , Proteína Adaptadora de Señalización NOD1/genética , Polimorfismo Genético , Adulto , Artritis Reumatoide/fisiopatología , Resorción Ósea/genética , Resorción Ósea/fisiopatología , Estudios de Casos y Controles , Progresión de la Enfermedad , Femenino , Regulación de la Expresión Génica , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Países Bajos , Valores de Referencia , Medición de Riesgo
20.
Cytokine ; 64(1): 433-40, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23727326

RESUMEN

BACKGROUND: IL-32 has been previously shown to promote inflammation in rheumatoid arthritis patients and to contribute to IL-1ß-induced ICAM-1 as well as other proinflammatory cytokines synthesis in human umbilical endothelial cells (HUVECs). Given the high rate of atherosclerosis in RA, these observations suggest that IL-32 may be involved in the inflammatory pathways of atherosclerosis. METHODS: mRNA and protein levels of IL-32 were determined in human atherosclerotic arterial vessel wall tissue by quantitative real-time PCR and immunohistochemistry. HUVEC and M1/M2 macrophages were stimulated with proinflammatory cytokines and TLR ligands to assess IL-32 mRNA induction. Human THP1 macrophages were transduced with AdIL-32γ, to investigate induction of several proatherosclerotic mediators. Finally, aortas from IL-32γ transgenic mice were studied and compared with aortas from age-matched wild-type mice. RESULTS: IL-32 expression was detectable in human atherosclerotic arterial vessel wall, with the expression of IL-32ß and IL-32γ mRNA significantly enhanced. TLR3-ligand Poly I:C in combination with IFNγ were the most potent inducers of IL-32 mRNA expression in both HUVEC and M1/M2 macrophages. Adenoviral overexpression of IL-32γ in human THP1 macrophages resulted in increased production of CCL2, sVCAM-1, MMP1, MMP9, and MMP13. The IL-32γ transgenic mice chow a normal fat diet exhibited vascular abnormalities resembling atherosclerosis. CONCLUSIONS: IL-32 acts as a proinflammatory factor and may be implicated in the inflammatory cascade contributing to atherosclerosis. By promoting the synthesis of matrix metalloproteinases, it may further contribute to plaque instability. Further studies are warranted to investigate whether IL-32 may serve as a potential therapeutic target in fighting atherosclerosis.


Asunto(s)
Aorta/inmunología , Aterosclerosis/inmunología , Inflamación/inmunología , Interleucinas/metabolismo , Animales , Aorta/citología , Aorta/metabolismo , Aterosclerosis/metabolismo , Quimiocina CCL2/biosíntesis , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Interferón gamma/metabolismo , Interleucinas/genética , Macrófagos/citología , Macrófagos/inmunología , Metaloproteinasa 1 de la Matriz/biosíntesis , Metaloproteinasa 13 de la Matriz/biosíntesis , Metaloproteinasa 9 de la Matriz/biosíntesis , Ratones , Ratones Transgénicos , ARN Mensajero/biosíntesis , Molécula 1 de Adhesión Celular Vascular/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA