Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Ann Rheum Dis ; 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38986577

RESUMEN

OBJECTIVES: Bone remodelling is a highly dynamic process dependent on the precise coordination of osteoblasts and haematopoietic-cell derived osteoclasts. Changes in core metabolic pathways during osteoclastogenesis, however, are largely unexplored and it is unknown whether and how these processes are involved in bone homeostasis. METHODS: We metabolically and transcriptionally profiled cells during osteoclast and osteoblast generation. Individual gene expression was characterised by quantitative PCR and western blot. Osteoblast function was assessed by Alizarin red staining. immunoresponsive gene 1 (Irg1)-deficient mice were used in various inflammatory or non-inflammatory models of bone loss. Tissue gene expression was analysed by RNA in situ hybridisation. RESULTS: We show that during differentiation preosteoclasts rearrange their tricarboxylic acid cycle, a process crucially depending on both glucose and glutamine. This rearrangement is characterised by the induction of Irg1 and production of itaconate, which accumulates intracellularly and extracellularly. While the IRG1-itaconate axis is dispensable for osteoclast generation in vitro and in vivo, we demonstrate that itaconate stimulates osteoblasts by accelerating osteogenic differentiation in both human and murine cells. This enhanced osteogenic differentiation is accompanied by reduced proliferation and altered metabolism. Additionally, supplementation of itaconate increases bone formation by boosting osteoblast activity in mice. Conversely, Irg1-deficient mice exhibit decreased bone mass and have reduced osteoproliferative lesions in experimental arthritis. CONCLUSION: In summary, we identify itaconate, generated as a result of the metabolic rewiring during osteoclast differentiation, as a previously unrecognised regulator of osteoblasts.

2.
Ann Rheum Dis ; 83(4): 518-528, 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38071515

RESUMEN

OBJECTIVES: Osteoclasts (OCs) are myeloid-derived multinucleated cells uniquely able to degrade bone. However, the exact nature of their myeloid precursors is not yet defined. METHODS: CD11c-diphtheria toxin receptor (CD11cDTR) transgenic mice were treated with diphtheria toxin (DT) or phosphate buffered saline (PBS) during serum transfer arthritis (STA) and human tumour necrosis factor transgenic (hTNFtg) arthritis and scored clinically and histologically. We measured cytokines in synovitis by quantitative polymerase chain reaction (qPCR). We performed ovariectomy in CD11cDTR mice treated with PBS or DT. We analysed CD11cDTR, CD11c-Cre/CX3CR1-STOP-DTR and Zbtb46-DTR-treated mice with DT using histomorphometry and OC of CD11c and Zbtb46 fate reporter mice by fluorescent imaging. We sorted murine and human OC precursors and stimulated them with macrophage colony-stimulating factor (M-CSF) and receptor activator of nuclear factor kappa-B ligand (RANKL) to generate OCs. RESULTS: Targeting CD11c+ cells in vivo in models of inflammatory arthritis (STA and hTNFtg) ameliorates arthritis by reducing inflammatory bone destruction and OC generation. Targeting CD11c-expressing cells in unchallenged mice removes all OCs in their long bones. OCs do not seem to be derived from CD11c+ cells expressing CX3CR1+, but from Zbtb46+conventional dendritic cells (cDCs) as all OCs in Zbtb46-Tomato fate reporter mice are Tomato+. In line, administration of DT in Zbtb46-DTR mice depletes all OCs in long bones. Finally, human CD1c-expressing cDCs readily differentiated into bone resorbing OCs. CONCLUSION: Taken together, we identify DCs as important OC precursors in bone homeostasis and inflammation, which might open new avenues for therapeutic interventions in OC-mediated diseases.


Asunto(s)
Artritis , Osteoclastos , Femenino , Ratones , Humanos , Animales , Citocinas/metabolismo , Diferenciación Celular , Artritis/metabolismo , Células Dendríticas/metabolismo , Ligando RANK/metabolismo
3.
Bone Joint Res ; 11(7): 484-493, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35801532

RESUMEN

AIMS: Insufficient treatment response in rheumatoid arthritis (RA) patients requires novel treatment strategies to halt disease progression. The potential benefit of combination of cytokine-inhibitors in RA is still unclear and needs further investigation. To explore the impact of combined deficiency of two major cytokines, namely interleukin (IL)-1 and IL-6, in this study double deficient mice for IL-1αß and IL-6 were investigated in different tumour necrosis factor (TNF)-driven inflammatory bone disorders, namely peripheral arthritis and sacroiliitis, as well as systemic bone loss. METHODS: Disease course, histopathological features of arthritis, and micro-CT (µCT) bone analysis of local and systemic bone loss were assessed in 15-week-old IL1-/-IL6-/- hTNFtg in comparison to IL1-/- hTNFtg, IL6-/- hTNFtg, and hTNFtg mice. µCT bone analysis of single deficient and wild-type mice was also performed. RESULTS: Combined deficiency of IL-1/IL-6 markedly ameliorated TNF-mediated arthritis and bilateral sacroiliitis, but without additive benefits compared to single IL-1 deficiency. This finding confirms the important role of IL-1 and the marginal role of IL-6 in TNF-driven pathways of local joint damage, but questions the efficacy of potential combinatorial therapies of IL-1 and IL-6 in treatment of RA. In contrast, combined deficiency of IL-1/IL-6 led to an additive protective effect on TNF-driven systemic bone loss compared to single IL-1 and IL-6 deficiency. This finding clearly indicates a common contribution of both IL-1 and IL-6 in TNF-driven systemic bone loss, and points to a discrepancy of cytokine dependency in local and systemic TNF-driven mechanisms of inflammatory arthritis. CONCLUSION: Combinatorial treatments in RA might provide different benefits to inflammatory local arthritis and systemic comorbidities. Cite this article: Bone Joint Res 2022;11(7):484-493.

4.
Ann Rheum Dis ; 80(6): 714-726, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33602797

RESUMEN

Animal models for inflammatory arthritides such as rheumatoid arthritis (RA) and psoriatic arthritis are widely accepted and frequently used to identify pathological mechanisms and validate novel therapeutic strategies. Unfortunately, many publications reporting on these animal studies lack detailed description and appropriate assessment of the distinct histopathological features of arthritis: joint inflammation, cartilage damage and bone erosion. Therefore, the European consortium BeTheCure, consisting of 38 academic and industrial partners from 15 countries, set as goal to standardise the histological evaluation of joint sections from animal models of inflammatory arthritis. The consensual approach of a task force including 16 academic and industrial scientists as well as laboratory technicians has resulted in the development of the Standardised Microscopic Arthritis Scoring of Histological sections ('SMASH') recommendations for a standardised processing and microscopic scoring of the characteristic histopathological features of arthritis, exemplified by four different rodent models for arthritis: murine collagen-induced arthritis, collagen-antibody-induced arthritis, human tumour necrosis factor transgenic Tg197 mice and rat pristane-induced arthritis, applicable to any other inflammatory arthritis model. Through standardisation, the SMASH recommendations are designed to improve and maximise the information derived from in vivo arthritis experiments and to promote reproducibility and transparent reporting on such studies. In this manuscript, we will discuss and provide recommendations for analysis of histological joint sections: identification of the regions of interest, sample preparation, staining procedures and quantitative scoring methods. In conclusion, awareness of the different features of the arthritis pathology in animal models of inflammatory arthritis is of utmost importance for reliable research outcome, and the standardised histological processing and scoring methods in these SMASH recommendations will help increase uniformity and reproducibility in preclinical research on inflammatory arthritis.


Asunto(s)
Artritis Experimental , Artritis Reumatoide , Animales , Artritis Experimental/patología , Artritis Reumatoide/patología , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , Ratas , Reproducibilidad de los Resultados
5.
Aging Cell ; 19(11): e13244, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33085187

RESUMEN

Bone loss is one of the consequences of aging, leading to diseases such as osteoporosis and increased susceptibility to fragility fractures and therefore considerable morbidity and mortality in humans. Here, we identify microRNA-146a (miR-146a) as an essential epigenetic switch controlling bone loss with age. Mice deficient in miR-146a show regular development of their skeleton. However, while WT mice start to lose bone with age, animals deficient in miR-146a continue to accrue bone throughout their life span. Increased bone mass is due to increased generation and activity of osteoblasts in miR-146a-deficient mice as a result of sustained activation of bone anabolic Wnt signaling during aging. Deregulation of the miR-146a target genes Wnt1 and Wnt5a parallels bone accrual and osteoblast generation, which is accompanied by reduced development of bone marrow adiposity. Furthermore, miR-146a-deficient mice are protected from ovariectomy-induced bone loss. In humans, the levels of miR-146a are increased in patients suffering fragility fractures in comparison with those who do not. These data identify miR-146a as a crucial epigenetic temporal regulator which essentially controls bone homeostasis during aging by regulating bone anabolic Wnt signaling. Therefore, miR-146a might be a powerful therapeutic target to prevent age-related bone dysfunctions such as the development of bone marrow adiposity and osteoporosis.


Asunto(s)
MicroARNs/genética , Osteoporosis/genética , Animales , Resorción Ósea/genética , Resorción Ósea/patología , Diferenciación Celular/fisiología , Epigénesis Genética , Femenino , Masculino , Ratones , MicroARNs/metabolismo , Osteoblastos/citología , Osteoporosis/patología , Proteína Wnt-5a/metabolismo , Proteína Wnt1/metabolismo
7.
Exp Mol Med ; 51(7): 1-11, 2019 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-31285419

RESUMEN

Rheumatoid arthritis (RA) is an autoimmune disease characterized by persistent synovial inflammation. The major drivers of synovial inflammation are cytokines and chemokines. Among these molecules, TNF activates fibroblast-like synoviocytes (FLSs), which leads to the production of inflammatory mediators. Here, we show that TNF regulates the expression of the transcription factor interferon regulatory factor 1 (IRF1) in human FLSs as well as in a TNF transgenic arthritis mouse model. Transcriptomic analyses of IRF1-deficient, TNF-stimulated FLSs define the interferon (IFN) pathway as a major target of IRF1. IRF1 expression is associated with the expression of IFNß, which leads to the activation of the JAK-STAT pathway. Blocking the JAK-STAT pathway with the Janus kinase inhibitor (JAKinib) baricitinib or tofacitinib reduces the expression of IFN-regulated genes (IRGs) in TNF-activated FLSs. Therefore, we conclude that TNF induces a distinct inflammatory cascade, in which IRGs are key elements, in FLSs. The IFN-signature might be a promising biomarker for the efficient and personalized use of new treatment strategies for RA, such as JAKinibs.


Asunto(s)
Artritis Reumatoide/inmunología , Factor 1 Regulador del Interferón/metabolismo , Interferones/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Artritis Reumatoide/metabolismo , Artritis Reumatoide/patología , Azetidinas/uso terapéutico , Biomarcadores/metabolismo , Femenino , Expresión Génica , Humanos , Inflamación , Factor 1 Regulador del Interferón/genética , Interferones/genética , Inhibidores de las Cinasas Janus/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Piperidinas/uso terapéutico , Purinas , Pirazoles , Pirimidinas/uso terapéutico , Pirroles/uso terapéutico , Sulfonamidas/uso terapéutico , Membrana Sinovial/inmunología , Membrana Sinovial/metabolismo , Membrana Sinovial/patología , Sinoviocitos/metabolismo , Factor de Necrosis Tumoral alfa/genética
8.
J Bone Miner Res ; 34(9): 1632-1645, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31063606

RESUMEN

In rheumatoid arthritis (RA), chronic joint inflammation leading to bone and cartilage damage is the major cause of functional impairment. Whereas reduction of synovitis and blockade of joint damage can be successfully achieved by disease modifying antirheumatic therapies, bone repair upon therapeutic interventions has only been rarely reported. The aim of this study was to use fluorodeoxyglucose ([18 F]FDG) and [18 F]fluoride µPET/CT imaging to monitor systemic inflammatory and destructive bone remodeling processes as well as potential bone repair in an established mouse model of chronic inflammatory, erosive polyarthritis. Therefore, human tumor necrosis factor transgenic (hTNFtg) mice were treated with infliximab, an anti-TNF antibody, for 4 weeks. Before and after treatment period, mice received either [18 F]FDG, for detecting inflammatory processes, or [18 F]fluoride, for monitoring bone remodeling processes, for PET scans followed by CT scans. Standardized uptake values (SUVmean ) were analyzed in various joints and histopathological signs of arthritis, joint damage, and repair were assessed. Longitudinal PET/CT scans revealed a significant decrease in [18 F]FDG SUVs in affected joints demonstrating complete remission of inflammatory processes due to TNF blockade. In contrast, [18 F]fluoride SUVs could not discriminate between different severities of bone damage in hTNFtg mice. Repeated in vivo CT images proved a structural reversal of preexisting bone erosions after anti-TNF therapy. Accordingly, histological analysis showed complete resolution of synovial inflammation and healing of bone at sites of former bone erosion. We conclude that in vivo multimodal [18 F]FDG µPET/CT imaging allows to quantify and monitor inflammation-mediated bone damage and reveals not only reversal of synovitis but also bone repair upon TNF blockade in experimental arthritis. © 2019 The Authors. Journal of Bone and Mineral Research Published by Wiley Periodicals, Inc.


Asunto(s)
Huesos/diagnóstico por imagen , Huesos/patología , Fluorodesoxiglucosa F18/química , Inflamación/patología , Imagen Multimodal , Tomografía Computarizada por Tomografía de Emisión de Positrones , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Regeneración Ósea , Remodelación Ósea , Cartílago/diagnóstico por imagen , Cartílago/patología , Humanos , Articulaciones/diagnóstico por imagen , Articulaciones/patología , Estudios Longitudinales , Ratones Endogámicos C57BL , Ratones Transgénicos , Osteoclastos/patología , Membrana Sinovial/diagnóstico por imagen , Membrana Sinovial/patología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
9.
J Cell Mol Med ; 22(11): 5278-5285, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30133119

RESUMEN

Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease, characterized by synovial infiltration of various inflammatory cells. Chemokines are involved in controlling the recruitment of different cell types into the synovial membrane. The role of CCR6 in the development of arthritis so far remains unclear. In this study, we investigated the role of CCR6 in the pathogenesis of arthritis using three different murine arthritis models. Compared to WT animals, CCR6-/- mice developed less clinical signs of arthritis in the collagen-induced arthritis model but not in the K/BxN serum transfer arthritis model and in the human tumour necrosis factor transgenic arthritis model, suggesting a defect in adaptive effector functions but intact innate effector functions in the development of arthritis in CCR6-/- animals. In line with this, anti-collagen antibody levels were significantly reduced in CCR6-/- mice compared with WT mice. Moreover, we demonstrate enhanced osteoclastogenesis in vitro in CCR6-/- mice compared with WT mice. However, we did not detect differences in bone mass under steady state conditions in vivo between WT and CCR6-deficient mice. These data suggest that CCR6 is crucially involved in adaptive but not in innate immunity-driven arthritis. CCR6 or its chemokine ligand CCL20 might represent a possible new target for the treatment of RA.


Asunto(s)
Artritis Experimental/genética , Artritis Reumatoide/genética , Enfermedades Autoinmunes/genética , Quimiocina CCL20/genética , Receptores CCR6/genética , Animales , Artritis Experimental/inmunología , Artritis Experimental/patología , Artritis Reumatoide/inmunología , Artritis Reumatoide/patología , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Quimiocina CCL20/inmunología , Humanos , Inmunidad Innata/genética , Ratones , Receptores CCR6/inmunología , Membrana Sinovial/inmunología , Membrana Sinovial/patología
10.
Ann Rheum Dis ; 77(10): 1490-1497, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29959183

RESUMEN

OBJECTIVES: Bone destruction in rheumatoid arthritis is mediated by osteoclasts (OC), which are derived from precursor cells of the myeloid lineage. The role of the two monocyte subsets, classical monocytes (expressing CD115, Ly6C and CCR2) and non-classical monocytes (which are CD115 positive, but low in Ly6C and CCR2), in serving as precursors for OC in arthritis is still elusive. METHODS: We investigated CCR2-/- mice, which lack circulating classical monocytes, crossed into hTNFtg mice for the extent of joint damage. We analysed monocyte subsets in hTNFtg and K/BxN serum transfer arthritis by flow cytometry. We sorted monocyte subsets and analysed their potential to differentiate into OC and their transcriptional response in response to RANKL by RNA sequencing. With these data, we performed a gene ontology enrichment analysis and gene set enrichment analysis. RESULTS: We show that in hTNFtg arthritis local bone erosion and OC generation are even enhanced in the absence of CCR2. We further show the numbers of non-classical monocytes in blood are elevated and are significantly correlated with histological signs of joint destruction. Sorted non-classical monocytes display an increased capacity to differentiate into OCs. This is associated with an increased expression of signal transduction components of RANK, most importantly TRAF6, leading to an increased responsiveness to RANKL. CONCLUSION: Therefore, non-classical monocytes are pivotal cells in arthritis tissue damage and a possible target for therapeutically intervention for the prevention of inflammatory joint damage.


Asunto(s)
Artritis Experimental/fisiopatología , Artritis Reumatoide/fisiopatología , Resorción Ósea/fisiopatología , Monocitos/fisiología , Osteoclastos/fisiología , Animales , Artritis Experimental/complicaciones , Artritis Reumatoide/complicaciones , Resorción Ósea/etiología , Diferenciación Celular , Modelos Animales de Enfermedad , Citometría de Flujo , Ratones , Ligando RANK/metabolismo , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Receptores CCR2/metabolismo , Transducción de Señal/fisiología , Factor 6 Asociado a Receptor de TNF/metabolismo
11.
J Autoimmun ; 82: 74-84, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28545737

RESUMEN

Synovial fibroblasts are key cells orchestrating the inflammatory response in arthritis. Here we demonstrate that loss of miR-146a, a key epigenetic regulator of the innate immune response, leads to increased joint destruction in a TNF-driven model of arthritis by specifically regulating the behavior of synovial fibroblasts. Absence of miR-146a in synovial fibroblasts display a highly deregulated gene expression pattern and enhanced proliferation in vitro and in vivo. Deficiency of miR-146a induces deregulation of tumor necrosis factor (TNF) receptor associated factor 6 (TRAF6) in synovial fibroblasts, leading to increased proliferation. In addition, loss of miR-146a shifts the metabolic state of fibroblasts towards glycolysis and augments the ability of synovial fibroblasts to support the generation of osteoclasts by controlling the balance of osteoclastogenic regulatory factors receptor activator of NF-κB ligand (RANKL) and osteoprotegerin (OPG). Bone marrow transplantation experiments confirmed the importance of miR-146a in the radioresistant mesenchymal compartment for the control of arthritis severity, in particular for inflammatory joint destruction. This study therefore identifies microRNA-146a as an important local epigenetic regulator of the inflammatory response in arthritis. It is a central element of an anti-inflammatory feedback loop in resident synovial fibroblasts, who are orchestrating the inflammatory response in chronic arthritis. MiR-146a restricts their activation, thereby preventing excessive tissue damage during arthritis.


Asunto(s)
Artritis/genética , Artritis/metabolismo , Fibroblastos/metabolismo , Articulaciones/metabolismo , Articulaciones/patología , MicroARNs/genética , Animales , Artritis/patología , Artritis Experimental , Resorción Ósea/genética , Proliferación Celular , Modelos Animales de Enfermedad , Fibroblastos/patología , Expresión Génica , Regulación de la Expresión Génica , Humanos , Ratones , Ratones Transgénicos , Interferencia de ARN , Membrana Sinovial/citología , Membrana Sinovial/metabolismo , Factor 6 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
12.
Dis Model Mech ; 9(11): 1329-1338, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27638666

RESUMEN

Chronic inflammation of articular joints causing bone and cartilage destruction consequently leads to functional impairment or loss of mobility in affected joints from individuals affected by rheumatoid arthritis (RA). Even successful treatment with complete resolution of synovial inflammatory processes does not lead to full reversal of joint functionality, pointing to the crucial contribution of irreversibly damaged structural components, such as bone and cartilage, to restricted joint mobility. In this context, we investigated the impact of the distinct components, including synovial inflammation, bone erosion or cartilage damage, as well as the effect of blocking tumor necrosis factor (TNF) on functional impairment in human-TNF transgenic (hTNFtg) mice, a chronic inflammatory erosive animal model of RA. We determined CatWalk-assisted gait profiles as objective quantitative measurements of functional impairment. We first determined body-weight-independent gait parameters, including maximum intensity, print length, print width and print area in wild-type mice. We observed early changes in those gait parameters in hTNFtg mice at week 5 - the first clinical signs of arthritis. Moreover, we found further gait changes during chronic disease development, indicating progressive functional impairment in hTNFtg mice. By investigating the association of gait parameters with inflammation-mediated joint pathologies at different time points of the disease course, we found a relationship between gait parameters and the extent of cartilage damage and bone erosions, but not with the extent of synovitis in this chronic model. Next, we observed a significant improvement of functional impairment upon blocking TNF, even at progressed stages of disease. However, blocking TNF did not restore full functionality owing to remaining subclinical inflammation and structural microdamage. In conclusion, CatWalk gait analysis provides a useful tool for quantitative assessment of functional impairment in inflammatory destructive arthritis. Our findings indicate that cartilage damage and bone erosion, but not synovial inflammation, are the most important determinants for progressive functional impairment in this chronic erosive arthritis model.


Asunto(s)
Artritis Experimental/patología , Artritis Experimental/fisiopatología , Huesos/patología , Huesos/fisiopatología , Cartílago Articular/patología , Cartílago Articular/fisiopatología , Inflamación/patología , Membrana Sinovial/fisiología , Envejecimiento , Animales , Peso Corporal , Femenino , Marcha , Humanos , Inflamación/fisiopatología , Modelos Lineales , Ratones Endogámicos C57BL , Ratones Transgénicos , Membrana Sinovial/patología , Membrana Sinovial/fisiopatología , Factor de Necrosis Tumoral alfa/metabolismo , Microtomografía por Rayos X
13.
Arthritis Res Ther ; 18: 63, 2016 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-26970742

RESUMEN

BACKGROUND: Our aim was to investigate the role of nicotinic acetylcholine receptors (nAChRs) in in-vitro osteoclastogenesis and in in-vivo bone homeostasis. METHODS: The presence of nAChR subunits as well as the in-vitro effects of nAChR agonists were investigated by ex vivo osteoclastogenesis assays, real-time polymerase chain reaction, Western blot and flow cytometry in murine bone marrow-derived macrophages differentiated in the presence of recombinant receptor activator of nuclear factor kappa B ligand (RANKL) and macrophage colony-stimulating factor (M-CSF). The bone phenotype of mice lacking various nAChR subunits was investigated by peripheral quantitative computed tomography and histomorphometric analysis. Oscillations in the intracellular calcium concentration were detected by measuring the Fura-2 fluorescence intensity. RESULTS: We could demonstrate the presence of several nAChR subunits in bone marrow-derived macrophages stimulated with RANKL and M-CSF, and showed that they are capable of producing acetylcholine. nAChR ligands reduced the number of osteoclasts as well as the number of tartrate-resistant acidic phosphatase-positive mononuclear cells in a dose-dependent manner. In vitro RANKL-mediated osteoclastogenesis was reduced in mice lacking α7 homomeric nAChR or ß2-containing heteromeric nAChRs, while bone histomorphometry revealed increased bone volume as well as impaired osteoclastogenesis in male mice lacking the α7 nAChR. nAChR ligands inhibited RANKL-induced calcium oscillation, a well-established phenomenon of osteoclastogenesis. This inhibitory effect on Ca(2+) oscillation subsequently led to the inhibition of RANKL-induced NFATc1 and c-fos expression after long-term treatment with nicotine. CONCLUSIONS: We have shown that the activity of nAChRs conveys a marked effect on osteoclastogenesis in mice. Agonists of these receptors inhibited calcium oscillations in osteoclasts and blocked the RANKL-induced activation of c-fos and NFATc1. RANKL-mediated in-vitro osteoclastogenesis was reduced in α7 knockout mice, which was paralleled by increased tibial bone volume in male mice in vivo.


Asunto(s)
Remodelación Ósea/fisiología , Huesos/fisiología , Osteoclastos/metabolismo , Receptores Nicotínicos/metabolismo , Animales , Western Blotting , Citometría de Flujo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa
14.
Nat Med ; 21(9): 1085-90, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26236992

RESUMEN

Myostatin (also known as growth and differentiation factor 8) is a secreted member of the transforming growth factor-ß (TGF-ß) family that is mainly expressed in skeletal muscle, which is also its primary target tissue. Deletion of the myostatin gene (Mstn) in mice leads to muscle hypertrophy, and animal studies support the concept that myostatin is a negative regulator of muscle growth and regeneration. However, myostatin deficiency also increases bone formation, mainly through loading-associated effects on bone. Here we report a previously unknown direct role for myostatin in osteoclastogenesis and in the progressive loss of articular bone in rheumatoid arthritis (RA). We demonstrate that myostatin is highly expressed in the synovial tissues of RA subjects and of human tumor necrosis factor (TNF)-α transgenic (hTNFtg) mice, a model for human RA. Myostatin strongly accelerates receptor activator of nuclear factor κB ligand (RANKL)-mediated osteoclast formation in vitro through transcription factor SMAD2-dependent regulation of nuclear factor of activated T-cells (NFATC1). Myostatin deficiency or antibody-mediated inhibition leads to an amelioration of arthritis severity in hTNFtg mice, chiefly reflected by less bone destruction. Consistent with these effects in hTNFtg mice, the lack of myostatin leads to increased grip strength and less bone erosion in the K/BxN serum-induced arthritis model in mice. The results strongly suggest that myostatin is a potent therapeutic target for interfering with osteoclast formation and joint destruction in RA.


Asunto(s)
Artritis Reumatoide/terapia , Diferenciación Celular , Miostatina/fisiología , Osteoclastos/fisiología , Animales , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Humanos , Factor Estimulante de Colonias de Macrófagos/farmacología , Ratones , Miostatina/antagonistas & inhibidores , Factores de Transcripción NFATC/metabolismo , Osteoclastos/citología , Osteogénesis , Ligando RANK/farmacología
15.
Sci Signal ; 8(381): ra60, 2015 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-26082436

RESUMEN

Oxidative stress produced in response to infection or sterile injury activates the innate immune response. We found that extracellular vesicles (EVs) isolated from the plasma of patients with rheumatoid arthritis or secreted from cells subjected to oxidative stress contained oxidized phospholipids that stimulated cells expressing Toll-like receptor 4 (TLR4) in a manner dependent on its co-receptor MD-2. EVs from healthy subjects or reconstituted synthetic EVs subjected to limited oxidation gained the ability to stimulate TLR4-expressing cells, whereas prolonged oxidation abrogated this property. Furthermore, we found that 15-lipoxygenase generated hydro(pero)xylated phospholipids that stimulated TLR4-expressing cells. Molecular modeling suggested that the mechanism of activation of TLR4 by oxidized phospholipids in EVs was structurally similar to that of the TLR4 ligand lipopolysaccharide (LPS). This was supported by experiments showing that EV-mediated stimulation of cells required MD-2, that mutations that block LPS binding to TLR4 abrogated the stimulatory effect of EVs, and that EVs induced TLR4 dimerization. On the other hand, analysis of gene expression profiles showed that genes encoding factors that resolve inflammation were more abundantly expressed in responses to EVs than in response to LPS. Together, these data suggest that EVs act as an oxidative stress-induced endogenous danger signal that underlies the pervasive role of TLR4 in inflammatory diseases.


Asunto(s)
Artritis Reumatoide/inmunología , Micropartículas Derivadas de Células/inmunología , Estrés Oxidativo/inmunología , Receptor Toll-Like 4/inmunología , Artritis Reumatoide/genética , Artritis Reumatoide/patología , Micropartículas Derivadas de Células/genética , Femenino , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Lipopolisacáridos/farmacología , Antígeno 96 de los Linfocitos/genética , Antígeno 96 de los Linfocitos/inmunología , Masculino , Estrés Oxidativo/efectos de los fármacos , Receptor Toll-Like 4/agonistas , Receptor Toll-Like 4/genética
17.
Arthritis Rheum ; 65(3): 608-17, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23280418

RESUMEN

OBJECTIVE: To investigate how tumor necrosis factor (TNF)-inhibiting therapy affects bone destruction and inflammation in a TNF-driven mouse model of rheumatoid arthritis. METHODS: In order to evaluate the influence of TNF on osteoclastogenesis in vitro, different concentrations of TNF were added to spleen cell-derived monocytes in the absence or presence of different concentrations of RANKL. In addition, the effects of TNF inhibition on osteoclast precursors as well as local bone destruction in vivo were assessed by treating TNF-transgenic mice with different doses of adalimumab. RESULTS: TNF stimulated osteoclastogenesis mainly by increasing the number of osteoclast precursor cells in vitro. This TNF effect was independent of the presence of RANKL. In the hTNF-transgenic mouse model of destructive arthritis, low-dose TNF-inhibiting therapy with adalimumab had no effect on synovial inflammation but significantly inhibited local bone destruction and the generation of osteoclasts. This inhibition was accompanied by a reduction in the number of c-Fms-positive osteoclast precursor cells in the bone marrow and a reduction of the osteoclast precursor pools in the blood and inflamed synovial membrane of hTNF-transgenic mice. CONCLUSION: Low-dose TNF-inhibiting therapy significantly reduces bone erosions by reducing the number of circulating and joint-invading osteoclast precursors. This effect is uncoupled from its antiinflammatory action.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Artritis Reumatoide/tratamiento farmacológico , Sinovitis/tratamiento farmacológico , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/farmacología , Adalimumab , Animales , Antirreumáticos/farmacología , Artritis Reumatoide/inmunología , Artritis Reumatoide/patología , Huesos/efectos de los fármacos , Huesos/patología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Osteoclastos/efectos de los fármacos , Osteoclastos/patología , Ligando RANK/genética , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Células Madre/efectos de los fármacos , Células Madre/patología , Membrana Sinovial/efectos de los fármacos , Membrana Sinovial/patología , Sinovitis/inmunología , Sinovitis/patología , Factor de Necrosis Tumoral alfa/genética
18.
Wien Klin Wochenschr ; 124(5-6): 170-6, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22210441

RESUMEN

INTRODUCTION: Rheumatoid arthritis is a chronic autoimmune disease characterised by inflammation of joints with cartilage and bone destruction leading to progressive disability. While the cause of rheumatoid arthritis is not known and the disease cannot be cured, conventional disease modifying antirheumatic drugs and biologicals are effective treatments for many patients. However, new therapies are needed in order to achieve better relief from rheumatoid arthritis symptoms than currently possible and to fully prevent joint damage. 18ß-Glycyrrhetinic acid is not only used frequently in traditional Chinese medicine, but has been reported to target some of the inflammatory mediators involved in the pathogenesis of rheumatoid arthritis. Moreover, it has been reported that liquorice, which contains high levels of 18ß-Glycyrrhetinic acid, reduces inflammation and articular damage in collagen induced arthritis. Therefore, we studied the effects of 18ß-Glycyrrhetinic acid in a Tumor necrosis factor (TNF) dependent mouse model of rheumatoid arthritis. MATERIAL AND METHODS: HTNFtg mice were treated with 18ß-Glycyrrhetinic acid from day 28 after birth every second or third day for 2 weeks, or 3 times a week for six weeks. TNF inhibitor treated animals served as positive control. RESULTS: Clinical scores of arthritis were not altered in animals treated with 18ß-Glycyrrhetinic acid compared to placebo treated animals. Histological data also indicate no effects of 18ß-Glycyrrhetinic acid on inflammatory joint destruction. TNF inhibitors, however markedly reduced not only clinical signs of TNF triggered joint inflammation but also histological signs of erosive disease. Therefore, in contrast to previous reports our data indicate that 18ß-Glycyrrhetinic acid does not provide a new therapeutic option for treating patients with rheumatoid arthritis.


Asunto(s)
Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/patología , Modelos Animales de Enfermedad , Ácido Glicirretínico/análogos & derivados , Factor de Necrosis Tumoral alfa/genética , Animales , Antirreumáticos/uso terapéutico , Ácido Glicirretínico/uso terapéutico , Humanos , Ratones , Ratones Transgénicos , Resultado del Tratamiento
19.
Ann Rheum Dis ; 71(6): 1004-11, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22258493

RESUMEN

OBJECTIVE: To elucidate the mechanisms involved in cartilage damage in an experimental model of rheumatoid arthritis (RA) by specifically addressing the time course of extracellular matrix degradation and the contribution of cell-matrix interactions for initiation and perpetuation of this process. METHODS: The human tumour necrosis factor (TNF) transgenic (hTNFtg) mouse model of RA was used to analyse the time course of pannus attachment to the cartilage and cartilage destruction, respectively, and crossed hTNFtg mice with interleukin (IL)-1(-/-) animals were used to investigate the role of IL-1 on these TNF-induced mechanisms in vivo. In addition, an in vitro attachment assay using synovial fibroblasts (SFs) from hTNFtg mice and freshly isolated articular cartilage was used to determine the role of proteoglycan loss in attachment of SFs and the role of the transmembrane heparan sulfate proteoglycan syndecan-4. RESULTS: In vivo analyses of hTNFtg mice showed that proteoglycan loss induced by IL-1 precedes and constitutes an important prerequisite for these processes as, in hTNFtg mice, IL-1 deficiency protected from the loss of cartilage proteoglycans and almost completely prevented the attachment and subsequent invasion of inflamed synovial tissue into cartilage. In vitro studies confirmed that loss of cartilage proteoglycans is required for attachment of SFs and that syndecan-4 is prominently involved in SF attachment and activation. CONCLUSIONS: The results of this study suggest that the loss of cartilage proteoglycans is an early event in the course of destructive arthritis that facilitates the attachment of the inflamed synovial membrane and also initiates matrix degradation and inflammation through cell-matrix interactions.


Asunto(s)
Artritis Reumatoide/inmunología , Artritis Reumatoide/patología , Membrana Sinovial/inmunología , Membrana Sinovial/patología , Sinovitis/inmunología , Sinovitis/patología , Animales , Artritis Reumatoide/metabolismo , Huesos/inmunología , Huesos/metabolismo , Huesos/patología , Cartílago/inmunología , Cartílago/metabolismo , Cartílago/patología , Comunicación Celular/inmunología , Modelos Animales de Enfermedad , Matriz Extracelular/inmunología , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Fibroblastos/inmunología , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Interleucina-1/inmunología , Interleucina-1/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteoglicanos/metabolismo , Sindecano-4/metabolismo , Membrana Sinovial/metabolismo , Sinovitis/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
20.
Ann Rheum Dis ; 70(6): 1122-9, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21415051

RESUMEN

OBJECTIVE: Reduced vitamin D intake has been linked to increased susceptibility to develop rheumatoid arthritis (RA) and vitamin D deficiency is associated with increased disease activity in RA patients. The pathophysiological role of vitamin D in joint inflammation is, however, unclear. METHODS: To determine the influence of absent vitamin D signalling in chronic arthritis, vitamin D receptor (VDR)-deficient mice were crossed with human tumour necrosis factor (TNF) transgenic mice (hTNFtg), which spontaneously develop chronic arthritis. RESULTS: Clinical signs and symptoms of chronic arthritis were aggravated in hTNFtg mice lacking functional VDR signalling. Moreover, synovial inflammation was clearly increased in VDR(-/-)hTNFtg mice as compared to hTNFtg mice and was associated with an increased macrophage influx in inflamed joints. In vitro, VDR-deficient monocytes were proinflammatory and hyper-responsive to TNF stimulation associated with prolonged mitogen-activated protein kinase activation and cytokine secretion. Also, VDR(-/-) monocytes showed enhanced potential to differentiate into bone resorbing osteoclasts in vitro. In line, VDR(-/-)hTNFtg mice had significantly increased cartilage damage and synovial bone erosions. CONCLUSIONS: VDR plays an important role in limiting the inflammatory phenotype in a mouse model of RA. Absent VDR signalling causes a proinflammatory monocyte phenotype associated with increased inflammation, cartilage damage and bone erosion.


Asunto(s)
Artritis Experimental/metabolismo , Artritis Reumatoide/metabolismo , Receptores de Calcitriol/fisiología , Animales , Artritis Experimental/patología , Artritis Reumatoide/patología , Densidad Ósea/fisiología , Resorción Ósea/metabolismo , Resorción Ósea/fisiopatología , Cartílago Articular/metabolismo , Células Cultivadas , Citocinas/biosíntesis , Macrófagos/patología , Macrófagos/fisiología , Ratones , Ratones Noqueados , Ratones Transgénicos , Osteoclastos/fisiología , Proteoglicanos/metabolismo , Receptores de Calcitriol/deficiencia , Transducción de Señal/fisiología , Sinovitis/metabolismo , Sinovitis/patología , Factor de Necrosis Tumoral alfa/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...